Preprint
Review

This version is not peer-reviewed.

Urinary Tract Microbiome Association with Diseases and Autoimmunity: Plausible Potential for Therapeutics and Accompanying Challenges

A peer-reviewed article of this preprint also exists.

Submitted:

28 October 2024

Posted:

30 October 2024

You are already at the latest version

Abstract

The urinary tract, once considered sterile, is now understood to host a diverse community of microorganisms known as the urinary microbiome. This microbiome, which is made up of bacteria, fungi, and viruses, is important for preserving urological health and has been linked to the pathogenesis of a number of urinary tract disorders. In this review, we offer a consolidated overview of the urinary tract bacterial population, including its composition, diversity, and factors influencing its dynamics. We explore its involvement in urinary tract infections, interstitial cystitis/bladder pain syndrome (IC/BPS), urinary stone formation, and other urological diseases, highlighting the importance of understanding its correlation with disease pathology for the development of therapeutic strategies. Additionally, we explore its role in autoimmune diseases of the urinary tract and the potential of leveraging the urinary microbiome for targeted interventions in the treatment of urinary tract infections. Longitudinal studies are emphasized for their ability to elucidate microbial dynamics, establish causality, and ensure consistency and reproducibility across research endeavors. These efforts underscore the necessity for continued research and multidisciplinary collaborations in this rapidly evolving field to advance our understanding and therapeutic strategies for urinary tract disorders.

Keywords: 
;  ;  ;  ;  ;  ;  

1. Introduction

The establishment of the Human Microbiome Project (HMP) in 2008 marked the beginning of a new chapter in our knowledge of the microbial populations that live inside of us. [1]. The population of bacteria that thrives in the urinary system, encompassing kidneys, ureters, bladder, and urethra, constitutes the urinary tract (UT) microbiome[2,3]. Although the urinary system was once thought to be sterile, advances in molecular biology techniques have shown that a wide variety of bacteria, fungi, and viruses may be present there [4]. The UT microbiome preserves the integrity of the associated epithelium, protect against infections and foster the appropriate operation of the immune system[5]. It maintains the urinary tract homeostasis through various mechanisms, including the regulation of pH levels, production of antimicrobial peptides, and competition for resources, which collectively help to establish and preserve a balanced urinary tract environment[6]. The human immune system and the epithelial cells lining the UT interact with the microbiota, which may impact the inflammatory and immunological responses[7]. Stimulation of host defense by modulating immune system can play crucial roles in defending against pathogens and maintaining tissue integrity.
The microbial composition of the urinary tract microbiota is influenced by various factors, including host genetics, anatomy, immune function, and external factors such as diet and antibiotic use[8]. The microbiota typically includes bacteria, but fungi and viruses may also be present in smaller quantities[9]. Roughly 90% of samples with no growth by normal urine culture contain live bacteria, according to studies using both 16S rRNA amplicon sequencing and advanced urine culture [10]. In healthy individuals, the urinary tract microbiota is often dominated by a few bacterial species, primarily from the genera Lactobacillus[11]. Numerous urological conditions, including UT infections, incontinence, bladder pain syndrome and urinary stone formation have been related to changes in the microbial community composition[12,13]. Several bacteria and fungi can cause UT infections, but the most frequent culprits are uropathogenic bacteria Escherichia coli, Klebsiella pneumoniae, Proteus mirabili,s Staphylococcus saprophyticus, Enterococcus faecalis, Proteus mirabilis [14] and fungus Candida sp.[15,16]. The urinary tract microbiota is dynamic and can change in response to various factors, such as hormonal fluctuations, sexual activity, urinary tract infections, and antibiotic treatment[17,18]. These changes can affect the balance of beneficial and potentially pathogenic microorganisms. Overall, urobiota contributes to the maintenance of the bladder homeostasis[19] and dysbiosis in it leads to health issues[20]. The diversity of the UT microbiota communities and their impact on health, namely the distinction between eubiosis and dysbiosis, are the subject of ongoing research. The present goal of scientific study on the UT microbiota is to comprehend the potential function that it may play in the development of UTIs as well as other medical conditions including cancer and other UIs [4]. Understanding the urinary tract microbiota has important clinical implications for the diagnosis and management of urinary tract infections and other urinary tract disorders[18,21]. For example, changes in the urinary microbiota may influence susceptibility to infections, treatment outcomes, and recurrence rates.

2. Methods to Study the Microbiome

While conventional urine culture methods primarily targeted aerobic microorganisms and pathogens like Escherichia coli, molecular techniques offer a more comprehensive view of the urinary microbiota[22,23]. Advent of cutting-edge molecular techniques have helped us get a better picture of the microbial population in the UT system. Urine microbiome can now be studied by two major methods; microbial culturing and metagenomic sequencing[24].

2.1. Microbial Culturing

To study UT microbiota, urine samples are usually tested for the presence of urinary pathogens using culture-dependent techniques. Standard urine culture, which includes plating urine onto agar plates containing 5% sheep blood agar as well as MacConkey agar plates and incubation aerobically at 35°C for 24 hours to produce quantitative colony counts, is the primary approach used to diagnose UTIs [25]. Nevertheless, the detection of bacteria in urine is severely limited by this approach, as the diagnostic criteria are ≥105. This traditional method for diagnosing urinary tract infections (UTIs), often misses low bacterial counts. Enhanced quantitative urine culture (EQUC) offers a more sensitive approach by utilizing larger urine volumes, multiple incubation durations, diverse culture media, varied incubation atmospheres (CO2-enriched, aerobic, anaerobic, microaerobic)[26]. Compared to standard culture, EQUC can detect 88% of non-E. coli uropathogens and 67% of all uropathogens missed by standard culture[27]. However, it still has limitations and this can lead to misdiagnosis and inappropriate treatment.

2.2. Metagenomic Sequencing

The entire range of persistent microbial communities in the human UT and UGT is undetectable, even by EQUC[28]. With next generation sequencing (NGS) based metagenomic sequencing techniques, researchers have evaluated the entire microbial composition without bias. Currently, 16S rRNA amplicon sequencing and shotgun metagenomic sequencing are the two main methods of metagenomic sequencing [29]. 16S rRNA Amplicon Sequencing involves deep sequencing of amplicons encompassing variable regions of the 16S rRNA gene, enabling ecological structure assessment, community profiling, and sequence identification through subsequent bioinformatics analyses [30]. While this method offers sensitive detection of low-abundance microbial DNA with minimal host contamination, it suffers from inherent bias in primer binding, limiting its application to taxonomic analysis and relative abundance assessments [31,32]. Conversely, shotgun metagenomics sequences all DNA molecules present in a sample, providing an in-depth analysis of the entire sequence space. Despite its potential for detailed examination, it often encounters host contamination, necessitating sufficient sequencing depth to discern microbial populations accurately[33]. Strategies to enhance microbial DNA enrichment through refined sample preparation and DNA extraction methods are crucial for minimizing host contamination. The comprehensive analysis of the metagenome facilitates the determination of taxonomic composition, community structure, and functional attributes of the local microbiota's genetic potential. Nonetheless, 16S rRNA sequencing remains prevalent in human urine microbiome studies, primarily due to its utility in community taxonomic profiling[34].

3. Variability of UT Microbiome

Variability in the composition and diversity of the microbiome stems from a myriad of factors encompassing age, gender, diet, lifestyle, and overall health status[35]. Predominantly, bacterial infections of the UT afflict adults, with microbial analyses revealing distinct microbial profiles across age and gender cohorts[17,36]. In individuals around 18 years old, prevalent taxa include Lactobacillus, Corynebacterium, Escherichia, and Streptococcus sp., whereas 28-year-old males exhibited Lactobacillus, Sneathia, Veillonella, Corynebacterium, and Prevotella. Females aged 27–67 years showcased a microbial landscape dominated by Lactobacillus, Prevotella, Gardnerella, Peptoniphilus, and Dialister[4]. Notably, both healthy males and females spanning various age ranges exhibit diverse bacterial taxa in midstream urine samples, including Lactobacillus, Klebsiella, Corynebacterium, and Staphylococcus, among others[37,38]. Gender-specific differences are discernible, likely influenced by physiological and hormonal variations, alongside differential excretion of urinary chemicals such as creatinine, citrate, calcium, and oxalate, which may modulate microbial colonization patterns [39]. Studies indicate that midstream urine samples from females demonstrate greater bacterial diversity compared to those from males, with Lactobacillus species prominently represented in fertile women alongside Actinobacteria and Bacteroidetes phyla[40]. Despite gender-specific distinctions, core genera such as Lactobacillus, Corynebacterium, and Streptococcus persist in the UT microbiome across genders and life stages, albeit in varying abundances [41,42]. Women bear a disproportionate burden of urinary tract infections (UTIs), with about 50% experiencing UTIs at some point in their lives, with heightened risks among postmenopausal and elderly individuals[43,44]. Conditions like interstitial cystitis/bladder pain syndrome (IC/BPS) and UT obstructions also influence UT microbiome dynamics, with pathogenic bacteria associated with UTIs and structural abnormalities disrupting microbial composition[45].
Beyond host-related factors, medications, dietary patterns, hygiene practices, and environmental variables exert substantial influence on the UT microbiome. Medications like antibiotics and cancer treatments, dietary habits, fluid intake, hygiene practices, and environmental exposures all shape microbial ecology and colonization patterns[46,47]. Comprehensive understanding of these multifaceted influences is crucial for elucidating the urinary microbiome's implications in health and disease and devising strategies for UT health maintenance.

4. Role in Health and Disease

The UT microbiome serves a pivotal role in maintaining health by contributing to protective mechanisms akin to those observed in the gut microbiome. Through niche occupation and resource competition, beneficial bacteria inhibit the colonization of pathogenic counterparts, thereby preventing microbial imbalances and UT infections [21]. Additionally, interactions between the UT microbiome and the local immune system are instrumental in modulating immune activity, crucial for preserving immunological homeostasis and averting inflammation or autoimmune responses[48,49,50]. Metabolic versatility allows microbes to metabolize various substances, including host-produced molecules and dietary components, affecting the nutritional and chemical landscape of the surrounding environment. The composition of the UT microbiome can influence the body response to treatments, such as medications for UT infections, suggesting the potential for developing more effective therapeutic interventions by elucidating microbiota functions in treatment outcomes[51]. Alterations in UT microbiome composition may serve as biomarkers for specific diseases or conditions, offering insights into personalized treatment strategies and providing valuable diagnostic or prognostic information[52]. The UT microbiome exhibits lower pH levels in a healthy state, which can rise in disease states. Presence of various probiotic strains like Lactobacillus rhamnosus, Lactobacillus fermentum, and Lactobacillus reuteri are beneficial and exert their antibacterial activity primarily through the production of lactic acid during carbohydrate metabolism in the epithelium's glycosaminoglycan layer. The resulting decrease in pH (≤4.5) creates an inhospitable environment for many pathogenic bacteria (Figure 1)[53]. Additionally, they produce other antibacterial substances like hydrogen peroxide and bacteriocin. Hence, probiotics and prebiotics represent promising avenues for UT illness management alongside personalized antibiotic therapies, informed by a deeper understanding of urine microbiome dynamics and composition [53].
Changes in the composition of the UT microbiome, known as dysbiosis, have been linked to various diseases, including UTIs, interstitial cystitis or bladder pain syndrome (IC/BPS), overactive bladder syndrome (OAB), urinary incontinence (UI), and kidney stones[28]. Additionally, the gut microbiota plays a significant role in regulating the development of certain UT illnesses through a mechanism known as the gut-bladder axis[54,55]. NGS studies have revealed a strong causal relationship between these disorders and the influence of the microbiome on both the bladder and gut. Consequently, dysbiosis of both the gut and urine microbiota may represent a significant contributing factor in the development of diseases affecting the urinary system [56]. Some of these have been elaborated below to highlight the role of microbiota in disease.

UTIs

UTIs can generally be classified as higher UTIs or lower UTIs. Upper UTIs involve infections in the kidneys or ureters, while lower UTIs refer to infections in the bladder or urethra[57]. E. coli bacteria are the main cause of UTIs[58]. Although this bacterium is naturally occurring in human bodies, it intermittently finds a way into the UT through the urethra and begins to proliferate in the bladder and ureters, leading to UT infections[59]. Another concept, referred to as the gut-vaginal bladder axis, suggests that when UTI causing strains enter the bladder by the gastrointestinal tract or vagina, they engage in polyinfection and quorum sensing [60]. Further evidence supports the role of vaginal microbiota in regulating UTI as well, because women with vaginosis caused by anaerobic G. vaginalis overgrowth have a higher risk of UTI than women with healthy microbial communities dominated by Lactobacillus [61]. In every age group, women are more likely than males to get an uncomplicated UTI. Age-matched men report an incidence rate of roughly 0.01 per person/year, whereas young, sexually active women report incidence rates ranging from 0.5 to 0.7 per person/year[62]. There is now a lot of research being done on the connection between the compositional dynamics of the urinary microbiome and the prevalence of UTIs [63].
Exposure to G. vaginalis leads to the outflow of latent intracellular reservoirs of E. coli in the bladder, hence increasing the risk of potentially fatal E. coli infections[64,65]. Exposure to G. vaginalis is sufficient to induce renal damage that persists after the microbe is eliminated from the urine system, as well as bladder epithelial apoptosis and exfoliation[64]. Therefore, UTI-associated disease in humans cannot be explained by a single invasion of an exogenous pathogenic bacterium. The complexity of UTI-associated diseases in humans underscores the need to consider both urinary microbiota imbalance and polymicrobial pathogenic causes for a comprehensive understanding of UTIs [45,66]. Thus, addressing UTIs requires a multifaceted approach that acknowledges the interplay between host physiology, microbial communities, and infectious agents.

Interstitial Cystitis or Bladder Pain Syndrome (IC/BPS)

Interstitial cystitis or bladder pain syndrome (IC/BPS) is a perplexing urological illness characterized by urgency of urine and discomfort in the bladder[67]. While the exact cause of IC/BPS remains unknown, recent research suggests a potential link between the UT microbiome and the development of the condition[68,69,70]. While traditionally, the microbiological diagnosis of UTIs relied on culture techniques, which may not adequately capture the diverse microbial communities present in patients with IC/BPS, recent advancements in metagenomic analysis have provided insights into the role of the urinary microbiome in this condition [71,72]. Xu et al. investigating the microbiome of individuals with IC/BPS have revealed significant correlations between urinary microbiota composition (upregulation of four opportunistic pathogenic genera, i.e. Serratia, Brevibacterium, Porphyromonas, and Citrobacter) and symptomatology[71]. Utilizing metagenomic techniques, Zheng et al. have identified alterations in the urinary microbiome of females (abundance of Lactobacillus and Escherichia-Shigella; decreased Bacteroides and Acinetobacter) vs males with IC/BPS compared to healthy controls[73]. Ceprnja et al. have further elaborated the contributing role of Gammaproteobacteria in IC by metagenomics and prediction through generalized Lotka-Volterra modeling[74].

Overactive Bladder Syndrome (OAB)

Overactive bladder syndrome (OAB) is a chronic medical disorder that significantly impacts the quality of life for both men and women, characterized by urgency of urination, frequency, and nocturia, with or without urgent incontinence, in the absence of obvious illness[75]. The multifactorial nature of OAB complicates its understanding, as the underlying causes may vary among individuals[76]. Previously, it was allied with neurological factors impacting bladder but now research has revealed that low-grade bacterial colonization or dysbiosis (imbalance) in the urinary tract might contribute to OAB symptoms in some individuals. The microbiomes of healthy women without OAB symptoms and those with OAB have revealed significant differences in the microbial composition of the bladder. Siddiqui et al. have used 16s rRNA technique to identify Streptococcus, Atopobium, Ureaplasma, Prevotella, Bacteroides in the OAB impacted patient[77]. Hilt et al.[78] have shown presence of Lactobacillus, Corynebacterium, Streptococcus, Actinomyces, Staphylococcus, Aerococcus, Gardnerella, Bifidobacterium, and Actinobaculum by using culture dependent techniques alongside 16s rRNA sequencing study of four women suffering from OAB. While Lactobacillus was less prevalent in the urine of women with OAB compared to healthy controls, the presence of Proteus was more frequently observed in OAB patients[79]. Bacterial presence may trigger inflammation in the bladder, leading to hypersensitivity and OAB symptoms. Certain bacteria might alter nerve signaling pathways involved in bladder control[75]. Using 70 patients, Li et al. identified certain bacterial genera (e.g., Porphyromona and Prevotella) to be significantly correlated with severity of OAB sub-symptoms[80]. A systematic review analysis by Sze et al. shows that no study reveals identical microbiota abundance, even among healthy individuals[81]. OAB patients exhibit a reduced bacterial diversity compared to controls and overall microbiome composition depends on the specimen collection method and the metagenomic sequencing technique used. OAB urine microbiome is more susceptible to alterations from gut or vaginal influences compared to controls[81,82]. Not all OAB cases are linked to changed UT microbiota and studies have identified a connection between OAB and the gastrointestinal tract microbiota as well[61]. Alterations in the gut microbiome composition have been linked to OAB and daily urine urgency, with individuals experiencing OAB symptoms exhibiting less microbial diversity compared to non-OAB groups. Specifically, the abundance of Bifidobacteriaceae was significantly lower in individuals with OAB and daily urine urgency.

Urinary Incontinence (UI)

UI refers to the involuntary loss of urine, a prevalent condition affecting millions worldwide[83]. While the causes of UI are diverse, a growing body of research suggests a complex interplay between the urinary tract microbiome and the development of UI, particularly urgency urinary incontinence (UUI)[84]. UUI has symptoms that resemble those of a urinary infection, including increased frequency and urgency of urination. Significant changes in the quantity and frequency of urinary microbiota have been discovered by both high-throughput sequencing and extended culture techniques [85]. A control cohort showed a decrease in Lactobacillus and an increase in Gardnerella in the UUI microbiome after using EQUC procedures to extract live bacteria from urine collected via transurethral catheter from women with UUI[86]. Culture revealed the presence of Actinobaculum, Actinomyces, Aerococcus, Arthrobacter, Corynebacterium, Gardnerella, Oligella, Staphylococcus, and Streptococcus in patients. Lactobacillus gasseri was more frequently detected in the UUI cohort, while Lactobacillus crispatus was most commonly found in controls[86]. Lactobacillus abundance was also allied with resistance to anticholinergic treatment for this condition[84]. Several investigations have suggested a potential connection between intestinal dysbiosis and UUI[61].

Kidney Stones

Kidney stones, also known as nephrolithiasis or renal calculi, are mineral deposits that form in the kidneys and can cause significant pain and discomfort when they obstruct urinary flow[87]. Nanobacteria have been linked to kidney stone formation, with 97.2% of the stones tested yielding positive results for nanobacteria[88]. Notably, only struvite stones exhibited the presence of common bacteria alongside nanobacteria. While apatite stones exhibited the highest nanobacteria antigen signals, the overall presence of nanobacteria was not affected by the stone type. In vitro experiments revealed that the isolated nanobacteria could indeed generate apatite stones, as demonstrated by the incorporation of calcium and strontium-85. Urease-producing bacteria, such as Proteus mirabilis, Klebsiella pneumoniae, Staphylococcus aureus, Pseudomonas aeruginosa, Providentia stuartii, Serratia, and Morganella morganii, are consistently associated with the formation and recurrence of struvite stones[89]. These bacteria produce urease, which breaks down urea, resulting in the formation of ammonia and carbon dioxide. This process leads to urine alkalinization and the subsequent formation of phosphate salts, contributing to stone formation in the kidneys. Moreover, there is a concurrent increase in acid-tolerant pathobionts among individuals with recurrent stone formation[90]. Additionally, specific enzymes involved in oxalate metabolism have been found to be elevated in those who suffer from kidney stones chronically.
Furthermore, the presence of biofilms, microbial communities encased in a protective matrix, on the surface of kidney stones may contribute to stone growth and recurrence. These biofilms can harbor bacteria and facilitate the adherence of crystals, promoting stone formation and providing a reservoir for recurrent infections [91]. When the urinary microbiome of kidney stone patients was investigated using 16S rRNA gene sequencing, it was found that the patients urine contained a different representation of bacteria linked to inflammation, such as Acinetobacter, and that the diversity of the urinary microbiota was significantly lower in these patients than in healthy controls. Thus, dysbiosis in the urinary and stomach microbiota appears to be a major factor in kidney stone formation, mediated by the rate at which oxalate is consumed or the control of the kidney's inflammatory response [92]. Certain bacteria in the gut and potentially the urinary tract may influence the breakdown and absorption of substances like oxalate, a major component of some kidney stones[93]. Clinical trials exploring the potential of using O. formigenes or its enzymes to decrease hyperoxaluria have exhibited encouraging findings. In vivo studies have demonstrated that Lactobacillus and Bifidobacterium species possess the capability to reduce oxalate levels[93].

Benign Prostatic Hyperplasia

Benign Prostatic Hyperplasia (BPH) is a condition characterized by the non-cancerous enlargement of the prostate gland, which can lead to urinary symptoms such as difficulty urinating, frequent urination, weak urine flow, and incomplete bladder emptying[94]. While BPH is primarily considered a condition related to changes in hormone levels and age, there is growing evidence suggesting a potential role for the microbiota in the development or exacerbation of BPH[95,96,97]. Propionibacterium acnes, commonly found in the skin microbiota and implicated in various inflammatory conditions like acne, has shown an association with BPH in some studies, suggesting a potential role in prostatic inflammation[98]. Okada et al. have also identified Burkholderia as a culprit for BPH[99]. Sarkar et al.[100] have also reported Kocuria palustris and Cellvibrio mixtus to be notably enriched in samples from patients with BPH. Jain et al. [101]revealed a unique microbial composition within BPH tissues, characterized by the notable presence of E. coli among other species such as Micrococcus spp, coagulase-positive Staphylococcus, Bacteroidetes, Proteobacteria, Firmicutes, and Actinobacteria. This finding suggests a potential role of these bacteria, particularly E. coli, in the inflammation and tissue damage associated with BPH. In vitro studies further demonstrated that E.coli associated with BPH can activate NF-κB signaling and induce DNA damage in prostate epithelial cells. Bajic et al. (2020) expanded the research, utilizing both mid-stream urine and transurethral catheterization samples. They utilized urine culture and 16S rRNA gene sequencing analysis, identifying Streptococcus, Veillonella, Gardnerella, Staphylococcus, and Candida as relevant microbiota[102]. They also found that an increase in the International Prostate Symptom Score (IPSS) was associated with significantly higher odds of detectable bacteria in catheterized urine, indicating the adequacy of catheterized urine in sampling the bladder microbiome

Glomerulonephritis

Glomerulonephritis, a condition characterized by inflammation in the kidney's glomeruli, can be triggered or exacerbated by certain microbial infections, particularly in cases of post-infectious glomerulonephritis[103]. Among the microbial agents responsible for this condition, Group A Streptococcus (Streptococcus pyogenes) is the most common culprit, often associated with throat or skin infections such as strep throat or impetigo, leading to what is known as post-streptococcal glomerulonephritis (PSGN)[103]. Although its incidence is now decreased in developed countries, it still occurs with notable frequency in densely populated and economically disadvantaged communities worldwide, where group A β-haemolytic streptococcal infections are prevalent[104]. Among children, PSGN remains the most common cause of acute glomerulonephritis. Bateman et al. [105]have stated that postinfectious glomerulonephritis represents an immune-mediated form of acute glomerulonephritis typically observed several weeks after an infection with Streptococcus pyogenes. Koyama et al.[106] have also reported its association with MRSA infection. It is hypothesized that post-MRSA glomerulonephritis may be induced by superantigens, leading to the production of high levels of cytokines and polyclonal activation of IgG and IgA. The formation of immune complexes containing IgA and IgG in the circulation subsequently contributes to the development of glomerulonephritis. Other bacteria, including Staphylococcus aureus, Streptococcus pneumoniae, Haemophilus influenzae, and Mycoplasma pneumoniae, have also been linked to cases of post-infectious glomerulonephritis, albeit less frequently[107,108,109]. In post-infectious glomerulonephritis, the immune response elicited by these microbial infections results in the deposition of immune complexes within the glomeruli, leading to inflammation and kidney tissue damage. However, not all cases of glomerulonephritis are directly linked to microbial infections, as various forms of the disease have different underlying causes.

Urinary Tract Cancer

Urinary tract cancer encompasses a range of malignancies that can affect different parts of the urinary system, including the kidneys, bladder, ureters, and urethra. While the role of the microbiota in urinary tract cancer is an area of ongoing research, there is growing evidence suggesting that alterations in the urinary microbiota may be associated with the development or progression of certain types of urinary tract cancer[110,111,112,113,114]. These differences include alterations in the abundance of specific bacterial species or taxa, as well as changes in overall microbial diversity can lead to cancer. Certain microbial metabolites produced by dysbiotic urinary microbiota may have carcinogenic properties or influence tumor progression. For example, metabolites such as nitrosamines and aromatic amines, which can be derived from dietary sources or microbial metabolism, have been implicated in bladder carcinogenesis [115]. Three key components have been identified to represent the tumor-promoting effect of E. coli in this context[116]. Firstly, the contamination of bladder tissues by E. coli increases the carcinogenic potential of nitrosamine precursors, potentially due to an elevation in nitrite production by the bacteria and sustained synthesis of nitrosamine through in-situ nitrosamine formation. Secondly, E. coli contamination accelerates urothelial cell proliferation, which may enhance the mutagenic effects of the carcinogenic agent. Thirdly, prolonged exposure to oxidative and nitrosative stress leads to DNA damage and mutation. These findings highlight the multifaceted mechanisms through which E. coli contamination in bladder tissues can contribute to the progression of cancer.
Heidler et al. [117]studied kidney cancer microbiota using amplicon metagenomics and found Aeromonas salmonicida, Pseudoalteromonas haloplanktis, Parageobacillus toebii, Trachelomonas volvocinopsis, Mycoplasma mycoides, and Halomicrobium mukohataei as common in the cancerous parts. Bacteria like Cyanophora paradoxa, Spirosoma navajo, Phaeocystis antarctica, Euglena mutabilis, and Mycoplasma vulturii were only found in the cancerous tissue. These bacteria can produce carcinogenic metabolites, induce chronic inflammation, or promote DNA damage, potentially contributing to carcinogenesis. However, the most extensively studied types of urinary tract cancer in relation to the microbiota is bladder cancer, where differences in the composition of the urinary microbiota between individuals with bladder cancer and healthy controls[118,119]. Increased abundance of certain bacteria, like Proteobacteria and Fusobacteria, has been linked to bladder cancer, while others like Lactobacillus may have protective effects. E. coli and other members of the Enterobacteriaceae family, have also been associated with bladder cancer[120].

Autoimmune Diseases

The urinary tract system organs are also affected by autoimmune diseases (Table 1). Although not all of them have been allied with impaired microbiota of urinary tract, there is evidence supporting role of bacteria in infection and autoimmunity trigger in some of the diseases, such as pyelonephritis.
Acute pyelonephritis is characterized by fever, back-pain, pus and foul smell of urine. Its primary culprits have been identified as E. coli and K. pneumoniae [133]. G. vaginalis abundance also exposes women to pyelonephritis[134]. Research has also shown that P. mirabilis and uropathogenic E. coli cooperate through metabolic interactions, allowing them to better colonize and persist within the urinary tract, potentially increasing the risk of pyelonephritis[135,136]. Keogh et al. (2016) revealed that in iron-limited environment, E. faecalis secretes L-ornithine to aid uropathogenic E. coli in acquiring iron, a nutrient crucial for bacterial growth and worsen infection during UTI, leading to pyelonephritis[137]. This suggests that these bacterial interactions can worsen infections. Staining of glomerular structures has revealed presence of H. pylori antigens in 100% of kidney samples from IgA vasculitis patients[138]. Haemophilus parainfluenzae has been detected in 35% of IgA vasculitis kidney biopsies, with a much lower prevalence (4%) in biopsies from patients with other kidney diseases[139]. Nephritis-associated plasmin receptor (NAPI-r), a group A streptococcal antigen has been detected in some patients with IgA vasculitis[140] as well as IgA nephropathy[141]. Kidney biopsies of some IgA nephropathy patients have also shown evidence of E. coli and H. influenza. This suggests that a direct bacterial infection with these species might contribute to IgA nephropathy and the exposure might trigger an autoimmune response that mistakenly attacks the glomeruli (filtering units) in the kidneys, leading to IgA nephropathy. H. pylori has also been implicated in the membranous nephropathy in almost two third of patients and some patients in Lupus nephritis [138]. However the pathogenic nature of the H. pylori presence remains yet to be elucidated in Lupus nephritis[142].
Aerobic culture of the seminal fluid of prostatitis patients revealed Enterobacteriaceae, enterococci and Staphylococcus aureus inpatients and absent from control group[143]. Nickel et al. [144]reported Burkholderia cenocepacia over represented in chronic prostatitis, using initial and mid-stream urine. Shoskes et al. [145]used urine DNA to identify higher counts of Clostridia compared with controls in prostatitis patients. Mandar et al. [146]further revealed that men with prostatitis had lower levels of health-promoting lactobacilli and a more diverse bacterial community (including proteobacteria) compared to healthy men[147].

Therapeutic Implications

The study of the urinary tract microbiota has significant therapeutic implications, particularly in the diagnosis, treatment, and prevention of UTIs and other urinary tract disorders[53]. Understanding the factors that influence the composition and stability of the urinary microbiota can inform preventive strategies for UTIs and other urinary tract disorders. Lifestyle modifications, dietary interventions, and hygiene practices aimed at promoting a healthy urinary microbiota may help reduce the risk of UTIs and maintain urinary tract health[148]. The development of UTI microbiome-targeted medicines has been encouraged by the discovery of the urinary microbiome and its link to disease. Antibacterial medication is the most widely used treatment for UTIs but broad-spectrum antibiotic treatment may have a deleterious effect on the beneficial bacterial flora of the host and leads to the pathogenic bacterial selective expansion. For some antimicrobials, prolonged usage of the drug can result in up to 50% of cases of bacterial resistance [53]. In an attempt to lower rates of antibiotic resistance and recurring infections, probiotics, which replicate purported defenses bolstered by the normal microbiome, have emerged as a therapeutic alternative for the prevention and treatment of UTIs [149]. The restoration of Lactobacillus communities is linked, in part, to the development of novel therapeutic approaches for urinary disorders[18]. Probiotics offer a promising avenue for restoring UT microbial balance and work in concert with the host immune system to produce compounds like vitamins and immunomodulators, acidify the mucosal surface and inhibit pathogen attachment [150,151]. Clinical studies have shown promising results, demonstrating the efficacy of probiotics in preventing and treating UTIs (Table 2) [152,153,154,155,156].
If specific bacterial components are found to trigger autoimmune responses, research could focus on developing therapies that target these specific immune cells or pathways to prevent them from attacking healthy tissues. The role of iron acquisition by bacteria like E. faecalis suggests that iron chelation therapies could be also explored as a way to starve harmful bacteria and potentially reduce their ability to worsen infections. The preservation of mucosal immunological homeostasis in the UT may be improved by microbiome transfer and restoration therapies that involve the inoculation of artificial communities of health-associated Lactobacilli. Probiotics can help restore the balance of the urinary microbiome, potentially reducing the risk of urinary tract infections and promoting overall urinary tract health [8]. Certain Lactobacillus strains, including L. rhamnosus GR1, L. fermentum RC-14, and L. reuteri B-54, have demonstrated efficacy in treating UTIs[157,158]. This positive impact is attributed to the release of lactic acid into the environment as these bacteria break down carbohydrates in the glycosaminoglycan layer of the lower urinary tract epithelium. Lactic acid contributes significantly to the antibacterial activity of Lactobacillus strains by lowering the pH (below 4.5), creating an unfavorable environment for most pathogenic bacteria[159]. Additionally, Lactobacillus species produce other metabolites such as bacteriocins and hydrogen peroxide, further enhancing their antimicrobial properties[160]. Lactobacillus species significantly suppress E. coli and help host urothelial cells produce type I interferon, which modifies the innate immunity in the bladder to produce a protective milieu for the host[120]. Lactobacilli leads to a change in the proportion of bacteria towards a more beneficial microbial makeup that guard against UTIs [161].
In addition to probiotics, exploring competitive alternatives like antibiotics is vital for addressing UTIs[162]. Competitive inhibition is where beneficial microorganisms outcompete uropathogens for resources, presents an effective strategy. These include bacteriocins and bacteriophages, which target specific bacterial strains without disrupting the UT microbiome equilibrium[163]. Human cathelicidins, defensins, and bacteriocins are examples of antimicrobial peptides that are secreted from the urothelium and have both antibacterial activity and immunomodulatory effects[164,165]. The presence of H. pylori in some autoimmune kidney diseases warrants further investigation and if a causal link is established, eradication of H. pylori with antibiotics could be a therapeutic option for some patients. Studies reporting synergism between strains such as E. coli and other bacteria highlight how bacterial cooperation can worsen infections. Understanding these interactions could lead to the development of drugs that disrupt these partnerships and potentially prevent infections that might trigger autoimmune responses.
To counter increasing multidrug resistance, bacteriocins and natural product based inhibitors to destroy rival organisms could be explored. An example of bacteriocin is colicins, which cause harm to target cells by inhibiting the formation of peptidoglycan, cellular nuclease degradation and membrane permeabilization via voltage-dependent channels[166]. They have been described as a means of preventing and lessening the development of biofilm on urinary catheters [167]. Several studies have reported successful outcomes using bacteriophages in treating UTIs caused by antibiotic-resistant bacteria. Bao et al. [168]have reported a synergistic approach involving non-active antibiotics and bacteriophages has been successfully employed to treat recurrent UTIs caused by extensively drug-resistant Klebsiella pneumoniae. This approach highlights the potential of combining different antimicrobial strategies to combat multidrug-resistant pathogens and underscores the importance of exploring novel treatment modalities in the face of increasing antibiotic resistance. Khawaldeh et al. have reported the successful treatment of a patient with a complicated UTI caused by multidrug-resistant Pseudomonas aeruginosa, which was resistant to antibiotics including gentamicin, ceftazidime, ciprofloxacin, and meropenem. Bacteriophages have also effectively removed mature biofilms formed by bacteria such as Escherichia coli and Proteus mirabilis, which are responsible for catheter-associated UTIs[169].
Preventive therapies include cranberry supplement containing compounds such as proanthocyanidins that may help prevent bacteria, particularly Escherichia coli, from adhering to the urinary tract lining[170]. Certain supplements or foods, such as vitamin C or foods high in citric acid, can acidify urine, creating an environment less conducive to bacterial growth[171]. Additionally, research suggests that D-Mannose may be effective in preventing UTIs and reducing the recurrence rate by interfering with the attachment of uropathogenic bacteria to UT epithelial cells (Figure 2)[172]. D-Mannose is a naturally occurring sugar that can inhibit bacterial adhesion to the UT lining and clinical studies have demonstrated the efficacy of D-Mannose supplementation in reducing UTI symptoms and recurrence, making it a promising alternative or adjunct therapy to antibiotics[173]. In addition, D-Mannose has been investigated for its potential in restoring UT microbiota balance. By preventing bacterial adhesion and colonization, D-Mannose may help maintain a healthy UT microbiota composition, reducing the risk of dysbiosis and UTI recurrence.

Future Perspective and Challenges

In future, microbiome studies have the potential to revolutionize the management of UTIs by offering personalized, community-based approaches. Understanding the normal urinary microbiome could aid in identifying microbial imbalances or dysbiosis that predispose individuals to UTIs, allowing for early intervention or prevention strategies. Co-occurrence network analysis to identify microbial species that frequently appear together to find cooperative/synergistic interactions could also be helpful. Culture independent methods have helped us gain insights at a deeper level but characterizing the diversity of microbes causing asymptomatic bacteriuria versus symptomatic infection could lead to more targeted treatment approaches, recognizing that not all bacteria need to be treated equally. Moreover, we also need long-term implications of antibiotic exposure on the urinary microbiome and recurrent infections need better characterization to optimize stewardship and prevent unintended consequences. Investigating microbial interactions and community dynamics may inform combination or sequential therapy approaches, enhancing treatment efficacy. Additionally, tracking microbial reservoirs could help eradicate sources of reinfection, particularly for recurrent UTIs associated with the re-emergence of previous infecting strains. Probiotics, synbiotics and phage therapy could be used to restore microbiome balance and train the immune system for long-term UTI prevention.
There are some challenges associated with UT microbiome research that need to be overcome in future. One challenge lies in standardizing urine collection and processing methods to ensure consistency and reproducibility across studies. Variations in collection techniques and laboratory protocols can introduce bias and affect the comparability of results. Standardizing these procedures will require collaboration among researchers, clinicians, and laboratory professionals to establish consensus guidelines and best practices. Another challenge is the need for robust computational analytics to analyze and interpret complex microbiome data effectively. Advances in bioinformatics tools and computational algorithms are essential for processing large-scale microbiome datasets, identifying microbial signatures associated with UTIs, and elucidating underlying mechanisms. Collaborative efforts between computational biologists, microbiologists, and clinicians are crucial for developing sophisticated analytical approaches and integrating multi-omics data for comprehensive insights. Longitudinal studies should also be conducted to enable researchers to track dynamic shifts in the urinary microbiome composition in response to various factors, including antibiotic treatment, lifestyle changes, and disease progression. This will help identify biomarkers predictive of UTI recurrence and treatment response, facilitating personalized risk prediction and prevention strategies.
Establishing causality between the urinary microbiota and UTI pathogenesis remains a complex and challenging task. While observational studies have identified associations between microbial dysbiosis and UTIs, establishing causality requires rigorous experimental designs, including intervention studies and mechanistic investigations. Critically appraising current evidence and addressing methodological limitations are essential steps toward elucidating the causal role of the microbiota in UTI pathogenesis. In addition, collaborative research efforts combining microbiome profiling, metabolomics, and immunological studies are essential for validating association and translating into clinical applications. We primarily focused on bacterial associations, but other aspects of the urinary tract microbiome, like fungi or viruses, might also play a role and need further investigation.
To facilitate standardized approaches and open data sharing in UT microbiome research, proposing frameworks and fostering multidisciplinary collaborations are imperative. Establishing consortia and research networks dedicated to UTI microbiome research can promote data harmonization, protocol standardization, and knowledge exchange among researchers worldwide. By fostering a collaborative ecosystem, we can overcome challenges, accelerate scientific discoveries, and ultimately improve UTI prevention, diagnosis, and treatment outcomes.

Conclusion

The urinary tract (UT) microbiome is a dynamic ecosystem that plays a significant role in both UT health and disease. Advancements in various culturing techniques and metagenomic sequencing have greatly contributed to our understanding of the UT microbiome community and its implications for disease. The UT microbiome is involved in a range of functions related to urinary tract health and pathology. By leveraging insights from microbiome research, these approaches have the potential to improve treatment outcomes and overcome challenges associated with antibiotic resistance in managing UTIs and other UT-related conditions. Novel therapeutic approaches utilizing microbiomes as probiotics or competitive alternatives show promise in minimizing UT microbiome-related diseases. Additionally, antimicrobial proteins offer supplementary therapeutic benefits to address antibiotic resistance in UT-related conditions. These innovative therapeutic strategies aim to restore microbial balance in the UT, potentially reducing the incidence of UTIs and other urinary tract-related disorders. Further research into the composition, dynamics, and functional significance of the UT microbiome is essential to fully comprehend its role in disease pathogenesis and to identify potential intervention targets. Other challenges in standardizing urine collection/processing methods, computational analytics, and establishing causality necessitate multidisciplinary collaborations for developing consensus guidelines, advanced analytical tools and translation of findings to clinical relevance.

Author Contributions

C.R.W., Z.B., M.O., K.M., Y.W., and S.S.H. prepared and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

C.R.W., M.O., and K.M. is employed by Shing Huei Group while Z.B. is employed by Alpha Genomics Private Limited. M.O., Y.W., and S.S.H declares no conflict of interest.

References

  1. Tang, J. Microbiome in the urinary system—a review. AIMS Microbiology 2017, 3, 143. [Google Scholar] [CrossRef] [PubMed]
  2. Hazen, J.E. The Enemy Within: An Investigation of the Intracellular Bacteria in Urinary Tract Infections. Washington University in St. Louis, 2022.
  3. Pratt, C. Modified reporting of positive urine cultures to reduce treatment of catheter-associated asymptomatic bacteriuria (CA-ASB) among inpatients: a randomized controlled trial. Memorial University of Newfoundland, 2021.
  4. Colella, M.; Topi, S.; Palmirotta, R.; D’Agostino, D.; Charitos, I.A.; Lovero, R.; Santacroce, L. An Overview of the Microbiota of the Human Urinary Tract in Health and Disease: Current Issues and Perspectives. Life 2023, 13, 1486. [Google Scholar] [CrossRef] [PubMed]
  5. Sirisinha, S. The potential impact of gut microbiota on your health: Current status and future challenges. Asian Pac J Allergy Immunol 2016, 34, 249–264. [Google Scholar]
  6. Jones, J.; Murphy, C.P.; Sleator, R.D.; Culligan, E.P. The urobiome, urinary tract infections, and the need for alternative therapeutics. Microbial Pathogenesis 2021, 161, 105295. [Google Scholar] [CrossRef]
  7. Duerkop, B.A.; Vaishnava, S.; Hooper, L.V. Immune responses to the microbiota at the intestinal mucosal surface. Immunity 2009, 31, 368–376. [Google Scholar] [CrossRef] [PubMed]
  8. Jones-Freeman, B.; Chonwerawong, M.; Marcelino, V.R.; Deshpande, A.V.; Forster, S.C.; Starkey, M.R. The microbiome and host mucosal interactions in urinary tract diseases. Mucosal Immunology 2021, 14, 779–792. [Google Scholar] [CrossRef]
  9. Rowan-Nash, A.D.; Korry, B.J.; Mylonakis, E.; Belenky, P. Cross-domain and viral interactions in the microbiome. Microbiology Molecular Biology Reviews 2019, 83, 10–1128. [Google Scholar] [CrossRef]
  10. Neugent, M.L.; Kumar, A.; Hulyalkar, N.V.; Lutz, K.C.; Nguyen, V.H.; Fuentes, J.L.; Zhang, C.; Nguyen, A.; Sharon, B.M.; Kuprasertkul, A.J.C.R.M. Recurrent urinary tract infection and estrogen shape the taxonomic ecology and function of the postmenopausal urogenital microbiome. 2022, 3.
  11. Kim, J.-M.; Park, Y.-J. Lactobacillus and urine microbiome in association with urinary tract infections and bacterial vaginosis. Urogenital Tract Infection 2018, 13, 7–13. [Google Scholar] [CrossRef]
  12. Marand, A.J.B.; Van Koeveringe, G.A.; Janssen, D.; Vahed, N.; Vögeli, T.-A.; Heesakkers, J.; Hajebrahimi, S.; Rahnama'i, M.S. Urinary microbiome and its correlation with disorders of the genitourinary system. 2021.
  13. Suarez Arbelaez, M.C.; Monshine, J.; Porto, J.G.; Shah, K.; Singh, P.K.; Roy, S.; Amin, K.; Marcovich, R.; Herrmann, T.R.; Shah, H.N. The emerging role of the urinary microbiome in benign noninfectious urological conditions: an up-to-date systematic review. World journal of urology 2023, 41, 2933–2948. [Google Scholar] [CrossRef]
  14. Rao, P.; Chennu, M.; Vennila, T.; Kosanam, S.; Ponsudha, P.; Suriyakrishnaan, K.; Alarfaj, A.A.; Hirad, A.H.; Sundaram, S.; Surendhar, P. Assessment of bacterial isolates from the urine specimens of urinary tract infected patient. BioMed Research International 2022, 2022. [Google Scholar]
  15. Yuan, F.; Huang, Z.; Yang, T.; Wang, G.; Li, P.; Yang, B.; Li, J. Pathogenesis of Proteus mirabilis in catheter-associated urinary tract infections. Urologia Internationalis 2021, 105, 354–361. [Google Scholar] [CrossRef] [PubMed]
  16. Pulipati, S.; Babu, P.S.; Narasu, M.L.; Anusha, N. An overview on urinary tract infections and effective natural remedies. J. Med. Plants 2017, 5, 50–56. [Google Scholar]
  17. Price, T.K.; Wolff, B.; Halverson, T.; Limeira, R.; Brubaker, L.; Dong, Q.; Mueller, E.R.; Wolfe, A.J. Temporal dynamics of the adult female lower urinary tract microbiota. MBio 2020, 11, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  18. Meštrović, T.; Matijašić, M.; Perić, M.; Čipčić Paljetak, H.; Barešić, A.; Verbanac, D. The role of gut, vaginal, and urinary microbiome in urinary tract infections: from bench to bedside. Diagnostics 2020, 11, 7. [Google Scholar] [CrossRef] [PubMed]
  19. Kawalec, A.; Zwolińska, D. Emerging role of microbiome in the prevention of urinary tract infections in children. International Journal of Molecular Sciences 2022, 23, 870. [Google Scholar] [CrossRef]
  20. Whiteside, S.A.; Razvi, H.; Dave, S.; Reid, G.; Burton, J.P. The microbiome of the urinary tract—a role beyond infection. Nature Reviews Urology 2015, 12, 81–90. [Google Scholar] [CrossRef]
  21. Neugent, M.L.; Hulyalkar, N.V.; Nguyen, V.H.; Zimmern, P.E.; De Nisco, N.J. Advances in understanding the human urinary microbiome and its potential role in urinary tract infection. MBio 2020, 11, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  22. Fritzenwanker, M.; Imirzalioglu, C.; Chakraborty, T.; Wagenlehner, F.M. Modern diagnostic methods for urinary tract infections. Expert review of anti-infective therapy 2016, 14, 1047–1063. [Google Scholar] [CrossRef]
  23. Xu, R.; Deebel, N.; Casals, R.; Dutta, R.; Mirzazadeh, M. A new gold rush: a review of current and developing diagnostic tools for urinary tract infections. Diagnostics 2021, 11, 479. [Google Scholar] [CrossRef]
  24. Gasiorek, M.; Hsieh, M.H.; Forster, C.S. Utility of DNA next-generation sequencing and expanded quantitative urine culture in diagnosis and management of chronic or persistent lower urinary tract symptoms. Journal of Clinical Microbiology 2019, 58, 10–1128. [Google Scholar] [CrossRef]
  25. Hilt, E.E.; McKinley, K.; Pearce, M.M.; Rosenfeld, A.B.; Zilliox, M.J.; Mueller, E.R.; Brubaker, L.; Gai, X.; Wolfe, A.J.; Schreckenberger, P.C. Urine is not sterile: use of enhanced urine culture techniques to detect resident bacterial flora in the adult female bladder. Journal of clinical microbiology 2014, 52, 871–876. [Google Scholar] [CrossRef] [PubMed]
  26. Deen, N.S.; Ahmed, A.; Tasnim, N.T.; Khan, N. Clinical relevance of expanded quantitative urine culture in health and disease. Frontiers in Cellular Infection Microbiology 2023, 13. [Google Scholar] [CrossRef] [PubMed]
  27. Price, T.K.; Dune, T.; Hilt, E.E.; Thomas-White, K.J.; Kliethermes, S.; Brincat, C.; Brubaker, L.; Wolfe, A.J.; Mueller, E.R.; Schreckenberger, P.C. The clinical urine culture: enhanced techniques improve detection of clinically relevant microorganisms. Journal of clinical microbiology 2016, 54, 1216–1222. [Google Scholar] [CrossRef] [PubMed]
  28. Ksiezarek, M.D. Comprehensive urogenital microbiome profiling: towards better understanding of female urinary tract in health and disease. 2022.
  29. Barraud, O.; Ravry, C.; François, B.; Daix, T.; Ploy, M.-C.; Vignon, P. Shotgun metagenomics for microbiome and resistome detection in septic patients with urinary tract infection. International journal of antimicrobial agents 2019, 54, 803–808. [Google Scholar] [CrossRef]
  30. Regueira-Iglesias, A.; Balsa-Castro, C.; Blanco-Pintos, T.; Tomás, I. Critical review of 16S rRNA gene sequencing workflow in microbiome studies: From primer selection to advanced data analysis. Molecular Oral Microbiology 2023, 38, 347–399. [Google Scholar] [CrossRef]
  31. Liao, Y. Emerging tools for uncovering genetic and transcriptomic heterogeneities in bacteria. Biophysical Reviews 2024, 1–16. [Google Scholar] [CrossRef]
  32. Abdi, G.; Tarighat, M.A.; Jain, M.; Tendulkar, R.; Tendulkar, M.; Barwant, M. Revolutionizing Genomics: Exploring the Potential of Next-Generation Sequencing. In Advances in Bioinformatics; Springer: 2024; pp. 1–33.
  33. Kumar, B.; Lorusso, E.; Fosso, B.; Pesole, G. A comprehensive overview of microbiome data in the light of machine learning applications: categorization, accessibility, and future directions. Frontiers in Microbiology 2024, 15, 1343572. [Google Scholar] [CrossRef]
  34. Moustafa, A.; Li, W.; Singh, H.; Moncera, K.J.; Torralba, M.G.; Yu, Y.; Manuel, O.; Biggs, W.; Venter, J.C.; Nelson, K.E. Microbial metagenome of urinary tract infection. Scientific reports 2018, 8, 4333. [Google Scholar] [CrossRef]
  35. Dasgupta, N.; Srivastava, A.; Rao, A.; Murugkar, V.; Shroff, R.; Das, G. Microbiome Diagnostics and Interventions in Health and Disease. Microbiome in Human Health Disease 2021, 157–215. [Google Scholar]
  36. Guliciuc, M.; Porav-Hodade, D.; Mihailov, R.; Rebegea, L.-F.; Voidazan, S.T.; Ghirca, V.M.; Maier, A.C.; Marinescu, M.; Firescu, D. Exploring the Dynamic Role of Bacterial Etiology in Complicated Urinary Tract Infections. Medicina 2023, 59, 1686. [Google Scholar] [CrossRef]
  37. Pohl, H.G.; Groah, S.L.; Pérez-Losada, M.; Ljungberg, I.; Sprague, B.M.; Chandal, N.; Caldovic, L.; Hsieh, M. The urine microbiome of healthy men and women differs by urine collection method. International Neurourology Journal 2020, 24, 41. [Google Scholar] [CrossRef] [PubMed]
  38. El-Alawi, S.H. Using culture independent approaches to gain insights into human urinary tract microbiome in healthy male and female individuals. 2021.
  39. Mueller, E.R.; Wolfe, A.J.; Brubaker, L. The female urinary microbiota. Current opinion in urology 2017, 27, 282. [Google Scholar] [CrossRef] [PubMed]
  40. Singer, M.; Koedooder, R.; Bos, M.P.; Poort, L.; Savelkoul, P.H.; Laven, J.S.; Morré, S.A.; Budding, A.E. The profiling of microbiota in vaginal and urine samples using 16s rRNA gene sequencing and IS-pro analysis. Bacterial interactions in the female genital tract 2018, 171. [Google Scholar]
  41. Tesei, D.; Jewczynko, A.; Lynch, A.M.; Urbaniak, C. Understanding the complexities and changes of the astronaut microbiome for successful long-duration space missions. Life 2022, 12, 495. [Google Scholar] [CrossRef]
  42. Fouts, D.E.; Pieper, R.; Szpakowski, S.; Pohl, H.; Knoblach, S.; Suh, M.-J.; Huang, S.-T.; Ljungberg, I.; Sprague, B.M.; Lucas, S.K. 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. Journal of translational medicine 2012, 10, 1–17. [Google Scholar] [CrossRef]
  43. Brady, S.S.; Bavendam, T.G.; Bradway, C.K.; Conroy, B.; Dowling-Castronovo, A.; Epperson, C.N.; Hijaz, A.K.; Hsi, R.S.; Huss, K.; Kim, M. Noncancerous genitourinary conditions as a public health priority: conceptualizing the hidden burden. Urology 2022, 166, 39–49. [Google Scholar] [CrossRef]
  44. Krishnaswamy, P.H.; Basu, M. Urinary tract infection in gynaecology and obstetrics. Obstetrics, Gynaecology Reproductive Medicine 2020, 30, 276–282. [Google Scholar] [CrossRef]
  45. Perez-Carrasco, V.; Soriano-Lerma, A.; Soriano, M.; Gutiérrez-Fernández, J.; Garcia-Salcedo, J.A. Urinary microbiome: yin and yang of the urinary tract. Frontiers in Cellular Infection Microbiology 2021, 11, 617002. [Google Scholar] [CrossRef]
  46. Patangia, D.V.; Anthony Ryan, C.; Dempsey, E.; Paul Ross, R.; Stanton, C. Impact of antibiotics on the human microbiome and consequences for host health. MicrobiologyOpen 2022, 11, e1260. [Google Scholar] [CrossRef]
  47. Josephs-Spaulding, J.; Krogh, T.J.; Rettig, H.C.; Lyng, M.; Chkonia, M.; Waschina, S.; Graspeuntner, S.; Rupp, J.; Møller-Jensen, J.; Kaleta, C. Recurrent urinary tract infections: unraveling the complicated environment of uncomplicated rUTIs. Frontiers in Cellular Infection Microbiology 2021, 11, 562525. [Google Scholar] [CrossRef]
  48. Lacerda Mariano, L.; Ingersoll, M.A. The immune response to infection in the bladder. Nature Reviews Urology 2020, 17, 439–458. [Google Scholar] [CrossRef] [PubMed]
  49. Sorić Hosman, I.; Cvitković Roić, A.; Lamot, L. A Systematic Review of the (Un) known Host Immune Response Biomarkers for Predicting Recurrence of Urinary Tract Infection. Frontiers in Medicine 2022, 9, 931717. [Google Scholar] [CrossRef] [PubMed]
  50. Liu, F.; Du, J.; Zhai, Q.; Hu, J.; Miller, A.W.; Ren, T.; Feng, Y.; Jiang, P.; Hu, L.; Sheng, J. The bladder microbiome, metabolome, cytokines, and phenotypes in patients with systemic lupus erythematosus. Microbiology Spectrum 2022, 10, e00212–00222. [Google Scholar] [CrossRef] [PubMed]
  51. Murray, B.O.; Flores, C.; Williams, C.; Flusberg, D.A.; Marr, E.E.; Kwiatkowska, K.M.; Charest, J.L.; Isenberg, B.C.; Rohn, J.L. Recurrent urinary tract infection: a mystery in search of better model systems. Frontiers in Cellular Infection Microbiology 2021, 11, 691210. [Google Scholar] [CrossRef]
  52. Hiergeist, A.; Gessner, A. Clinical implications of the microbiome in urinary tract diseases. Current Opinion in Urology 2017, 27, 93–98. [Google Scholar] [CrossRef]
  53. Aragon, I.M.; Herrera-Imbroda, B.; Queipo-Ortuño, M.I.; Castillo, E.; Del Moral, J.S.-G.; Gomez-Millan, J.; Yucel, G.; Lara, M.F. The urinary tract microbiome in health and disease. European urology focus 2018, 4, 128–138. [Google Scholar] [CrossRef]
  54. Brauer-Nikonow, A.; Zimmermann, M. How the gut microbiota helps keep us vitaminized. Cell Host Microbe 2022, 30, 1063–1066. [Google Scholar] [CrossRef] [PubMed]
  55. Salazar, A.M.; Neugent, M.L.; De Nisco, N.J.; Mysorekar, I.U. Gut-bladder axis enters the stage: Implication for recurrent urinary tract infections. Cell host microbe 2022, 30, 1066–1069. [Google Scholar] [CrossRef]
  56. Lee, K.W.; Song, H.Y.; Kim, Y.H. The microbiome in urological diseases. Investigative Clinical Urology 2020, 61, 338. [Google Scholar] [CrossRef]
  57. Tan, C.W.; Chlebicki, M.P. Urinary tract infections in adults. Singapore medical journal 2016, 57, 485. [Google Scholar] [CrossRef]
  58. Mao, B.-H.; Chang, Y.-F.; Scaria, J.; Chang, C.-C.; Chou, L.-W.; Tien, N.; Wu, J.-J.; Tseng, C.-C.; Wang, M.-C.; Chang, C.-C. Identification of Escherichia coli genes associated with urinary tract infections. Journal of clinical microbiology 2012, 50, 449–456. [Google Scholar] [CrossRef] [PubMed]
  59. Hickling, D.R.; Sun, T.T.; Wu, X.R. Anatomy and physiology of the urinary tract: relation to host defense and microbial infection. Urinary tract infections: Molecular pathogenesis clinical management.
  60. Yoo, J.-J.; Song, J.S.; Kim, W.B.; Yun, J.; Shin, H.B.; Jang, M.-A.; Ryu, C.B.; Kim, S.S.; Chung, J.C.; Kuk, J.C. Gardnerella vaginalis in recurrent urinary tract infection is associated with dysbiosis of the bladder microbiome. Journal of Clinical Medicine 2022, 11, 2295. [Google Scholar] [CrossRef] [PubMed]
  61. Choi, H.W.; Lee, K.W.; Kim, Y.H. Microbiome in urological diseases: Axis crosstalk and bladder disorders. Investigative Clinical Urology 2023, 64, 126. [Google Scholar] [CrossRef] [PubMed]
  62. Guay, D.R. Contemporary management of uncomplicated urinary tract infections. Drugs 2008, 68, 1169–1205. [Google Scholar] [CrossRef]
  63. Rowe, T.A.; Juthani-Mehta, M. Urinary tract infection in older adults. Aging health 2013, 9, 519–528. [Google Scholar] [CrossRef]
  64. Gilbert, N.M.; O’Brien, V.P.; Lewis, A.L. Transient microbiota exposures activate dormant Escherichia coli infection in the bladder and drive severe outcomes of recurrent disease. PLoS pathogens 2017, 13, e1006238. [Google Scholar] [CrossRef]
  65. O’Brien, V.P.; Lewis, A.L.; Gilbert, N.M. Bladder exposure to gardnerella activates host pathways necessary for Escherichia coli recurrent UTI. Frontiers in cellular infection microbiology 2021, 11, 788229. [Google Scholar] [CrossRef]
  66. Sathiananthamoorthy, S. Characterisation of the urinary microbial community and its association with lower urinary tract symptoms. UCL (University College London), 2018.
  67. Roth, S. " Interstitial cystitis" or" bladder pain syndrome"? Aktuelle Urologie 2008, 39, 163–164. [Google Scholar]
  68. Abernethy, M.G.; Rosenfeld, A.; White, J.R.; Mueller, M.G.; Lewicky-Gaupp, C.; Kenton, K. Urinary microbiome and cytokine levels in women with interstitial cystitis. Obstetrics Gynecology 2017, 129, 500–506. [Google Scholar] [CrossRef]
  69. Siddiqui, H.; Lagesen, K.; Nederbragt, A.J.; Jeansson, S.L.; Jakobsen, K.S. Alterations of microbiota in urine from women with interstitial cystitis. BMC microbiology 2012, 12, 1–11. [Google Scholar] [CrossRef]
  70. Abernethy, M.G.; Tsuei, A. The bladder microbiome and interstitial cystitis: is there a connection? Current Opinion in Obstetrics Gynecology 2021, 33, 469–473. [Google Scholar] [CrossRef] [PubMed]
  71. Xu, H.; Tamrat, N.E.; Gao, J.; Xu, J.; Zhou, Y.; Zhang, S.; Chen, Z.; Shao, Y.; Ding, L.; Shen, B. Combined signature of the urinary microbiome and metabolome in patients with interstitial cystitis. Frontiers in Cellular Infection Microbiology 2021, 11, 711746. [Google Scholar] [CrossRef]
  72. Nickel, J.C.; Stephens-Shields, A.J.; Landis, J.R.; Mullins, C.; van Bokhoven, A.; Lucia, M.S.; Henderson, J.P.; Sen, B.; Krol, J.E.; Ehrlich, G.D. A culture-independent analysis of the microbiota of female interstitial cystitis/bladder pain syndrome participants in the MAPP research network. Journal of clinical medicine 2019, 8, 415. [Google Scholar] [CrossRef]
  73. Zheng, Z.; Hu, J.; Li, W.; Ma, K.; Zhang, C.; Li, K.; Yao, Y. Integrated microbiome and metabolome analysis reveals novel urinary microenvironmental signatures in interstitial cystitis/bladder pain syndrome patients. Journal of Translational Medicine 2023, 21, 1–16. [Google Scholar] [CrossRef] [PubMed]
  74. Ceprnja, M.; Oros, D.; Melvan, E.; Svetlicic, E.; Skrlin, J.; Barisic, K.; Starcevic, L.; Zucko, J.; Starcevic, A. Modeling of urinary microbiota associated with cystitis. Frontiers in Cellular Infection Microbiology 2021, 11, 643638. [Google Scholar] [CrossRef]
  75. Peyronnet, B.; Mironska, E.; Chapple, C.; Cardozo, L.; Oelke, M.; Dmochowski, R.; Amarenco, G.; Gamé, X.; Kirby, R.; Van Der Aa, F. A comprehensive review of overactive bladder pathophysiology: on the way to tailored treatment. European urology focus 2019, 75, 988–1000. [Google Scholar] [CrossRef] [PubMed]
  76. Leron, E.; Weintraub, A.Y.; Mastrolia, S.A.; Schwarzman, P. Overactive bladder syndrome: evaluation and management. Current urology 2018, 11, 117–125. [Google Scholar] [CrossRef]
  77. Siddiqui, H.; Lagesen, K.; Nederbragt, A.J.; Eri, L.M.; Jeansson, S.L.; Jakobsen, K.S. Pathogens in urine from a female patient with overactive bladder syndrome detected by culture-independent high throughput sequencing: a case report. The open microbiology journal 2014, 8, 148. [Google Scholar] [CrossRef]
  78. Hilt, E.E.; McKinley, K.; Pearce, M.M.; Rosenfeld, A.B.; Zilliox, M.J.; Mueller, E.R.; Brubaker, L.; Gai, X.; Wolfe, A.J.; Schreckenberger, P.C. Urine is not sterile: use of enhanced urine culture techniques to detect resident bacterial flora in the adult female bladder. Journal of clinical microbiology 2014, 52, 871–876. [Google Scholar] [CrossRef]
  79. Curtiss, N.; Balachandran, A.; Krska, L.; Peppiatt-Wildman, C.; Wildman, S.; Duckett, J.J. A case controlled study examining the bladder microbiome in women with Overactive Bladder (OAB) and healthy controls. European Journal of Obstetrics Gynecology Reproductive Biology 2017, 214, 31–35. [Google Scholar] [CrossRef]
  80. Li, K.; Chen, C.; Zeng, J.; Wen, Y.; Chen, W.; Zhao, J.; Wu, P. Interplay between bladder microbiota and overactive bladder symptom severity: a cross-sectional study. BMC urology 2022, 22, 39. [Google Scholar] [CrossRef]
  81. Sze, C.; Pressler, M.; Lee, J.R.; Chughtai, B. The gut, vaginal, and urine microbiome in overactive bladder: a systematic review. International Urogynecology Journal 2022, 33, 1157–1164. [Google Scholar] [CrossRef] [PubMed]
  82. Jacobsen, G.E.; Amin, K. Our Knowledge of the Relationship of the Urinary Microbiome and Overactive Bladder: Past, Present, Future. Current Bladder Dysfunction Reports 2023, 18, 285–292. [Google Scholar] [CrossRef]
  83. Milsom, I.; Coyne, K.S.; Nicholson, S.; Kvasz, M.; Chen, C.-I.; Wein, A.J. Global prevalence and economic burden of urgency urinary incontinence: a systematic review. European urology focus 2014, 65, 79–95. [Google Scholar] [CrossRef]
  84. Govender, Y.; Gabriel, I.; Minassian, V.; Fichorova, R. The current evidence on the association between the urinary microbiome and urinary incontinence in women. Frontiers in cellular infection microbiology 2019, 9, 133. [Google Scholar] [CrossRef]
  85. Aoki, Y.; Brown, H.W.; Brubaker, L.; Cornu, J.N.; Daly, J.O.; Cartwright, R. Urinary incontinence in women. Nature reviews Disease primers 2017, 3, 1–20. [Google Scholar] [CrossRef]
  86. Pearce, M.M.; Hilt, E.E.; Rosenfeld, A.B.; Zilliox, M.J.; Thomas-White, K.; Fok, C.; Kliethermes, S.; Schreckenberger, P.C.; Brubaker, L.; Gai, X. The female urinary microbiome: a comparison of women with and without urgency urinary incontinence. MBio 2014, 5, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  87. Khan, S.R.; Pearle, M.S.; Robertson, W.G.; Gambaro, G.; Canales, B.K.; Doizi, S.; Traxer, O.; Tiselius, H.-G. Kidney stones. Nature reviews Disease primers 2016, 2, 1–23. [Google Scholar] [CrossRef]
  88. Çiftçioglu, N.; Björklund, M.; Kuorikoski, K.; Bergström, K.; Kajander, E.O. Nanobacteria: an infectious cause for kidney stone formation. Kidney international 1999, 56, 1893–1898. [Google Scholar] [CrossRef]
  89. Wang, Z.; Zhang, Y.; Zhang, J.; Deng, Q.; Liang, H. Recent advances on the mechanisms of kidney stone formation. International journal of molecular medicine 2021, 48, 1–10. [Google Scholar] [CrossRef]
  90. Suryavanshi, M.V.; Bhute, S.S.; Jadhav, S.D.; Bhatia, M.S.; Gune, R.P.; Shouche, Y.S. Hyperoxaluria leads to dysbiosis and drives selective enrichment of oxalate metabolizing bacterial species in recurrent kidney stone endures. Scientific reports 2016, 6, 1–15. [Google Scholar] [CrossRef] [PubMed]
  91. Bichler, K.-H.; Eipper, E.; Naber, K.; Braun, V.; Zimmermann, R.; Lahme, S. Urinary infection stones. International journal of antimicrobial agents 2002, 19, 488–498. [Google Scholar] [CrossRef] [PubMed]
  92. Xie, J.; Huang, J.-s.; Huang, X.-j.; Peng, J.-m.; Yu, Z.; Yuan, Y.-q.; Xiao, K.-f.; Guo, J.-n. Profiling the urinary microbiome in men with calcium-based kidney stones. BMC microbiology 2020, 20, 1–10. [Google Scholar] [CrossRef] [PubMed]
  93. Abratt, V.R.; Reid, S.J. Oxalate-degrading bacteria of the human gut as probiotics in the management of kidney stone disease. Advances in applied microbiology 2010, 72, 63–87. [Google Scholar] [PubMed]
  94. Kishorebabu, A.; Sree, S.N.; Chandralekha, S.P. A Review on Benign Prostatic Hyperplasia. World Journal of Current Medical Pharmaceutical Research 2019, 192–197. [Google Scholar]
  95. Kim, M.S.; Jung, S.I. The urinary tract microbiome in male genitourinary diseases: focusing on benign prostate hyperplasia and lower urinary tract symptoms. International Neurourology Journal 2021, 25, 3. [Google Scholar] [CrossRef]
  96. Takezawa, K.; Fujita, K.; Matsushita, M.; Motooka, D.; Hatano, K.; Banno, E.; Shimizu, N.; Takao, T.; Takada, S.; Okada, K. The Firmicutes/Bacteroidetes ratio of the human gut microbiota is associated with prostate enlargement. The Prostate 2021, 81, 1287–1293. [Google Scholar] [CrossRef]
  97. Li, J.; Li, Y.; Zhou, L.; Li, C.; Liu, J.; Liu, D.; Fu, Y.; Wang, Y.; Tang, J.; Zhou, L. The human microbiome and benign prostatic hyperplasia: Current understandings and clinical implications. Microbiological Research 2024, 127596. [Google Scholar] [CrossRef]
  98. Radej, S.; Szewc, M.; Maciejewski, R. Prostate infiltration by Treg and Th17 cells as an immune response to Propionibacterium acnes infection in the course of benign prostatic hyperplasia and prostate cancer. International Journal of Molecular Sciences 2022, 23, 8849. [Google Scholar] [CrossRef]
  99. Okada, K.; Takezawa, K.; Tsujimura, G.; Imanaka, T.; Kuribayashi, S.; Ueda, N.; Hatano, K.; Fukuhara, S.; Kiuchi, H.; Fujita, K. Localization and potential role of prostate microbiota. Frontiers in Cellular Infection Microbiology 2022, 12, 1048319. [Google Scholar] [CrossRef]
  100. Sarkar, P.; Malik, S.; Banerjee, A.; Datta, C.; Pal, D.K.; Ghosh, A.; Saha, A. Differential microbial signature associated with benign prostatic hyperplasia and prostate cancer. Frontiers in Cellular Infection Microbiology 2022, 12, 894777. [Google Scholar] [CrossRef] [PubMed]
  101. Jain, S.; Samal, A.G.; Das, B.; Pradhan, B.; Sahu, N.; Mohapatra, D.; Behera, P.K.; Satpathi, P.S.; Mohanty, A.K.; Satpathi, S. Escherichia coli, a common constituent of benign prostate hyperplasia-associated microbiota induces inflammation and DNA damage in prostate epithelial cells. The Prostate 2020, 80, 1341–1352. [Google Scholar] [CrossRef] [PubMed]
  102. Bajic, P.; Van Kuiken, M.E.; Burge, B.K.; Kirshenbaum, E.J.; Joyce, C.J.; Wolfe, A.J.; Branch, J.D.; Bresler, L.; Farooq, A.V. Male bladder microbiome relates to lower urinary tract symptoms. European urology focus 2020, 6, 376–382. [Google Scholar] [CrossRef] [PubMed]
  103. Satoskar, A.A.; Parikh, S.V.; Nadasdy, T. Epidemiology, pathogenesis, treatment and outcomes of infection-associated glomerulonephritis. Nature Reviews Nephrology 2020, 16, 32–50. [Google Scholar] [CrossRef] [PubMed]
  104. Mosquera, J.; Pedreañez, A. Acute post-streptococcal glomerulonephritis: analysis of the pathogenesis. International Reviews of Immunology 2021, 40, 381–400. [Google Scholar] [CrossRef]
  105. Bateman, E.; Mansour, S.; Okafor, E.; Arrington, K.; Hong, B.-Y.; Cervantes, J. Examining the efficacy of antimicrobial therapy in preventing the development of postinfectious glomerulonephritis: a systematic review and meta-analysis. Infectious Disease Reports 2022, 14, 176–183. [Google Scholar] [CrossRef]
  106. Koyama, A.; Kobayashi, M.; Yamaguchi, N.; Yamagata, K.; Takano, K.; Nakajima, M.; Irie, F.; Goto, M.; Igarashi, M.; Iitsuka, T. Glomerulonephritis associated with MRSA infection: a possible role of bacterial superantigen. Kidney international 1995, 47, 207–216. [Google Scholar] [CrossRef]
  107. Casuscelli, C.; Longhitano, E.; Maressa, V.; Di Carlo, S.; Peritore, L.; Di Lorenzo, S.; Calabrese, V.; Cernaro, V.; Santoro, D. Autoimmunity and Infection in Glomerular Disease. Microorganisms 2023, 11, 2227. [Google Scholar] [CrossRef]
  108. Tzvi-Behr, S.; Frishberg, Y.; Megged, O.; Weinbrand-Goichberg, J.; Becher-Cohen, R.; Terespolsky, H.; Rinat, C.; Choshen, S.; Ben-Shalom, E. Acute glomerulonephritis with concurrent suspected bacterial pneumonia–is it the tip of the iceberg? Pediatric Nephrology 2023, 1–5. [Google Scholar] [CrossRef]
  109. Skrzypczyk, P.; Ofiara, A.; Zacharzewska, A.; Pańczyk-Tomaszewska, M. Acute post-streptococcal glomerulonephritis–immune-mediated acute kidney injury–case report and literature review. Central European Journal of Immunology 2021, 46, 516–523. [Google Scholar]
  110. Yacouba, A.; Alou, M.T.; Lagier, J.-C.; Dubourg, G.; Raoult, D. Urinary microbiota and bladder cancer: A systematic review and a focus on uropathogens. In Proceedings of the Seminars in Cancer Biology; 2022; pp. 875–884. [Google Scholar]
  111. Karam, A.; Mjaess, G.; Albisinni, S.; El Daccache, Y.; Farah, M.; Daou, S.; Kazzi, H.; Hassoun, R.; Bou Kheir, G.; Aoun, F. Uncovering the role of urinary microbiota in urological tumors: a systematic review of literature. World Journal of Urology 2022, 40, 951–964. [Google Scholar] [CrossRef] [PubMed]
  112. Al-Karawi, A.S.; Abid, F.M.; Mustafa, A.; Abdulla, M. Revealing the Urinary Microbiota in Prostate Cancer: A Comprehensive Review Unveiling Insights into Pathogenesis and Clinical Application. Al-Salam Journal for Medical Science 2024, 3, 45–54. [Google Scholar]
  113. Tsai, K.-Y.; Wu, D.-C.; Wu, W.-J.; Wang, J.-W.; Juan, Y.-S.; Li, C.-C.; Liu, C.-J.; Lee, H.-Y. Exploring the association between gut and urine microbiota and prostatic disease including benign prostatic hyperplasia and prostate cancer using 16S rRNA sequencing. Biomedicines 2022, 10, 2676. [Google Scholar] [CrossRef] [PubMed]
  114. Gupta, S.; Kanwar, S.S. The influence of dysbiosis on kidney stones that risk up renal cell carcinoma (RCC). In Proceedings of the Seminars in cancer biology; 2021; pp. 134–138. [Google Scholar]
  115. Wu, P.; Zhang, G.; Zhao, J.; Chen, J.; Chen, Y.; Huang, W.; Zhong, J.; Zeng, J. Profiling the urinary microbiota in male patients with bladder cancer in China. Frontiers in cellular infection microbiology 2018, 8, 167. [Google Scholar]
  116. Saeed, A.; Imtiaz, S.; Riaz, S. Study of Urinary Tract Infection in Patients Suffering from Cancer. Journal of Cancer Research Reviews Reports 2020, 2, 1–15. [Google Scholar] [CrossRef]
  117. Heidler, S.; Lusuardi, L.; Madersbacher, S.; Freibauer, C. The microbiome in benign renal tissue and in renal cell carcinoma. Urologia internationalis 2020, 104, 247–252. [Google Scholar] [CrossRef]
  118. Bi, H.; Tian, Y.; Song, C.; Li, J.; Liu, T.; Chen, Z.; Chen, C.; Huang, Y.; Zhang, Y. Urinary microbiota–a potential biomarker and therapeutic target for bladder cancer. Journal of medical microbiology 2019, 68, 1471–1478. [Google Scholar] [CrossRef]
  119. Bajic, P.; Wolfe, A.J.; Gupta, G.N. The urinary microbiome: implications in bladder cancer pathogenesis and therapeutics. Urology 2019, 126, 10–15. [Google Scholar] [CrossRef]
  120. Friedrich, V.; Choi, H.W. The urinary microbiome: role in bladder cancer and treatment. Diagnostics 2022, 12, 2068. [Google Scholar] [CrossRef]
  121. Reggiani, F.; L'Imperio, V.; Calatroni, M.; Pagni, F.; Sinico, R.A. Goodpasture syndrome and anti-glomerular basement membrane disease. Clin Exp Rheumatol 2023, 41, 964–974. [Google Scholar] [CrossRef]
  122. Gesualdo, L.; Di Leo, V.; Coppo, R. The mucosal immune system and IgA nephropathy. Semin Immunopathol 2021, 43, 657–668. [Google Scholar] [CrossRef] [PubMed]
  123. Suzuki, S.; Nakatomi, Y.; Odani, S.; Sato, H.; Gejyo, F.; Arakawa, M.J.C.; Immunology, E. Circulating IgA, IgG, and IgM class antibody against Haemophilus parainfluenzae antigens in patients with IgA nephropathy. 1996, 104, 306–311.
  124. Sugino, H.; Sawada, Y.; Nakamura, M.J.I.j.o.m.s. IgA vasculitis: etiology, treatment, biomarkers and epigenetic changes. 2021, 22, 7538.
  125. Keri, K.C.; Blumenthal, S.; Kulkarni, V.; Beck, L.; Chongkrairatanakul, T. Primary membranous nephropathy: comprehensive review and historical perspective. Postgrad Med J 2019, 95, 23–31. [Google Scholar] [CrossRef] [PubMed]
  126. Almaani, S.; Meara, A.; Rovin, B.H. Update on Lupus Nephritis. Clin J Am Soc Nephrol 2017, 12, 825–835. [Google Scholar] [CrossRef]
  127. Dhillon, S.; Higgins, R.M. Interstitial nephritis. Postgrad Med J 1997, 73, 151–155. [Google Scholar] [CrossRef] [PubMed]
  128. Marcu, I.; Campian, E.C.; Tu, F.F. Interstitial Cystitis/Bladder Pain Syndrome. Semin Reprod Med 2018, 36, 123–135. [Google Scholar] [CrossRef]
  129. Karlsen, T.H.; Folseraas, T.; Thorburn, D.; Vesterhus, M. Primary sclerosing cholangitis - a comprehensive review. J Hepatol 2017, 67, 1298–1323. [Google Scholar] [CrossRef]
  130. Hanson, L.A.; Ahlstedt, S.; Fasth, A.; Hagberg, M.; Kaijser, B.; Mattsby-Baltzer, I.; Svanborg-Eden, C. Immunological aspects of pyelonephritis. Contrib Nephrol 1979, 16, 16–21. [Google Scholar] [CrossRef]
  131. Rifai, A.O.; Denig, K.M.; Caza, T.; Webb, S.M.; Rifai, S.; Khan, S.; Dahan, S.; Alamin, S.J.C.R.i.N. Antitubular Basement Membrane Antibody Disease Associated with Nivolumab Infusion and Concomitant Acute Pyelonephritis Leading to Acute Kidney Injury: a Case Report and Literature Review. 2023, 2023.
  132. Lu, J.C.; Shen, J.M.; Hu, X.C.; Peng, L.P.; Hong, Z.W.; Yao, B. Identification and preliminary study of immunogens involved in autoimmune prostatitis in human males. Prostate 2018. [CrossRef] [PubMed]
  133. Hyun, M.; Lee, J.Y.; Lim, K.R.; Kim, H.a. Clinical Characteristics of Uncomplicated Acute Pyelonephritis Caused by Escherichia coli and Klebsiella pneumoniae. Infectious Diseases Therapy 2024, 1–15. [Google Scholar] [CrossRef]
  134. Jirillo, E.; Palmirotta, R.; Colella, M.; Santacroce, L. A Bird’s-Eye View of the Pathophysiologic Role of the Human Urobiota in Health and Disease: Can We Modulate It? Pathophysiology 2024, 31, 52–67. [Google Scholar] [CrossRef]
  135. Kline, K.; Lewis, A. (UTI-0012-2012). Gram-positive uropathogens, polymicrobial urinary tract infection, and the emerging microbiota of the urinary tract. Microbiol Spectr 4: UTI-0012-2012: 2016. [CrossRef]
  136. Alteri, C.J.; Himpsl, S.D.; Mobley, H.L. Preferential use of central metabolism in vivo reveals a nutritional basis for polymicrobial infection. PLoS pathogens 2015, 11, e1004601. [Google Scholar] [CrossRef] [PubMed]
  137. Keogh, D.; Tay, W.H.; Ho, Y.Y.; Dale, J.L.; Chen, S.; Umashankar, S.; Williams, R.B.; Chen, S.L.; Dunny, G.M.; Kline, K.A.J.C.h.; et al. Enterococcal metabolite cues facilitate interspecies niche modulation and polymicrobial infection. 2016, 20, 493–503.
  138. Li, Q.; Lin, X.; Wu, Z.; He, L.; Wang, W.; Cao, Q.; Zhang, J.J.S.J.o.K.D. ; Transplantation. Immuno-histochemistry analysis of Helicobacter pylori antigen in renal biopsy specimens from patients with glomerulonephritis. 2013, 24, 751–758. [Google Scholar]
  139. Ogura, Y.; Suzuki, S.; Shirakawa, T.; Masuda, M.; Nakamura, H.; Iijima, K.; Yoshikawa, N. Haemophilus parainfluenzae antigen and antibody in children with IgA nephropathy and Henoch-Schönlein nephritis. American journal of kidney diseases 2000, 36, 47–52. [Google Scholar] [CrossRef] [PubMed]
  140. Masuda, M.; Nakanishi, K.; Yoshizawa, N.; Iijima, K.; Yoshikawa, N. Group A streptococcal antigen in the glomeruli of children with Henoch-Schönlein nephritis. American journal of kidney diseases 2003, 41, 366–370. [Google Scholar] [CrossRef]
  141. Schmitt, R.; Carlsson, F.; Mörgelin, M.; Tati, R.; Lindahl, G.; Karpman, D.J.T.A.j.o.p. Tissue deposits of IgA-binding streptococcal M proteins in IgA nephropathy and Henoch-Schönlein purpura. 2010, 176, 608–618.
  142. Kronbichler, A.; Kerschbaum, J.; Mayer, G. The influence and role of microbial factors in autoimmune kidney diseases: a systematic review. Journal of Immunology Research 2015, 2015. [Google Scholar] [CrossRef]
  143. Ivanov, I.B.; Kuzmin, M.D.; Gritsenko, V.A.J.I.j.o.a. Microflora of the seminal fluid of healthy men and men suffering from chronic prostatitis syndrome. 2009, 32, 462–467.
  144. Nickel, J.C.; Stephens, A.; Landis, J.R.; Chen, J.; Mullins, C.; van Bokhoven, A.; Lucia, M.S.; Melton-Kreft, R.; Ehrlich, G.D.; urology, M.R.N.J.T.J.o. Search for microorganisms in men with urologic chronic pelvic pain syndrome: a culture-independent analysis in the MAPP research network. 2015, 194, 127–135.
  145. Shoskes, D.A.; Altemus, J.; Polackwich, A.S.; Tucky, B.; Wang, H.; Eng, C.J.U. The urinary microbiome differs significantly between patients with chronic prostatitis/chronic pelvic pain syndrome and controls as well as between patients with different clinical phenotypes. 2016, 92, 26–32.
  146. Mändar, R.; Punab, M.; Korrovits, P.; Türk, S.; Ausmees, K.; Lapp, E.; Preem, J.K.; Oopkaup, K.; Salumets, A.; Truu, J.J.I.J.o.U. Seminal microbiome in men with and without prostatitis. 2017, 24, 211–216.
  147. Miyake, M.; Tatsumi, Y.; Ohnishi, K.; Fujii, T.; Nakai, Y.; Tanaka, N.; Fujimoto, K.J.P.I. Prostate diseases and microbiome in the prostate, gut, and urine. 2022, 10, 96–107.
  148. Venturini, S.; Reffo, I.; Avolio, M.; Basaglia, G.; Del Fabro, G.; Callegari, A.; Tonizzo, M.; Sabena, A.; Rondinella, S.; Mancini, W. The management of recurrent urinary tract infection: non-antibiotic bundle treatment. Probiotics Antimicrobial Proteins 2023, 1–9. [Google Scholar] [CrossRef]
  149. Pessoa, W.F.B.; Melgaço, A.C.C.; Almeida, M.E.; Santos, T.F.; Romano, C.C. Probiotics for urinary tract disease prevention and treatment. In Probiotics for Human Nutrition in Health and Disease; Elsevier: 2022; pp. 513–536.
  150. Ahmed, R.A.; Kamal, L.D.; Ahmed, H.S. The mechanisms of Lactobacillus activities: Probiotic importance of Lactobacillus species. Egyptian Academic Journal of Biological Sciences, E. Medical Entomology Parasitology 2021, 13, 45–63. [Google Scholar]
  151. Iannitti, T.; Palmieri, B. Therapeutical use of probiotic formulations in clinical practice. Clinical nutrition 2010, 29, 701–725. [Google Scholar] [CrossRef] [PubMed]
  152. Stapleton, A.E.; Au-Yeung, M.; Hooton, T.M.; Fredricks, D.N.; Roberts, P.L.; Czaja, C.A.; Yarova-Yarovaya, Y.; Fiedler, T.; Cox, M.; Stamm, W.E. Randomized, placebo-controlled phase 2 trial of a Lactobacillus crispatus probiotic given intravaginally for prevention of recurrent urinary tract infection. Clinical infectious diseases 2011, 52, 1212–1217. [Google Scholar] [CrossRef]
  153. Hosseini, M.; Yousefifard, M.; Ataei, N.; Oraii, A.; Razaz, J.M.; Izadi, A.J. The efficacy of probiotics in prevention of urinary tract infection in children: A systematic review and meta-analysis. Journal of pediatric urology 2017, 13, 581–591. [Google Scholar] [CrossRef]
  154. New, F.J.; Theivendrampillai, S.; Juliebø-Jones, P.; Somani, B. Role of probiotics for recurrent UTIs in the twenty-first century: a systematic review of literature. Current urology reports 2022, 23, 19–28. [Google Scholar] [CrossRef] [PubMed]
  155. Daniel, M.; Szymanik-Grzelak, H.; Turczyn, A.; Pańczyk-Tomaszewska, M. Lactobacillus rhamnosus PL1 and Lactobacillus plantarum PM1 versus placebo as a prophylaxis for recurrence urinary tract infections in children: a study protocol for a randomised controlled trial. BMC urology 2020, 20, 1–6. [Google Scholar] [CrossRef] [PubMed]
  156. Meena, J.; Thomas, C.C.; Kumar, J.; Raut, S.; Hari, P. Non-antibiotic interventions for prevention of urinary tract infections in children: A systematic review and meta-analysis of randomized controlled trials. European Journal of Pediatrics 2021, 180, 3535–3545. [Google Scholar] [CrossRef] [PubMed]
  157. Das, S.; Ameeruddin, S. Probiotics in common urological conditions: A narrative review. Longhua Chin. Med 2022, 5, 14. [Google Scholar] [CrossRef]
  158. Fazly Bazzaz, B.S.; Darvishi Fork, S.; Ahmadi, R.; Khameneh, B. Deep insights into urinary tract infections and effective natural remedies. African Journal of Urology 2021, 27, 1–13. [Google Scholar] [CrossRef]
  159. Ayivi, R.D.; Gyawali, R.; Krastanov, A.; Aljaloud, S.O.; Worku, M.; Tahergorabi, R.; Silva, R.C.d.; Ibrahim, S.A. Lactic acid bacteria: Food safety and human health applications. Dairy 2020, 1, 202–232. [Google Scholar] [CrossRef]
  160. Voidarou, C.; Alexopoulos, A.; Tsinas, A.; Rozos, G.; Tzora, A.; Skoufos, I.; Varzakas, T.; Bezirtzoglou, E. Effectiveness of bacteriocin-producing lactic acid bacteria and bifidobacterium isolated from honeycombs against spoilage microorganisms and pathogens isolated from fruits and vegetables. Applied Sciences 2020, 10, 7309. [Google Scholar] [CrossRef]
  161. Song, C.H.; Kim, Y.H.; Naskar, M.; Hayes, B.W.; Abraham, M.A.; Noh, J.H.; Suk, G.; Kim, M.J.; Cho, K.S.; Shin, M. Lactobacillus crispatus limits bladder uropathogenic E. coli infection by triggering a host type I interferon response. Proceedings of the National Academy of Sciences 2022, 119, e2117904119. [Google Scholar]
  162. Zagaglia, C.; Ammendolia, M.G.; Maurizi, L.; Nicoletti, M.; Longhi, C. Urinary tract infections caused by uropathogenic Escherichia coli strains—new strategies for an old pathogen. Microorganisms 2022, 10, 1425. [Google Scholar] [CrossRef]
  163. Kamel, M.; Aleya, S.; Alsubih, M.; Aleya, L. Microbiome Dynamics: A Paradigm Shift in Combatting Infectious Diseases. Journal of Personalized Medicine 2024, 14, 217. [Google Scholar] [CrossRef]
  164. Schwartz, L.; de Dios Ruiz-Rosado, J.; Stonebrook, E.; Becknell, B.; Spencer, J.D. Uropathogen and host responses in pyelonephritis. Nature Reviews Nephrology 2023, 19, 658–671. [Google Scholar] [CrossRef] [PubMed]
  165. Kenneally, C.; Murphy, C.P.; Sleator, R.D.; Culligan, E.P. The urinary microbiome and biological therapeutics: Novel therapies for urinary tract infections. Microbiological Research 2022, 259, 127010. [Google Scholar] [CrossRef] [PubMed]
  166. Abdulsalam, A.A.; Saleh, M.K. Effect of colicin from E. coli product on some species of gram-negative bacteria isolated from Urinary Tract infections. Journal of Education Scientific Studies.
  167. Roy, S.M.; Riley, M.A. Evaluation of the potential of colicins to prevent extraluminal contamination of urinary catheters by Escherichia coli. International journal of antimicrobial agents 2019, 54, 619–625. [Google Scholar] [CrossRef] [PubMed]
  168. Bao, J.; Wu, N.; Zeng, Y.; Chen, L.; Li, L.; Yang, L.; Zhang, Y.; Guo, M.; Li, L.; Li, J. Non-active antibiotic and bacteriophage synergism to successfully treat recurrent urinary tract infection caused by extensively drug-resistant Klebsiella pneumoniae. Emerging microbes infections 2020, 9, 771–774. [Google Scholar] [CrossRef] [PubMed]
  169. Zalewska-Piątek, B.; Piątek, R. Phage therapy as a novel strategy in the treatment of urinary tract infections caused by E. coli. Antibiotics 2020, 9, 304. [Google Scholar] [CrossRef]
  170. González de Llano, D.; Moreno-Arribas, M.V.; Bartolomé, B. Cranberry polyphenols and prevention against urinary tract infections: relevant considerations. Molecules 2020, 25, 3523. [Google Scholar] [CrossRef]
  171. Amábile-Cuevas, C.F. Ascorbate and Antibiotics, at Concentrations Attainable in Urine, Can Inhibit the Growth of Resistant Strains of Escherichia coli Cultured in Synthetic Human Urine. Antibiotics 2023, 12, 985. [Google Scholar] [CrossRef]
  172. Pani, A.; Valeria, L.; Dugnani, S.; Senatore, M.; Scaglione, F. Pharmacodynamics of D-mannose in the prevention of recurrent urinary infections. Journal of Chemotherapy 2022, 34, 459–464. [Google Scholar] [CrossRef]
  173. Ala-Jaakkola, R.; Laitila, A.; Ouwehand, A.C.; Lehtoranta, L. Role of D-mannose in urinary tract infections–a narrative review. Nutrition journal 2022, 21, 18. [Google Scholar] [CrossRef]
Figure 1. UT microbiome in healthy state showing lower pH and in disease state showing high pH.
Figure 1. UT microbiome in healthy state showing lower pH and in disease state showing high pH.
Preprints 137643 g001
Figure 2. Competitive inhibition of E. coli adhesion by D-Mannose.
Figure 2. Competitive inhibition of E. coli adhesion by D-Mannose.
Preprints 137643 g002
Table 1. Autoimmune diseases associated with urinary tract organs.
Table 1. Autoimmune diseases associated with urinary tract organs.
Autoimmune disease Primary organ/body part affected Autoantibodies Reference
Goodpasture syndrome Kidneys, lungs Anti-GBM antibodies [121]
IgA nephropathy Kidneys IgA autoantibodies, anti-OMHP antibodies [122,123]
IgA vasculitis Kidneys, lungs IgA1 autoantibody [124]
Membranous nephropathy Kidneys Anti-PLA2R antibodies [125]
Lupus nephritis Kidneys Anti-dsDNA, Anti-Sm, Anti-nuclear antibodies [126]
Interstitial nephritis Kidneys Various autoantibodies [127]
Interstitial cystitis Bladder Anti-urothelial and anti-nuclear antibodies [128]
Primary sclerosing cholangitis Bile ducts, can affect gallbladder ANCA, Anti-mitochondrial antibodies [129]
Pyelonephritis kidney Tamm-Horsfall protein antibodies, Tubulointerstitial nephritis antigen protein antibody [130,131]
Xanthogranulomatous pyelonephritis kidney - -
Prostatitis Prostate gland Prostate tissue immunodominant antigen (HPTIAs) protein antibodies [132]
Table 2. Interventional or observational clinical trials for UTI prevention, involving the use of probiotics. Data obtained from Clinicaltrials.gov (retrieved 1 June 2024).
Table 2. Interventional or observational clinical trials for UTI prevention, involving the use of probiotics. Data obtained from Clinicaltrials.gov (retrieved 1 June 2024).
# Name Status NCT ID Phase Drugs
1 Lactobacillus Probiotic for Prevention of UTI Completed NCT03151967 Phase 2, Phase 3 Lactobacillus crispatus CTV-05; Placebo
2 Targeted Pathogen Replacement With Novel Probiotic Treatment for Prevention of Recurrent UTIs in Children Completed NCT01696227 - Nissle 1917
3 Observational Study Evaluating the Number of Symptomatic Cystitis-like Episodes and Urinary Comfort of Women Consuming Cranberry, Cinnamon and Probiotic Strain Extracts Completed NCT04987164 - Cranberry, Cinnamon, Probiotics
4 Probiotic and Effects on Multi-Drug Resistant Urinary Tract Infection Completed NCT03644966 - Bifidobacterium infantis, Antibiotics
5 A Double-blinded, Randomized, Placebo-controlled, Parallel-group Study Evaluating the Effect of the Probiotic on Recurrent Urinary Tract Infection (UTI) in Adult Women Recently Treated for UTI. Completed NCT03366077 - Probiotic (name not mentioned)
6 Preventing Urinary Tract Infections in Infants and Young Children With Probiotic E. Coli Nissle: Recruiting NCT04608851 Phase 4 E. coli Nissle
7 A Novel Probiotic-antibiotic Combination to Prevent Recurrent Urinary Tract Infections Recruiting NCT06149676 Early Phase 1 Saccharomyces Boulardii 250 MG [Florastor]
8 Randomized, Double-blind, Placebo-controlled Study to Evaluate the Effect of a Probiotic on the Urinary Tract Microbiota in Women With Recurrent Urinary Tract Infections (rUTI). Recruiting NCT05895578 - Probiotic (name not mentioned) and antibiotic
9 A Clinical Trial to Determine the Extent to Which Probiotic Therapy Reduces Side Effects of Antibiotic Prophylaxis in Pediatric Neurogenic Bladder Patients With a History of Recurrent Urinary Tract Infections Unknown status NCT02044965 Phase 1, Phase 2 Trimethoprim; sulfamethoxazole; L. rhamnosus GR-1 and L. reuteri RC-14
10 Probiotics/Lactobacillus as a Prophylactic Aid in Recurrent Bacterial Cystitis in Women. A Randomized, Prospective, Double-Blinded, Placebo Controlled, Multi-Center Study. Unknown status NCT00781625 - UREX-Cap-5 (Lactobacillus rhamnosus GR-1, Lactobacillus reuteri RC-14)
11 Effectiveness of Prophylaxis of Urinary Tract Infections in Children With a Probiotic Containing Lactobacillus Rhamnosus PL1 and Lactobacillus Plantarum PM1, a Randomised Clinical Trial Unknown status NCT03462160 - Lactobacillus Rhamnosus PL1 and Lactobacillus Plantarum PM1
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated