Skip to main content
mBio logoLink to mBio
. 2020 Apr 28;11(2):e00218-20. doi: 10.1128/mBio.00218-20

Advances in Understanding the Human Urinary Microbiome and Its Potential Role in Urinary Tract Infection

Michael L Neugent a, Neha V Hulyalkar a, Vivian H Nguyen a, Philippe E Zimmern b, Nicole J De Nisco a,
Editor: Danielle A Garsinc
PMCID: PMC7188990  PMID: 32345639

Recent advances in the analysis of microbial communities colonizing the human body have identified a resident microbial community in the human urinary tract (UT). Compared to many other microbial niches, the human UT harbors a relatively low biomass. Studies have identified many genera and species that may constitute a core urinary microbiome. However, the contribution of the UT microbiome to urinary tract infection (UTI) and recurrent UTI (rUTI) pathobiology is not yet clearly understood. Evidence suggests that commensal species within the UT and urogenital tract (UGT) microbiomes, such as Lactobacillus crispatus, may act to protect against colonization with uropathogens.

KEYWORDS: metagenomics, microbial communities, microbiome, probiotics, urinary tract infection

ABSTRACT

Recent advances in the analysis of microbial communities colonizing the human body have identified a resident microbial community in the human urinary tract (UT). Compared to many other microbial niches, the human UT harbors a relatively low biomass. Studies have identified many genera and species that may constitute a core urinary microbiome. However, the contribution of the UT microbiome to urinary tract infection (UTI) and recurrent UTI (rUTI) pathobiology is not yet clearly understood. Evidence suggests that commensal species within the UT and urogenital tract (UGT) microbiomes, such as Lactobacillus crispatus, may act to protect against colonization with uropathogens. However, the mechanisms and fundamental biology of the urinary microbiome-host relationship are not understood. The ability to measure and characterize the urinary microbiome has been enabled through the development of next-generation sequencing and bioinformatic platforms that allow for the unbiased detection of resident microbial DNA. Translating technological advances into clinical insight will require further study of the microbial and genomic ecology of the urinary microbiome in both health and disease. Future diagnostic, prognostic, and therapeutic options for the management of UTI may soon incorporate efforts to measure, restore, and/or preserve the native, healthy ecology of the urinary microbiomes.

INTRODUCTION

The human microbiome is the sum of all genomic information belonging to the resident microbiota, the nonhuman life that colonizes the body. Through years of research, it has become clear that the human body harbors distinct microbial populations within various anatomical niches (13). A long understudied but important microbial niche is now being closely examined and functionally characterized. The human urinary tract harbors a resident microbial community (48). These relatively new data characterizing the human urinary microbiota defy decades-old theory and clinical practice of urinary tract sterility in urogenital health (5, 9). For the purposes of this review, the term “urinary tract” (UT) refers to the organ system involved in the production, transport, storage, and excretion of urine, namely, the kidney, ureter, bladder, and urethra. Discussion of a larger system, which includes the UT, is also relevant for the purposes of this review. Hence, the term “urogenital tract” (UGT) is used to refer to an organ system which includes the UT as well as the anatomical sites and organs involved in reproduction that may contribute to the urine microbial load, such as the vagina, cervix, periurethral skin, penis, pubic skin surfaces, and perineal area. Depending on the sampling method, the urinary microbiota may consist of species residing within the bladder, UT, or UGT. The urinary microbiota has been characterized and reproducibly measured. To date, >100 species from more than 50 genera are thought to reside in the human UT and UGT (46, 933). However, a consensus within the field regarding the composition of the urinary microbiota has not yet been reached. Furthermore, the role of this microbial community in urogenital health and disease, such as urinary tract infection (UTI), is not fully understood.

The UT is the site of the most common bacterial infection experienced in adults. The medical burden of UTI is substantial, with an estimated 1% of all clinical visits in the United States being attributed to UTI management, the cost of which exceeds $3.5 billion annually (3441). UTI disproportionately affects women, with approximately 50% of women experiencing a UTI in their lifetimes (37, 39). While UTIs are prevalent among young, sexually active populations, the risk of UTI in women also increases with age, leading to an elevated risk in postmenopausal and elderly women. UTIs can be caused by a variety of bacteria and fungi, but the most common pathogens are uropathogenic Escherichia coli (UPEC), followed by Klebsiella pneumoniae, Enterococcus faecalis, and Proteus mirabilis (34). The public’s perception of UTI treatment assumes that it is a simple matter of clearing the infection with antibiotic therapy (36, 42). However, antibiotic resistance and/or allergy is frequent, and UTIs often recur, becoming prolonged cycles of infection, known as recurrent UTI (rUTI), that dramatically reduce quality of life (37, 43). UTIs that ascend to the kidneys can lead to pyelonephritis and life-threatening urosepsis. Some cases of rUTI can last decades and, when refractory to antibiotic therapy, may ultimately require removal of the bladder (34).

Current treatments for UTI and rUTI rely primarily on antibiotic therapy to eliminate the pathogen and achieve UT sterility (34, 43). Despite advances in confirming the existence of the urinary microbiome, conventional antimicrobial strategies for treating UTI and rUTI do not include the preservation or restoration of the microbial community that exists in the host healthy state. This may be because little functional knowledge exists about whether or how the urinary microbiota confers protection against infection. However, the microbial communities resident in many body sites are known to play critical roles in preserving host physiology and health. Disruptions in the resident microbiota are associated with defects in host health, such as inflammatory bowel disease, bacterial vaginosis, cancer, and various metabolic diseases (4448). The host and resident microbial communities are now believed to be components of a larger composite organism, the holobiont (49, 50). Given such a reciprocal relationship, changes in host physiology or microbial ecology likely affect the system as a whole. This review focuses on the current advances in understanding the microbial and genomic ecology of the human urinary microbiome in the context of how it contributes to host health. A particular focus is placed on the microbiota present in female urine, a microbial community with great potential to inform novel strategies for the diagnosis and therapy of UTI, a disease that affects more than 150 million people per year worldwide (34, 41).

THE HUMAN BODY IS A COLLECTION OF NICHES WITH UNIQUE MICROBIAL SIGNATURES

The past century has seen significant progress in the functional characterization of the human microbiome. As early as 1908, the concept of a healthy microbiota was proposed by Metchnikoff with his suggestion of enriching the diet with a species of Lactobacillus thought to be beneficial in the human gut (46). In 1921, Cannon first characterized compositional changes in the gut flora by diet (46). Today, recent studies have estimated that the female and male human bodies are colonized by 44 × 1012 and 38 × 1012 individual microbial cells, respectively (51). With the advent of next-generation sequencing (NGS) technologies, significant progress in investigating the human microbiome has been made (52). The deployment of such technologies allows for high-throughput culture-independent characterization of the microbiome with minimal bias (53).

The evolution of metagenomics as a tractable and robust tool has fundamentally transformed our ability to characterize the taxonomic and genomic ecology of microbial ecosystems (52). Armed with metagenomics and advanced culturing techniques, numerous research endeavors have improved our understanding of not only which microbes reside within a particular ecosystem but also which genes, metabolic pathways, and physiological adaptations are needed to survive and thrive within a particular ecosystem (1, 2). Rather than searching for core taxa that may be enriched in a particular microbial ecosystem, the research focus is now switching to include the identification of the core gene sets and metabolic pathways needed for a microbial community to function within a niche (54). The genetic signature of a resident microbiota may encode functions that the host cannot perform, giving a competitive advantage to the host. At the time of writing of this review, the Human Microbiome Project (HMP) had profiled the resident microbiota at 18 distinct body sites of more than 250 healthy young individuals using both 16S rRNA gene amplicon sequencing and whole-genome shotgun metagenomics (13, 55). Among the accomplishments of the HMP is the first detailed metagenomic characterization of microbial niches throughout the human body. A critical observation made from these data is that each body site sampled seems to have a unique microbial signature and that no microbial taxa were found to be universally present across all body sites and individuals (1). For example, the mouth, gut, vagina, and skin microbial ecologies are each distinct from one another, and these sites harbor microbial populations that have evolved to survive in the unique chemical, metabolic, and immune environment of each niche (1, 51, 5671) (Fig. 1).

FIG 1.

FIG 1

Microbial niches and their environmental characteristics across the human body. Niche conditions for nasal (5658), oral (51, 5961), skin (51, 62, 63), gastrointestinal (51, 64, 65), urinary tract (6669), and vaginal (70, 71) sites are summarized. Abbreviations: Temp, temperature; O2, oxygen tension (sea-level average = 760 mm Hg); MD, microbial density (reported in relevant units). This figure was created with Biorender.

THE HUMAN URINARY MICROBIOME: AN UNEXPECTED NICHE COMES INTO FOCUS

Given the advances in our current understanding of the human microbiome, it seems reasonable to propose the existence of microbial communities in any body site which encounters the outside world. However, the decades-old dogma of UT sterility, particularly in the bladder, has only now begun to be recognized as disproven (4). Two seminal studies, by Nelson et al. (33) and Siddiqui et al. (5), used metagenomic approaches to demonstrate that the urine of healthy individuals is not sterile and that a UGT microbiome may exist in healthy female and male adults.

The environmental niche of the urinary tract.

The human UT can be broadly divided into upper and lower compartments. The upper compartment is comprised of the kidneys and ureters, and the lower compartment contains the bladder and urethra (Fig. 1) (72). The luminal surface of the lower urinary tract, bladder, and proximal urethra, called the urothelium, is a transitional epithelium coated by a thin glycosaminoglycan (GAG) layer (7277). The apical, differentiated cells of the urothelium, called umbrella cells, function to form an impermeable barrier between urinary waste products and underlying body tissues (78). Umbrella cell transmembrane proteins called uroplakins contribute to the barrier function of the urothelium by forming dense plaques on the apical surface of the umbrella cells (73). Because of the sheer force exerted by urinary flow, the ability to bind uroplakins via type 1 fimbriae is a critical virulence factor of UPEC; however, it is unknown if the UT microbiota also utilizes uroplakins as adhesion sites (79, 80). The spatial organization of the UT microbiota has not been assessed.

