The Urobiome: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

The microbiome of the urinary tract plays a significant role in maintaining health through the impact on bladder homeostasis. Urobiome is of great importance in maintaining the urothelial integrity and preventing urinary tract infection, as well as promoting local immune function.

  • microbiome
  • urinary tract infection
  • pediatric diseases

1. Introduction

In recent years, much attention has been paid to the human microbiome and its importance in health and disease [1][2][3][4]. It is estimated that the human body is colonized by 10–100 trillion microbial cells containing 100-fold more genes than the human genome [5]. The term ‘microbiota’ refers to the group of microorganisms associated with a specific biologic niche, mainly bacteria, but also protozoa, viruses, fungi, and archaea [2][6]. Correspondingly, the term ‘microbiome’ relates to the group of microbial genomes in a specific environment [2][6].
Urobiota helps maintain bladder homeostasis in terms of maintaining the integrity of the urinary tract epithelium, protecting against infections, regulating neurotransmission, and promoting the proper functioning of the immune system [7].
The composition, characteristics, and role of the microbiota of the urinary tract are still under investigation [7][8]. The majority of the urobiome studies have focused on the adult population, and the alterations of the microbial community structure have been linked to several urological diseases such as urinary tract infections, incontinence, overactive bladder, urolithiasis, and prostate or bladder cancer [9][10]. The number of studies in the pediatric population is rather limited and involved smaller numbers of subjects, thus the characteristic of the urobiome in children, its role in a child’s health, and pediatric urologic diseases are not completely understood [11].

2. The Urobiome Investigation

2.1. Diagnostic Techniques

From the 1950s, the urinary tract was believed sterile under normal conditions. This approach has changed with the start of the Human Microbiome Project, the first large-scale mapping of the human microbiome using culture-independent methods. Standard urine cultures detect aerobic and fast-growing bacteria, while slow-growing anaerobic microorganisms or bacteria that require different growth conditions remain undetected [12][13].
Relatively recently, due to advances in bacterial assessment, it has been shown that the bladder is not a sterile environment [12]. In particular, new diagnostic techniques such as PCR, expanded quantitative urine culture (EQUC), whole-genome sequencing (WGS), and next-generation sequencing (NGS) including 16S ribosomal RNA gene sequencing allowed the discovery of uncultured microorganisms and revealed the existence of a urinary microbiome [12][13][14].

2.2. Challenges in the Urobiome Research

Although new diagnostic approaches have facilitated a qualitative and quantitative identification of individual bacteria species with very high accuracy, there are some significant concerns due to the limitations of the methodology, the low biomass of the urinary microbiota, and its immediacy to other bacterial niches with higher microbial biomass [12][14]. The techniques that have been used to obtain urine samples to investigate the urinary microbiota include the midstream clean-catch urine, transurethral catheterization, or suprapubic aspiration [14][15]. The choice of a specific method of a urine sample collection may affect the obtained results [14] with difficulties in distinguishing bacteria from the bladder from microbial contamination from the skin, vulvovaginal and perineal flora [12][14][15]. Additionally, urethral or bladder tissue samples, obtained by urethral swabs, bladder biopsy, or scraping, might be used in urobiome research [16]. Pohl et al. showed that the urobiome of healthy men and women differs according to sampling method (voiding versus catheterized samples). In addition, their results indicate that the microbiome of the urethra and bladder are different. In the urethral samples, the most abundant genera were Veillonella, Staphylococcus, and Neisseria, while in bladder samples Lactobacillus, Streptococcus, and Gardnerella [17]. According to Wolfe et al., suprapubic aspiration sampling minimizes contamination from non-urinary sites and provides the best view of the bladder bacteria [18]. The recommended sampling method to use in studies investigating the microbiome of the urinary tract is transurethral catheterization [18][19]. However, the use of invasive techniques to collect urine for urobiome research in the pediatric population raises an ethical concern. It is worth underlining that urine cultures of clean-catch urine samples have good efficacy in diagnosing urinary tract infections in children [20][21], and the contamination rate of urine collected via urethral catheterization is lower but not significantly different from that of clean-catch [21]. Interestingly, Bundgaard-Nielsen et al., who investigated the impact of the collection method on the urobiome composition, did not report any interpersonal daily or day-to-day deviations in microbiota composition in women, girls, or boys [22].
Another methodological challenge is the inability of the sequencing technique to differentiate DNA from living versus dead bacteria [11][12]. To assess the viability of identified microorganisms, modified clinical cultivation procedures were implemented, and an enhanced quantitative urine culture (EQUC) protocol, which uses multiple culture media and various incubation conditions to cultivate bacteria that do not grow under standard conditions, was developed [15][18][19]. Considering a non-invasive urine collection method for the urobiome studies, Ozer et al. using first voided or midstream urine samples showed no significant difference regarding 16s ribosomal RNA analysis [23].
Furthermore, sequencing of 16S ribosomal RNA gene cannot provide information below the species level, in particular data about the strain level or presence of virulence factors. These details should be taken into consideration, as not all strains belonging to the same species exhibit pathogenic behavior [24]. For instance, several virulence genes are characteristic of uropathogenic Escherichia coli (E. coli) and are not present in other E. coli strains. These genes encode virulence factors essential in UTI development, such as adhesins (fimH gene encoding type 1 fimbrial tip adhesin), siderophores, and toxins (hlyA gene encoding α-hemolysin, cnf1 gene encoding cytotoxic necrotizing factor) [25][26].

3. The Urobiome Origin and Composition

3.1. The Origin of Urinary Microbiota

The origin of urinary microbiota is still not entirely clear. A urinary microbiome was identified even in neonates [27]. The fetus is considered sterile during normal pregnancy and acquires bacteria through transmission from the mother at delivery [28]. The maternal microbiome has a strong influence on the neonatal skin, oral mucosa, and nasopharyngeal microbiome development, and might similarly influence newborns’ urobiome [29]. The composition and function of the early infant microbiota are primarily determined by birth mode, maternal microbiota, exposure to antibiotics, and feeding practices in early life [30]. It is hypothesized that the main origins of urinary microbiota are microbial communities of the gastrointestinal tract and vagina, majorly because of the anatomical proximity. Evidence suggests that the infant microbiota assembles and stabilizes 2–3 years after birth [28][30]. In adolescents, pubertal hormonal changes may influence the maturation of the microbiome of the urinary tract [11]. However, the effect of age on the urobiome of healthy children has been scarcely analyzed.

3.2. Urobiome Composition Depending on the Child’s Age

Kinneman et al. aimed to examine the urinary microbiome of eighty-five children younger than 48 months who presented to the Emergency Department mainly because of fever. Urine samples were collected via transurethral catheterization. The urinary microbiome was identified in every child. The most abundant families detected in urine samples were tissierellaceae, prevotellaeae, veillonellaceae, enterobacteriaceae, and comamonadaceae, while the five most abundant genera were Prevotella, Peptoniphilus, Escherichia, Veillonella, and Finegoldia. There were no significant differences according to gender, probably because urine samples were collected via catheterization, which minimized periurethral and perineal contamination. Nine children were diagnosed with urinary tract infection, in this group decreased urobiome diversity was observed [27].
Two studies investigated the urobiome in toilet trained children [31][32]. Fredsgaard et al. described the voided urinary microbiota in a group of thirty asymptomatic prepubertal children aged 6 to 10 years old. Bacterial DNA was extracted from all “clean-catch” midstream urine samples. The voided urinary microbiota differed significantly according to gender. The urine of girls was dominated by Prevotella (18.2%), Porphyromonas (12.9%), Ezakiella (8.1%), Prevotella (7.4%), and Dialister (7.0%). In boys, the most abundant genus was Porphyromonas (22.4%), followed by Ezakiella (12.0%), Campylobacter (11.6%), Prevotella (8.6%), and Dialister (3.7%). The authors suggest that the discrepancy in the urobiome composition between girls and boys could be due to gender-specific anatomy and differences in sex hormones levels in the prepubertal period [31].
Another study conducted by Kassiri et al. focused on prepubertal boys aged from 3 months to 8 years. The catheterized urine samples did not reveal a clear predominance of a particular bacterial genus. The majority of urine samples showed the presence of Staphylococcus and Varibaculum species, and to a lesser extent Peptoniphilus and Actinobaculum. The authors support the opinion that the development of the urinary microbiota starts and evolves in early life and becomes more stable in adulthood, similarly to the microbiota of the gastrointestinal tract. However, the study group consisted of patients who required elective urologic procedures, therefore the composition of urobiome may not adequately represent the bacteria present in the urinary tract of children without urological pathologies [32].
From puberty, the shift in the composition of the microbiota of the urinary tract may reflect physiological changes. Presumably, the urobiome of adolescents begins to resemble the urobiome of healthy adults. However, no studies were performed on this topic, and further investigations regarding alterations of the urinary microbiota in this age group are needed.

4. Dysbiosis and Urinary Tract Infection

An imbalance in urinary microbiota may cause an overgrowth of pathogenic bacteria. Changes in urobiome composition may impact disease susceptibility and pathophysiology [3]. Dysbiosis of the urobiome has been related to an increased risk of urinary tract infection, nephrolithiasis, and dysfunction of the lower urinary tract [9][10][33]. Although most studies focused on adults, the number of investigations evaluating the relationship between the urobiome, dysbiosis, and urinary tract diseases in children increases [11][24][34][35].
The findings of recent studies in this field may have significant clinical implications. Strategies of manipulating and reshaping disease-prone microbiomes may serve as an alternative or supportive option in the management of pediatric urological diseases. The modifications of the urobiome include microbial supplements (probiotics or synbiotics), foods or substrates (diet or prebiotics), microbial suppression or elimination (antibiotics) strategies [3].
Urinary tract infection is one of the most common bacterial infections in children [36]. The predominant pathogen is E. coli which accounts for 90% of primary UTIs in girls and 80% of primary UTIs in boys [36][37][38][39]. Other bacterial pathogens include gram-negative Klebsiella, Enterobacter, Proteus and Citrobacter [37][38], and gram-positive Enterococcus and Staphylococcus saprophyticus [38]. The clinical manifestation of UTI has a broad spectrum and may present as infection of the lower or upper urinary tract. According to patient comorbidities, UTIs can be divided into uncomplicated and complicated. In children, complicated UTIs are mainly associated with congenital anomalies of kidneys and urinary tract. A serious problem is the recurrence of urinary tract infections, defined as three or more infections per year [40]. Noteworthy, different microbial communities are associated with UTIs in specific patient groups (Table 1).
Table 1. UTIs classification according to complicating factors [40][41].
UTI Characteristic Related Microorganisms
uncomplicated occurs in immunocompetent patients with anatomically and functionally normal urinary tract uropathogenic E. coli
(causative factor in 80%)
complicated occurs in patients with anatomical or neurological abnormalities of urinary tract (i.e., hydronephrosis, vesicoureteral reflux), in patients with compromised immunity, or if foreign bodies are present in patient’s urinary tract (i.e., calculi, catheters) E. coli (often presenting combination of many virulence factors, or multi-drug resistant profile),
non-E. coli infection
(i.e., Klebsiella spp., Enterococcus spp., Pseudomonas aeruginosa)
Most UTIs result from ascending infection, and periurethral colonization with uropathogenic bacteria is the first step in the development of UTI [38]. Among several host defense mechanisms, a diverse urobiome has been associated with a protective role. According to Kinneman et al., children with UTIs had a significantly decreased alpha diversity, and the composition of the microbiome clustered separately compared with children without UTIs [27]. There is no consensus regarding the taxonomy of healthy urobiota. Additionally, the proportion of species forming the urobiome is dynamic. Commensal bacteria of the urinary tract are believed to serve as a barrier for uropathogens by blocking access to urothelium, producing antimicrobial compounds, or out-competing pathogenic bacteria for common resources [42]. It is suggested that probably most bacteria causing UTIs are part of the resident urinary tract bacteria and reveal their uropathogenicity due to an imbalance in normal urobiome composition [43]. UTIs develop as a result of several host-microbial interactions, and host susceptibility and bacterial virulence factors are crucial elements of that interplay. The interaction between the uroplakin receptors expressed at superficial urothelial cells and bacterial type 1 fimbrial adhesin fimH may serve as an example [43]. The process of E. coli invasion and formation of an intercellular population of bacteria starts after uropathogenic E. coli fimbriae connect with uroplakin Ia/Ib [44].
The reservoirs of uropathogens are the gastrointestinal tract and vagina [45][46]. Paalanne et al. conducted a case-control study that assessed differences in the gut microbiome between children with UTI and healthy controls. The biodiversity of the intestinal microbiome of children was similar in both groups, but there were differences at the family and genus levels. The genus Enterobacter was more abundant in the UTI patients, and the family peptostreptococcaceae was more abundant in controls. These findings suggest that the intestinal environment and its microbial community are associated with the risk of UTI in the pediatric population [47].
Similarly, the vaginal microbiota might impact host susceptibility to UTI and can either protect against or increase the risk of UTIs in girls. Gorbachinsky et al. demonstrated that vulvovaginitis may cause UTIs by changing the perineal microbiome and increased colonization of uropathogens [48]. Previous studies indicated that the vaginal microbiome alters with age. In prepubertal age, a variety of anaerobes, diphtheroids, coagulase-negative staphylococci, and E. coli dominate, while the postmenarcheal vaginal microbiome is dominated by Lactobacillus spp. [11][29]. In contrast, a prospective longitudinal study among perimenarcheal girls conducted by Hickey et al. demonstrated that bacteria producing lactic acid were dominant in the vaginal microbiota of most girls well before the onset of menarche [49]. A protective effect of Lactobacillus spp. is associated with the following mechanisms: maintaining the vagina’s characteristic low pH (mainly due to lactic acid production), releasing antimicrobial compounds such as hydrogen peroxide and bacteriocins, out-competing pathogenic bacteria, preventing the adhesion of pathogens to epithelial cells, and modulation of the host immune system by activation of the Toll-like receptor pathway and interleukins production [11][29][50][51].

This entry is adapted from the peer-reviewed paper 10.3390/ijms23020870

References

  1. Ogunrinola, G.A.; Oyewale, J.O.; Oshamika, O.O.; Olasehinde, G.I. The Human Microbiome and Its Impacts on Health. Int. J. Microbiol. 2020, 2020, 8045646.
  2. Cho, I.; Blaser, M.J. The human microbiome: At the interface of health and disease. Nat. Rev. Genet. 2012, 13, 260–270.
  3. Johnson, C.L.; Versalovic, J. The human microbiome and its potential importance to pediatrics. Pediatrics 2012, 129, 950–960.
  4. Wang, B.; Yao, M.; Lv, L.; Ling, Z.; Li, L. The Human Microbiota in Health and Disease. Engineering 2017, 3, 71–82.
  5. Amon, P.; Sanderson, I. What is the microbiome? Arch. Dis. Child. Educ. Pract. Ed. 2017, 102, 258–261.
  6. Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The Human Microbiome Project. Nature 2007, 449, 804–810.
  7. Ackerman, A.L.; Chai, T.C. The Bladder is Not Sterile: An Update on the Urinary Microbiome. Curr. Bladder Dysfunct. Rep. 2019, 14, 331–341.
  8. Li, J.K.M.; Chiu, P.K.F.; Ng, C.F. The impact of microbiome in urological diseases: A systematic review. Int. Urol. Nephrol. 2019, 51, 1677–1697.
  9. Aragón, I.M.; Herrera-Imbroda, B.; Queipo-Ortuño, M.I.; Castillo, E.; Del Moral, J.S.G.; Gómez-Millán, J.; Yucel, G.; Lara, M.F. The Urinary Tract Microbiome in Health and Disease. Eur. Urol. Focus 2018, 4, 128–138.
  10. Magistro, G.; Stief, C.G. The Urinary Tract Microbiome: The Answer to All Our Open Questions? Eur. Urol. Focus 2019, 5, 36–38.
  11. Gerber, D.; Forster, C.S.; Hsieh, M. The Role of the Genitourinary Microbiome in Pediatric Urology: A Review. Curr. Urol. Rep. 2018, 19, 13.
  12. Thomas-White, K.; Brady, M.; Wolfe, A.J.; Mueller, E.R. The Bladder Is Not Sterile: History and Current Discoveries on the Urinary Microbiome. Curr. Bladder Dysfunct. Rep. 2016, 11, 18–24.
  13. 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.
  14. Perez-Carrasco, V.; Soriano-Lerma, A.; Soriano, M.; Gutiérrez-Fernández, J.; Garcia-Salcedo, J.A. Urinary Microbiome: Yin and Yang of the Urinary Tract. Front. Cell. Infect. Microbiol. 2021, 11, 421.
  15. 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, e00218-20.
  16. Brubaker, L.; Gourdine, J.-P.F.; Siddiqui, N.Y.; Holland, A.; Halverson, T.; Limeria, R.; Pride, D.; Ackerman, L.; Forster, C.S.; Jacobs, K.M.; et al. Forming consensus to advance urobiome research. Msystems 2021, 6, e01371-20.
  17. 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. Int. Neurourol. J. 2020, 24, 41–51.
  18. Wolfe, A.J.; Toh, E.; Shibata, N.; Rong, R.; Kenton, K.; FitzGerald, M.P.; Mueller, E.R.; Schreckenberger, P.; Dong, Q.; Nelson, D.E.; et al. Evidence of uncultivated bacteria in the adult female bladder. J. Clin. Microbiol. 2012, 50, 1376–1383.
  19. Karstens, L.; Asquith, M.; Caruso, V.; Rosenbaum, J.T.; Fair, D.A.; Braun, J.; Gregory, W.T.; Nardos, R.; McWeeney, S.K. Community profiling of the urinary microbiota: Considerations for low-biomass samples. Nat. Rev. Urol. 2018, 15, 735–749.
  20. Mulcrone, A.E.; Parikh, M.; Ahmad, F.A. Reducing infant catheterization in the emergency department through clean-catch urine collection. J. Am. Coll. Emerg. Physicians Open 2020, 1, 1533–1541.
  21. Herreros, M.L.; Gili, P.; del Valle, R.; Barrios, A.; Pacheco, M.; Sánchez, A. Urine collection methods for infants under 3 months of age in clinical practice. Pediatr. Nephrol. 2020, 36, 3899–3904.
  22. Bundgaard-Nielsen, C.; Ammitzbøll, N.; Isse, Y.A.; Muqtar, A.; Jensen, A.M.; Leutscher, P.D.C.; Arenholt, L.T.S.; Hagstrøm, S.; Sørensen, S. Voided Urinary Microbiota Is Stable Over Time but Impacted by Post Void Storage. Front. Cell. Infect. Microbiol. 2020, 10, 435.
  23. Ozer, M.S.; Yildiz, H.A.; Incir, C.; Deger, M.D.; Bozkurt, O.; Ergor, G.; Tuncok, Y.; Esen, N.; Esen, A.A. Urinary microbiota; Which non-ınvasive urine collection method should we use? Int. J. Clin. Pract. 2021, 75, e14193.
  24. Cole, E.; Shaikh, N.; Forster, C.S. The pediatric urobiome in genitourinary conditions: A narrative review. Pediatr. Nephrol. 2021, 36, 1–10.
  25. Hashemizadeh, Z.; Kalantar-Neyestanaki, D.; Mansouri, S. Association between virulence profile, biofilm formation and phylogenetic groups of Escherichia coli causing urinary tract infection and the commensal gut microbiota: A comparative analysis. Microb. Pathog. 2017, 110, 540–545.
  26. Klein, R.D.; Hultgren, S.J. Urinary tract infections: Microbial pathogenesis, host-pathogen interactions and new treatment strategies. Nat. Rev. Microbiol. 2020, 18, 211–226.
  27. Kinneman, L.; Zhu, W.; Wong, W.S.W.; Clemency, N.; Provenzano, M.; Vilboux, T.; Jane’t, K.; Seo-Mayer, P.; Levorson, R.; Kou, M.; et al. Assessment of the urinary microbiome in children younger than 48 months. Pediatr. Infect. Dis. J. 2020, 39, 565–570.
  28. Roswall, J.; Olsson, L.M.; Kovatcheva-Datchary, P.; Nilsson, S.; Tremaroli, V.; Simon, M.C.; Kiilerich, P.; Akrami, R.; Krämer, M.; Uhlén, M.; et al. Developmental trajectory of the healthy human gut microbiota during the first 5 years of life. Cell Host Microbe 2021, 29, 765–776.
  29. Curley, T.; Forster, C.S. Recurrent UTIs in Girls: What Is the Role of the Microbiome? Urology 2021, 151, 94–97.
  30. Robertson, R.C.; Manges, A.R.; Finlay, B.B.; Prendergast, A.J. The Human Microbiome and Child Growth—First 1000 Days and Beyond. Trends Microbiol. 2019, 27, 131–147.
  31. Fredsgaard, L.; Thorsteinsson, K.; Bundgaard-Nielsen, C.; Ammitzbøll, N.; Leutscher, P.; Chai, Q.; Jensen, A.M.; Sørensen, S.; Pedersen, L.M.; Hagstrøm, S.; et al. Description of the voided urinary microbiota in asymptomatic prepubertal children—A pilot study. J. Pediatr. Urol. 2021, 17, 545.e1–545.e8.
  32. Kassiri, B.; Shrestha, E.; Kasprenski, M.; Antonescu, C.; Florea, L.D.; Sfanos, K.S.; Wang, M.H. A Prospective Study of the Urinary and Gastrointestinal Microbiome in Prepubertal Males. Urology 2019, 131, 204–210.
  33. Lee, K.W.; Song, H.Y.; Kim, Y.H. The microbiome in urological diseases. Investig. Clin. Urol. 2020, 61, 338–348.
  34. Kilis-Pstrusinska, K.; Rogowski, A.; Bienkowski, P. Bacterial colonization as a possible source of overactive bladder symptoms in pediatric patients: A literature review. J. Clin. Med. 2021, 10, 1645.
  35. Zwolińska, D. Probiotics in the prophylaxis of recurrent urinary tract infections in children. Pediatria I Medycyna Rodzinna 2017, 13, 335–343.
  36. Tullus, K.; Shaikh, N. Urinary tract infections in children. Lancet 2020, 395, 1659–1668.
  37. Kutasy, B.; Coyle, D.; Fossum, M. Urinary Tract Infection in Children: Management in the Era of Antibiotic Resistance—A Pediatric Urologist’s View. Eur. Urol. Focus 2017, 3, 207–211.
  38. Hodson, E.M.; Craig, J.C. Urinary Tract Infections in Children. In Pediatric Nephrology, 7th ed.; Avner, E.D., Harmon, W.E., Niaudet, P., Yoshikawa, N., Emma, F., Goldstein, S.L., Eds.; Springer: Heidelberg, Germany, 2016; pp. 1695–1714.
  39. Lemberger, U.; Quhal, F.; Bruchbacher, A.; Shariat, S.F.; Hiess, M. The microbiome in urinary tract infections in children—An update. Curr. Opin. Urol. 2021, 31, 147–154.
  40. A’t Hoen, L.A.; Bogaert, G.; Radmayr, C.; Dogan, H.S.; Nijman, R.J.M.; Quaedackers, J.; Rawashdeh, Y.F.; Silay, M.S.; Tekgul, S.; Bhatt, N.R.; et al. Update of the EAU/ESPU guidelines on urinary tract infections in children. J. Pediatr. Urol. 2021, 17, 200–207.
  41. Luna-Pineda, V.M.; Ochoa, S.A.; Cruz-Córdova, A.; Cázares-Domínguez, V.; Reyes-Grajeda, J.P.; Flores-Oropeza, M.A.; Arellano-Galindo, J.; Hernández-Castro, R.; Flores-Encarnación, M.; Ramírez-Vargas, A.; et al. Features of urinary Escherichia coli isolated from children with complicated and uncomplicated urinary tract infections in Mexico. PLoS ONE 2018, 13, e0204934.
  42. Whiteside, S.A.; Razvi, H.; Dave, S.; Reid, G.; Burton, J.P. The microbiome of the urinary tract-a role beyond infection. Nat. Rev. Urol. 2015, 12, 81–90.
  43. Hernández-Hernández, D.; Padilla-Fernández, B.; Ortega-González, M.Y.; Castro-Díaz, D.M. Recurrent Urinary Tract Infections and Asymptomatic Bacteriuria in Adults. Curr. Bladder Dysfunct. Rep. 2021, 16, 1–12.
  44. Szymańska, B.; Matuszewski, M.; Dembowski, J.; Piwowar, A. Initial evaluation of uroplakins UPIIIa and UPII in selected benign urological diseases. Biomolecules 2021, 11, 1816.
  45. Khan, A.; Jhaveri, R.; Seed, P.C.; Arshad, M. Update on associated risk factors, diagnosis, and management of recurrent urinary tract infections in children. J. Pediatr. Infect. Dis. Soc. 2019, 8, 152–159.
  46. 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 2021, 11, 7.
  47. Paalanne, N.; Husso, A.; Salo, J.; Pieviläinen, O.; Tejesvi, M.V.; Koivusaari, P.; Pirttilä, A.M.; Pokka, T.; Mattila, S.; Jyrkäs, J.; et al. Intestinal microbiome as a risk factor for urinary tract infections in children. Eur. J. Clin. Microbiol. Infect. Dis. 2018, 37, 1881–1891.
  48. Gorbachinsky, I.; Sherertz, R.; Russell, G.; Krane, L.S.; Hodges, S.J. Altered perineal microbiome is associated with vulvovaginitis and urinary tract infection in preadolescent girls. Ther. Adv. Urol. 2014, 6, 224–229.
  49. Hickey, R.J.; Zhou, X.; Settles, M.L.; Erb, J.; Malone, K.; Hansmann, M.A.; Shew, M.L.; Van Der Pol, B.; Fortenberry, J.D.; Forney, L.J. Vaginal microbiota of adolescent girls prior to the onset of menarche resemble those of reproductive-age women. MBio 2015, 6, e00097-15.
  50. Ng, Q.X.; Peters, C.; Venkatanarayanan, N.; Goh, Y.Y.; Ho, C.Y.X.; Yeo, W.S. Use of Lactobacillus spp. to prevent recurrent urinary tract infections in females. Med. Hypotheses 2018, 114, 49–54.
  51. Sihra, N.; Goodman, A.; Zakri, R.; Sahai, A.; Malde, S. Nonantibiotic prevention and management of recurrent urinary tract infection. Nat. Rev. Urol. 2018, 15, 750–776.
More
This entry is offline, you can click here to edit this entry!
Video Production Service