- Research
- Open access
- Published:
Comparison of phenotypic and genetic traits of ESBL-producing UPEC strains causing recurrent or single episode UTI in postmenopausal women
Annals of Clinical Microbiology and Antimicrobials volume 24, Article number: 11 (2025)
Abstract
Background
Recurrent urinary tract infections (rUTIs) occur in over 20% of patients, with postmenopausal women (over 50 years old) carrying the highest risk for recurrence compared to younger women. Virulence factors such as type 1 fimbriae adhesin FimH, the outer membrane protease OmpT, and the secreted pore-forming toxin α-hemolysin (HlyA) have been shown to support the formation of intracellular bacterial communities (IBCs) within bladder epithelial cells (BECs), facilitating persistence. This study aims to characterize the virulence expression and intracellular persistence of ESBL-producing uropathogenic E. coli (E-UPEC) strains isolated from postmenopausal women with recurrent or single episode infections.
Methods
Study strains included 72 E-UPEC strains collected from patients (36 recurrent; 36 single episode) with a confirmed UTI diagnosis and control UPEC strains (CFT073 and UTI89). Patient demographics and clinical course were collected. Presence of hlyA, ompT, and fimH genes were confirmed by colony PCR, and qRT-PCR was performed using extracted RNA from a subset of 18 strains (12 recurrent; 6 single episode) grown in Luria-Bertani media and isolated from infected BECs to characterize gene expression. Bladder cell line 5637 was infected with study strains at MOI 15 for 2 h, treated with amikacin for 2 h to remove extracellular bacteria, then lysed to enumerate intracellular CFU counts.
Results
No differences in clinical characteristics between patient groups were observed. Overall prevalence of fimH, ompT, and hlyA was 99% (71/72), 82% (59/72), and 26% (19/72) respectively; presence of all three genes did not differ between recurrent and single-episode strains. Notably, all recurrent strains had significantly more intracellular CFUs compared to single episode strains (median 16,248 CFU/mL vs. 4,118 CFU/mL, p = 0.018). Intracellular expression ompT was significantly increased (p = 0.0312) in the recurrent group compared to LB media, while fimH was significantly decreased (p = 0.0365) in the single episode group compared to expression in LB media.
Conclusion
Our findings indicate strain-specific ability to persist inside BECs with the recurrent strains exhibiting increased ompT expression inside BECs and higher intracellular bacterial burden compared to strains causing single episode UTI. These results emphasize the potential microbial contributions to recurrence in postmenopausal women and warrant future investigations on the impact of antibiotic therapy and host response on IBC-supportive UPEC virulence.
Background
Urinary tract infection (UTI) is one of the most common infections worldwide and responsible for over 11 million outpatient visits and costing over 4 billion dollars annually in the United States [1,2,3]. Women experience the highest risk for contracting UTIs with over 20% of female patients experiencing recurrence [4, 5]. Among those with recurrent UTIs, the subpopulations most affected are women in postmenopausal age and when extended spectrum beta-lactamase (ESBL) producing uropathogenic Escherichia coli (UPEC) is the causative pathogen [3, 6,7,8]. In addition to carrying a multidrug-resistant phenotype, UPEC strains can form intracellular bacterial communities (IBCs) which are biofilm-like structures within bladder epithelial cells (BECs) that confer protection of the bacteria from antibiotic and immune cell exposure [9,10,11,12]. These IBCs go through a cycle of reinfection of surrounding BECs, allowing the bacteria to spread deeper within the bladder tissue of postmenopausal women [4, 9]. When left untreated or incompletely treated, these bladder infections, also known as cystitis, can progress to infections of the kidney (pyelonephritis) and urosepsis may ensue [9, 13]. UPEC virulence factors that have been identified to support IBC formation within BECs include type 1 fimbrial adhesin FimH which facilitates attachment to BECs, pore-forming toxin α-hemolysin (HlyA) which allows UPEC to escape lysosomal compartments within BECs, and outer membrane protease OmpT which can degrade host antimicrobial peptides and contributes to intracellular biofilm formation [9, 14,15,16]. The fimH gene is only encoded on the chromosome, while hlyA and ompT genes can both be encoded on the chromosome as well as on plasmids [17,18,19,20]. Overall prevalence of fimH, hlyA, and ompT ranges from 86 to 100%, 21–47%, 67–94%, respectively [21,22,23,24,25]. However, the prevalence of these genes among UPEC strains isolated from postmenopausal women and particularly among those with recurrence is unknown. Significant reduction in IBCs has been shown in both in vitro and in vivo models of bladder infection with UPEC strains upon deletion of fimH, hlyA, and/or ompT, supporting their role in UPEC pathogenesis [14,15,16, 19, 26]. While patient factors such as comorbid conditions and genetic polymorphisms as contributing factors to recurrence have been well described, whether microbial characteristics differ between UPEC strains causing recurrent or single episode infections among postmenopausal women has not been well studied [7, 27, 28]. Therefore, we hypothesized that UPEC isolates from postmenopausal women with recurrent episodes are better able to survive inside of BECs than single episode strains due to robust virulence factor expression supporting IBC formation during infection. In this study, we examined the clinical characteristics of postmenopausal women who experienced UTI caused by ESBL-producing UPEC strains and profiled the expression of fimH, hlyA, and ompT as well as the in vitro intracellular persistence in BECs. Our results indicate marked differences in virulence factor expression and intracellular persistence between strains causing single versus recurrent episodes of UTI. This study emphasizes the importance of investigating the contribution of microbial virulence to recurrent UTIs.
Methods
Study design
This study was conducted at a community-teaching hospital on patients hospitalized during 2014–2021 under an IRB-approved protocol #HS-17-00943. Informed consent was waived since no interventions were made. Microbiology reports were reviewed to identify patients who had a urine culture positive for ESBL-producing UPEC strain. ESBL phenotype was confirmed by the clinical microbiology laboratory by disk diffusion using ceftazidime and ceftazidime/clavulanic acid discs in accordance with CLSI guidelines [29]. Electronic medical records were reviewed to screen for eligibility based on the following inclusion criteria: age 50 years or older (postmenopause), female sex, diagnosis of UTI based on symptomology, urinalysis, and/or discharge diagnosis, and the bacteria strain had been saved as part of a longitudinal surveillance program to evaluate antimicrobial resistance trends at the institution. Single episode was defined as the absence of either a UTI diagnosis or positive urinalysis within 6 months of the “index” episode while recurrence was defined as two symptomatic episodes in 6 months or 3 symptomatic episodes in 12 months. Episodes greater than 30 days apart were defined as separate UTI episodes. Patients were excluded if younger than 50 years old, male, or without UTI diagnosis. The following demographic and clinical characteristics were obtained from the medical records: age, ethnicity, UTI diagnosis, history of UTI within 12 months, comorbid conditions such as diabetes, dementia, kidney disease, immunocompromised status, and functional quadriplegia, presence of a chronic Foley catheter, antibiotic prescribed, and discharge disposition [28, 30, 31]. Saved bacteria strains were subjected to molecular and phenotypic analysis. All collected data was managed with the Research Electronic Data Capture (REDCap) software hosted at the University of Southern California, a secure web-based application designed to support data capture for research studies [32]. A subset of the UPEC strains matched by age were selected to represent recurrent and single episode strains in a 2:1 ratio to evaluate intracellular survival using an in vitro bladder infection model.
Presence of IBC-supportive genes
Polymerase chain reaction (PCR) was performed to identify the presence of IBC-supportive genes fimH, hlyA, and ompT in ESBL-UPEC isolated from study patients. These IBC-supportive genes were chosen due to significant decreases in intracellular CFUs of deletion mutants observed in both cell line and mouse models of UTI infection [14, 16, 26, 33]. Colony PCR using a single colony of each index strain from CHROMagar plates incubated at 37 °C overnight was used with Green GoTaq mastermix (Promega) and 250 nM primer concentration per the manufacturer’s instructions to perform an initial screen for the presence of fimH, hlyA, and ompT in the genome. If genes were not detected by colony PCR, colonies of bacteria from incubated CHROMagar plates were grown overnight at 37 °C, 250 rpm in LB Lennox broth for genomic DNA extraction using the QIAmp Mini extraction kit per the manufacturer’s instructions. Extracted genomic DNA (250 ng) was used with 250 nM primer concentration to perform PCR using the Green GoTaq mastermix (Promega) in 20 uL reaction mix per the manufacturer’s instructions to verify absence of the genes. UPEC reference strain UTI89 (cystitis strain) was used as a positive control given that it contains all three genes [15, 33, 34]. A previously sequenced UPEC strain with confirmed absence of ompT and hlyA was used as a negative control. The cysG gene was used as a reference, and the PCR products were visualized on a 1.5% agarose gel using gel electrophoresis (Bio Rad) [35]. Primer sequences and conditions are listed in Supplemental Table 1.
Intracellular CFU assay
UPEC strains were selected to represent those causing recurrent and single episode respectively as well as presence or absence of IBC-supportive genes (hlyA and ompT; fimH was near universally present) to examine their propensity for intracellular survival and persistence. The assay was performed following published protocol with modifications [10]. Briefly, human bladder epithelial cell line 5637 (ATCC HTB9) was selected for in vitro modeling of UTI in accordance with previous studies, and cultivated in RPMI + 10% FBS at 37 °C, 5% CO2 for up to nine passages [14, 16, 26, 33, 36]. Colonies of bacteria were incubated at 37 °C, 250 rpm in LB Lennox broth for 12–14 h, washed twice in chilled DPBS, and resuspended in RPMI media without fetal bovine serum (Gibco). 1.3 × 105 of BECs in RPMI + 10% FBS were seeded in 48-well plates 16 h prior to the start of the experiment. At the start of the experiments, monolayer was confirmed by light microscope and cells were washed once in warmed DPBS. Bacteria in RPMI media were adjusted to OD600 0.05, diluted 1/20 in 5 mL of RPMI + 10% FBS, then 400 uL of diluted bacteria was added to 48-well plate of washed BECs (final MOI 15). Infection was synchronized by centrifugation at 600 xg, 25 C for 5 minutes then incubated at 37 C, 5% CO2 for 2 h. Infecting dilution was also plated on LB agar in duplicate for CFU counts to confirm MOI. Extracellular bacteria were washed three times using warmed DPBS and 400 uL of RPMI + 10% FBS + 450 ug/mL amikacin was added to the wells and incubated at 37° C, 5% CO2 for 2 h to kill remaining extracellular bacteria. Amikacin was used instead of gentamicin given high gentamicin resistance observed in the clinical strains and low cell penetration [37]. BECs were then washed twice using DPBS, lysed using 0.1% Triton X-100, and diluted and plated on LB agar in duplicate. Plates were incubated at 37° C overnight and counted. Intracellular counts were normalized to initial CFU at the start of infection. UTI89 and CFT073 were used as positive controls as their intracellular persistence was previously described [14, 26]. The experiment was performed in six technical replicates and biological duplicates per strain.
Expression of IBC-supportive genes in LB and during in vitro infection
To assess expression of IBC-supportive genes in Luria-Bertani (LB) media, bacteria were grown in LB Lennox overnight at 37° C, 250 rpm, diluted then grown to mid-log phase (OD600 0.3) then extracted using the Purelink Mini RNA extraction kit per manufacturer instructions (Invitrogen). To assess intracellular expression of IBC-supportive genes, the intracellular CFU assay was performed above with modifications. 2 × 106 BECs were seeded in a 6 well plate and infected approximately 16 h later at MOI 50 to ensure sufficient yield of intracellular bacterial RNA [38,39,40]. After the incubation with amikacin, wells were washed and lysed with lysis buffer, then extracted using the Purelink Mini RNA extraction kit per manufacturer instructions (Invitrogen). All RNA was treated with Turbo DNAse and cleaned using RNA Clean and Concentrate (Zymo). Purity and concentration of RNA was determined using threshold 260/280 and 260/230 values of 2.0 and above obtained using Nanodrop (ThermoFisher). Removal of genomic DNA contamination was verified using qPCR consisting of a 10 uL reaction with 18.75 ng/uL RNA (three times final concentration) and 300 nM cysG primers. A total of 1 ug RNA in addition to iScript mastermix (BioRad) was used to create cDNA, which was subsequently diluted to a final concentration of 6.25 ng/uL in nuclease free water. 25 ng of cDNA was used in a 10 uL reaction with SsoAdvanced SYBER mastermix (Biorad) and 300 nM final primer concentration. The cysG gene was used as a reference, and the ΔΔCT method was used for relative expression analysis using UPEC reference strains UTI89 (cystitis strain) and CFT073 (pyelonephritis strain) as a positive controls given that they both contain all three genes [14, 15, 33,34,35]. The experiments were performed in biological duplicates.
Statistical analysis
Chi-squared test, Fisher’s exact test, and descriptive statistics on clinical data were performed using SPSS Statistics (IBM, version 29). Fisher’s exact test, Mann-Whitney test, paired t-test, and correlation analysis on experiemental data were performed using software GraphPad Prism 10 (Graphpad, San Diego, CA, version 10.4.1).
Results
Patient characteristics
A total of 72 patients were evaluated for this study. Postmenopausal women with at least one UTI diagnosis were grouped based on a history of single or recurrent episodes and were compared with respect to demographics and comorbid conditions particularly those known to predispose to UTI development (Table 1). The age of the study patients ranged from 50 to 95 years old with an overall median age of 73.5 years. Patients in the recurrent group were slightly older compared to those with single episode (median age of 78 vs. 72 years). The majority of the patients identified as White (50.0%, 36/72) followed by Hispanic (29.2%, 21/72). The majority of the patients were diagnosed with cystitis (84.7%, 61/72), followed by pyelonephritis (8.3%, 6/72) and urosepsis (2.8%, 2/72). Three patients (Other, 4.2%) had abnormal urinalysis but no mention of a diagnosis in the medical records. In the recurrent arm, the median number of previous episodes within the last 12 months was 3 (IQR 2, 4). The overall prevalence of comorbid conditions among study patients were 25% (18/72) for diabetes, 23.6% (17/72) kidney disease, 20.8% (15/72) immunocompromised status, 20.8% (15/72) dementia, 4.2% (3/72) functional quadrepelgia, and 4.2% (3/72) with a chronic urinary catheter. The prevalence of each comorbid condition was similar between study groups except for diabetes where there were twice as many patients in the recurrent compared to the single episode group (33.3%, 12/36 vs. 16.7%, 6/36); all three patients with chronic urinary catheters were in the recurrent arm. Most patients were admitted to the hospital (66.7%, 48/72) then discharged home after receiving antibiotic therapy (55.6%, 40/72). Ertapenem was the most frequently prescribed antibiotic (37.5%, 27/72), followed by ceftriaxone (33.3%, 24/72) and nitrofurantoin (22.2%, 16/72). Antibiotic therapies were similar between the study groups.
Prevalence of IBC-supportive genes
We investigated the molecular determinants of IBC formation using PCR to compare the prevalence of fimH, hlyA, and ompT among strains in each study group. The fimH and ompT genes were observed to be the most prevalent among the 72 UPEC strains isolated from postmenopausal women with 98.6% (71/72) and 81.9% (59/72) of strains possessing the genes respectively, whereas the hlyA gene was present in 26.4% (19/72) of strains. The majority of strains had both fimH and ompT (55.6%, 40/72), with a smaller subset of strains carrying all three genes (25.0%, 18/72), or only fimH (16.7%, 12/72). The prevalence of all three genes did not differ between strains from patients with recurrent or single episode UTI (Fig. 1; Fisher’s exact test, fimH p value > 0.999, ompT p value = 0.5414, hlyA p value = 0.1073). In the single episode group, one strain possessed only ompT, and another strain had only hlyA and fimH.
Presence/absence of virulence genes in UPEC strains isolated from 72 postmenopausal women. PCR was performed to amplify genes, then visualized using gel electrophoresis. UTI89 used as positive control; cysG used as the reference gene. Fisher’s exact test performed to compare the prevalence of each gene in recurrent and single episode strains (fimH p value > 0.999, ompT p value = 0.5414, hlyA p value = 0.1073)
Intracellular persistence of UPEC strains
We next sought to characterize the IBC phenotype between recurrent and single episode strains using a modified intracellular CFU assay. Twelve strains from recurrent patients and six strains from patients with single episodes were selected for further investigation based on the presence of ompT with or without the presence of hlyA to represent the population of isolated strains, and to enrich for the recurrent group (study group of interest) for the detection of potential differences between the groups. Three out of the twelve selected strains from the recurrent arm as well as four out of six selected strains from the single episode arm also contained the hlyA gene in addition to fimH and ompT genes. Overall, recurrent strains showed four-fold greater intracellular CFU counts compared to single-episode strains following 5 h of infection (median 16,248 CFU/mL vs. 4,118 CFU/mL, p = 0.0182) (Fig. 2). The range of intracellular CFU counts also varied widely between recurrent and single episode strains (7,172–129,300 average CFU/mL versus 1,848–33,258 CFU/mL), indicating differential ability to persist intracellularly within bladder epithelial cells.
Comparison of intracellular CFU counts of clinical UPEC from recurrent and single episode patients. 12 recurrent strains, and 6 single episode strains were used to infect bladder cell line 5637 at MOI 15 for two hours. Extracellular bacteria were washed away and cells treated with 450 ug/mL amikacin for two hours, then lysed and counted. Mann-Whitney test performed
Expression of IBC-supportive genes during infection and laboratory conditions
Since we observed differential intracellular persistence between recurrent and single episode strains, we investigated whether the expression of IBC-supportive virulence genes varied between recurrent and single episode strains during infection of BECs and when grown in laboratory media. Notably, we observed gene expression differences when grown in LB media compared to during infection in BECs and between strains causing single or recurrent episodes. When strains were grown in LB media, expression of fimH and ompT was similar between the two groups, p = 0.7503 and 0.8201 respectively (Supplemental Fig. 1). However, when the gene expression data was compared between intracellular populations of bacteria and growth in LB media, ompT expression was significantly increased in the recurrent group (Fig. 3, p = 0.0312), supporting its role in virulence. Interestingly, among the single episode strains, fimH was significantly decreased in the intracellular population compared to growth in LB media (Fig. 3, p = 0.0365) while recurrent strains did not differ in expression between LB and intracellular environments (Fig. 3). No difference in hlyA expression was observed between LB and infection for both groups. A trend towards higher intracellular expression of fimH and ompT was observed in recurrent strains when directly compared to that in single episode strains, with median ΔΔCq values 3- and 2-fold higher, p = 0.0529 and 0.0831, respectively (Supplemental Fig. 1). Taken together, these results suggest strain differences between those causing recurrent versus single episodes of UTI in postmenopausal women in both the intracellular expression of IBC-supportive genes and the intracellular persistence phenotype.
Normalized ompT, fimH, and hlyA expression in LB media and during BEC infection (ΔΔCq). Bacteria incubated to mid-log phase in LB for RNA extraction; BEC infection performed at MOI 50 as described for intracellular RNA extraction. Normalized to cysG and control strains, UTI89 and CFT073. Paired t-test performed
Exploratory correlation analysis between intracellular CFUs and intracellular expression of ompT and fimH
Given the observed increase in intracellular gene expression, we performed exploratory correlation analysis between intracellular gene expression and intracellular CFUs produced by all 18 strains using Spearman correlation. Intracellular expression of ompT and fimH displayed a modest positive correlation with intracellular CFUs (Supplemental Fig. 2A; r = 0.23, 0.39 (p values = 0.367 and 0.247)). However, removal of the recurrent strain outlier yielded a stronger correlation with only fimH expression as statistically significant (Supplemental Fig. 2B; r = 0.42, 0.53 (p values = 0.095 and 0.031)). These preliminary results suggest the potential contribution of both ompT and fimH to intracellular survival of UPEC in bladder epithelial cells.
Discussion
Recurrent UTIs have been attributed in part to the development of persistent bacterial reservoirs from invading UPEC forming intracellular bacterial communities (IBCs), as the cycle of IBC formation facilitates spread of the bacteria deep into the bladder tissue [12, 41]. This phenomenon has been observed in the bladders of postmenopausal women, who have been previously identified as high risk for developing recurrent UTIs [4]. Although observed within the bladders of postmenopausal women, the role of microbial determinants in IBC formation in this high risk population remains unclear. This study sought to examine the clinical characteristics of postmenopausal women who experienced recurrent versus single episode UTIs caused by ESBL-producing UPEC strains and compare the virulence gene expression and differential intracellular persistence of the infected strains. Patient factors known to increase risk of recurrence such as immunocompromised status and kidney disease were similar between groups, with twice as many diabetic patients in the recurrent arm though the difference was not statistically significant [3, 28, 30, 31].
With respect to microbial characteristics, the IBC-supportive genes fimH, and ompT were both highly prevalent across study strains, irrespective of recurrent or single episode UTI. The prevalence of each gene was consistent with published literature [21, 24, 25, 42,43,44]. Notably, we have shown that expression of ompT by the recurrent strains was significantly increased during infection compared to when grown in standard laboratory media. Additionally, a trend towards higher expression was observed in recurrent strains compared to single episode strains but will need to be confirmed with a larger sample size. These results highlighted the potential importance of OmpT in the intracellular environment, which is consistent with previous literature linking OmpT to biofilm formation [24, 45]. As the IBC structure has previously been observed to be biofilm-like, the increase in intracellular ompT expression may support the formation and/or maintenance of IBCs within BECs [11, 46]. Interestingly, type 1 fimbrial adhesin FimH expression was increased during in vitro infection in BECs among recurrent strains compared to expression during infection with single episode strains, and was significantly decreased in single episode strains during infection compared to expression in LB media. These results suggest that the role of FimH in the intracellular process of IBC formation may be strain dependent and relies on the microbial production of FimH which can be regulated by environmental cues such as osmolarity [47, 48]. Previous studies have also shown in both UPEC and K. pneumoniae that mutations in the FimH protein directly impact IBC formation and bladder colonization [33, 49]. Mutations supporting increased attachment and IBC formation may work in concert with other virulence factors to persist in the bladder tissue. When directly comparing intracellular expression of ompT and fimH between recurrent and single episode strains, there was no significant difference between the groups although a trend towards increased expression in the recurrent group was observed. Notably, significantly greater intracellular CFUs were found with recurrent strains compared to single episode strains. This study is the first to describe a significant difference in intracellular persistence of ESBL-producing UPEC isolated from postmenopausal women in association with UTI recurrence. Differential ability to persist intracellularly may be attributed to adaptive changes in virulence gene expression and production that develop in vivo. A modest positive correlation was observed between intracellular CFUs and intracellular expression of ompT and fimH for all 18 strains though not statistically significant, and show potential contributions of intracellular gene expression changes to adaptation in the bladder environment. For example, differential production of capsule production, alterations in metabolism, and the accumulation of various mutations supporting overall virulence, resistance, and metabolism changes over time has been observed in K. pneumoniae strains which successfully persisted in the bladder to cause chronic infection [50, 51]. Virulence gene expression changes in UPEC strains may support intracellular survival and adaptation to their particular host. Previous studies investigating virulence and metabolic gene expression in UPEC isolated from females with active UTI showed large variations in fimH expression in human urine as well as expression differences between voided urine and active infection [52, 53]. Urine composition and microbiome can vary between pre- and post-menopausal women, and between women in the same life stage [54,55,56,57]. Adaptation to each host may provide evolutionary and fitness benefits to infecting UPEC strains, as expression profiles vary significantly between laboratory media and urine as well as between patients given the variation in available nutrients and metabolites [52, 53, 57, 58]. Further studies with larger sample sizes are needed to confirm the direct contribution of OmpT and FimH, and to elucidate whether UPEC virulence factors change over time to better support chronic infection and persistence in the bladders of postmenopausal women.
The primary strength of this study lies in the investigation of the molecular characteristics of the bacteria causing the infections to complement the clinical characteristics of the patients experiencing recurrent UTIs. We are the first to show that strains causing recurrent infections in postmenopausal women have differential abilities to form IBCs compared to strains causing single episodes. The intracellular CFU counts for UPEC strain CFT073 were also consistent with previous studies using a bladder epithelial cell monolayer model, validating our experimental approach [14, 16, 37]. This work emphasizes the importance of using molecular determinants such as expression of bacterial protease OmpT in addition to patient factors in devising therapeutic strategies targeting IBC formation to prevent recurrent infection.
Several limitations to this study are notable. This retrospective study had a small sample size and the patients were selected from a single hospital site, although the patient demographics match a recently published large single-site epidemiology study on 374,171 women experiencing recurrent UTIs [3]. Patients in the single episode study group were identified based on single site visit for UTIs on medical records; however, it is possible that those patients could visit other clinic sites for UTI diagnosis and treatment. Infections were performed in an in vitro static model of the bladder using a monolayer of BECs which only consists on one layer of epithelium. We plan to investigate the development of IBCs using strains from these recurrent UTI patients in a 3D model of the bladder using primary human bladder epithelial cells to examine the adaptation of identified UPEC virulence factors over time in bladder tissue.
Conclusions
ESBL-UPEC strains causing recurrent UTIs in postmenopausal women exhibit differential expression of IBC-supportive virulence factors and intracellular persistence phenotypes as demonstrated in this study. Future investigations will explore the antivirulence potential of current and new treatments on the expression of IBC-supportive virulence factors and their efficacy in the clearance of IBCs generated by ESBL-UPEC from postmenopausal women.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- UTI:
-
Urinary tract infection
- UPEC:
-
Uropathogenic Escherichia coli
- ESBL:
-
Extended spectrum beta-lactamase
- BECs:
-
Bladder epithelial cells
- IBCs:
-
Intracellular bacterial communities
- CFUs:
-
Colony forming units
References
Carey AJ, Tan CK, Ipe DS, Sullivan MJ, Cripps AW, Schembri MA et al. Urinary tract infection of mice to model human disease: practicalities, implications and limitations. Crit Rev Microbiol. 2015;1–20.
Simmering JE, Tang F, Cavanaugh JE, Polgreen LA, Polgreen PM. The increase in hospitalizations for urinary tract infections and the Associated costs in the United States, 1998–2011. Open Forum Infect Dis. 2017;4(1):ofw281.
Ackerson BK, Tartof SY, Chen LH, Contreras R, Reyes IAC, Ku JH et al. Risk factors for recurrent urinary tract infections among women in a large integrated health care organization in the United States. J Infect Dis. 2024;jiae331.
De Nisco NJ, Neugent M, Mull J, Chen L, Kuprasertkul A, de Souza Santos M, et al. Direct detection of tissue-resident Bacteria and chronic inflammation in the bladder wall of Postmenopausal Women with recurrent urinary tract infection. J Mol Biol. 2019;431(21):4368–79.
Litwin MS, Saigal CS, Yano EM, Avila C, Geschwind SA, Hanley JM, et al. Urologic diseases in america project: analytical methods and principal findings. J Urol. 2005;173(3):933–7.
Goyal D, Dean N, Neill S, Jones P, Dascomb K. Risk factors for community-acquired extended-spectrum beta-lactamase–producing Enterobacteriaceae Infections—A retrospective study of symptomatic urinary tract infections. Open Forum Infect Dis. 2019;6(2):ofy357.
Ahn ST, Lee HS, Han DE, Lee DH, Kim JW, Park MG, et al. What are the risk factors for recurrent UTI with repeated ESBL-producing Enterobacteriaceae? A retrospective cohort study. J Infect Chemother. 2023;29(1):72–7.
Jorgensen S, Zurayk M, Yeung S, Terry J, Dunn M, Nieberg P, et al. Risk factors for early return visits to the emergency department in patients with urinary tract infection. Am J Emerg Med. 2018;36(1):12–7.
Klein RD, Hultgren SJ. Urinary tract infections: microbial pathogenesis, host–pathogen interactions and new treatment strategies. Nat Rev Microbiol. 2020;18(4).
Mulvey MA, Schilling JD, Hultgren SJ. Establishment of a persistent Escherichia coli reservoir during the acute phase of a bladder infection. Infect Immun. 2001;69(7):4572–9.
Anderson GG, Palermo JJ, Schilling JD, Roth R, Heuser J, Hultgren SJ. Intracellular bacterial biofilm-like pods in urinary tract infections. Science. 2003;301(5629):105–7.
Glover M, Moreira CG, Sperandio V, Zimmern P. Recurrent urinary tract infections in healthy and nonpregnant women. Urol Sci. 2014;25(1):1–8.
Flores-Mireles AL, Walker JN, Caparon M, Hultgren SJ. Urinary tract infections: Epidemiology, mechanisms of infection and treatment options. Nat Rev Microbiol. 2015;13(5):269–84.
Naskar M, Parekh VP, Abraham MA, Alibasic Z, Kim MJ, Suk G et al. α-Hemolysin promotes uropathogenic E. coli persistence in bladder epithelial cells via abrogating bacteria-harboring lysosome acidification. PLOS Pathog. 2023;19(5):e1011388.
Brannon JR, Thomassin JL, Desloges I, Gruenheid S, Le Moual H. Role of uropathogenic Escherichia coli OmpT in the resistance against human cathelicidin LL-37. FEMS Microbiol Lett. 2013;345(1):64–71.
He XL, Wang Q, Peng L, Qu YR, Puthiyakunnon S, Liu XL et al. Role of uropathogenic Escherichia coli outer membrane protein T in pathogenesis of urinary tract infection. Pathog Dis [Internet]. 2015 Apr 1 [cited 2023 Feb 3];73(3). Available from: http://academic.oup.com/femspd/article/doi/10.1093/femspd/ftv006/529669/Role-of-uropathogenic-Escherichia-coli-outer
Wurpel DJ, Beatson SA, Totsika M, Petty NK, Schembri MA. Chaperone-Usher Fimbriae of Escherichia coli. Wandersman C, editor. PLoS ONE. 2013;8(1):e52835.
Kolenda R, Sidorczuk K, Noszka M, Aleksandrowicz A, Khan MM, Burdukiewicz M et al. Genome placement of alpha-haemolysin cluster is associated with alpha-haemolysin sequence variation, adhesin and iron acquisition factor profile of Escherichia coli. Microb Genomics [Internet]. 2021 Dec 24 [cited 2024 Oct 17];7(12). Available from: https://www.microbiologyresearch.org/content/journal/mgen/https://doiorg.publicaciones.saludcastillayleon.es/10.1099/mgen.0.000743
Desloges I, Taylor JA, Leclerc J, Brannon JR, Portt A, Spencer JD et al. Identification and characterization of OmpT-like proteases in uropathogenic Escherichia coli clinical isolates. MicrobiologyOpen [Internet]. 2019 Nov [cited 2023 May 3];8(11). Available from: https://onlinelibrary.wiley.com/doi/10.1002/mbo3.915
Sarowska J, Futoma-Koloch B, Jama-Kmiecik A, Frej-Madrzak M, Ksiazczyk M, Bugla-Ploskonska G, et al. Virulence factors, prevalence and potential transmission of extraintestinal pathogenic Escherichia coli isolated from different sources: recent reports. Gut Pathog. 2019;11(1):10.
Whelan S, Lucey B, Finn K. Uropathogenic Escherichia coli (UPEC)-Associated urinary tract infections: the molecular basis for challenges to Effective Treatment. Microorganisms. 2023;11(9):2169.
Yamamoto S. Molecular epidemiology of uropathogenic Escherichia coli. J Infect Chemother. 2007;13(2):68–73.
Kanamaru S, Kurazono H, Ishitoya S, Terai A, Habuchi T, Nakano M, et al. Distribution and genetic association of putative uropathogenic virulence factors iron, Iha, kpsMT, ompT and Usp in Escherichia coli isolated from urinary tract infections in Japan. J Urol. 2003;170(6):2490–3.
Baldiris-Avila R, Montes-Robledo A, Buelvas-Montes Y, Phylogenetic classification B-F, capacity. virulence factors, and antimicrobial resistance in uropathogenic Escherichia coli (UPEC). Curr Microbiol. 2020;77(11):3361–70.
Hernández-Chiñas U, Ahumada-Cota RE, Navarro-Ocaña A, Chávez-Berrocal ME, Molina-López J, Rocha-Ramírez LM, et al. Phenotypic and genotypic characteristics of Escherichia coli strains isolated during a longitudinal follow-up study of chronic urinary tract infections. Front Public Health. 2023;11:1240392.
Wright KJ, Seed PC, Hultgren SJ. Development of intracellular bacterial communities of uropathogenic Escherichia coli depends on type 1 pili. Cell Microbiol. 2007;9(9):2230–41.
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. Front Med. 2022;9:931717.
Hu KK, Boyko EJ, Scholes D, Normand E, Chen CL, Grafton J, et al. Risk factors for urinary tract infections in Postmenopausal Women. Arch Intern Med. 2004;164(9):989.
CLSI CLSInstitute. Methods for Dilution Antimicrobial susceptibility tests for Bacteria that grow aerobically; approved standard — Ninth Edition. CLSI document M07-A9. Clin Lab Standars Inst. 2012;32(2).
Mody L, Juthani-Mehta M. Urinary tract infections in older women: a clinical review. JAMA. 2014;311(8):844.
Alghoraibi H, Asidan A, Aljawaied R, Almukhayzim R, Alsaydan A, Alamer E, et al. Recurrent urinary tract infection in adult patients, risk factors, and efficacy of low dose prophylactic antibiotics therapy. J Epidemiol Glob Health. 2023;13(2):200–11.
Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—A metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inf. 2009;42(2):377–81.
Chen SL, Hung CS, Pinkner JS, Walker JN, Cusumano CK, Li Z, et al. Positive selection identifies an in vivo role for FimH during urinary tract infection in addition to mannose binding. Proc Natl Acad Sci. 2009;106(52):22439–44.
Russell SK, Harrison JK, Olson BS, Lee HJ, O’Brien VP, Xing X, et al. Uropathogenic Escherichia coli infection-induced epithelial trained immunity impacts urinary tract disease outcome. Nat Microbiol. 2023;8(5):875–88.
Zhou K, Zhou L, Lim Q, ’En, Zou R, Stephanopoulos G, Too HP. Novel reference genes for quantifying transcriptional responses of Escherichia coli to protein overexpression by quantitative PCR. BMC Mol Biol. 2011;12(1):18.
Murray BO, Flores C, Williams C, Flusberg DA, Marr EE, Kwiatkowska KM, et al. Recurrent urinary tract infection: a mystery in search of Better Model systems. Front Cell Infect Microbiol. 2021;11:691210.
González MJ, Zunino P, Scavone P, Robino L. Selection of effective antibiotics for Uropathogenic Escherichia coli intracellular bacteria reduction. Front Cell Infect Microbiol. 2020;10:542755.
Li S, Pang Y, Zhang S, Li Q, Wang Q, Wang L, et al. Transcriptomic analysis reveals that the small protein MgtS contributes to the virulence of uropathogenic Escherichia coli. Microb Pathog. 2021;152:104765.
Westermann AJ, Gorski SA, Vogel J. Dual RNA-seq of pathogen and host. Nat Rev Microbiol. 2012;10(9):618–30.
Ho CH, Fan CK, Wu CC, Yu HJ, Liu HT, Chen KC, et al. Enhanced uropathogenic Escherichia coli-induced infection in uroepithelial cells by sugar through TLR-4 and JAK/STAT1 signaling pathways. J Microbiol Immunol Infect. 2021;54(2):193–205.
Hannan TJ, Totsika M, Mansfield KJ, Moore KH, Schembri MA, Hultgren SJ. Host–pathogen checkpoints and population bottlenecks in persistent and intracellular uropathogenic Escherichia coli bladder infection. FEMS Microbiol Rev. 2012;36(3):616–48.
Zhao L, Chen X, Zhu X, Yang W, Dong L, Xu X, et al. Prevalence of virulence factors and antimicrobial resistance of uropathogenic Escherichia coli in Jiangsu Province (China). Urology. 2009;74(3):702–7.
Mirzahosseini H, Najmeddin F, Najafi A, Ahmadi A, Sharifnia H, Khaledi A, et al. Correlation of biofilm formation, virulence factors, and phylogenetic groups among Escherichia coli strains causing urinary tract infection: a global systematic review and meta-analysis. J Res Med Sci. 2023;28(1):66.
Wang MC, Tseng CC, Wu AB, Huang JJ, Sheu BS, Wu JJ. Different roles of host and bacterial factors in Escherichia coli extra-intestinal infections. Clin Microbiol Infect. 2009;15(4):372–9.
Schembri MA, Kjærgaard K, Klemm P. Global gene expression in Escherichia coli biofilms. Mol Microbiol. 2003;48(1):253–67.
Duraiswamy S, Chee JLY, Chen S, Yang E, Lees K, Chen SL. Purification of Intracellular Bacterial Communities during Experimental Urinary Tract Infection Reveals an Abundant and Viable Bacterial Reservoir. Roy CR, editor. Infect Immun. 2018;86(4):e00740–17.
Schwan WR. Regulation of fim genes in uropathogenic Escherichia coli. World J Clin Infect Dis. 2011;1(1):17.
Bessaiah H, Anamalé C, Sung J, Dozois CM. What flips the switch? signals and stress regulating extraintestinal pathogenic Escherichia coli Type 1 Fimbriae (Pili). Microorganisms. 2021;10(1):5.
Lopatto EDB, Pinkner JS, Sanick DA, Potter RF, Liu LX, Bazán Villicaña J, et al. Conformational ensembles in Klebsiella pneumoniae FimH impact uropathogenesis. Proc Natl Acad Sci. 2024;121(39):e2409655121.
Ernst CM, Braxton JR, Rodriguez-Osorio CA, Zagieboylo AP, Li L, Pironti A et al. Adaptive evolution of virulence and persistence in carbapenem-resistant Klebsiella pneumoniae. Nat Med. 2020;26(5).
Kalu M, Tan K, Algorri M, Jorth P, Wong-Beringer A. In-human multiyear evolution of carbapenem-resistant Klebsiella pneumoniae causing chronic colonization and intermittent urinary tract infections: a case study. mSphere. 2022.
Subashchandrabose S, Hazen TH, Brumbaugh AR, Himpsl SD, Smith SN, Ernst RD, et al. Host-specific induction of Escherichia coli fitness genes during human urinary tract infection. Proc Natl Acad Sci. 2014;111(51):18327–32.
Sintsova A, Frick-Cheng AE, Smith S, Pirani A, Subashchandrabose S, Snitkin ES, et al. Genetically diverse uropathogenic Escherichia coli adopt a common transcriptional program in patients with UTIs. eLife. 2019;8:e49748.
Neugent ML, Kumar A, Hulyalkar NV, Lutz KC, Nguyen VH, Fuentes JL, et al. Recurrent urinary tract infection and estrogen shape the taxonomic ecology and function of the postmenopausal urogenital microbiome. Cell Rep Med. 2022;3(10):100753.
Ammitzbøll N, Bau BPJ, Bundgaard-Nielsen C, Villadsen AB, Jensen AM, Leutscher PDC, et al. Pre- and postmenopausal women have different core urinary microbiota. Sci Rep. 2021;11(1):2212.
Van Der Hugenholtz F, Terpstra ML, Borgdorff H, Van Houdt R, Bruisten S, et al. Urine and vaginal microbiota compositions of postmenopausal and premenopausal women differ regardless of recurrent urinary tract infection and renal transplant status. Sci Rep. 2022;12(1):2698.
Papp SB, Hogins J, Mekala S, Christie A, Chavez J, Reitzer L, et al. Daily and weekly urine variations in bacterial growth susceptibility in Postmenopausal Women with no history of urinary tract infection: a pilot study. Urology. 2024;194:82–8.
Frick-Cheng AE, Sintsova A, Smith SN, Krauthammer M, Eaton KA, Mobley HLT. The gene expression profile of uropathogenic Escherichia coli in women with uncomplicated urinary tract infections is recapitulated in the mouse model. mBio. 2020;11(4):e01412–20.
Tseng CC, Huang JJ, Ko WC, Yan JJ, Wu JJ. Decreased predominance of papG class II allele in Escherichia coli strains isolated from adults with acute pyelonephritis and urinary tract abnormalities. J Urol. 2001;166(5):1643–6.
Derakhshan S, Ahmadi S, Ahmadi E, Nasseri S, Aghaei A. Characterization of Escherichia coli isolated from urinary tract infection and association between virulence expression and antimicrobial susceptibility. BMC Microbiol. 2022;22(1):89.
Acknowledgements
Bacterial strain UTI89, isolated from a patient diagnosed with cystitis, was graciously provided by the laboratory of Dr. Scott Hultgren. Human bladder epithelial cell line 5637 (HTB9) as well as strain CFT073 (ATCC 700928), isolated from a patient diagnosed with pyelonephritis, were obtained from the American Type Culture Collection (ATCC).
Funding
None of the authors received funding for this study.
Author information
Authors and Affiliations
Contributions
M.K. was responsible for the conceptualization, execution of experiments, data analysis, preparation of all figures and tables, and writing of the manuscript. P. J. assisted in the conceptualization, data analysis, and writing of the manuscript. A.W.B. was responsible for the funding, conceptualization, data analysis, and writing of the manuscript. All authors read, reviewed, and approved the manuscript for publication.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Electronic medical record data was collected under IRB approval (HS-17-00943). Informed consent was waived as no interventions were made.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
12941_2025_779_MOESM1_ESM.tiff
Supplemental Fig. 1. Normalized ompT and fimH expression in (A) LB media and (B) during BEC infection (ΔΔCq). Bacteria incubated to mid-log phase in LB for RNA extraction; BEC infection performed at MOI 50 as described for intracellular RNA extraction. Normalized to cysG and control strains, UTI89 and CFT073. Mann-Whitney t-test performed.
12941_2025_779_MOESM3_ESM.tiff
Supplemental Fig. 2. Spearman correlation matrix between average intracellular CFUs and average intracellular expression of ompT and fimH for (A) all strains and (B) without recurrent strain outlier. Spearman r value listed for each comparison with blue as positive correlation and red as negative correlation. P values less than 0.05 are starred.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Kalu, M., Jorth, P. & Wong-Beringer, A. Comparison of phenotypic and genetic traits of ESBL-producing UPEC strains causing recurrent or single episode UTI in postmenopausal women. Ann Clin Microbiol Antimicrob 24, 11 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12941-025-00779-7
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12941-025-00779-7