Factors influencing microbial colonization include environmental characteristics, such as pH, oxygen tension, osmolarity, nutrient availability, adhesion sites, and immune interaction (81). Urine pH varies among individuals and is usually acidic, although healthy urine pH can range from 5 to 8 (8284). Given that many bacteria possess strictly aerobic or anaerobic metabolism, oxygen availability in the UT may play a role in shaping the ecology and spatial organization of the UT microbiota (69, 85, 86). Arterial blood, for example, exhibits an observed range of oxygen tension of 75 to 100 mm Hg in healthy adults, while the UT oxygen tension ranges from 0.47 to 51.5 mm Hg (Fig. 1) (69, 85, 86). Shannon et al. recently reported that variation in the bladder urinary oxygen tension is correlated with compositional changes in the urinary microbiota (69). Importantly, to support microbial growth, the UT must contain a replenishable nutrient source (83, 87). In a healthy voiding state, the UT is a chemostat, constantly filling and voiding urine. The flux of new urine presumably supplies nutrients to support resident microbes (88). Human urine is composed of many soluble elements, including electrolytes, osmolytes, amino acids, and carbohydrates. To date, a catalogue of more than 2,600 compounds has been detected in urine (83, 87). However, there are other possible sources of nutrients within the UT. The urothelium is covered by a thin layer of GAGs that serves to lubricate and protect the underlying tissue (7375, 89). The composition of the human UT GAG layer is not fully understood. However, vaginal and gut mucosal epithelia are coated with a layer of excreted, viscous material comprised primarily of amino acids, mucins, various GAGs, and other complex carbohydrates (75, 8992). Interestingly, many human commensals, such as species of the genera Lactobacillus, Bifidobacterium, and Streptococcus, known to colonize the UT express enzymes that degrade various extracellular GAGs into smaller metabolizable sugars (9395). The metabolic specializations of UT-resident communities require further mechanistic studies to yield insights that may be clinically translated (83).

How do we study the human urinary microbiome?

Efforts to study and characterize the human urinary microbiome have resulted in the development of techniques aimed at accurately capturing a microbial community that is challenging to study (Table 1). The human urinary microbiome generally has low biomass in the absence of a UTI, harboring anywhere from <100 to <105 CFU per milliliter of urine (39). Therefore, efforts to profile a resident microbial community must take into account limited starting materials in urine samples, a concept that is well reviewed by Karstens et al. (67). A major consideration for any study assessing the microbiome is the sampling technique used. Three major sampling techniques have been used over the past 10 years to obtain urine samples proposed to represent the urinary microbiota and involve the collection of midstream clean-catch urine (CC) and the collection of urine through a transurethral catheter (TUC) and via suprapubic aspiration (SPA) (Table 1). Each of these methods has advantages and disadvantages. Most sampling techniques introduce perturbations to local microbial communities. With any sampling technique, it is important to consider that sampling order can result in the contamination of sequential sampling sites.

TABLE 1.

Methods and resources for studying the urinary microbiomea

Topic Methods Referencesb
Study design Cross sectional 10, 21, 53, 137139
Longitudinal 2, 44, 53, 103, 137, 138
Sample collection Voided/midstream clean catch 5, 9, 16, 17, 2732, 67
Transurethral catheterization 4, 9, 10, 13, 16, 18, 19, 22, 24, 31, 67
Suprapubic aspiration 9, 67
Data and metadata acquisition Culture based 4, 13, 19
16S rRNA amplicon sequencing 4, 5, 9, 10, 1318, 21, 22, 24, 2633, 67
Whole-genome metagenomics 11, 13, 15, 25, 53, 67
Data analysis Taxonomic profiling 4, 5, 9, 10, 1318, 21, 22, 24, 2633, 140
Functional analysis 11, 13, 15, 25, 53
Meta-analysis 53, 134, 135, 138, 141, 142
Data democratization Data curation, structure, public availability and ethics 53, 134, 142144
a

The table presents a summary of selected topics, methods, and references relevant to studying the human UT and UGT microbiome.

b

References 53, 67, 134, 137 to 139, and 140 to 144 consist of relevant technical reviews.

For CC, urine is obtained after it has passed through the entire lower urinary tract, and the advantage of the use of CC is that it may be collected by the least invasive means (9, 29). However, this method can suffer from skin, vaginal, and perineal contamination of the urine sample if the periurethral area is not sterilized before urine collection (67). A higher likelihood of contamination lends CC more toward use in the study of the UGT microbiota. Groups studying the spatial organization of the UT-specific microbiota have used a TUC to sample the bladder and urethral microbiota. The use of a TUC has the advantage of more specifically sampling the urinary microbial communities than the use of CC does. However, the insertion of a transurethral catheter to access the bladder disrupts and samples the urethral microbiota (67). One technique to decouple the perturbed urethral microbiota from the bladder-resident microbiota is to take urethral swabs to assess contamination. Finally, suprapubic aspiration (SPA) is the most accurate method to specifically sample the bladder-resident microbiota. However, it is also the most invasive and can be difficult to justify ethically, particularly among elderly populations (6, 9). During SPA, the bladder is accessed via insertion of a needle through the skin of the suprapubic area, just above the pubic bone, and urine is collected directly from the bladder (9). Indeed, Wolfe et al. found in a 2014 study that, when the use of a TUC and SPA was compared, SPA appeared to be the best method to minimize vulvovaginal and urethral contamination of the urine samples for the purposes of studying the bladder microbiome (9). However, the use of a TUC has also been reported to produce results similar to those produced by the use of SPA (6). Balancing the advantages and disadvantages in sampling techniques is a critical step in choosing methodologies for future studies.

Urine microbial culturing.

Typically, culture-dependent methods are used to test for the presence of uropathogens in urine samples. A critical advantage of urine culturing techniques is the ability to identify and verify viable bacterial populations, which have been done with samples collected with a TUC from the female UT by Hilt et al. (4). The diagnosis of UTI mainly relies on standard urine culture, a method that involves plating urine onto agar plates containing 5% sheep blood agar as well as MacConkey agar plates and incubating aerobically at 35°C for 24 h to obtain quantitative colony counts (4). However, this method has significant limitations in terms of the detection of bacteria in urine, with diagnostic thresholds being ≥105 culturable CFU/ml (96). Studies utilizing both 16S rRNA amplicon sequencing and advanced urine culture indicate the presence of viable bacteria in approximately 90% of samples with no growth by standard urine culture (4) (Table 1). New approaches, like enhanced quantitative urine culture (EQUC), have advanced our understanding of both pathogenic and commensal species within the UT and UGT microbiota (4, 9, 24). This technique includes the use of various combinations of different urine volumes, incubation times, and culture media, as well as aerobic, anaerobic, microaerobic, and CO2-enriched atmospheres (19). In contrast, the standard urine culture was designed to detect the major uropathogen E. coli and other bacterial species with similar growth requirements. Direct comparison of these methods in a 2016 study by Price et al. revealed that standard urine culture missed 67% of all uropathogens and 88% of non-E. coli uropathogens detected by the expanded-spectrum EQUC (19). To facilitate the use of EQUC in clinical laboratories, Price et al. developed the streamlined EQUC protocol, which involves plating 100 μl of urine onto blood agar plates, colistin-nalidixic acid agar plates, and MacConkey agar plates in 5% CO2 for 48 h (19). The streamlined EQUC protocol was reported to detect 84% of potential uropathogens cultured by the expanded EQUC protocol, whereas standard urine culture detected 33% of potential uropathogens (19). In a 2015 study comparing the urinary microbiomes of women with and without stress urinary incontinence sampled by the use of a TUC, Pearce et al. found that Lactobacillus, Corynebacterium, Streptococcus, Actinomyces, and Staphylococcus were the most prevalent genera isolated from urine by EQUC, followed by Aerococcus, Gardnerella, Bifidobacterium, and Actinobaculum (24). Thomas-White et al. reported that EQUC captured approximately 72% of the genera detected by metagenomics. EQUC did not detect anaerobes from the phyla Actinobacteria (Propionimicrobium, Varibaculum, Atopobium), Firmicutes (Peptoniphilus, Megasphaera, Finegoldia), or Bacteroidetes (Prevotella) (13).

Metagenomic sequencing of the urinary microbiome.

Even EQUC cannot detect the complete spectrum of resident microbial populations within the human UT and UGT. To assess the complete microbial composition without culture bias, researchers have employed NGS-based metagenomic sequencing approaches (35, 9, 10, 12, 13, 15, 27, 29, 31). Two major routes of metagenomic sequencing are currently used: 16S rRNA amplicon sequencing and whole-genome shotgun metagenomic sequencing (WGMS) (3, 15, 53) (Table 1). Both techniques rely on NGS technologies. However, 16S rRNA amplicon sequencing uses deep sequencing of amplicons containing the variable regions of the 16S rRNA gene. Downstream bioinformatic analysis allows for sequence identification, community profiling, and ecological structure assessment. An advantage and a disadvantage of 16S rRNA amplicon sequencing are its use of PCR amplification. As a result, 16S rRNA metagenomic sequencing possesses a high sensitivity to detect low-abundance microbial DNA without host contamination (15). However, 16S rRNA amplicon sequencing does not capture genomic sequence information outside the primer-specified amplicon and suffers from intrinsic bias in primer binding, which is not universally efficient across all taxa. Therefore, 16S rRNA amplicon sequencing can reasonably be used only for taxonomic assessment and measurements of relative abundance (15, 67). In contrast, WGMS sequences all the DNA present in a sample. Given a sufficient sequencing depth, WGMS has the potential to comprehensively analyze the whole metagenomic sequence space. WGMS experiments usually suffer from some degree of host contamination (53). For example, Moustafa et al. found that human genomic contamination accounted for 1.3% to 99.9% of the sequencing reads obtained from clinical urine samples (15). It is therefore necessary to sequence samples to a sufficient read depth to account for host contamination and appropriately sample the microbial community (53). Sample preparation as well as DNA extraction techniques can be optimized to enrich for microbial DNA (97, 98). With the whole metagenome available for analysis, it is possible to obtain the community structure and taxonomic profile as well as to functionally characterize the genetic potential of the resident microbiota (53). However, since community taxonomic profiling has been the major goal of most studies of the human urinary microbiome, 16S rRNA sequencing has been the major modality used to date (6).

The taxonomic profile of the human UT and UGT microbiomes.

Many taxonomic profiling studies have characterized the human urinary microbiome (4, 5, 911, 1319, 2133). The majority of these studies have used 16S rRNA amplicon sequencing and advanced culture techniques like EQUC to assess taxonomic enrichment between healthy and disease states (4, 5, 9) (Table 1). The major taxa identified in the healthy urinary microbiome are species generally considered to be fastidious, slow-growing microbes (5, 6, 9). Most belong to five major phyla, Firmicutes, Bacteroidetes, Actinobacteria, Fusobacteria, and Proteobacteria (5), and frequently include the genera Lactobacillus, Corynebacterium, Prevotella, Staphylococcus, and Streptococcus (6). Many studies have observed a high level of variability between individuals (31). As a result, the members of a core microbial community have not yet been defined.

Several studies using CC or mixed sampling techniques have identified differences between the male and female urinary microbiota (15, 17, 31). For example, Fouts et al. reported that the male urinary microbiota is significantly enriched with the genus Corynebacterium, which is typically associated with the skin microbiome. This finding could be related to contamination from skin when CC is used (31). Interestingly, numerous studies have found that the female urinary microbiomes are frequently dominated by the genus Lactobacillus (6). Data from a study by Thomas-White et al., using EQUC and whole-genome sequencing of bacterial isolates from urine collected with a TUC, suggest that the vaginal and UT microbiomes are interconnected. While it is possible that contamination during sampling could confound these results, the authors of this study demonstrate that the UT and vaginal microbiotas not only share clonal pathogens but also share commensal organisms associated with vaginal health (13). In healthy women of reproductive age, the vaginal microbiome is usually dominated by species belonging to the genus Lactobacillus (99). Lactobacillus crispatus, L. iners, L. gasseri, and L. jensenii are the most prevalent lactobacilli found in the vaginal microbiome (100). The vaginal microbiome is known to play a key role in the maintenance of vaginal pH and the prevention of various urogenital diseases (47, 99, 101). Evidence suggests that vaginal lactobacilli confer a level of protection against bacterial vaginosis, sexually transmitted diseases, and UTIs (47, 99). Continuing research focuses on deciphering the mechanistic basis of protection (47, 100103).

The temporal stability of the microbial populations within the human UT and UGT remains relatively unknown, as most of the studies performed to date have employed a cross-sectional cohort design (Table 1) (73). Future work is needed to understand how the urinary microbial community changes with both short- and long-term observation intervals. The bulk of studies characterizing the urinary microbiome have focused on urological disorders that disproportionately affect women, such as UTI, urinary incontinence, and pelvic organ prolapse (6, 7, 10, 12, 13, 16, 22, 24). This bias has led to the more thorough characterization of the female urinary microbiome. Although the urinary microbiome has now been taxonomically profiled, the high variability, the lack of a clear association with the host phenotype, and unknown temporal dynamics have left significant areas of opportunity for functional analysis by WGMS and longitudinal observation.

THE FEMALE URINARY MICROBIOME IN THE CONTEXT OF UTI AND rUTI

UTIs are one of the most commonly diagnosed infections in women and are second only to respiratory tract infections in postmenopausal and elderly women (39). Uncomplicated UTIs are not associated with catheterization or UT abnormality. The incidence of uncomplicated UTI is higher among women than among men across all age groups. Young, sexually active women report incidence rates ranging from 0.5 to 0.7 per person-year, while age-matched men report an incidence rate of about 0.01 per person-year (39). The relationship between the compositional dynamics of the urinary microbiome and the incidence of UTIs is currently an active area of research.

The pathobiology of rUTI is not completely understood; however, the disease can be modeled by an oscillating pattern of relapsed infection interspersed with periods of remission between infections (38). There are two main models to explain the etiology of rUTI: repeated ascending infections from a reservoir outside the UT or reemergence from a persistent population residing within the UT (104). Evidence for both models exists (105, 106). For example, Forde et al. recently showed the dynamics of an E. coli sequence type 131 strain over a 5-year period from an elderly female patient with rUTI and provided evidence of an intestinal reservoir for recurrence (107). On the other hand, Hannan et al. (108) and De Nisco et al. (109) showed the existence of persistent uropathogen populations within the bladder walls of mouse models and human patients, respectively, and a study by Schreiber et al. observed a high incidence of same-strain rUTI in a cohort of 14 women with frequent rUTI (110). Identifying the mechanistic signals that trigger the reemergence of persistent bacterial populations within the UT is critical to understanding the pathogenesis of rUTIs. Interestingly, in a 2017 report, Gilbert and colleagues showed that transient exposures to the vaginal microbiota, particularly Gardnerella vaginalis, can awaken quiescent UPEC reservoirs, leading to reinoculation of the bladder lumen, epithelial cell death and exfoliation, and kidney damage (111).

The relationship between the urinary microbiota compositional dynamics and UTI pathobiology is beginning to be closely studied in humans (15). There is evidence that both the infection itself and treatment strategies, such as antibiotic therapy, affect the urinary microbiome (15, 112, 113). To date, few metagenomic studies have directly profiled the urinary microbiome during UTI; as a result, it remains unclear how the urinary microbial community changes during and after infection. Advanced culturing techniques have been used to characterize the viable urinary microbiota during infection. The current progress in this area has largely been in establishing robust procedures for use in research and clinical laboratories (19). However, Price et al. have reported a differential abundance of several species between patients experiencing UTI and healthy controls (19). For example, K. pneumoniae, Streptococcus agalactiae, Aerococcus urinae, E. faecalis, E. coli, Staphylococcus aureus, and Streptococcus anginosus were found to be enriched in the UTI group compared to the no-UTI group (19). While it is valuable to identify and culture uropathogenic species, characterization of the whole ecology of the urinary microbiome during UTI currently requires the use of NGS-based metagenomics approaches. In 2014, a report on the first use of WGMS of clinical urine samples from 23 patients suspected of having a UTI was published (25). In this study, Hasman et al. detected polymicrobial communities in many of the samples that they analyzed (25). The authors were also able to identify, annotate, and quantify the abundance of antimicrobial resistance genes in these microbial communities (25). In a 2018 study, Moustafa et al. used both 16S rRNA amplicon sequencing and WGMS on 121 CC samples to profile the bacterial, viral, and eukaryotic components of the UGT microbiome (15). Their analysis identified three microbial signatures within patient urine. Two signatures were not associated with UTI and were dominated by Actinobacteria and Firmicutes. The UTI-associated microbiota signature was dominated by Proteobacteria and included many canonical uropathogens, such as UPEC, Klebsiella, Pseudomonas, and Enterobacter. Importantly, the analysis of UTI-associated metagenomes also detected several novel taxa associated with infection, such as Acidovorax, Rhodanobacter, and Oligella. The authors also noted the detection of mammalian viruses and bacteriophage, indicating that further study of the viral component of the urinary microbiome is needed to fully characterize the ecosystem (15). A distinct lack of WGMS analysis exists for urine from rUTI patients. Future, longitudinally designed WGMS studies would greatly aid in the fundamental understanding of the microbial ecology and pathobiology of rUTIs.

Clinical labs often dismiss urine cultures exhibiting multiple colony types, as they consider these results to be from periurethral or vaginal contamination. However, work characterizing polymicrobial infections of the UT has highlighted the potential importance of these previously underappreciated culture results. Polymicrobial UTIs occur more frequently in elderly or immunocompromised populations (114). A polymicrobial ecosystem facilitates the development of synergistic microbial relationships. Interestingly, murine models of polymicrobial UTI have shown that P. mirabilis and Staphylococcus saprophyticus act synergistically during coinfection to increase the incidence of ascending pyelonephritis (115). Similarly, it has been shown that P. mirabilis and UPEC act synergistically in murine coinfection through metabolic interactions that mutually enhance bacterial colonization and persistence (116). In a 2016 report, Keogh et al. showed a remarkable synergistic relationship between UPEC and E. faecalis, also a known gut commensal, in iron-limiting environments (117). E. faecalis secretes a metabolic cue, l-ornithine, that stimulates the biosynthesis of iron uptake machinery by UPEC (117). Taken together, these studies highlight the pathogenic potential of synergistic polymicrobial relationships within the UT and UGT, which may potentially be targeted for new UTI therapies (114).

Current rUTI therapeutic strategies include long-term antibiotic management, topical estrogens, and surgical interventions, like electrofulguration of trigonitis (37, 43, 104, 118). However, long-term or frequent antibiotic therapy remains the frontline treatment strategy (43, 104). Interestingly, Schilling et al. showed in a 2002 report that 10 days of therapy with trimethoprim-sulfamethoxazole, a common first-line antibiotic for the treatment of UTI, was insufficient to eradicate bacteria from bladder reservoirs in a mouse model of UTI (119). Emphasis is now being placed on using appropriate strategies of antimicrobial selection and stewardship for the treatment of UTI and rUTI (120). A legacy of the dogma of UT sterility is the use of antibiotics to treat related but potentially benign conditions, like asymptomatic bacteriuria (ASB), a common clinical observation of a positive urine culture in the absence of symptoms (121). Future whole-genome metagenome studies must shed light on how current paradigms of antibiotic therapy select for antimicrobial resistance and aim to differentiate the metagenomic signatures of UTI and ASB. In a 2019 report, Mulder et al. used CC to assess the effect of antibiotic therapy on the urinary microbiome composition of elderly people and found that antibiotic therapy was associated with an altered composition of the urinary microbiome (112). Interestingly, they noted that the genus Lactobacillus, which may be associated with UGT health, was significantly depleted in patients with a recent history of antibiotic use, while uropathogenic species were significantly enriched (112).

Modulation of the UGT microbiome with probiotics has shown promise as a possible route toward novel UTI therapeutics (122). A few studies suggest that the vaginal microbiome may support UGT health by acting as a reservoir for protective commensal species against UTI and rUTI (13, 122). The female UT, UGT, and vaginal microbiotas are frequently enriched in the genus Lactobacillus (6, 9, 17, 22). In the vaginal microbial niche, lactobacilli are known to act as a protective microbial population through the secretion of lactic acid, which modulates the local chemical environment (71, 103, 122125). The antimicrobial and protective properties of Lactobacillus-produced lactic acid have been associated with species that secrete large quantities of the d-(−)-isomer, such as L. crispatus (71). Edwards et al. have shown that a vaginal L. crispatus isolate confers protection against Chlamydia trachomatis infection, while Lactobacillus iners, which does not have the ability to synthesize d-(−)-lactic acid, does not confer protection (47). In a 2011 randomized, placebo-controlled clinical study, Stapleton et al. found a moderate reduction in the rUTI incidence in patients given a vaginal L. crispatus probiotic compared to that in patients given a placebo (122). These observations suggest a protective role for L. crispatus and imply a differential host benefit among colonizing Lactobacillus species.

A microbiome-aware future for management and prevention of UTI and rUTI.

With growing knowledge of the role that the urinary microbiota plays in urogenital health, it is becoming clear that UTI and rUTI treatment paradigms may need to consider the state of the urinary microbiome. The traditional goal of achieving UT sterility in the management of UTI and rUTI may destroy beneficial, protective microbial populations as well as pathogens. Without the beneficial microbiota, the UT may be thrown into a dysbiotic, sensitized state at risk for colonization by uropathogens. Probiotics are therefore a logical route for the development of novel therapies. However, a mechanistic understanding of how the urinary microbiota directly influences host health (and vice versa) must be gained before optimal probiotic microbial consortiums may be formulated. Recently, Lev-Sagie et al. reported that vaginal microbiome transplantation from healthy donors may be a viable therapeutic alternative for women with intractable bacterial vaginosis (48). Given that the vaginal and UT microbial niches are predicted to be highly interconnected, vaginal microbiome transplantation may also be an option for the treatment of rUTI (13). Understanding the relationship between urinary microbial populations and UTI risk is a critical step in developing not only therapeutic options but also diagnostic and prognostic tools. The development of molecular ecological signatures of health may allow the accurate identification of dysbiosis and allow at-risk communities to be screened for proactive interventions.

FUTURE DIRECTIONS IN STUDYING THE HUMAN UT AND UGT MICROBIOME

Gaining translational insights from the study of the human urinary microbiome will require concentrated efforts to fill gaps in our current understanding. To date, the majority of urinary microbiome studies have used cross-sectional cohort designs in order to compare individuals with a disease state to healthy controls. While they are extremely valuable, cross-sectional cohort designs are often confounded by interpersonal variability and are not able to capture temporal variation. Temporal stability is an important feature of a microbial niche and is incorporated into measurements of dysbiosis (44). Recent evidence shows that some, if not much, of the variability seen in other microbial niches can be attributed to temporal variation (103, 126). Employing longitudinal cohorts with participants enrolled to provide urine samples over a rationally defined time course would establish the baseline temporal variation in the urinary microbiota and shed light on the contribution of periodic changes in host physiology, such as menstruation and UTI, on the urinary microbiome (Table 1).

The use of 16S rRNA amplicon sequencing has been the major strategy in profiling the human urinary microbiome for 10 years, yet a core microbiome has not been established for the microbial niches of the bladder, UT, or UGT. Rather than identifying a core taxonomic enrichment, it may be more informative to establish a core genetic enrichment, which requires the use of WGMS. The gut microbiome is, by far, the most-studied human microbial niche (51). In a 2010 report of the MetaHIT project, Qin et al. reported the identification of 3.3 million nonredundant microbial genes from an estimated 1,150 microbial species from human fecal samples using WGMS (54). The catalogue of microbial genes within the human gut microbiome is approximately 150 times the size of the human genome and possesses a diverse range of functional potential. Of particular interest, the authors identified a core, minimal metagenome possessed by the human gut microbiota. Encoded within the gut minimal metagenome are genes and metabolic pathways that complement host physiology, such as the fermentation of sugar to short-chain fatty acids. Similarly, identifying the genetic potential of the urinary microbiome using WGMS will bring insight into which genes or metabolic pathways are required in the UT and UGT to establish host health. Clinical medicine will also benefit from the integration of WGMS, as the genetic assessment of antimicrobial resistance could enrich antimicrobial stewardship practices (25). A current area of research opportunity within the field deals with modeling the community dynamics of UT-resident microbial populations during infection. Commonly used sequencing depths are sometimes not sufficient to profile less abundant, UT microbial populations during UTI. Deeper sequencing of metagenomes and calculation of absolute abundances may help ascertain the fate of putative commensal populations during a UTI (67).

Host-microbe interactions in the UT have been defined by the model systems available to the research community. A critical feature of a model system is that it recapitulates the relevant physiology. To date, the modeling of human UT diseases, such as UTI, has relied primarily on mouse models, in which a TUC is used to inject high-density solutions of a uropathogen into the bladder. These models have been indispensable in defining the mechanisms of bacterial pathogenesis and the host response during UTI (108, 111, 127132). New model systems, including 3-dimensional cultured organoids and genetically engineered mouse models, that may contribute to the generation of tractable model systems for the mechanistic study of UTI and rUTI are being reported (129, 133). The incorporation of model systems that allow for the study of the immune system in relation to the urinary microbiome will be of great importance to the field. Host immune interactions with the urinary microbiome are currently not well understood and may lead to critical insight into UTI and rUTI pathogenesis as well as maintenance of the urinary microbiome ecological structure.

The public availability of human metagenomic data sets is crucial for translational progress, as it allows for large-scale meta-analyses and independent scrutinization of published results (134). In a 2017 report, Duvallet et al. performed a meta-analysis of 28 published, publicly available gut microbiome data sets from human case-control cohorts (135). They found consistent disease-associated changes in the microbiome composition across multiple independently produced data sets. They also identified common genus-level changes that were shared among multiple disease states, indicating a likely association with nonspecific changes in host health rather than specific clinical associations (135). Data democratization will be critical to the development of consensus microbial community structures of the human UT and UGT in health and disease (Table 1).

As the field moves toward a greater understanding of the urinary microbiome, many patients wait for the advancement that will improve their quality of life. A 2010 study found a significant impact of urinary tract infections on morale and a strong link between UTI incidence and depression among elderly women (136). It is the hopes of many that novel therapies for the management of UTI and rUTI are on the horizon. An understanding of how the urinary microbiome functions in health and disease is critical to this pursuit.

Footnotes

Citation Neugent ML, Hulyalkar NV, Nguyen VH, Zimmern PE, De Nisco NJ. 2020. Advances in understanding the human urinary microbiome and its potential role in urinary tract infection. mBio 11:e00218-20. https://doi.org/10.1128/mBio.00218-20.

REFERENCES

  • 1.Human Microbiome Project Consortium. 2012. Structure, function and diversity of the healthy human microbiome. Nature 486:207–214. doi: 10.1038/nature11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lloyd-Price J, Mahurkar A, Rahnavard G, Crabtree J, Orvis J, Hall AB, Brady A, Creasy HH, McCracken C, Giglio MG, McDonald D, Franzosa EA, Knight R, White O, Huttenhower C. 2017. Strains, functions and dynamics in the expanded Human Microbiome Project. Nature 550:61–66. doi: 10.1038/nature23889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Human Microbiome Project Consortium. 2012. A framework for human microbiome research. Nature 486:215–221. doi: 10.1038/nature11209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hilt EE, McKinley K, Pearce MM, Rosenfeld AB, Zilliox MJ, Mueller ER, Brubaker L, Gai X, Wolfe AJ, Schreckenberger PC. 2014. Urine is not sterile: use of enhanced urine culture techniques to detect resident bacterial flora in the adult female bladder. J Clin Microbiol 52:871–876. doi: 10.1128/JCM.02876-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Siddiqui H, Nederbragt AJ, Lagesen K, Jeansson SL, Jakobsen KS. 2011. Assessing diversity of the female urine microbiota by high throughput sequencing of 16S rDNA amplicons. BMC Microbiol 11:244. doi: 10.1186/1471-2180-11-244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Brubaker L, Wolfe AJ. 2017. The female urinary microbiota, urinary health and common urinary disorders. Ann Transl Med 5:34. doi: 10.21037/atm.2016.11.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Aragón IM, Herrera-Imbroda B, Queipo-Ortuño MI, Castillo E, Del Moral JS-G, Gómez-Millán J, Yucel G, Lara MF. 2018. The urinary tract microbiome in health and disease. Eur Urol Focus 4:128–138. doi: 10.1016/j.euf.2016.11.001. [DOI] [PubMed] [Google Scholar]
  • 8.Magistro G, Stief CG. 2019. The urinary tract microbiome: the answer to all our open questions? Eur Urol Focus 5:36–38. doi: 10.1016/j.euf.2018.06.011. [DOI] [PubMed] [Google Scholar]
  • 9.Wolfe AJ, Toh E, Shibata N, Rong R, Kenton K, Fitzgerald M, Mueller ER, Schreckenberger P, Dong Q, Nelson DE, Brubaker L. 2012. Evidence of uncultivated bacteria in the adult female bladder. J Clin Microbiol 50:1376–1383. doi: 10.1128/JCM.05852-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Price TK, Hilt EE, Thomas-White K, Mueller ER, Wolfe AJ, Brubaker L. 2020. The urobiome of continent adult women: a cross-sectional study. BJOG 127:193–201. doi: 10.1111/1471-0528.15920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Barraud O, Ravry C, Francois B, Daix T, Ploy MC, Vignon P. 2019. Shotgun metagenomics for microbiome and resistome detection in septic patients with urinary tract infections. Int J Antimicrob Agents 54:803–808. doi: 10.1016/j.ijantimicag.2019.09.009. [DOI] [PubMed] [Google Scholar]
  • 12.Thomas-White KJ, Gao X, Lin H, Fok CS, Ghanayem K, Mueller ER, Dong Q, Brubaker L, Wolfe AJ. 2018. Urinary microbes and postoperative urinary tract infection risk in urogynecologic surgical patients. Int Urogynecol J 29:1797–1805. doi: 10.1007/s00192-018-3767-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Thomas-White K, Forster SC, Kumar N, Van Kuiken M, Putonti C, Stares MD, Hilt EE, Price TK, Wolfe AJ, Lawley TD. 2018. Culturing of female bladder bacteria reveals an interconnected urogenital microbiota. Nat Commun 9:1557. doi: 10.1038/s41467-018-03968-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Shrestha E, White JR, Yu SH, Kulac I, Ertunc O, De Marzo AM, Yegnasubramanian S, Mangold LA, Partin AW, Sfanos KS. 2018. Profiling the urinary microbiome in men with positive versus negative biopsies for prostate cancer. J Urol 199:161–171. doi: 10.1016/j.juro.2017.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Moustafa A, Li W, Singh H, Moncera KJ, Torralba MG, Yu Y, Manuel O, Biggs W, Venter JC, Nelson KE, Pieper R, Telenti A. 2018. Microbial metagenome of urinary tract infection. Sci Rep 8:4333. doi: 10.1038/s41598-018-22660-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Thomas-White KJ, Kliethermes S, Rickey L, Lukacz ES, Richter HE, Moalli P, Zimmern P, Norton P, Kusek JW, Wolfe AJ, Brubaker L, National Institute of Diabetes and Digestive and Kidney Diseases Urinary Incontinence Treatment Network. 2017. Evaluation of the urinary microbiota of women with uncomplicated stress urinary incontinence. Am J Obstet Gynecol 216:55.e1–55.e16. doi: 10.1016/j.ajog.2016.07.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gottschick C, Deng Z-L, Vital M, Masur C, Abels C, Pieper DH, Wagner-Döbler I. 2017. The urinary microbiota of men and women and its changes in women during bacterial vaginosis and antibiotic treatment. Microbiome 5:99. doi: 10.1186/s40168-017-0305-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Thomas-White KJ, Hilt EE, Fok C, Pearce MM, Mueller ER, Kliethermes S, Jacobs K, Zilliox MJ, Brincat C, Price TK, Kuffel G, Schreckenberger P, Gai X, Brubaker L, Wolfe AJ. 2016. Incontinence medication response relates to the female urinary microbiota. Int Urogynecol J 27:723–733. doi: 10.1007/s00192-015-2847-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Price TK, Dune T, Hilt EE, Thomas-White KJ, Kliethermes S, Brincat C, Brubaker L, Wolfe AJ, Mueller ER, Schreckenberger PC. 2016. The clinical urine culture: enhanced techniques improve detection of clinically relevant microorganisms. J Clin Microbiol 54:1216–1222. doi: 10.1128/JCM.00044-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Karstens L, Asquith M, Davin S, Stauffer P, Fair D, Gregory WT, Rosenbaum JT, McWeeney SK, Nardos R. 2016. Does the urinary microbiome play a role in urgency urinary incontinence and its severity? Front Cell Infect Microbiol 6:78. doi: 10.3389/fcimb.2016.00078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Groah SL, Perez-Losada M, Caldovic L, Ljungberg IH, Sprague BM, Castro-Nallar E, Chandel NJ, Hsieh MH, Pohl HG. 2016. Redefining healthy urine: a cross-sectional exploratory metagenomic study of people with and without bladder dysfunction. J Urol 196:579–587. doi: 10.1016/j.juro.2016.01.088. [DOI] [PubMed] [Google Scholar]
  • 22.Pearce MM, Zilliox MJ, Rosenfeld AB, Thomas-White KJ, Richter HE, Nager CW, Visco AG, Nygaard IE, Barber MD, Schaffer J, Moalli P, Sung VW, Smith AL, Rogers R, Nolen TL, Wallace D, Meikle SF, Gai X, Wolfe AJ, Brubaker L, Pelvic Floor Disorders Network. 2015. The female urinary microbiome in urgency urinary incontinence. Am J Obstet Gynecol 213:347.e1–347.e11. doi: 10.1016/j.ajog.2015.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Willner D, Low S, Steen JA, George N, Nimmo GR, Schembri MA, Hugenholtz P. 2014. Single clinical isolates from acute uncomplicated urinary tract infections are representative of dominant in situ populations. mBio 5:e01064-13. doi: 10.1128/mBio.01064-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pearce MM, Hilt EE, Rosenfeld AB, Zilliox MJ, Thomas-White K, Fok C, Kliethermes S, Schreckenberger PC, Brubaker L, Gai X, Wolfe AJ. 2014. The female urinary microbiome: a comparison of women with and without urgency urinary incontinence. mBio 5:e01283-14. doi: 10.1128/mBio.01283-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hasman H, Saputra D, Sicheritz-Ponten T, Lund O, Svendsen CA, Frimodt-Møller N, Aarestrup FM. 2014. Rapid whole-genome sequencing for detection and characterization of microorganisms directly from clinical samples. J Clin Microbiol 52:139–146. doi: 10.1128/JCM.02452-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fricke WF, Maddox C, Song Y, Bromberg JS. 2014. Human microbiota characterization in the course of renal transplantation. Am J Transplant 14:416–427. doi: 10.1111/ajt.12588. [DOI] [PubMed] [Google Scholar]
  • 27.Lewis DA, Brown R, Williams J, White P, Jacobson SK, Marchesi JR, Drake MJ. 2013. The human urinary microbiome; bacterial DNA in voided urine of asymptomatic adults. Front Cell Infect Microbiol 3:41. doi: 10.3389/fcimb.2013.00041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hooton TM, Roberts PL, Cox ME, Stapleton AE. 2013. Voided midstream urine culture and acute cystitis in premenopausal women. N Engl J Med 369:1883–1891. doi: 10.1056/NEJMoa1302186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Siddiqui H, Lagesen K, Nederbragt AJ, Jeansson SL, Jakobsen KS. 2012. Alterations of microbiota in urine from women with interstitial cystitis. BMC Microbiol 12:205. doi: 10.1186/1471-2180-12-205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Nelson DE, Dong Q, Van der Pol B, Toh E, Fan B, Katz BP, Mi D, Rong R, Weinstock GM, Sodergren E, Fortenberry JD. 2012. Bacterial communities of the coronal sulcus and distal urethra of adolescent males. PLoS One 7:e36298. doi: 10.1371/journal.pone.0036298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fouts DE, Pieper R, Szpakowski S, Pohl H, Knoblach S, Suh MJ, Huang ST, Ljungberg I, Sprague BM, Lucas SK, Torralba M, Nelson KE, Groah SL. 2012. Integrated next-generation sequencing of 16S rDNA and metaproteomics differentiate the healthy urine microbiome from asymptomatic bacteriuria in neuropathic bladder associated with spinal cord injury. J Transl Med 10:174. doi: 10.1186/1479-5876-10-174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Dong Q, Nelson DE, Toh E, Diao L, Gao X, Fortenberry JD, Van der Pol B. 2011. The microbial communities in male first catch urine are highly similar to those in paired urethral swab specimens. PLoS One 6:e19709. doi: 10.1371/journal.pone.0019709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Nelson DE, Van Der Pol B, Dong Q, Revanna KV, Fan B, Easwaran S, Sodergren E, Weinstock GM, Diao L, Fortenberry JD. 2010. Characteristic male urine microbiomes associate with asymptomatic sexually transmitted infection. PLoS One 5:e14116. doi: 10.1371/journal.pone.0014116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Flores-Mireles AL, Walker JN, Caparon M, Hultgren SJ. 2015. Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nat Rev Microbiol 13:269–284. doi: 10.1038/nrmicro3432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Foxman B. 2003. Epidemiology of urinary tract infections: incidence, morbidity, and economic costs. Dis Mon 49:53–70. doi: 10.1067/mda.2003.7. [DOI] [PubMed] [Google Scholar]
  • 36.Foxman B. 2014. Urinary tract infection syndromes: occurrence, recurrence, bacteriology, risk factors, and disease burden. Infect Dis Clin North Am 28:1–13. doi: 10.1016/j.idc.2013.09.003. [DOI] [PubMed] [Google Scholar]
  • 37.Gaitonde S, Malik RD, Zimmern PE. 2019. Financial burden of recurrent urinary tract infections in women: a time-driven activity-based cost analysis. Urology 128:47–54. doi: 10.1016/j.urology.2019.01.031. [DOI] [PubMed] [Google Scholar]
  • 38.Jhang JF, Kuo HC. 2017. Recent advances in recurrent urinary tract infection from pathogenesis and biomarkers to prevention. Ci Ji Yi Xue Za Zhi 29:131–137. doi: 10.4103/tcmj.tcmj_53_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rowe TA, Juthani-Mehta M. 2013. Urinary tract infection in older adults. Aging Health 9:519–528. doi: 10.2217/ahe.13.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dielubanza EJ, Schaeffer AJ. 2011. Urinary tract infections in women. Med Clin North Am 95:27–41. doi: 10.1016/j.mcna.2010.08.023. [DOI] [PubMed] [Google Scholar]
  • 41.Stamm WE, Norrby SR. 2001. Urinary tract infections: disease panorama and challenges. J Infect Dis 183(Suppl 1):S1–S4. doi: 10.1086/318850. [DOI] [PubMed] [Google Scholar]
  • 42.Waller TA, Pantin SAL, Yenior AL, Pujalte G. 2018. Urinary tract infection antibiotic resistance in the United States. Prim Care 45:455–466. doi: 10.1016/j.pop.2018.05.005. [DOI] [PubMed] [Google Scholar]
  • 43.Malik RD, Wu YR, Christie AL, Alhalabi F, Zimmern PE. 2018. Impact of allergy and resistance on antibiotic selection for recurrent urinary tract infections in older women. Urology 113:26–33. doi: 10.1016/j.urology.2017.08.070. [DOI] [PubMed] [Google Scholar]
  • 44.Lloyd-Price J, Abu-Ali G, Huttenhower C. 2016. The healthy human microbiome. Genome Med 8:51. doi: 10.1186/s13073-016-0307-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, Takahashi M, Fukuda NN, Murakami S, Miyauchi E, Hino S, Atarashi K, Onawa S, Fujimura Y, Lockett T, Clarke JM, Topping DL, Tomita M, Hori S, Ohara O, Morita T, Koseki H, Kikuchi J, Honda K, Hase K, Ohno H. 2013. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504:446–450. doi: 10.1038/nature12721. [DOI] [PubMed] [Google Scholar]
  • 46.Postler TS, Ghosh S. 2017. Understanding the holobiont: how microbial metabolites affect human health and shape the immune system. Cell Metab 26:110–130. doi: 10.1016/j.cmet.2017.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Edwards VL, Smith SB, McComb EJ, Tamarelle J, Ma B, Humphrys MS, Gajer P, Gwilliam K, Schaefer AM, Lai SK, Terplan M, Mark KS, Brotman RM, Forney LJ, Bavoil PM, Ravel J. 2019. The cervicovaginal microbiota-host interaction modulates Chlamydia trachomatis infection. mBio 10:e01548-19. doi: 10.1128/mBio.01548-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lev-Sagie A, Goldman-Wohl D, Cohen Y, Dori-Bachash M, Leshem A, Mor U, Strahilevitz J, Moses AE, Shapiro H, Yagel S, Elinav E. 2019. Vaginal microbiome transplantation in women with intractable bacterial vaginosis. Nat Med 25:1500–1504. doi: 10.1038/s41591-019-0600-6. [DOI] [PubMed] [Google Scholar]
  • 49.Simon JC, Marchesi JR, Mougel C, Selosse MA. 2019. Host-microbiota interactions: from holobiont theory to analysis. Microbiome 7:5. doi: 10.1186/s40168-019-0619-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sanchez-Canizares C, Jorrin B, Poole PS, Tkacz A. 2017. Understanding the holobiont: the interdependence of plants and their microbiome. Curr Opin Microbiol 38:188–196. doi: 10.1016/j.mib.2017.07.001. [DOI] [PubMed] [Google Scholar]
  • 51.Sender R, Fuchs S, Milo R. 2016. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol 14:e1002533. doi: 10.1371/journal.pbio.1002533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Gilbert JA, Blaser MJ, Caporaso JG, Jansson JK, Lynch SV, Knight R. 2018. Current understanding of the human microbiome. Nat Med 24:392–400. doi: 10.1038/nm.4517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Quince C, Walker AW, Simpson JT, Loman NJ, Segata N. 2017. Shotgun metagenomics, from sampling to analysis. Nat Biotechnol 35:833–844. doi: 10.1038/nbt.3935. [DOI] [PubMed] [Google Scholar]
  • 54.Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, Mende DR, Li J, Xu J, Li S, Li D, Cao J, Wang B, Liang H, Zheng H, Xie Y, Tap J, Lepage P, Bertalan M, Batto J-M, Hansen T, Le Paslier D, Linneberg A, Nielsen HB, Pelletier E, Renault P, Sicheritz-Ponten T, Turner K, Zhu H, Yu C, Li S, Jian M, Zhou Y, Li Y, Zhang X, Li S, Qin N, Yang H, Wang J, Brunak S, Doré J, Guarner F, Kristiansen K, Pedersen O, Parkhill J, Weissenbach J, MetaHIT Consortium, Bork P, Ehrlich SD, Wang J. 2010. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464:59–65. doi: 10.1038/nature08821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. 2007. The Human Microbiome Project. Nature 449:804–810. doi: 10.1038/nature06244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.England RJ, Homer JJ, Knight LC, Ell SR. 1999. Nasal pH measurement: a reliable and repeatable parameter. Clin Otolaryngol 24:67–68. doi: 10.1046/j.1365-2273.1999.00223.x. [DOI] [PubMed] [Google Scholar]
  • 57.Keck T, Leiacker R, Riechelmann H, Rettinger G. 2000. Temperature profile in the nasal cavity. Laryngoscope 110:651–654. doi: 10.1097/00005537-200004000-00021. [DOI] [PubMed] [Google Scholar]
  • 58.Smith-Vaughan H, Byun R, Nadkarni M, Jacques NA, Hunter N, Halpin S, Morris PS, Leach AJ. 2006. Measuring nasal bacterial load and its association with otitis media. BMC Ear Nose Throat Disord 6:10. doi: 10.1186/1472-6815-6-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Aframian DJ, Davidowitz T, Benoliel R. 2006. The distribution of oral mucosal pH values in healthy saliva secretors. Oral Dis 12:420–423. doi: 10.1111/j.1601-0825.2005.01217.x. [DOI] [PubMed] [Google Scholar]
  • 60.Eskow RN, Loesche WJ. 1971. Oxygen tensions in the human oral cavity. Arch Oral Biol 16:1127–1128. doi: 10.1016/0003-9969(71)90218-4. [DOI] [PubMed] [Google Scholar]
  • 61.Geneva II, Cuzzo B, Fazili T, Javaid W. 2019. Normal body temperature: a systematic review. Open Forum Infect Dis 6:ofz032. doi: 10.1093/ofid/ofz032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Du Plessis JL, Stefaniak AB, Wilhelm KP. 2018. Measurement of skin surface pH. Curr Probl Dermatol 54:19–25. doi: 10.1159/000489514. [DOI] [PubMed] [Google Scholar]
  • 63.Oh J, Byrd AL, Deming C, Conlan S, NISC Comparative Sequencing Program, Kong HH, Segre JA. 2014. Biogeography and individuality shape function in the human skin metagenome. Nature 514:59–64. doi: 10.1038/nature13786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Evans DF, Pye G, Bramley R, Clark AG, Dyson TJ, Hardcastle JD. 1988. Measurement of gastrointestinal pH profiles in normal ambulant human subjects. Gut 29:1035–1041. doi: 10.1136/gut.29.8.1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zheng L, Kelly CJ, Colgan SP. 2015. Physiologic hypoxia and oxygen homeostasis in the healthy intestine. A review in the theme: cellular responses to hypoxia. Am J Physiol Cell Physiol 309:C350–C360. doi: 10.1152/ajpcell.00191.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Fransen F, Melchers MJB, Lagarde CMC, Meletiadis J, Mouton JW. 2017. Pharmacodynamics of nitrofurantoin at different pH levels against pathogens involved in urinary tract infections. J Antimicrob Chemother 72:3366–3373. doi: 10.1093/jac/dkx313. [DOI] [PubMed] [Google Scholar]
  • 67.Karstens L, Asquith M, Caruso V, Rosenbaum JT, Fair DA, Braun J, Gregory WT, Nardos R, McWeeney SK. 2018. Community profiling of the urinary microbiota: considerations for low-biomass samples. Nat Rev Urol 15:735–749. doi: 10.1038/s41585-018-0104-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Schell-Chaple HM, Liu KD, Matthay MA, Puntillo KA. 2018. Rectal and bladder temperatures vs forehead core temperatures measured with SpotOn monitoring system. Am J Crit Care 27:43–50. doi: 10.4037/ajcc2018865. [DOI] [PubMed] [Google Scholar]
  • 69.Shannon MB, Limeira R, Johansen D, Gao X, Lin H, Dong Q, Wolfe AJ, Mueller ER. 2019. Bladder urinary oxygen tension is correlated with urinary microbiota composition. Int Urogynecol J 30:1261–1267. doi: 10.1007/s00192-019-03931-y. [DOI] [PubMed] [Google Scholar]
  • 70.Hill DR, Brunner ME, Schmitz DC, Davis CC, Flood JA, Schlievert PM, Wang-Weigand SZ, Osborn TW. 2005. In vivo assessment of human vaginal oxygen and carbon dioxide levels during and post menses. J Appl Physiol (1985) 99:1582–1591. doi: 10.1152/japplphysiol.01422.2004. [DOI] [PubMed] [Google Scholar]
  • 71.Amabebe E, Anumba D. 2018. The vaginal microenvironment: the physiologic role of lactobacilli. Front Med (Lausanne) 5:181. doi: 10.3389/fmed.2018.00181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Abelson B, Sun D, Que L, Nebel RA, Baker D, Popiel P, Amundsen CL, Chai T, Close C, DiSanto M, Fraser MO, Kielb SJ, Kuchel G, Mueller ER, Palmer MH, Parker-Autry C, Wolfe AJ, Damaser MS. 2018. Sex differences in lower urinary tract biology and physiology. Biol Sex Differ 9:45. doi: 10.1186/s13293-018-0204-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ingersoll MA, Albert ML. 2013. From infection to immunotherapy: host immune responses to bacteria at the bladder mucosa. Mucosal Immunol 6:1041–1053. doi: 10.1038/mi.2013.72. [DOI] [PubMed] [Google Scholar]
  • 74.Nickel JC, Emerson L, Cornish J. 1993. The bladder mucus (glycosaminoglycan) layer in interstitial cystitis. J Urol 149:716–718. doi: 10.1016/s0022-5347(17)36191-8. [DOI] [PubMed] [Google Scholar]
  • 75.Anand M, Wang C, French J, Isaacson-Schmid M, Wall LL, Mysorekar IU. 2012. Estrogen affects the glycosaminoglycan layer of the murine bladder. Female Pelvic Med Reconstr Surg 18:148–152. doi: 10.1097/SPV.0b013e31824b76bd. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Cornish J, Lecamwasam JP, Harrison G, Vanderwee MA, Miller TE. 1988. Host defence mechanisms in the bladder. II. Disruption of the layer of mucus. Br J Exp Pathol 69:759–770. [PMC free article] [PubMed] [Google Scholar]
  • 77.Hutch JA. 1970. The role of urethral mucus in the bladder defense mechanism. J Urol 103:165–167. doi: 10.1016/s0022-5347(17)61914-1. [DOI] [PubMed] [Google Scholar]
  • 78.Khandelwal P, Abraham SN, Apodaca G. 2009. Cell biology and physiology of the uroepithelium. Am J Physiol Renal Physiol 297:F1477–F1501. doi: 10.1152/ajprenal.00327.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sokurenko EV, Chesnokova V, Dykhuizen DE, Ofek I, Wu XR, Krogfelt KA, Struve C, Schembri MA, Hasty DL. 1998. Pathogenic adaptation of Escherichia coli by natural variation of the FimH adhesin. Proc Natl Acad Sci U S A 95:8922–8926. doi: 10.1073/pnas.95.15.8922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jones CH, Pinkner JS, Roth R, Heuser J, Nicholes AV, Abraham SN, Hultgren SJ. 1995. FimH adhesin of type 1 pili is assembled into a fibrillar tip structure in the Enterobacteriaceae. Proc Natl Acad Sci U S A 92:2081–2085. doi: 10.1073/pnas.92.6.2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gritz EC, Bhandari V. 2015. The human neonatal gut microbiome: a brief review. Front Pediatr 3:17. doi: 10.3389/fped.2015.00017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Brumfitt W, Hamilton-Miller JM, Cooper J, Raeburn A. 1990. Relationship of urinary pH to symptoms of ‘cystitis.’ Postgrad Med J 66:727–729. doi: 10.1136/pgmj.66.779.727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Reitzer L, Zimmern P. 2019. Rapid growth and metabolism of uropathogenic Escherichia coli in relation to urine composition. Clin Microbiol Rev 33:e00101-19. doi: 10.1128/CMR.00101-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Simerville JA, Maxted WC, Pahira JJ. 2005. Urinalysis: a comprehensive review. Am Fam Physician 71:1153–1162. [PubMed] [Google Scholar]
  • 85.Landes RR, Leonhardt KO, Duruman N. 1964. A clinical study of the oxygen tension of the urine and renal structures. II. J Urol 92:171–178. doi: 10.1016/S0022-5347(17)63916-8. [DOI] [PubMed] [Google Scholar]
  • 86.Leonhardt KO, Landes RR. 1963. Oxygen tension of the urine and renal structures. Preliminary report of clinical findings. N Engl J Med 269:115–121. doi: 10.1056/NEJM196307182690301. [DOI] [PubMed] [Google Scholar]
  • 87.Bouatra S, Aziat F, Mandal R, Guo AC, Wilson MR, Knox C, Bjorndahl TC, Krishnamurthy R, Saleem F, Liu P, Dame ZT, Poelzer J, Huynh J, Yallou FS, Psychogios N, Dong E, Bogumil R, Roehring C, Wishart DS. 2013. The human urine metabolome. PLoS One 8:e73076. doi: 10.1371/journal.pone.0073076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Forsyth VS, Armbruster CE, Smith SN, Pirani A, Springman AC, Walters MS, Nielubowicz GR, Himpsl SD, Snitkin ES, Mobley H. 2018. Rapid growth of uropathogenic Escherichia coli during human urinary tract infection. mBio 9:e00186-18. doi: 10.1128/mBio.00186-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lilly JD, Parsons CL. 1990. Bladder surface glycosaminoglycans is a human epithelial permeability barrier. Surg Gynecol Obstet 171:493–496. [PubMed] [Google Scholar]
  • 90.Kati LM, Feldner PC, de Castro RA, Kobayashi EY, Sartori MGF, Nader HB, Castello Girão MJB. 2010. Analysis of glycosaminoglycans in the parametrium and vaginal apex of women with and without uterine prolapse. J Womens Health (Larchmt) 19:1341–1344. doi: 10.1089/jwh.2009.1567. [DOI] [PubMed] [Google Scholar]
  • 91.Martin R, Martin C, Escobedo S, Suarez JE, Quiros LM. 2013. Surface glycosaminoglycans mediate adherence between HeLa cells and Lactobacillus salivarius Lv72. BMC Microbiol 13:210. doi: 10.1186/1471-2180-13-210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Moghissi K, Neuhaus OW, Stevenson CS. 1960. Composition and properties of human cervical mucus. I. Electrophoretic separation and identification of proteins. J Clin Invest 39:1358–1363. doi: 10.1172/JCI104154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hynes WL, Walton SL. 2000. Hyaluronidases of Gram-positive bacteria. FEMS Microbiol Lett 183:201–207. doi: 10.1111/j.1574-6968.2000.tb08958.x. [DOI] [PubMed] [Google Scholar]
  • 94.Kawai K, Kamochi R, Oiki S, Murata K, Hashimoto W. 2018. Probiotics in human gut microbiota can degrade host glycosaminoglycans. Sci Rep 8:10674. doi: 10.1038/s41598-018-28886-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zuniga M, Monedero V, Yebra MJ. 2018. Utilization of host-derived glycans by intestinal Lactobacillus and Bifidobacterium species. Front Microbiol 9:1917. doi: 10.3389/fmicb.2018.01917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Wait RB. 1984. Urinary tract infection during pregnancy. Asymptomatic bacteriuria, acute cystitis, and acute pyelonephritis. Postgrad Med 75:153–161. doi: 10.1080/00325481.1984.11698643. [DOI] [PubMed] [Google Scholar]
  • 97.Feehery GR, Yigit E, Oyola SO, Langhorst BW, Schmidt VT, Stewart FJ, Dimalanta ET, Amaral-Zettler LA, Davis T, Quail MA, Pradhan S. 2013. A method for selectively enriching microbial DNA from contaminating vertebrate host DNA. PLoS One 8:e76096. doi: 10.1371/journal.pone.0076096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Marotz CA, Sanders JG, Zuniga C, Zaramela LS, Knight R, Zengler K. 2018. Improving saliva shotgun metagenomics by chemical host DNA depletion. Microbiome 6:42. doi: 10.1186/s40168-018-0426-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SS, McCulle SL, Karlebach S, Gorle R, Russell J, Tacket CO, Brotman RM, Davis CC, Ault K, Peralta L, Forney LJ. 2011. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci U S A 108:4680–4687. doi: 10.1073/pnas.1002611107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.France MT, Mendes-Soares H, Forney LJ. 2016. Genomic comparisons of Lactobacillus crispatus and Lactobacillus iners reveal potential ecological drivers of community composition in the vagina. Appl Environ Microbiol 82:7063–7073. doi: 10.1128/AEM.02385-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ceccarani C, Foschi C, Parolin C, D'Antuono A, Gaspari V, Consolandi C, Laghi L, Camboni T, Vitali B, Severgnini M, Marangoni A. 2019. Diversity of vaginal microbiome and metabolome during genital infections. Sci Rep 9:14095. doi: 10.1038/s41598-019-50410-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mendes-Soares H, Suzuki H, Hickey RJ, Forney LJ. 2014. Comparative functional genomics of Lactobacillus spp. reveals possible mechanisms for specialization of vaginal lactobacilli to their environment. J Bacteriol 196:1458–1470. doi: 10.1128/JB.01439-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Gajer P, Brotman RM, Bai G, Sakamoto J, Schutte UM, Zhong X, Koenig SS, Fu L, Ma ZS, Zhou X, Abdo Z, Forney LJ, Ravel J. 2012. Temporal dynamics of the human vaginal microbiota. Sci Transl Med 4:132ra52. doi: 10.1126/scitranslmed.3003605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Glover M, Moreira CG, Sperandio V, Zimmern P. 2014. Recurrent urinary tract infections in healthy and nonpregnant women. Urol Sci 25:1–8. doi: 10.1016/j.urols.2013.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Chen SL, Wu M, Henderson JP, Hooton TM, Hibbing ME, Hultgren SJ, Gordon JI. 2013. Genomic diversity and fitness of E. coli strains recovered from the intestinal and urinary tracts of women with recurrent urinary tract infection. Sci Transl Med 5:184ra60. doi: 10.1126/scitranslmed.3005497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Rosen DA, Hooton TM, Stamm WE, Humphrey PA, Hultgren SJ. 2007. Detection of intracellular bacterial communities in human urinary tract infection. PLoS Med 4:e329. doi: 10.1371/journal.pmed.0040329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Forde BM, Roberts LW, Phan MD, Peters KM, Fleming BA, Russell CW, Lenherr SM, Myers JB, Barker AP, Fisher MA, Chong TM, Yin WF, Chan KG, Schembri MA, Mulvey MA, Beatson SA. 2019. Population dynamics of an Escherichia coli ST131 lineage during recurrent urinary tract infection. Nat Commun 10:3643. doi: 10.1038/s41467-019-11571-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Hannan TJ, Mysorekar IU, Hung CS, Isaacson-Schmid ML, Hultgren SJ. 2010. Early severe inflammatory responses to uropathogenic E. coli predispose to chronic and recurrent urinary tract infection. PLoS Pathog 6:e1001042. doi: 10.1371/journal.ppat.1001042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.De Nisco NJ, Neugent M, Mull J, Chen L, Kuprasertkul A, de Souza Santos M, Palmer KL, Zimmern P, Orth K. 2019. Direct detection of tissue-resident bacteria and chronic inflammation in the bladder wall of postmenopausal women with recurrent urinary tract infection. J Mol Biol 431:4368–4379. doi: 10.1016/j.jmb.2019.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Schreiber HL IV, Conover MS, Chou WC, Hibbing ME, Manson AL, Dodson KW, Hannan TJ, Roberts PL, Stapleton AE, Hooton TM, Livny J, Earl AM, Hultgren SJ. 2017. Bacterial virulence phenotypes of Escherichia coli and host susceptibility determine risk for urinary tract infections. Sci Transl Med 9:eaaf1283. doi: 10.1126/scitranslmed.aaf1283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Gilbert NM, O'Brien VP, Lewis AL. 2017. Transient microbiota exposures activate dormant Escherichia coli infection in the bladder and drive severe outcomes of recurrent disease. PLoS Pathog 13:e1006238. doi: 10.1371/journal.ppat.1006238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Mulder M, Radjabzadeh D, Hassing RJ, Heeringa J, Uitterlinden AG, Kraaij R, Stricker BH, Verbon A. 2019. The effect of antimicrobial drug use on the composition of the genitourinary microbiota in an elderly population. BMC Microbiol 19:9. doi: 10.1186/s12866-018-1379-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Goneau LW, Hannan TJ, MacPhee RA, Schwartz DJ, Macklaim JM, Gloor GB, Razvi H, Reid G, Hultgren SJ, Burton JP. 2015. Subinhibitory antibiotic therapy alters recurrent urinary tract infection pathogenesis through modulation of bacterial virulence and host immunity. mBio 6:e00356-15. doi: 10.1128/mBio.00356-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kline KA, Lewis AL. 2016. Gram-positive uropathogens, polymicrobial urinary tract infection, and the emerging microbiota of the urinary tract. Microbiol Spectr 4:UTI-0012-2012. doi: 10.1128/microbiolspec.UTI-0012-2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Hjelm E, Lundell-Etherden I, Mårdh PA. 1987. Ascending urinary tract infections in rats induced by Staphylococcus saprophyticus and Proteus mirabilis. Acta Pathol Microbiol Immunol Scand B 95:347–350. doi: 10.1111/j.1699-0463.1987.tb03136.x. [DOI] [PubMed] [Google Scholar]
  • 116.Alteri CJ, Himpsl SD, Mobley HL. 2015. Preferential use of central metabolism in vivo reveals a nutritional basis for polymicrobial infection. PLoS Pathog 11:e1004601. doi: 10.1371/journal.ppat.1004601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Keogh D, Tay WH, Ho YY, Dale JL, Chen S, Umashankar S, Williams RBH, Chen SL, Dunny GM, Kline KA. 2016. Enterococcal metabolite cues facilitate interspecies niche modulation and polymicrobial infection. Cell Host Microbe 20:493–503. doi: 10.1016/j.chom.2016.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Crivelli JJ, Alhalabi F, Zimmern PE. 2019. Electrofulguration in the advanced management of antibiotic-refractory recurrent urinary tract infections in women. Int J Urol 26:662–668. doi: 10.1111/iju.13963. [DOI] [PubMed] [Google Scholar]
  • 119.Schilling JD, Lorenz RG, Hultgren SJ. 2002. Effect of trimethoprim-sulfamethoxazole on recurrent bacteriuria and bacterial persistence in mice infected with uropathogenic Escherichia coli. Infect Immun 70:7042–7049. doi: 10.1128/iai.70.12.7042-7049.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Abbo LM, Hooton TM. 2014. Antimicrobial stewardship and urinary tract infections. Antibiotics (Basel) 3:174–192. doi: 10.3390/antibiotics3020174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Cai T, Bartoletti R. 2017. Asymptomatic bacteriuria in recurrent UTI—to treat or not to treat. GMS Infect Dis 5:Doc09. doi: 10.3205/id000035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Stapleton AE, Au-Yeung M, Hooton TM, Fredricks DN, Roberts PL, Czaja CA, Yarova-Yarovaya Y, Fiedler T, Cox M, Stamm WE. 2011. Randomized, placebo-controlled phase 2 trial of a Lactobacillus crispatus probiotic given intravaginally for prevention of recurrent urinary tract infection. Clin Infect Dis 52:1212–1217. doi: 10.1093/cid/cir183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Pfau A, Sacks T. 1977. The bacterial flora of the vaginal vestibule, urethra and vagina in the normal premenopausal woman. J Urol 118:292–295. doi: 10.1016/S0022-5347(17)57976-8. [DOI] [PubMed] [Google Scholar]
  • 124.Heinemann C, Reid G. 2005. Vaginal microbial diversity among postmenopausal women with and without hormone replacement therapy. Can J Microbiol 51:777–781. doi: 10.1139/w05-070. [DOI] [PubMed] [Google Scholar]
  • 125.Chaban B, Links MG, Jayaprakash TP, Wagner EC, Bourque DK, Lohn Z, Albert AY, van Schalkwyk J, Reid G, Hemmingsen SM, Hill JE, Money DM. 2014. Characterization of the vaginal microbiota of healthy Canadian women through the menstrual cycle. Microbiome 2:23. doi: 10.1186/2049-2618-2-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Johnson AJ, Vangay P, Al-Ghalith GA, Hillmann BM, Ward TL, Shields-Cutler RR, Kim AD, Shmagel AK, Syed AN, Personalized Microbiome Class Students, Walter J, Menon R, Koecher K, Knights D. 2019. Daily sampling reveals personalized diet-microbiome associations in humans. Cell Host Microbe 25:789–802.e5. doi: 10.1016/j.chom.2019.05.005. [DOI] [PubMed] [Google Scholar]
  • 127.Ingersoll MA, Kline KA, Nielsen HV, Hultgren SJ. 2008. G-CSF induction early in uropathogenic Escherichia coli infection of the urinary tract modulates host immunity. Cell Microbiol 10:2568–2578. doi: 10.1111/j.1462-5822.2008.01230.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.O'Brien VP, Dorsey DA, Hannan TJ, Hultgren SJ. 2018. Host restriction of Escherichia coli recurrent urinary tract infection occurs in a bacterial strain-specific manner. PLoS Pathog 14:e1007457. doi: 10.1371/journal.ppat.1007457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Mora-Bau G, Platt AM, van Rooijen N, Randolph GJ, Albert ML, Ingersoll MA. 2015. Macrophages subvert adaptive immunity to urinary tract infection. PLoS Pathog 11:e1005044. doi: 10.1371/journal.ppat.1005044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Wiles TJ, Bower JM, Redd MJ, Mulvey MA. 2009. Use of zebrafish to probe the divergent virulence potentials and toxin requirements of extraintestinal pathogenic Escherichia coli. PLoS Pathog 5:e1000697. doi: 10.1371/journal.ppat.1000697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Yu L, O'Brien VP, Livny J, Dorsey D, Bandyopadhyay N, Colonna M, Caparon MG, Roberson ED, Hultgren SJ, Hannan TJ. 2019. Mucosal infection rewires TNFa signaling dynamics to skew susceptibility to recurrence. Elife 8:46677. doi: 10.7554/eLife.46677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.O'Brien VP, Hannan TJ, Yu L, Livny J, Roberson EDO, Schwartz DJ, Souza S, Mendelsohn CL, Colonna M, Lewis AL, Hultgren SJ. 2016. A mucosal imprint left by prior Escherichia coli bladder infection sensitizes to recurrent disease. Nat Microbiol 2:16196. doi: 10.1038/nmicrobiol.2016.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Horsley H, Dharmasena D, Malone-Lee J, Rohn JL. 2018. A urine-dependent human urothelial organoid offers a potential alternative to rodent models of infection. Sci Rep 8:1238. doi: 10.1038/s41598-018-19690-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Langille MGI, Ravel J, Fricke WF. 2018. “Available upon request”: not good enough for microbiome data! Microbiome 6:8. doi: 10.1186/s40168-017-0394-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Duvallet C, Gibbons SM, Gurry T, Irizarry RA, Alm EJ. 2017. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat Commun 8:1784. doi: 10.1038/s41467-017-01973-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Eriksson I, Gustafson Y, Fagerstrom L, Olofsson B. 2010. Do urinary tract infections affect morale among very old women? Health Qual Life Outcomes 8:73. doi: 10.1186/1477-7525-8-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Casals-Pascual C, Gonzalez A, Vazquez-Baeza Y, Song SJ, Jiang L, Knight R. 10 January 2020. Microbial diversity in clinical microbiome studies: sample size and statistical power considerations: statistical power for microbiome studies. Gastroenterology doi: 10.1053/j.gastro.2019.11.305. [DOI] [PubMed] [Google Scholar]
  • 138.Debelius J, Song SJ, Vazquez-Baeza Y, Xu ZZ, Gonzalez A, Knight R. 2016. Tiny microbes, enormous impacts: what matters in gut microbiome studies? Genome Biol 17:217. doi: 10.1186/s13059-016-1086-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hudson JI, Pope HG Jr, Glynn RJ. 2005. The cross-sectional cohort study: an underutilized design. Epidemiology 16:355–359. doi: 10.1097/01.ede.0000158224.50593.e3. [DOI] [PubMed] [Google Scholar]
  • 140.Lopez-Garcia A, Pineda-Quiroga C, Atxaerandio R, Perez A, Hernandez I, Garcia-Rodriguez A, Gonzalez-Recio O. 2018. Comparison of Mothur and QIIME for the analysis of rumen microbiota composition based on 16S rRNA amplicon sequences. Front Microbiol 9:3010. doi: 10.3389/fmicb.2018.03010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Thomas T, Gilbert J, Meyer F. 2012. Metagenomics—a guide from sampling to data analysis. Microb Inform Exp 2:3. doi: 10.1186/2042-5783-2-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Li PE, Lo CC, Anderson JJ, Davenport KW, Bishop-Lilly KA, Xu Y, Ahmed S, Feng S, Mokashi VP, Chain PS. 2017. Enabling the democratization of the genomics revolution with a fully integrated web-based bioinformatics platform. Nucleic Acids Res 45:67–80. doi: 10.1093/nar/gkw1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Shamarina D, Stoyantcheva I, Mason CE, Bibby K, Elhaik E. 2017. Communicating the promise, risks, and ethics of large-scale, open space microbiome and metagenome research. Microbiome 5:132. doi: 10.1186/s40168-017-0349-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.McGuire AL, Colgrove J, Whitney SN, Diaz CM, Bustillos D, Versalovic J. 2008. Ethical, legal, and social considerations in conducting the Human Microbiome Project. Genome Res 18:1861–1864. doi: 10.1101/gr.081653.108. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from mBio are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES