Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Oct 9.
Published in final edited form as: Dev Biol. 2022 Nov 8;493:29–39. doi: 10.1016/j.ydbio.2022.11.003

Effects of Aging on Urinary Tract Epithelial Homeostasis and Immunity

Marianne M Ligon 1,*, Chetanchandra S Joshi 2,*, Bisiayo E Fashemi 1, Arnold M Salazar 1, Indira U Mysorekar 1,2,3,4,#
PMCID: PMC11463731  NIHMSID: NIHMS2025996  PMID: 36368522

Abstract

A global increase in older individuals creates an increasing demand to understand numerous healthcare challenges related to aging. This population is subject to changes in tissue physiology and the immune response network. Older individuals are particularly susceptible to infectious diseases, with one of the most common being urinary tract infections (UTIs). Postmenopausal and older women have the highest risk of recurrent UTIs (rUTIs); however, why rUTIs become more frequent after menopause and during old age is incompletely understood. This increased susceptibility and severity among older individuals may involve functional changes to the immune system with age. Aging also has substantial effects on the epithelium and the immune system that led to impaired protection against pathogens, yet heightened and prolonged inflammation. How the immune system and its responses to infection changes within the bladder mucosa during aging has largely remained poorly understood. In this review, we highlight our understanding of bladder innate and adaptive immunity and the impact of aging and hormones and hormone therapy on bladder epithelial homeostasis and immunity. In particular, we elaborate on how the cellular and molecular immune landscape within the bladder can be altered during aging as aged mice develop bladder tertiary lymphoid tissues (bTLT), which are absent in young mice leading to profound age-associated change to the immune landscape in bladders that might drive the significant increase in UTI susceptibility. Knowledge of host factors that prevent or promote infection can lead to targeted treatment and prevention regimens. This review also identifies unique host factors to consider in the older, female host for improving rUTI treatment and prevention by dissecting the age-associated alteration of the bladder mucosal immune system.

Keywords: Aging, Urinary Tract Infection, tertiary lymphoid tissue, Autophagy, ROS, CXCL13, TNFα, Estrogen, D-mannose

Introduction

On a global scale, the average age of the population is increasing. It is estimated that by 2050, the population over the age of 65 will double, and this group will grow to make up more than 20% of the total population1. Given this worldwide shift in age demographics, there is an increasing need to provide healthcare and treatments capable of meeting the needs of an aging population. This population is particularly susceptible to infectious diseases, the most common being respiratory infections, urinary tract infections (UTIs), and skin and soft tissue infections2. In addition to being more susceptible to contracting these infections, older individuals also suffer greater morbidity and mortality due to increased severity when these infections occur. This is the case in the urinary tract, where aging is associated with significant changes to host physiology and immunity. Women are particularly prone to multiple conditions of the urinary tract as they age, and women over the age of 50 are highly susceptible to bladder disorders including recurrent urinary tract infections (rUTIs), urinary incontinence, and overactive bladder (OAB), among others1,36. These bladder diseases all have a chronic inflammatory component as well as overlapping symptoms, known as lower urinary tract symptoms (LUTS)7,8.

This review will address the current state of knowledge about age-associated changes to the underlying biology of the bladder that may contribute to several diseases, with a particular focus on UTIs. We also elaborate further on the innate and adaptive immune responses used by the bladder mucosa, how aging of the urothelium influences these immune responses at baseline and during an infection, and potential therapeutic strategies based on these responses.

Diseases and disorders of the bladder in women

Among the many diseases of the bladder, susceptibility may vary between men and women due to differing anatomy and physiology9. While men are more likely to develop bladder cancer, for instance, other bladder disorders in men are frequently mediated by the prostate rather than the bladder alone10. In women, dysfunction and disorders of the bladder are more likely to be bladder-intrinsic but may also be secondary to changes in the vagina. The most common bladder disorder among all women is bacterial infection of the bladder or kidneys, usually referred to as urinary tract infections (UTIs)11. The majority of UTIs in women are limited to the bladder (cystitis) without complicating factors such as anatomic abnormality, systemic symptoms such as fever, or indwelling catheters11. Other common afflictions of the bladder in women include: (1) interstitial cystitis/bladder pain syndrome (IC/BPS), (2) overactive bladder (OAB), (3) stress incontinence, and (4) incomplete bladder emptying. IC/BPS is a little-understood, sterile inflammatory disease characterized by urothelial permeability defects, inflammation, ulceration, scarring and fibrosis of the bladder, hemorrhage upon distension, and reduced bladder capacity3,5,7,12,13. It is most commonly diagnosed in women in their 40s. OAB is characterized by uninhibited detrusor contractions with or without incontinence. Over 15 % of women report OAB and 11.0 % report OAB with urge urinary incontinence in the United States14,15. Kuo and colleagues have recently shown that urinary TNF-α levels are significantly higher individuals with voiding dysfunction including OAB16. We have previously also demonstrated that TNF-α levels in plasma samples can reliably differentiate OAB relative to controls and can be used to distinguish OAB from the other conditions. Stress incontinence refers to when the urinary sphincter and pelvic floor muscles are too weak to prevent urine leakage during high intraabdominal pressure such as coughing, laughing, or sneezing. Women may also experience mixed incontinence condition with symptoms of OAB and stress incontinence4,10,14,15,1720. Only 3% of women under 35 years of age experience symptoms of incontinence, but nearly 25% of women over age 65 report urinary incontinence21,22. Women generally suffer a higher rate of incontinence (urge, mixed, stress, and other) than men however, stress urinary incontinence is the most common type in women23. Furthermore, OAB has been postulated to result from inflamm-aging, with levels of nerve growth factor (NGF) and the chemokines CCL2 and CXCL1 in the urine increasing with age and OAB severity8,24,25. The common association of bladder disorders with both aging and chronic inflammation suggests that an underlying driver of pathology may be age-associated inflammation24,26. Treatment for bladder diseases such as OAB may involve invasive procedures or surgeries that further increase the risk of UTIs and other infections in older women.

Urinary Tract Infections

More than 10% of women in the US experience UTIs each year, and 60% of women will be diagnosed with a UTI during their lifetime2729 Click or tap here to enter text.. Furthermore, the economic impact of UTIs is over $3 billion, and UTIs account for at least 15% of all antibiotic prescriptions11,27,3032. UTIs may also ascend to the kidneys, causing pyelonephritis, then the blood stream, causing bacteremia and urosepsis. Recurrence of UTIs is a common concern, as 25–50% of infections will recur within 6 to 12 months after an initial infection, termed recurrent UTIs (rUTIs)33,34. Repeated treatment of rUTIs with antibiotics may lead to antibiotic resistance, disruption of the microbiota, and adverse effects of the drugs. Older patients are at increased risk of toxicity from certain antibiotics due to interactions with other medications and decreased renal function. Thus, there is an urgent need to identify new targets for UTI treatment and prophylaxis in women with rUTIs.

Risk factors for rUTIs vary among age demographics. The incidence of UTIs over the lifespan forms a J-shaped curve35. Some risk factors can be present at any age, such as uncontrolled diabetes although the prevalence of this co-morbidity also increases with age. Infant girls are at risk for UTIs from urinary tract abnormalities and vesicoureteral reflux36. After puberty, UTI risk increases and is frequently associated with sexual intercourse in adult women9. These risks plateau until menopause when UTIs become more frequent37. Postmenopausal and older women are particularly susceptible to rUTIs; in one study in a primary care setting, 53% of women over 55 had at least one UTI recurrence within a year compared to only 36% of those under 5538.

Postmenopausal vaginal atrophy and a change to the vaginal microbiota are related to reduced estrogen and increase the risk of UTIs39. Estrogen also plays a protective role against UTIs and has direct protective effects on urothelial barrier function4045. Vaginal estrogen therapy, which has low systemic absorption, is effective in reducing UTIs in postmenopausal women and reduces inflammation in the urinary tract4652. Anterior vaginal wall prolapse (cystocoele) and other causes of incomplete bladder emptying promote UTIs since uropathogens can more easily grow in stagnant urine within the bladder. Older women are also more likely to have incontinence, have had urogenital surgery, or be catheterized, all of which are associated with higher rates of UTIs and rUTIs37. Currently, there is a limited understanding of the extent to which age alone, or its associated co-morbidities, may have biologic effects on the bladder that mediate the increased risk of rUTIs.

Pathogenesis of UTIs in the bladder

UTIs are caused by different uropathogenic strains of bacteria. Infections from E. coli are the most common cause of UTIs, as uropathogenic E. coli (UPEC) infections alone account for approximately 80% of all UTIs30. Infections are can also be caused by other gram-negative bacteria such as, Klebsiella pneumoniae, Proteus mirabilis, Enterobacter species, Citrobacter species, and Pseudomonas aeruginosa, as well as gram-positive bacteria including group B Streptococcus (GBS), other Streptococci, Staphylococcus aureus, coagulase-negative Staphylococci, and Enterococcus species, among others29,53. Though UPEC still accounts for the majority of UTIs in older women, they are more likely to be infected with the less common uropathogens than younger women54. The pathogenic cycle of UPEC during an infection is well studied in vivo in a young mouse model and in vitro (Figure 1A). UPEC typically originate in the gut and go on to colonize the urethra, subsequently migrating into the bladder5559.

Figure 1: UPEC infection and immune response in young and aged bladders.

Figure 1:

(A) Schematic representation of events during UPEC infection cycle in the bladder. First, UPEC attach to uroplakins present on the superficial urothelial cells and invade. Next, a chain of competing responses from UPEC and the host determines the fate of infection. (1) UPEC is expelled into the bladder lumen via RAB27B+ fusiform vesicles. (2) UPEC escape into the cytosol and establish intracellular bacterial communities. (3) UPEC enter in the autophagosome where they subvert the autophagy pathway and form quiescent intracellular reservoirs (QIRs), and seed recurrent UTIs. (4) Innate immune cells infiltrate the site of infection and kill the bacteria. (5) Superficial cells loaded with IBCs are exfoliated into the bladder lumen thereby clearing the large bacterial load.

(B) Representation of epithelial and immune changes in the aged bladder. The aged bladder elaborates high levels of pro-inflammatory cytokines, IL-6, IL-1β, and TNF-α, and reduced levels of Estrogen relative to young bladders. The aged bladder is characterized by formation of tertiary lymphoid tissue (bTLT) with a bone fide germinal center with B cell and T cell zones and follicular dendritic cell (FDC) network. Within the bTLT, naïve B cells are recruited, undergo somatic hypermutation and respond to antigens. B cells may die by apoptosis or differentiate into memory B cells and antibody-secreting plasma cells which produce secretory IgA (sIgA). The aged urothelium harbors significantly higher numbers of QIRs and lysosomal accumulation and is increasingly permeable.

In women, UPEC can also colonize the vaginal epithelium, which may lead to an ascending infection60,61. When UPEC ultimately reach the bladder epithelium, the bacteria bind to the surface of the urinary tract using type 1 pili and pyelonephritis-associated (P) pili62. Located at the tip of UPEC type 1 pili are FimH adhesins, which bind uroplakins to initiate vesicle-dependent endocytosis62, allowing the internalization of UPEC into superficial urothelial cells, the outermost epithelial cell layer that forms the bladder’s watertight barrier. Bacteria enter into fusiform vesicles, where they release a phospholipase that destroys membrane integrity and permits bacterial escape into the cytoplasm63,64. After internalization, UPEC multiply within the protected intracellular niche forming biofilm-like intracellular bacterial communities (IBCs)65. IBCs may fill nearly an entire superficial cell, and a single IBC can consist of approximately 102-103 bacterial cells66. UPEC may then break out of the superficial cells, causing cell death, and go on to infect other urothelial cells where they can form quiescent intracellular reservoirs (QIRs) within autophagosomes, and can remerge to cause continued rounds of infection63,6769. QIRs are highly resistant to antibiotics and appear to be “hidden” from the immune system within this intracellular niche. Spontaneous reactivation of QIRs is not fully understood, but there is evidence that damage to the urothelium can trigger the release of UPEC back into the urine where the bacteria can replicate and re-infect. The urothelium can be damaged by other, transient bacteria, such as Gardnerella vaginalis, or be exfoliated by physical or chemical means to stimulate reactivation of latent infection67,70,71. Thus, UPEC have evolved mechanisms to live long-term in the bladder and continue to cause infections without re-inoculation from the intestinal reservoir. This finding has major implications for the treatment of UPEC infections in humans. Complete treatment of UTIs may thus be dependent on how fully we understand how to inhibit the formation of these reservoirs or if we can induce their reemergence to allow the host immune response to kill the extracellular bacteria.

Innate bladder defenses

The urinary bladder has physical barriers that prevent infection in addition to several other innate mechanisms72 similar to those in other mucosal surfaces. Urine flow can flush many, but not all, bacteria out of the lower urinary tract. Thus, in response to UPEC infection, the host activates several pathways to offset tissue damage and expel the bacteria. This includes a surge in the secretion of pro-inflammatory cytokines such as interleukin-6 (IL-6), exfoliation of infected superficial epithelial cells, and recruitment of monocytes and neutrophils. Exfoliation of superficial cells into the urine aids in the overall reduction of the bacterial burden in the bladder by removing cells with bacteria adhered their surface or containing IBCs and other intracellular bacteria. The urothelium also has cell-autonomous defense mechanisms to limit bacterial colonization of the bladder and prevent the establishment of UTIs. One early defense strategy is the expulsion of bacteria that have invaded the cell before they can form large IBCs69,7375. A redox stress response is activated as part of the host response to UPEC infection and has been shown to directly drive UPEC expulsion, a major urothelial defense against UPEC69. Joshi et al. 2021 recently demonstrated that the anti-redox pathway driven by NRF2, a transcription factor is rapidly activated to quench reactive oxygen species and thereby oxidative stress in part by directly activating the Rab GTPase, RAB27b which has been shown to play a central role in modulating UPEC entry into the urothelial cells as well as exocytosis of UPEC into the bladder lumen63,69,74,76,77 (Figure 1A). How this response changes with age is being explored in new work from our group and our observations suggest that the activation of the anti-redox pathway is compromised in aged bladders78.

As an innate defense, the urinary tract also secretes antimicrobial peptides such as Tamm–Horsfall protein (also called uromodulin), which is produced in the kidneys and is the most abundant protein in the urine79. Tamm–Horsfall protein competitively inhibits UPEC adhesion to uroplakins80. Mice lacking Tamm–Horsfall protein show 10 to 100-fold higher bacterial counts of UPEC positive for type 1 pili or P. mirabilis in their urine8183.

Like the gut, a healthy urinary tract also produces secretory IgA (sIgA)84,85. sIgA is produced by plasma cells in the lamina propria and actively secreted by epithelial cells at the mucosal surface85. The primary function of sIgA is to limit microbial access to the epithelium and control the commensal microbiota on the mucosal surface86,87. sIgA carries out this function via promoting agglutination-based clearance of invading pathogens88. Interestingly, aged bladders were shown to support increased local sIgA production in urine from aged mice, and the increased levels of sIgA89 were similar to elevated serum and intestinal IgA levels during aging90,91. Although IgA levels appear to be increased during UTI, it is uncertain whether the increase is antigen specific, protective, or dysregulated during a UTI92. Studies have suggested that sIgA has a very limited role in the defense against UTI84,93. However, new work is revealing that treatment with methanamine hippurate prevents breakthrough infections and may do so through increasing levels of urinary IgA94. Thus, the role of sIgA in bladder mucosal immunity requires further investigation95.

The aged urothelium has also been demonstrated to be increasingly permeable96. Given that flexibility and impermeability are two essential features of the urothelium, alteration in these could severely impact cellular homeostasis.

Autophagy and Aging in the bladder

Urothelial superficial cells utilize a network of autophagic vesicles directed by Rab GTPase proteins for membrane recycling of uroplakins97100. Autophagy is a conserved cellular process that recycles macromolecules and organelles in response to starvation or stress101. This complex process involving many components may also be used to capture and degrade intracellular pathogens, termed xenophagy102. While autophagy may be a useful early defense against invading UPEC, these bacteria also utilize autophagosomes to establish QIRs that can cause recurrent infections103,104. Mice that are hypomorphic for the essential autophagy gene Atg16L1 have faster UPEC clearance and form fewer QIRs than wildtype mice, indicating that UPEC normally benefit from the hosts’ initial autophagy response and sequestration in autophagosome105. The early clearance of UPEC in these mice was found to be due to heightened inflammation via increased production of IL-1β by macrophages, while the reduced QIR formation was intrinsic to loss of Atg16L1 in the urothelium104,106. While several pathogens have evolved to evade autophagy machinery, escape its compartments, or prevent lysosomal degradation, UPEC appear to use autophagosomes for long-term persistence within the bladder. UPEC are attracted to autophagosomes within urothelial cells to scavenge iron from ferritin107, which requires autophagic degradation (ferritinophagy) to release free iron for use by the cell from its ferritin cage where it is stored.

However, autophagy becomes dysregulated during aging with heightened oxidative stress accumulation108, affecting cellular processes like cell division, cytoskeleton architecture, membrane turnover, and vesicle trafficking. Impaired autophagy in the epithelium creates an energetic burden that promotes stem cell exhaustion and quiescence, a hallmark of premature aging109 observed in muscle satellite cells110, hematopoietic stem cells111, and neuronal stem cells109. Given that the urothelial innate immune response heavily depends on vesicle recycling, autophagy, and lysosome activity, the change in any of these cellular processes severely impacts the urothelial ability to respond to infection. The impact of the lysosome, autophagy, and metabolism on redox stress has been recently explored by our group and others in the aged urothelium78,112. Our studies demonstrate a significant block to autophagic flux and lysosomal degradation with low acid phosphatase activity indicative of inefficient cellular debris/waste removal. These findings align with prior studies showing accumulation of lipofuscin in aged urothelial cells113 and expanded endolysosomal compartments with accumulation of undigested material in the urothelium of aged rats96. Concomitant decrease in cellular respiration and mitochondrial bioenergetics in aged urothelial cell cultures have recently reported112. Further, a strong association with age and increase in oxidative stress63,112 has been reported supporting that aged urothelial cells exhibit dysfunctional lysosomes and accumulate metabolic waste products and cellular debris96. Decreased lysosomal function could lead to dysregulated autophagy and change the redox status of the urothelium. Accordingly, we note an increase in LC3I/II ratio with age and concomitant block in autophagy flux suggesting that there is age associated accumulation of autophagosomes63. Since UPEC utilize autophagosomes for QIR formation, the aged urothelium may be more conducive to harboring latent UPEC within QIRs. Indeed, there increased formation of intracellular bacterial communities (IBCs) and QIRs in aged mouse bladders resulting in increased incidence of recurrent UTIs63.

Overall, altered vesicular homeostasis, lysosomal function, and permeability barrier could affect the energetics of the tissue to influence its regeneration capacity and immune response to an infection. Given that these cellular functions are changed in aging tissue, a hampered urothelial response to invading pathogens is to be expected. Therefore, recurrence and persistence of urinary tract infections in older women are likely linked with aging bladder physiology. This opens several interesting questions for further investigation.

Immune responses in the bladder and rUTIs

Older individuals are more susceptible to infection and suffer disproportionate morbidity and mortality from infections than younger patients. The underlying mechanisms for this increased susceptibility and severity are not well understood but may involve functional changes to the immune system with age, known as immunosenescence. This state is characterized by several changes: (1) increased myeloid output and decreased lymphoid output from the bone marrow; (2) declining naïve repertoire of adaptive immune cells (decreased diversity of T and B cell receptors) with the oligoclonal expansion of antigen-experienced cells; (3) an increased rate of autoimmune disease; (4) poor control of latent infections and increased susceptibility to new infections, and (5) chronic low-grade inflammation of unknown origin, termed inflamm-aging114,115. While UTIs are one of the most common infections in both older men and women, it is not well understood how immunosenescence affects the bladder.

The bladder’s susceptibility to rUTIs has long suggested that eliciting effective adaptive immune responses may not be common. Several studies have demonstrated that repeat infection with the same UPEC strain in mice results in lower bladder bacterial titers during the challenge infection116,117. However, sterilizing immunity has not been observed, and latent UPEC reservoirs (QIRs) continue to form upon challenge infections. Furthermore, mice that have previously had severe, chronic UTIs are more susceptible to future UTIs than those that resolve their initial infection118, suggesting that divergent immune responses may dictate future responses to reinfection. T cell responses contribute to antigen-specific immunity in UTIs, but research has also found that bladder macrophages may be responsible for inhibiting these responses116,117. Interestingly, the abundant γδT cells in the bladder rapidly produce IL-17 upon infection, which aids in bacterial clearance119. It is not known if these cells are responding in an antigen-specific manner, as this response was demonstrated in naive mice. Recent work has shown that Type 17 immunity plays an important role in bladder defense implicating ILC3s in priming rapid responses to UPEC challenge120. This study showed presence of type 17 immune cell transcriptional factor, Rorc+ cells in the bladder. Whether the ILC3 response in increased or decreased with age is not known.

Of note, adaptive immune responses are markedly different if the infection is limited to the bladder or if it ascends to the kidneys. Kidney infection induces a robust, systemic adaptive immune response, including pathogen-specific IgG121. In humans, pyelonephritis is also marked by higher urine IgA concentrations. While antibody responses to UPEC may be antigen-specific, whether they are protective has not been demonstrated. Nevertheless, anti-UPEC vaccines are currently being investigated and several are already in use outside the U.S.70,122124. It is important to note that these vaccines are used as immunotherapy in patients with chronic UTIs rather than acting to prevent initial infection of the urinary tract.

Age-associated Tertiary lymphoid tissue formation in the bladder

The immune system is made up of lymphoid cells that inhabit nearly all tissues in the body and are frequently migratory between body tissues, lymphatics, and blood. The primary lymphoid organs produce new immune cells and include the bone marrow (where all hematopoietic cells are generated and B cells mature), and the thymus (which regulates T cell maturation). Secondary lymphoid organs (SLOs) include the spleen, lymph nodes, Peyer’s patches, and other, organized mucosa-associated lymphoid tissues. SLOs harbor large numbers of densely packed immune cells and function to facilitate adaptive immune responses. These organs are highly structured to facilitate a multitude of intercellular interactions between T cells, B cell, and antigen presenting cells that must take place in order to generate adaptive immune responses to pathogens125. Tertiary lymphoid tissues (TLTs) are capable of generating adaptive immune responses, and these responses typically act at their tissue-specific location. TLTs form ectopically at sites of chronic inflammation and antigenic stimulation rather than at pre-defined locations during embryogenesis126129. TLTs have been found in numerous tissues such as the lung130133, kidneys134136, intestines137140, and meninges141, and in varied disease states, including chronic infection, cancer, and autoimmune diseases. Interestingly, TLTs appear to be more common in the liver, intestines, and kidneys of aged mice135,142144, indicating that an aspect of the aging process may stimulate the development of TLTs. Whether immune responses arising within TLTs are pathogenic (such as in autoimmune disease) or protective (as in some cancers and in some infections) depends on both the tissue location and type of insult. Why TLTs form during aging and what type of responses these TLTs mediate is not fully understood.

TLTs and TLT-like structures have been reported in the bladder, but little detail in the description of their structure, function, causes, and consequences had been explored. Mice given repeat UTIs have larger, more distinct T cell influx at 24 hpi116,117. A subset of C3H/HeN mice with persistent bacteriuria, chronic cystitis, and pyelonephritis also have CD45+ aggregates, though specific cell types in these structures have not yet been defined145,146. Transgenic mice with constitutive Cox-2 expression in the urothelium develop bladder cancer and lymphoid aggregates containing B and T cells147. Furthermore, small lymphoid aggregates were observed in one small study of aged mice by routine and electron microscopy148. TLTs have also been identified in a small sample of muscle-invasive bladder cancer149. Despite these occurrences, TLTs in the bladder had been largely uncharacterized and their function entirely unexplored. A recent report from our group showed that in unperturbed, aged bladders, expanded numbers of B and T cells organize into structures termed bladder tertiary lymphoid tissue (bTLT)150 (Figure 1B). These bTLT form in a sex specific manner in aged female but not aged male bladders151. bTLT in aged mice serve as centers for B cell recruitment, activation, and differentiation into plasma cells and express multiple B cell-associated genes (Cxcr5, Cxcl13, Cd19, fclr5)150,151. B cell-associated pathways are enriched likely due to the redistribution of pools of B cells from the periphery to mucosal surface thus altering the mucosal landscape152. Age-dependent bTLT form in aged germ-free mice as well suggesting that microbial antigens are not required for their formation.

However aged TNFα−/− mice have fewer and smaller bTLTs, indicating that bTLTs require age-dependent TNFα to form. TNFα is required for the maintenance or growth of bTLTs once they had formed. The effect of anti-TNFα treatment on the composition, organization, or functionality of bTLTs remains to be determined. In addition, testing whether anti-TNFα treatment at an earlier age, such as 9 or 12 months when bTLTs begin to form, could prevent the growth of bTLTs and formation of new ones over a more extended period of time. These future studies would help further elucidate the mechanisms by which age-associated TNF promotes bTLT formation in female mice. TLT development is known to be induced by LTi cells, an ILC subtype that express surface, lymphotoxin, LTα1β2153 and LT signaling is potentiated by TNFα. Indeed, aged bladders also express increased lymphotoxin than bladders from young mice150. Increased LT signaling through its receptor, LTβR has been described in chronic cystitis154. Recent studies suggest that ILC3s are an important source of LT120 and it would be interesting to determine whether the numbers of ILC3 are increased in aged bladders. Taken together, emerging data suggests a profound change in the bladder mucosal immune system that is fundamental to aging in this tissue.

Given our findings that aged female bladders harbor increased bacterial reservoirs and are susceptible to rUTIs concomitant with formation of bTLT would suggest that presence of these structures are ‘pathogenic’ to the aged female. Indeed, human cohort studies would support this hypothesis as bTLT occur in postmenopausal women and are significantly associated increased frequency of rUTIs and shorter time interval to next recurrent episode150. bTLT in postmenopausal bladders have been shown to be associated with co-localization of E. coli species155. Presence of TLTs may potentiate overexuberant or ineffective immune responses that promote inflammation rather than resolution of UTIs. Identification of these structures provides a possible explanation for the increased occurrence of sex- and age- dependent conditions in aging females and establish a foundation for therapeutic interventions to limit bTLT formation.

Hormones, Aging, and Immunity

Recurrent UTIs afflict a large proportion of postmenopausal women, suggesting that lower estrogen levels predispose older women to a significant risk of chronic and recurrent UTIs. Estrogen receptors can be found throughout the lower urogenital tract, including the vagina, bladder, urethra, and pelvic floor musculature156,157, indicating that estrogen plays a broader role in urogenital physiology. Although the change is more dramatic in humans, mice do experience a reduction in estrogen levels during a “menopause” period around the time they lose their reproductive fitness. Interestingly, this “reproductive senescence” occurs around 9 to 12 months in mice, which coincides with the early appearance of bTLT150. Since the urothelium is estrogen-responsive, processes that occur during reproductive senescence and atrophy of the ovaries and uterus may trigger changes that lead to bTLT formation. TLT structures have been described in bladders of prepubertal females. Miosevic and colleagues have shown that prepubertal females may harbor lymphoid lesions similar to those noted in postmenopausal women and these are correlated with number of UTIs in the prior year and bladder wall thickness158,159. Together, these studies suggest that hormonal ranges may have significant implications for TLT formation and pathogenesis.

Building on this observation, in vivo data demonstrates that estrogen protects against UTIs and helps to repair urothelial permeability40,44,160. Hass and colleagues have also shown that estrogen deficiency has a negative impact on urothelial barrier function44. Furthermore, an ovariectomized mouse model that mimics the postmenopausal condition with or without exogenous estrogen supplementation demonstrated that mice with lower estrogen levels harbored significantly higher QIRs, implying a high risk of recurrent UTIs40,41. The studies by Lüthje et al. and Wang et al. showed that estrogen imparts protection against UTIs by modulating mucosal bladder defenses40,41, supporting the use of exogenous estrogen as prophylactic therapy for rUTIs51,161164.

Targeted use of VET has been shown to effectively reduce rUTIs and shows overall improvement of lower urinary tract symptoms (LUTS)46,47,165168. The effect of the hormone on local microbiota could be one of the factors influencing the response of the bladder. Similar to the age-associated changes that occur in the vaginal microbiome, an age-associated change in the urinary microbiome has also been observed. Both pre- and postmenopausal women show dominance of Lactobacillus in both the vagina and bladder, but Lactobacillus has a relative abundance of 77.8% in premenopausal women compared to 42.0% in postmenopausal women169. VET increases the dominance of Lactobacillus in both the vagina and bladder of postmenopausal women47,170. Microbiome analysis of urines from 34 premenopausal and 20 postmenopausal women revealed higher alpha diversity with higher abundance of the genera Gardnerella and Prevotella compared with premenopausal women who mostly presented with Lactobacillus dominated urotypes169. Jung et al., recently showed that VET leads to increase in Lactobacilli species in the urobiome of postmenopausal women with rUTI51. Neugent and colleagues very recently showed strong correlations between presence of Bifidobacteria and Lactobacilli and urinary estrogen in women without a UTI history. Collectively, they suggest that rUTI and estrogen levels shape the urobiome171.

Additionally, estrogen modulates the thickness of the urothelial glycosaminoglycan (GAG) layer and regulates the expression of GAG-sulfation enzyme HS6ST1 over the course of UPEC infection42. Our work has previously shown that HS6ST1 also modulates activation of Bmp4 signaling post-infection172. These data imply that estrogen could influence BMP4-regulated regeneration and the thickness of the urothelial GAG layer post-infection. In this manner, estrogen appears to modulate cellular physiology and influence the local microbiota of the vagina and bladder.

Other studies have determined the effects of sex and hormones on UTIs in mice, finding that male mice are less likely to clear experimental UTI than female mice and that these effects are at least partially mediated by testosterone173,174. Testosterone was recently found to impact the immune response to UTI175; this finding suggests that both sex and hormones may indeed have a substantial impact on the aging immune system within the bladder. However, this hypothesis remains to be formally tested. Regarding B cells, age-associated B cells accumulate earlier and in greater quantities in female mice compared to male mice176,177. Furthermore, these cells are found in models of autoimmune disease, which again is more common in females than males. These findings agree with the model demonstrating higher levels of inflammation response in females than males9, particularly in relation to aging. The interaction between age, sex, hormone levels, and reproductive senescence on the bladder is clearly complex and needs more work to be better understood.

Therapeutic Interventions

In most cases, a UTI is spontaneously resolved within days without medical intervention. However, an unresolved infection can lead to pyelonephritis and can even be fatal178,179. The recommended antibiotic therapy for uncomplicated UTIs includes the use of fosfomycin, nitrofurantoin, amoxicillin-clavulanate, and cephalosporins180. Bacterial resistance to antibiotics such as these is a growing issue of concern for the treatment of UTI. There is also no effective vaccine available that can prevent UTIs. Thus, several alternative therapeutic strategies are being pursued as treatments for UTIs and have been reviewed extensively elsewhere69.

In addition, the U.S. FDA-approved anti-inflammatory drug dimethyl fumarate (DMF) has showed some promise, as mice treated with DMF showed decreased UPEC count in the bladder and urine69. Another strategy is to block UPEC adhesion to superficial cells using the simple sugar D-mannose, which has reportedly lowered the risk of rUTIs when administered daily in a clinical trial181,182. Moreover, in a retrospective cohort study of patients with rUTIs and who harboured bTLT lesions were treated with D-mannose resulting in significant decrease in UTI incidence182. Recent studies from our group suggest that D-mannose not only plays an extrinsic role but also plays important urothelial cell-intrinsic roles: we show that D-mannose treatment of aged mice promotes autophagic flux and lysosomal degradation thus improving outcomes78.

Methenamine salts are FDA-approved in the U.S. for long-term prophylactic use in treating patients with rUTIs and have demonstrated effectiveness as antiseptics in preventing UTI in adults 60 years of age and older, regardless of kidney function status183. Methenamine was well tolerated and helpful in reducing UTIs, antibiotic prescriptions, and hospitalization in renal transplant recipients with rUTIs184 and appears to be effective as a means of improving barrier function of the urothelium94. Importantly, there are no known resistance mechanisms to methenamine. Therapeutic approaches such as these should be considered for more widespread use and as alternatives to conventional antibiotics in the treatment of adults with UTIs, especially rUTIs.

Interventions could improve outcomes by either reducing UTIs through antibiotic and drug treatments but also via reducing TLT lesions. Indeed, Zimmern and colleagues have shown that electrofulguration by cauterization of CC/TLT led to much improved outcomes in women with antibiotic-refractory rUTIs185. Milosevic and colleagues have also shown that regression of the TLT lesions improved rUTIs159. Finally, recent work from our group has revealed that vaginal estrogen therapy (VET) in aged female mice promotes regression of bTLT in part by reducing levels of TNFα and CXCL13 and this is associated with reduced rUTIs45.

Concluding remarks

Recurring UTIs will continue to be a burden on our aging population. Considering the increasing prevalence of antibiotic resistance among uropathogens, developing alternative or non-antibiotic approaches to treat and prevent UTIs is essential. Comprehensive knowledge of host factors that prevent or promote infection continues to be essential to develop new targeted treatment and prevention regimens. Since it is evident that aging affects the bladder at multiple levels, a multilayered response and treatment strategy should be pursued to effectively address rUTIs that disproportionately affect the older population. Future work to further dissect the impact of age on UTIs should also include examination of bTLT when addressing the chronic and rUTIs in older individuals. We have highlighted a new framework for studying the changing immune landscape of the bladder during old age that can be translated to clinical outcomes and potential therapies for women with rUTIs. This new concept of the interplay between aging, infection, and local immune responses within the bladder generates many new important questions for the field.

Table 1:

Cytokine/chemokines that are associated with various bladder disease conditions

Cytokines/Chemokines Model (cell culture, mouse, rat, human/clinical, etc) Tissue/Cell type (whole bladder, urine, urothelium, macrophages, bTLT etc) Disease condition (UTI, OAB, cystitis, etc) Reference(s)
mRNA levels: ↑IL-1β, IL-2, IL-4, IL-6, TNF-α/β; no change in IL-1α, IL-10 and IFN-γ Wistar rats Whole bladder CYP-induced cystitis 186
Protein level: ↑IL-1β, IL-10, IL-6; No change IL-4, TNF-α Wistar rats Whole bladder CYP-induced cystitis 186
↑IL-1β, CXCL1 (KC) and CCL2 (MCP-1) C57BL/6 & CB6F1 Urine UTI (Proteus mirabilis HI4320) (Escherichia coli CFT073) 187
↑IL-6; IL-8 Human (1–121mos.) Urine and serum acute UTI; Pyelonephritis (febrile UTI) 188,189
↑CXCL10, iNOS, CCL5, CXCL5, CXCL9, IL-1α, IL-1β, IL-6 C57BL/6 whole bladder & Urine UTI (CFT073, 83972 & GBS) 190
↑IL-1β, IL-ra, IL-8, GM-CSF Cell culture Uroepithelial cells (T24 & 5637) GBS cystitis 190
↑IL-8 Human Urine UTI 191
↑NGF, MCP-1, sCD40L, MIP-1β, IL-12p70/p40, IL-5, EGF, GRO-α, sIL-2Rα, IL-10 Human Urine OAB 24,25,192
↑CXCL1, CXCL8, CXCL10 UTI
↑IL-1β, IL-1α, TNF-α, IL-6 Human Urine Bacterial cystitis or microhematuria 193
↑MCP-1, TARC, PARC, Fas/TNFRSF6; ↓IL-5, IL-6, IL-7, GM-CSF Human Urine OAB 194
↑MCP-2, MCP-3, TNF-β, G-CSF, eotaxin-3 Human Urine OAB and UTI 194
↑BDNF, NGF, MCP-1, GAG Human Urine OAB 195
↑TNF-α, MIP-1β; ↓IL-4 Human Plasma OAB 8
↑CXCL1, CXCL8, IL-6 Human Urine Acute cystitis 196
↑IL-1β, IL-1ra, IL-6, IL-8, IL-10, TNF-α, GCSF, MCP-1, MIP-1α, MIP-1β, RANTES, VEGF, IL-4, IL-7, IL-9, IL-12 p70, IL-17A, eotaxin, GM-CSF, IP-10, FGF, IFN-γ, PDGF-BB Human Urine Acute UTI 197
↑VEGF, IL-1α, IL-6 Human Urine HIC and NHIC 198
↑IL-6 Human Urine OAB 199
↑IL-6, IL-8 Human Urine rUTI (UPEC) 200
↑IL-6, IL-8, BDNF; ↓IL-2, NGF, VEGF Human Urine rUTI with PRP injections 201
↑TNF-α Mouse Bladder Age 89
↑CXCL13 Mouse Bladder Age 89
↓IL-8 Human Bladder Interstitial cystitis/BPS 202
↑IL-8 Human Urine Interstitial cystitis/BPS 203
↑MCP-1, RANTES, CXCL10/IP-10, Eotaxin-1 Human Urine Interstitial cystitis/BPS 204
↑IL-6, IL-8 Human Urine UTI 205

Highlights.

  • Aging is associated with increased susceptibility to rUTIs

  • Aged mice develop bladder tertiary lymphoid tissues (bTLT)

  • bTLT are associated with significant increase in UTI frequency in mice and women.

  • Urothelial permeability and urobiome species change with age and menopausal status

  • Multimodal therapies can lead to bTLT regression and improved rUTI outcomes

Acknowledgments

This work was supported in part by NIH grants, R01DK100644, R01AG052494, P20DK119840, and R56AG064634 (to IUM), NIH training grants, T32AI007172 and T32GM007200 (to MML); and T32-AI007172 (to BEF). Figure created using BioRender®. We thank Dr. Robert Lawrence for valuable comments and editing.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of Interests

IUM serves on the scientific advisory board of Luca Biologics.

References

  • 1.Kline KA & Bowdish DM Infection in an aging population. Curr Opin Microbiol 29, 63–67 (2016). 10.1016/j.mib.2015.11.003 [DOI] [PubMed] [Google Scholar]
  • 2.Mouton CP, Bazaldua OV, Pierce B & Espino DV Common infections in older adults. Health care food & nutrition focus 18 (2001). [PubMed] [Google Scholar]
  • 3.Koziol JA, Adams HP & Frutos A Discrimination between the ulcerous and the nonulcerous forms of interstitial cystitis by noninvasive findings. J Urol 155, 87–90 (1996). [PubMed] [Google Scholar]
  • 4.Nygaard I et al. Prevalence of symptomatic pelvic floor disorders in US women. JAMA 300, 1311–1316 (2008). 10.1001/jama.300.11.1311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Simon LJ, Landis JR, Erickson DR & Nyberg LM The Interstitial Cystitis Data Base Study: concepts and preliminary baseline descriptive statistics. Urology 49, 64–75 (1997). [DOI] [PubMed] [Google Scholar]
  • 6.Suskind AM et al. Incidence and Management of Uncomplicated Recurrent Urinary Tract Infections in a National Sample of Women in the United States. Urology 90, 50–55 (2016). 10.1016/j.urology.2015.11.051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Grundy L, Caldwell A & Brierley SM Mechanisms Underlying Overactive Bladder and Interstitial Cystitis/Painful Bladder Syndrome. Front Neurosci 12, 931 (2018). 10.3389/fnins.2018.00931 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ma E et al. A multiplexed analysis approach identifies new association of inflammatory proteins in patients with overactive bladder. Am J Physiol Renal Physiol 311, F28–34 (2016). 10.1152/ajprenal.00580.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Deltourbe L, Lacerda Mariano L, Hreha TN, Hunstad DA & Ingersoll MA The impact of biological sex on diseases of the urinary tract. Mucosal immunology 15 (2022). 10.1038/s41385-022-00549-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Maserejian NN et al. Incidence of lower urinary tract symptoms in a population-based study of men and women. Urology 82, 560–564 (2013). 10.1016/j.urology.2013.05.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Foxman B The epidemiology of urinary tract infection. Nat Rev Urol 7, 653–660 (2010). 10.1038/nrurol.2010.190 [DOI] [PubMed] [Google Scholar]
  • 12.Maeda D et al. Hunner-Type (Classic) Interstitial Cystitis: A Distinct Inflammatory Disorder Characterized by Pancystitis, with Frequent Expansion of Clonal B-Cells and Epithelial Denudation. PLoS One 10, e0143316 (2015). 10.1371/journal.pone.0143316 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hurst RE et al. Increased bladder permeability in interstitial cystitis/painful bladder syndrome. Translational andrology and urology 4 (2015). 10.3978/j.issn.2223-4683.2015.10.03 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Reynolds WS, Fowke J & Dmochowski R The Burden of Overactive Bladder on US Public Health. Current bladder dysfunction reports 11 (2016). 10.1007/s11884-016-0344-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Eapen RS & Radomski SB Review of the epidemiology of overactive bladder. Research and reports in urology 8 (2016). 10.2147/RRU.S102441 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chow PM & Kuo HC Performance of urinary biomarkers in differentiating dysfunctional voiding in women with overactive bladder syndrome: a prospective pilot study. International urology and nephrology 54 (2022). 10.1007/s11255-022-03265-7 [DOI] [PubMed] [Google Scholar]
  • 17.Anger JT et al. The prevalence of urinary incontinence among community dwelling men: results from the National Health and Nutrition Examination survey. The Journal of urology 176 (2006). 10.1016/j.juro.2006.07.029 [DOI] [PubMed] [Google Scholar]
  • 18.Carpenter L & Campain NJ Overactive bladder: not just a normal part of getting older. British journal of nursing (Mark Allen Publishing) 31 (2022). 10.12968/bjon.2022.31.18.S16 [DOI] [PubMed] [Google Scholar]
  • 19.Patel UJ, Godecker AL, Giles DL & Brown HW Updated Prevalence of Urinary Incontinence in Women: 2015–2018 National Population-Based Survey Data. Female pelvic medicine & reconstructive surgery 28 (2022). 10.1097/SPV.0000000000001127 [DOI] [PubMed] [Google Scholar]
  • 20.Milsom I & Gyhagen M The prevalence of urinary incontinence. Climacteric: the journal of the International Menopause Society 22 (2019). 10.1080/13697137.2018.1543263 [DOI] [PubMed] [Google Scholar]
  • 21.Hung KJ, Awtrey CS & Tsai AC Urinary incontinence, depression, and economic outcomes in a cohort of women between the ages of 54 and 65 years. Obstetrics and gynecology 123 (2014). 10.1097/AOG.0000000000000186 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bresee C et al. Prevalence and correlates of urinary incontinence among older community-dwelling women. Female pelvic medicine & reconstructive surgery 20 (2014). 10.1097/SPV.0000000000000093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Alperin M et al. Foundational Science and Mechanistic Insights for a Shared Disease Model: An Expert Consensus. Female Pelvic Med Reconstr Surg 28, 347–350 (2022). 10.1097/spv.0000000000001216 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Tyagi P et al. Association of inflammaging (inflammation + aging) with higher prevalence of OAB in elderly population. Int Urol Nephrol 46, 871–877 (2014). 10.1007/s11255-013-0621-x [DOI] [PubMed] [Google Scholar]
  • 25.Tyagi P et al. Urine cytokines suggest an inflammatory response in the overactive bladder: a pilot study. International urology and nephrology 42 (2010). 10.1007/s11255-009-9647-5 [DOI] [PubMed] [Google Scholar]
  • 26.Franceschi C et al. Inflammaging 2018: An update and a model. Seminars in immunology 40 (2018). 10.1016/j.smim.2018.10.008 [DOI] [PubMed] [Google Scholar]
  • 27.Foxman B, Barlow R, D’Arcy H, Gillespie B & Sobel JD Urinary tract infection: self-reported incidence and associated costs. Annals of epidemiology 10 (2000). 10.1016/s1047-2797(00)00072-7 [DOI] [PubMed] [Google Scholar]
  • 28.Medina M & Castillo P An introduction to the epidemiology and burden of urinary tract infections. Ther Adv Urol 11 (2019). 10.1177/1756287219832172 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Foxman B & Brown P Epidemiology of urinary tract infections: transmission and risk factors, incidence, and costs. Infectious disease clinics of North America 17 (2003). 10.1016/s0891-5520(03)00005-9 [DOI] [PubMed] [Google Scholar]
  • 30.Dielubanza EJ & Schaeffer AJ Urinary tract infections in women. The Medical Clinics of North America 95, 27–41 (2011). 10.1016/j.mcna.2010.08.023 [DOI] [PubMed] [Google Scholar]
  • 31.Foxman B Epidemiology of urinary tract infections: incidence, morbidity, and economic costs. Am J Med 113 Suppl 1A, 5S–13S (2002). [DOI] [PubMed] [Google Scholar]
  • 32.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 infectious diseases 4 (2017). 10.1093/ofid/ofw281 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Brubaker L, Carberry C, Nardos R, Carter-Brooks C & Lowder JL American Urogynecologic Society Best-Practice Statement: Recurrent Urinary Tract Infection in Adult Women. Female Pelvic Med Reconstr Surg (2018). 10.1097/SPV.0000000000000550 [DOI] [PubMed] [Google Scholar]
  • 34.Ikäheimo R et al. Recurrence of urinary tract infection in a primary care setting: analysis of a 1-year follow-up of 179 women. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America 22 (1996). 10.1093/clinids/22.1.91 [DOI] [PubMed] [Google Scholar]
  • 35.Geerlings SE Clinical Presentations and Epidemiology of Urinary Tract Infections. Microbiology spectrum 4 (2016). 10.1128/microbiolspec.UTI-0002-2012 [DOI] [PubMed] [Google Scholar]
  • 36.Shaikh N, Morone NE, Bost JE & Farrell MH Prevalence of urinary tract infection in childhood: a meta-analysis. The Pediatric infectious disease journal 27 (2008). 10.1097/INF.0b013e31815e4122 [DOI] [PubMed] [Google Scholar]
  • 37.Stamm WE, Raz R & Vol. 28 723–725 (Clin Infect Dis, 1999). [DOI] [PubMed] [Google Scholar]
  • 38.Ikaheimo R et al. Recurrence of urinary tract infection in a primary care setting: analysis of a 1-year follow-up of 179 women. Clin Infect Dis 22, 91–99 (1996). [DOI] [PubMed] [Google Scholar]
  • 39.Stapleton AE The Vaginal Microbiota and Urinary Tract Infection. Microbiol Spectr 4 (2016). 10.1128/microbiolspec.UTI-0025-2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lüthje P et al. Estrogen Supports Urothelial Defense Mechanisms. Science Translational Medicine 5, 190ra180 (2013). 10.1126/scitranslmed.3005574 [DOI] [PubMed] [Google Scholar]
  • 41.Wang C et al. Vol. 81 733–739 (Infect Immun, 2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Anand M et al. Estrogen affects the glycosaminoglycan layer of the murine bladder. Female Pelvic Med Reconstr Surg 18, 148–152 (2012). 10.1097/SPV.0b013e31824b76bd [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lüthje P, Hirschberg AL & Brauner A Estrogenic action on innate defense mechanisms in the urinary tract. Maturitas 77, 32–36 (2014). 10.1016/j.maturitas.2013.10.018 [DOI] [PubMed] [Google Scholar]
  • 44.Hass MA, Nichol P, Lee L & Levin RM Estrogen modulates permeability and prostaglandin levels in the rabbit urinary bladder. Prostaglandins, leukotrienes, and essential fatty acids 80 (2009). 10.1016/j.plefa.2008.11.010 [DOI] [PubMed] [Google Scholar]
  • 45.Fashemi BE, Wang C, Chappidi RR, Morsy H & Mysorekar IU Supraphysiologic Vaginal Estrogen Therapy in Aged Mice Mitigates Age-associated Bladder Inflammatory Response to Urinary Tract Infections. Urogynecology (in press) (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Meister MR, Wang C, Lowder JL & Mysorekar IU Vaginal Estrogen Therapy Is Associated With Decreased Inflammatory Response in Postmenopausal Women With Recurrent Urinary Tract Infections. Female Pelvic Medicine & Reconstructive Surgery 27, e39–e44 (2021). 10.1097/SPV.0000000000000790 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Raz R & Stamm WE A controlled trial of intravaginal estriol in postmenopausal women with recurrent urinary tract infections. N Engl J Med 329, 753–756 (1993). 10.1056/nejm199309093291102 [DOI] [PubMed] [Google Scholar]
  • 48.Lenger SM et al. Feasibility and Research Insights From a Randomized Controlled Trial for Recurrent Urinary Tract Infection Prevention in Postmenopausal Women Using Vaginal Estrogen Therapy. Female pelvic medicine & reconstructive surgery 28 (2022). 10.1097/SPV.0000000000001171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Vaughan MH et al. The Urinary Microbiome in Postmenopausal Women with Recurrent Urinary Tract Infections. The Journal of urology 206 (2021). 10.1097/JU.0000000000001940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Anglim B, Phillips C, Shynlova O & Alarab M The effect of local estrogen therapy on the urinary microbiome composition of postmenopausal women with and without recurrent urinary tract infections. International urogynecology journal 33 (2022). 10.1007/s00192-021-04832-9 [DOI] [PubMed] [Google Scholar]
  • 51.Jung CE et al. Impact of Vaginal Estrogen on the Urobiome in Postmenopausal Women With Recurrent Urinary Tract Infection. Female pelvic medicine & reconstructive surgery 28 (2022). 10.1097/SPV.0000000000001051 [DOI] [PubMed] [Google Scholar]
  • 52.Fox KA, Lokken EM, Reed SD & Rahn DD Evaluation of systemic estrogen for preventing urinary tract infections in postmenopausal women. Menopause 28 (2021). [DOI] [PubMed] [Google Scholar]
  • 53.Flores-Mireles AL, Walker JN, Caparon M & Hultgren SJ Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nature reviews. Microbiology 13 (2015). 10.1038/nrmicro3432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Thomas-White KJ et al. Evaluation of the urinary microbiota of women with uncomplicated stress urinary incontinence. Am J Obstet Gynecol 216, 55 e51–55 e16 (2017). 10.1016/j.ajog.2016.07.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Terlizzi ME, Gribaudo G, Maffei ME & Vol. 8 (Front Microbiol, 2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Worby CJ et al. Longitudinal multi-omics analyses link gut microbiome dysbiosis with recurrent urinary tract infections in women. Nature microbiology 7 (2022). 10.1038/s41564-022-01107-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Worby CJ, Olson BS, Dodson KW, Earl AM & Hultgren SJ Establishing the role of the gut microbiota in susceptibility to recurrent urinary tract infections. The Journal of clinical investigation 132 (2022). 10.1172/JCI158497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Salazar AM, Neugent ML, De Nisco NJ & Mysorekar IU Gut-bladder axis enters the stage: Implication for recurrent urinary tract infections. Cell host & microbe 30 (2022). 10.1016/j.chom.2022.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Spaulding CN & Hultgren SJ Adhesive Pili in UTI Pathogenesis and Drug Development. Pathogens 5 (2016). 10.3390/pathogens5010030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Brannon JR et al. Invasion of vaginal epithelial cells by uropathogenic Escherichia coli. Nature communications 11 (2020). 10.1038/s41467-020-16627-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Robinson CK, Saenkham-Huntsinger P, Hanson BS, Adams LG & Subashchandrabose S Vaginal Inoculation of Uropathogenic Escherichia coli during Estrus Leads to Genital and Renal Colonization. Infection and immunity 90 (2022). 10.1128/iai.00532-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zhou G et al. Uroplakin Ia is the urothelial receptor for uropathogenic Escherichia coli: evidence from in vitro FimH binding. 114, 4095–4103 (2001). [DOI] [PubMed] [Google Scholar]
  • 63.Joshi CS, Cegelski L & Mysorekar IU PITing it forward: A new link in the journey of uropathogenic E. coli in the urothelium. Cell reports 39 (2022). 10.1016/j.celrep.2022.110758 [DOI] [PubMed] [Google Scholar]
  • 64.Pang Y et al. Bladder epithelial cell phosphate transporter inhibition protects mice against uropathogenic Escherichia coli infection. Cell reports 39 (2022). 10.1016/j.celrep.2022.110698 [DOI] [PubMed] [Google Scholar]
  • 65.Anderson GG et al. Intracellular bacterial biofilm-like pods in urinary tract infections. Science 301, 105–107 (2003). 10.1126/science.1084550 [DOI] [PubMed] [Google Scholar]
  • 66.Yang E et al. Isolation of Single Intracellular Bacterial Communities Generated from a Murine Model of Urinary Tract Infection for Downstream Single-cell Analysis. Journal of visualized experiments: JoVE (2019). 10.3791/58829 [DOI] [PubMed] [Google Scholar]
  • 67.Mysorekar IU & Hultgren SJ Mechanisms of uropathogenic Escherichia coli persistence and eradication from the urinary tract. Proceedings of the National Academy of Sciences of the United States of America 103, 14170–14175 (2006). 10.1073/pnas.0602136103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wang C, Symington JW & Mysorekar IU ATG16L1 and pathogenesis of urinary tract infections. Autophagy 8, 1693–1694 (2012). 10.4161/auto.21600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Joshi CS, Mora A, Felder PA & Mysorekar IU NRF2 promotes urothelial cell response to bacterial infection by regulating reactive oxygen species and RAB27B expression. Cell Reports 37, 109856 (2021). 10.1016/j.celrep.2021.109856 [DOI] [PubMed] [Google Scholar]
  • 70.Klein RD & Hultgren SJ Urinary tract infections: microbial pathogenesis, host–pathogen interactions and new treatment strategies. Nature Reviews Microbiology 18, 211–226 (2020). 10.1038/s41579-020-0324-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Gilbert NM, O’Brien VP & Lewis AL Transient microbiota exposures activate dormant Escherichia coli infection in the bladder and drive severe outcomes of recurrent disease. PLoS pathogens 13 (2017). 10.1371/journal.ppat.1006238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Munoz JA, Uhlemann AC & Barasch J Innate Bacteriostatic Mechanisms Defend the Urinary Tract. Annual review of physiology 84 (2022). 10.1146/annurev-physiol-052521-121810 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Miao Y, Wu J & Abraham SN Ubiquitination of Innate Immune Regulator TRAF3 Orchestrates Expulsion of Intracellular Bacteria by Exocyst Complex. Immunity 45, 94–105 (2016). 10.1016/j.immuni.2016.06.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Miao Y, Li G, Zhang X, Xu H & Abraham SN A TRP Channel Senses Lysosome Neutralization by Pathogens to Trigger Their Expulsion. Cell 161, 1306–1319 (2015). 10.1016/j.cell.2015.05.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Song J et al. Vol. 106 14966–14971 (Proc Natl Acad Sci U S A, 2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ireton K, Van Ngo H & Bhalla M Interaction of microbial pathogens with host exocytic pathways. Cellular microbiology 20 (2018). 10.1111/cmi.12861 [DOI] [PubMed] [Google Scholar]
  • 77.Miao Y et al. Collaboration between Distinct Rab Small GTPase Trafficking Circuits Mediates Bacterial Clearance from the Bladder Epithelium. Cell Host Microbe 22, 330–342 e334 (2017). 10.1016/j.chom.2017.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Joshi CS et al. D-mannose ameliorates age-associated cellular senescence in the bladder urothelium and NLRP3/Gasdermin/IL-1β -driven pyroptotic epithelial cell shedding. bioRkiv (pre-print) (2022). 10.1101/2022.09.06.506836 [DOI] [Google Scholar]
  • 79.Serafini-Cessi F, Malagolini N & Cavallone D Tamm-Horsfall glycoprotein: biology and clinical relevance. American journal of kidney diseases: the official journal of the National Kidney Foundation 42 (2003). 10.1016/s0272-6386(03)00829-1 [DOI] [PubMed] [Google Scholar]
  • 80.Pak J, Pu Y, Zhang ZT, Hasty DL & Wu XR Tamm-Horsfall protein binds to type 1 fimbriated Escherichia coli and prevents E. coli from binding to uroplakin Ia and Ib receptors. The Journal of biological chemistry 276 (2001). 10.1074/jbc.M008610200 [DOI] [PubMed] [Google Scholar]
  • 81.Bates JM et al. Tamm-Horsfall protein knockout mice are more prone to urinary tract infection: rapid communication. Kidney international 65 (2004). 10.1111/j.1523-1755.2004.00452.x [DOI] [PubMed] [Google Scholar]
  • 82.Lanne B et al. Glycoconjugate receptors for P-fimbriated Escherichia coli in the mouse. An animal model of urinary tract infection. The Journal of biological chemistry 270 (1995). 10.1074/jbc.270.15.9017 [DOI] [PubMed] [Google Scholar]
  • 83.Raffi HS, Bates JM, Laszik Z & Kumar S Tamm-horsfall protein protects against urinary tract infection by proteus mirabilis. The Journal of urology 181 (2009). 10.1016/j.juro.2009.01.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.James-Ellison MY, Roberts R, Verrier-Jones K, Williams JD & Topley N Mucosal immunity in the urinary tract: changes in sIgA, FSC and total IgA with age and in urinary tract infection. Clinical nephrology 48 (1997). [PubMed] [Google Scholar]
  • 85.Mestecky J & McGhee JR Immunoglobulin A (IgA): molecular and cellular interactions involved in IgA biosynthesis and immune response. Advances in immunology 40 (1987). 10.1016/s0065-2776(08)60240-0 [DOI] [PubMed] [Google Scholar]
  • 86.Corthésy B Role of secretory IgA in infection and maintenance of homeostasis. Autoimmunity reviews 12 (2013). 10.1016/j.autrev.2012.10.012 [DOI] [PubMed] [Google Scholar]
  • 87.Mathias A, Pais B, Favre L, Benyacoub J & Corthésy B Role of secretory IgA in the mucosal sensing of commensal bacteria. Gut microbes 5 (2014). 10.4161/19490976.2014.983763 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Mantis NJ, Rol N & Corthésy B Secretory IgA’s complex roles in immunity and mucosal homeostasis in the gut. Mucosal immunology 4 (2011). 10.1038/mi.2011.41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ligon MM et al. Single cell and tissue-transcriptomic analysis of murine bladders reveals age- and TNFalpha-dependent but microbiota-independent tertiary lymphoid tissue formation. Mucosal Immunol 13, 908–918 (2020). 10.1038/s41385-020-0290-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Radl J, Sepers JM, Skvaril F, Morell A & Hijmans W Immunoglobulin patterns in humans over 95 years of age. Clinical and experimental immunology 22 (1975). [PMC free article] [PubMed] [Google Scholar]
  • 91.Tietz NW, Shuey DF, Wekstein DR & Vol. 38 1167–1185 (1992). [PubMed] [Google Scholar]
  • 92.Thoreux K, Owen RL & Schmucker DL Intestinal lymphocyte number, migration and antibody secretion in young and old rats. Immunology 101, 161–167 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Bakun M et al. Urine proteomes of healthy aging humans reveal extracellular matrix (ECM) alterations and immune system dysfunction. Age (Dordr) 36 (2014). 10.1007/s11357-013-9562-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Sawhill JL et al. The Impact of Methenamine Hippurate Treatment on Urothelial Integrity and Bladder Inflammation in Aged Female Mice and Women With Urinary Tract Infections. Female pelvic medicine & reconstructive surgery 28 (2022). 10.1097/SPV.0000000000001185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lacerda Mariano L & Ingersoll MA The immune response to infection in the bladder. Nature Reviews Urology 17, 439–458 (2020). 10.1038/s41585-020-0350-8 [DOI] [PubMed] [Google Scholar]
  • 96.Truschel ST et al. Age-related endolysosome dysfunction in the rat urothelium. PloS One 13, e0198817 (2018). 10.1371/journal.pone.0198817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Chen Y et al. Rab27b is associated with fusiform vesicles and may be involved in targeting uroplakins to urothelial apical membranes. Proc Natl Acad Sci U S A 100, 14012–14017 (2003). 10.1073/pnas.2436350100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Guo X et al. Involvement of vps33a in the fusion of uroplakin-degrading multivesicular bodies with lysosomes. Traffic 10, 1350–1361 (2009). 10.1111/j.1600-0854.2009.00950.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kreft ME, Jezernik K, Kreft M & Romih R Apical plasma membrane traffic in superficial cells of bladder urothelium. Ann N Y Acad Sci 1152, 18–29 (2009). 10.1111/j.1749-6632.2008.04004.x [DOI] [PubMed] [Google Scholar]
  • 100.Behrends C, Sowa ME, Gygi SP & Harper JW Network organization of the human autophagy system. Nature 466, 68–76 (2010). 10.1038/nature09204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Klionsky DJ et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12, 1–222 (2016). 10.1080/15548627.2015.1100356 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Bauckman KA, Owusu-Boaitey N & Mysorekar IU Selective autophagy: xenophagy. Methods 75, 120–127 (2015). 10.1016/j.ymeth.2014.12.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Mysorekar IU & Hultgren SJ Mechanisms of uropathogenic Escherichia coli persistence and eradication from the urinary tract. Proc Natl Acad Sci U S A 103, 14170–14175 (2006). 10.1073/pnas.0602136103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wang C, Symington JW & Mysorekar IU ATG16L1 and pathogenesis of urinary tract infections. Autophagy 8, 1693–1694 (2012). 10.4161/auto.21600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wang C, Symington JW, Mysorekar IU & Vol. 8 1693–1694 (Autophagy, 2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Symington JW et al. Vol. 8 1388–1399 (Mucosal Immunol, 2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Bauckman KA & Mysorekar IU Ferritinophagy drives uropathogenic Escherichia coli persistence in bladder epithelial cells. Autophagy, 0 (2016). 10.1080/15548627.2016.1160176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Cannizzo ES et al. Age-related oxidative stress compromises endosomal proteostasis. Cell reports 2 (2012). 10.1016/j.celrep.2012.06.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wang C et al. Vol. 16 532–542 (Nat Neurosci, 2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Zecchini S et al. Autophagy controls neonatal myogenesis by regulating the GH-IGF1 system through a NFE2L2- and DDIT3-mediated mechanism. Autophagy 15 (2019). 10.1080/15548627.2018.1507439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ho TT et al. Autophagy maintains the metabolism and function of young and old stem cells. Nature 543, 205–210 (2017). 10.1038/nature21388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.de Rijk MM et al. Aging-Associated Changes in Oxidative Stress Negatively Impacts the Urinary Bladder Urothelium. Int Neurourol J 26, 111–118 (2022). 10.5213/inj.2142224.112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Perše M, Injac R & Erman A Oxidative status and lipofuscin accumulation in urothelial cells of bladder in aging mice. PloS one 8 (2013). 10.1371/journal.pone.0059638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Aiello A et al. Vol. 10 (Front Immunol, 2019). [Google Scholar]
  • 115.Fulop T et al. Immunosenescence and Inflamm-Aging As Two Sides of the Same Coin: Friends or Foes? Front Immunol 8, 1960 (2017). 10.3389/fimmu.2017.01960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Mora-Bau G et al. Macrophages Subvert Adaptive Immunity to Urinary Tract Infection. PLoS Pathog 11, e1005044 (2015). 10.1371/journal.ppat.1005044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Thumbikat P, Waltenbaugh C, Schaeffer AJ, Klumpp DJ & Vol. 176 3080–3086 (J Immunol, 2006). [DOI] [PubMed] [Google Scholar]
  • 118.O’Brien VP et al. A mucosal imprint left by prior Escherichia coli bladder infection sensitizes to recurrent disease. Nat Microbiol 2, 16196 (2016). 10.1038/nmicrobiol.2016.196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sivick KE, Schaller MA, Smith SN, Mobley HLT & Vol. 184 2065–2075 (J Urol, 2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Riding AM et al. Group 3 innate lymphocytes make a distinct contribution to type 17 immunity in bladder defence. iScience 25 (2022). 10.1016/j.isci.2022.104660 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Spencer JD, Schwaderer AL, Becknell B, Watson J & Hains DS The innate immune response during urinary tract infection and pyelonephritis. Pediatric nephrology (Berlin, Germany) 29 (2014). 10.1007/s00467-013-2513-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Nickel JC, Saz-Leal P & Doiron RC Could sublingual vaccination be a viable option for the prevention of recurrent urinary tract infection in Canada? A systematic review of the current literature and plans for the future. Canadian Urological Association journal = Journal de l’Association des urologues du Canada 14 (2020). 10.5489/cuaj.6690 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Forsyth VS et al. Optimization of an Experimental Vaccine To Prevent Escherichia coli Urinary Tract Infection. mBio 11 (2020). 10.1128/mBio.00555-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Ramírez Sevilla C, Gómez Lanza E, Manzanera JL, Martín JAR & Sanz MAB Active immunoprophyilaxis with uromune® decreases the recurrence of urinary tract infections at three and six months after treatment without relevant secondary effects. BMC infectious diseases 19 (2019). 10.1186/s12879-019-4541-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Girard J-P, Moussion C & Förster R HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nature Reviews Immunology 12, 762–773 (2012). 10.1038/nri3298 [DOI] [PubMed] [Google Scholar]
  • 126.Pitzalis C, Jones GW, Bombardieri M & Jones SA Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat Rev Immunol 14, 447–462 (2014). 10.1038/nri3700 [DOI] [PubMed] [Google Scholar]
  • 127.Jones GW & Jones SA Ectopic lymphoid follicles: inducible centres for generating antigen-specific immune responses within tissues. Immunology 147, 141–151 (2016). 10.1111/imm.12554 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Jones GW, Hill DG & Jones SA Understanding Immune Cells in Tertiary Lymphoid Organ Development: It Is All Starting to Come Together. Front Immunol 7, 401 (2016). 10.3389/fimmu.2016.00401 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Drayton DL, Liao S, Mounzer RH & Ruddle NH Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol 7, 344–353 (2006). 10.1038/ni1330 [DOI] [PubMed] [Google Scholar]
  • 130.Eddens T et al. Pneumocystis-Driven Inducible Bronchus-Associated Lymphoid Tissue Formation Requires Th2 and Th17 Immunity. Cell Rep 18, 3078–3090 (2017). 10.1016/j.celrep.2017.03.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Khader SA et al. IL-23 is required for long-term control of Mycobacterium tuberculosis and B cell follicle formation in the infected lung. J Immunol 187, 5402–5407 (2011). 10.4049/jimmunol.1101377 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Rangel-Moreno J et al. Inducible bronchus-associated lymphoid tissue (iBALT) in patients with pulmonary complications of rheumatoid arthritis. J Clin Invest 116, 3183–3194 (2006). 10.1172/JCI28756 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Rangel-Moreno J et al. The development of inducible bronchus-associated lymphoid tissue depends on IL-17. Nat Immunol 12, 639–646 (2011). 10.1038/ni.2053 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Chang A et al. In situ B cell-mediated immune responses and tubulointerstitial inflammation in human lupus nephritis. J Immunol 186, 1849–1860 (2011). 10.4049/jimmunol.1001983 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Huang Y et al. Identification of novel genes associated with renal tertiary lymphoid organ formation in aging mice. PLoS One 9, e91850 (2014). 10.1371/journal.pone.0091850 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Sato Y et al. Heterogeneous fibroblasts underlie age-dependent tertiary lymphoid tissues in the kidney. JCI Insight 1, e87680 (2016). 10.1172/jci.insight.87680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.McDonald KG et al. CC chemokine receptor 6 expression by B lymphocytes is essential for the development of isolated lymphoid follicles. Am J Pathol 170, 1229–1240 (2007). 10.2353/ajpath.2007.060817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Wang C, McDonald KG, McDonough JS, Newberry RD & Murine isolated lymphoid follicles contain follicular B lymphocytes with a mucosal phenotype. 291 (2006). 10.1152/ajpgi.00525.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wang C, McDonough JS, McDonald KG, Huang C & Newberry RD Alpha4beta7/MAdCAM-1 interactions play an essential role in transitioning cryptopatches into isolated lymphoid follicles and a nonessential role in cryptopatch formation. J Immunol 181, 4052–4061 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Wang C et al. Effect of α4β7 blockade on intestinal lymphocyte subsets and lymphoid tissue development. Inflammatory bowel diseases 16, 1751–1762 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Serafini B et al. Detection of ectopic B-cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis. Brain Pathol 14, 164–174 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Sato Y et al. Heterogeneous fibroblasts underlie age-dependent tertiary lymphoid tissues in the kidney. JCI Insight 1 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Singh P et al. Lymphoid neogenesis and immune infiltration in aged liver. Hepatology 47, 1680–1690 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.McDonald KG, Leach MR, Huang C, Wang C & Newberry RD Aging impacts isolated lymphoid follicle development and function. Immun Ageing 8, 1 (2011). 10.1186/1742-4933-8-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Hannan TJ, Mysorekar IU, Hung CS, Isaacson-Schmid ML & Hultgren SJ Early severe inflammatory responses to uropathogenic E. coli predispose to chronic and recurrent urinary tract infection. PLoS Pathog 6, e1001042 (2010). 10.1371/journal.ppat.1001042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yu L et al. Vol. 8 (Elife, 2019). [Google Scholar]
  • 147.Wang X et al. Overexpression of cyclooxygenase-2 (COX-2) in the mouse urinary bladder induces the expression of immune- and cell proliferation-related genes. Mol Carcinog 48, 1–13 (2009). 10.1002/mc.20449 [DOI] [PubMed] [Google Scholar]
  • 148.Phillips JI Inflammatory plasma cell infiltration of the urinary bladder in the aging C57BL/Icrfa(t) mouse. Invest Urol 19, 75–78 (1981). [PubMed] [Google Scholar]
  • 149.Koti M et al. Tertiary Lymphoid Structures Associate with Tumour Stage in Urothelial Bladder Cancer. Bladder Cancer 3, 259–267 (2017). 10.3233/BLC-170120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Ligon MM et al. Bladder mucosal cystitis cystica lesions are tertiary lymphoid tissues that correlate with recurrent UTI frequency in postmenopausal women. J Urology (In press) (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hamade A et al. Sex differences in the aging murine urinary bladder and influence on the tumor immune microenvironment of a carcinogen-induced model of bladder cancer. Biol Sex Differ 13, 19 (2022). 10.1186/s13293-022-00428-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Menees KB et al. Sex- and age-dependent alterations of splenic immune cell profile and NK cell phenotypes and function in C57BL/6J mice. Immunity & ageing: I & A 18 (2021). 10.1186/s12979-021-00214-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Evans I & Kim MY Involvement of lymphoid inducer cells in the development of secondary and tertiary lymphoid structure. BMB Rep 42, 189–193 (2009). [DOI] [PubMed] [Google Scholar]
  • 154.Shen M et al. Lymphotoxin β receptor activation promotes bladder cancer in a nuclear factor-κB-dependent manner. Molecular medicine reports 11 (2015). 10.3892/mmr.2014.2826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.De Nisco NJ 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 431, 4368–4379 (2019). 10.1016/j.jmb.2019.04.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Robinson D, Toozs-Hobson P & Cardozo L The effect of hormones on the lower urinary tract. Menopause international 19 (2013). 10.1177/1754045313511398 [DOI] [PubMed] [Google Scholar]
  • 157.Krause M, Wheeler TL 2nd, Snyder TE & Richter HE Local Effects of Vaginally Administered Estrogen Therapy: A Review. J Pelvic Med Surg 15, 105–114 (2009). 10.1097/SPV.0b013e3181ab4804 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Vrljicak K et al. Positive feedback loop for cystitis cystica: the effect of recurrent urinary tract infection on the number of bladder wall mucosa nodules. Acta Clin Croat 52, 444–447 (2013). [PubMed] [Google Scholar]
  • 159.Milosevic D et al. Cystitis cystica and recurrent urinary tract infections in children. Coll Antropol 34, 893–897 (2010). [PubMed] [Google Scholar]
  • 160.Imamov O et al. Estrogen receptor beta-deficient female mice develop a bladder phenotype resembling human interstitial cystitis. Proceedings of the National Academy of Sciences of the United States of America 104 (2007). 10.1073/pnas.0703410104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Jung C & Brubaker L The etiology and management of recurrent urinary tract infections in postmenopausal women. Climacteric: the journal of the International Menopause Society 22 (2019). 10.1080/13697137.2018.1551871 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Mody L & Juthani-Mehta M Urinary tract infections in older women: a clinical review. JAMA 311, 844–854 (2014). 10.1001/jama.2014.303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Ferrante KL, Wasenda EJ, Jung CE, Adams-Piper ER & Lukacz ES Vaginal Estrogen for the Prevention of Recurrent Urinary Tract Infection in Postmenopausal Women: A Randomized Clinical Trial. Female Pelvic Med Reconstr Surg 27, 112–117 (2021). 10.1097/spv.0000000000000749 [DOI] [PubMed] [Google Scholar]
  • 164.Lawson WG Chronic urinary tract infection in a female child: Recovery after vaginal estrogen therapy. The Journal of Pediatrics 43, 263–266 (1953). 10.1016/S0022-3476(53)80397-5 [DOI] [PubMed] [Google Scholar]
  • 165.Fait T Menopause hormone therapy: latest developments and clinical practice. Drugs Context 8, 212551–212551 (2019). 10.7573/dic.212551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Perrotta C, Aznar M, Mejia R, Albert X & Ng CW Oestrogens for preventing recurrent urinary tract infection in postmenopausal women. Obstet Gynecol 112, 689–690 (2008). 10.1097/AOG.0b013e318185f7a5 [DOI] [PubMed] [Google Scholar]
  • 167.Thomas-White K et al. Vaginal estrogen therapy is associated with increased Lactobacillus in the urine of postmenopausal women with overactive bladder symptoms. American journal of obstetrics and gynecology 223 (2020). 10.1016/j.ajog.2020.08.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Šimunić V et al. Local estrogen treatment in patients with urogenital symptoms. International Journal of Gynecology & Obstetrics 82, 187–197 (2003). 10.1016/S0020-7292(03)00200-5 [DOI] [PubMed] [Google Scholar]
  • 169.Ammitzbøll N et al. Pre- and postmenopausal women have different core urinary microbiota. Scientific Reports 11, 1–10 (2021). 10.1038/s41598-021-81790-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Gustafsson RJ et al. The Lactobacillus flora in vagina and rectum of fertile and postmenopausal healthy Swedish women. BMC Women’s Health 11, 1–8 (2011). 10.1186/1472-6874-11-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Neugent ML et al. Recurrent urinary tract infection and estrogen shape the taxonomic ecology and function of the postmenopausal urogenital microbiome. Cell reports. Medicine 3 (2022). 10.1016/j.xcrm.2022.100753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Mysorekar IU, Mulvey MA, Hultgren SJ & Gordon JI Molecular regulation of urothelial renewal and host defenses during infection with uropathogenic Escherichia coli. J Biol Chem 277, 7412–7419 (2002). 10.1074/jbc.M110560200 [DOI] [PubMed] [Google Scholar]
  • 173.Olson PD, Hruska KA & Hunstad DA Androgens Enhance Male Urinary Tract Infection Severity in a New Model. J Am Soc Nephrol 27, 1625–1634 (2016). 10.1681/ASN.2015030327 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Olson PD et al. Androgen exposure potentiates formation of intratubular communities and renal abscesses by Escherichia coli. Kidney Int 94, 502–513 (2018). 10.1016/j.kint.2018.04.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Ho CH et al. Testosterone suppresses uropathogenic Escherichia coli invasion and colonization within prostate cells and inhibits inflammatory responses through JAK/STAT-1 signaling pathway. PloS one 12 (2017). 10.1371/journal.pone.0180244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Hao Y, O’Neill P, Naradikian MS, Scholz JL & Cancro MP A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice. Blood 118, 1294–1304 (2011). 10.1182/blood-2011-01-330530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Rubtsov AV et al. Toll-like receptor 7 (TLR7)-driven accumulation of a novel CD11c(+) B-cell population is important for the development of autoimmunity. Blood 118, 1305–1315 (2011). 10.1182/blood-2011-01-331462 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Naber KG Treatment options for acute uncomplicated cystitis in adults. The Journal of antimicrobial chemotherapy 46 Suppl 1 (2000). [PubMed] [Google Scholar]
  • 179.Ragnarsdóttir B & Svanborg C Susceptibility to acute pyelonephritis or asymptomatic bacteriuria: host-pathogen interaction in urinary tract infections. Pediatric nephrology (Berlin, Germany) 27 (2012). 10.1007/s00467-011-2089-1 [DOI] [PubMed] [Google Scholar]
  • 180.Tan CW & Chlebicki MP Urinary tract infections in adults. Singapore Med J 485–490 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Kranjcec B, Papes D & Altarac S D-mannose powder for prophylaxis of recurrent urinary tract infections in women: a randomized clinical trial. World J Urol 32, 79–84 (2014). 10.1007/s00345-013-1091-6 [DOI] [PubMed] [Google Scholar]
  • 182.Chiu K, Zhang F, Sutcliffe S, Mysorekar IU & Lowder JL Recurrent Urinary Tract Infection Incidence Rates Decrease in Women With Cystitis Cystica After Treatment With d-Mannose: A Cohort Study. Female Pelvic Med Reconstr Surg 28, e62–e65 (2022). 10.1097/SPV.0000000000001144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Snellings MS et al. Effectiveness of Methenamine for UTI Prevention in Older Adults. The Annals of pharmacotherapy 54 (2020). 10.1177/1060028019886308 [DOI] [PubMed] [Google Scholar]
  • 184.Hollyer I, Varias F, Ho B & Ison MG Safety and efficacy of methenamine hippurate for the prevention of recurrent urinary tract infections in adult renal transplant recipients: A single center, retrospective study. Transplant infectious disease: an official journal of the Transplantation Society 21 (2019). 10.1111/tid.13063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Crivelli JJ, Alhalabi F & Zimmern PE Electrofulguration in the advanced management of antibiotic-refractory recurrent urinary tract infections in women. Int J Urol 26, 662–668 (2019). 10.1111/iju.13963 [DOI] [PubMed] [Google Scholar]
  • 186.Malley SE & Vizzard MA Changes in urinary bladder cytokine mRNA and protein after cyclophosphamide-induced cystitis. Physiological genomics 9 (2002). 10.1152/physiolgenomics.00117.2001 [DOI] [PubMed] [Google Scholar]
  • 187.Armbruster CE, Smith SN, Mody L & Mobley HLT Urine Cytokine and Chemokine Levels Predict Urinary Tract Infection Severity Independent of Uropathogen, Urine Bacterial Burden, Host Genetics, and Host Age. Infection and immunity 86 (2018). 10.1128/IAI.00327-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Sheu JN et al. Serum and urine levels of interleukin-6 and interleukin-8 in children with acute pyelonephritis. Cytokine 36 (2006). 10.1016/j.cyto.2007.02.006 [DOI] [PubMed] [Google Scholar]
  • 189.Sheu JN et al. Relationship between serum and urine interleukin-6 elevations and renal scarring in children with acute pyelonephritis. Scandinavian journal of urology and nephrology 43 (2009). 10.1080/00365590802478742 [DOI] [PubMed] [Google Scholar]
  • 190.Tan CK et al. Genome-wide mapping of cystitis due to Streptococcus agalactiae and Escherichia coli in mice identifies a unique bladder transcriptome that signifies pathogen-specific antimicrobial defense against urinary tract infection. Infection and immunity 80 (2012). 10.1128/IAI.00023-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Oregioni O et al. Urinary interleukin-8 is elevated in urinary tract infections independently of the causative germs. Cytokine 31 (2005). 10.1016/j.cyto.2005.06.009 [DOI] [PubMed] [Google Scholar]
  • 192.Tyagi P et al. Elevated CXC chemokines in urine noninvasively discriminate OAB from UTI. American journal of physiology. Renal physiology 311 (2016). 10.1152/ajprenal.00213.2016 [DOI] [PubMed] [Google Scholar]
  • 193.Davidoff R, Yamaguchi R, Leach GE, Park E & Lad PM Multiple urinary cytokine levels of bacterial cystitis. The Journal of urology 157 (1997). [PubMed] [Google Scholar]
  • 194.Ghoniem G et al. Differential profile analysis of urinary cytokines in patients with overactive bladder. International urogynecology journal 22 (2011). 10.1007/s00192-011-1401-8 [DOI] [PubMed] [Google Scholar]
  • 195.Alkis O, Zumrutbas AE, Toktas C, Aybek H & Aybek Z The use of biomarkers in the diagnosis and treatment of overactive bladder: Can we predict the patients who will be resistant to treatment? Neurourology and urodynamics 36 (2017). 10.1002/nau.22939 [DOI] [PubMed] [Google Scholar]
  • 196.Rodhe N, Löfgren S, Strindhall J, Matussek A & Mölstad S Cytokines in urine in elderly subjects with acute cystitis and asymptomatic bacteriuria. Scandinavian journal of primary health care 27 (2009). 10.1080/02813430902757634 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Sundac L et al. Protein-based profiling of the immune response to uropathogenic Escherichia coli in adult patients immediately following hospital admission for acute cystitis. Pathogens and disease 74 (2016). 10.1093/femspd/ftw062 [DOI] [PubMed] [Google Scholar]
  • 198.Furuta A et al. Comparison of inflammatory urine markers in patients with interstitial cystitis and overactive bladder. International urogynecology journal 29 (2018). 10.1007/s00192-017-3547-5 [DOI] [PubMed] [Google Scholar]
  • 199.Gill K, Horsley H, Swamy S, Khasriya R & Malone-Lee J A prospective observational study of urinary cytokines and inflammatory response in patients with Overactive Bladder Syndrome. BMC urology 21 (2021). 10.1186/s12894-021-00809-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Czaja CA et al. Prospective cohort study of microbial and inflammatory events immediately preceding Escherichia coli recurrent urinary tract infection in women. The Journal of infectious diseases 200 (2009). 10.1086/600385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Jhang JF, Ho HC, Hsu YH, Jiang YH & Kuo HC Bladder Ultrastructure and Urinary Cytokine Abnormality in Patients with Recurrent Urinary Tract Infection and the Changes after Intravesical Platelet-Rich Plasma Injections. Biomedicines 10 (2022). 10.3390/biomedicines10020245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Tseng-Rogenski S & Liebert M Interleukin-8 is essential for normal urothelial cell survival. American journal of physiology. Renal physiology 297 (2009). 10.1152/ajprenal.90733.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Erickson DR et al. Do the National Institute of Diabetes and Digestive and Kidney Diseases cystoscopic criteria associate with other clinical and objective features of interstitial cystitis? The Journal of urology 173 (2005). 10.1097/01.ju.0000146466.71311.ab [DOI] [PubMed] [Google Scholar]
  • 204.Jiang YH et al. Urine cytokines as biomarkers for diagnosing interstitial cystitis/bladder pain syndrome and mapping its clinical characteristics. American journal of physiology. Renal physiology 318 (2020). 10.1152/ajprenal.00051.2020 [DOI] [PubMed] [Google Scholar]
  • 205.Hedges S & Svanborg C The mucosal cytokine response to urinary tract infections. International journal of antimicrobial agents 4 (1994). 10.1016/0924-8579(94)90039-6 [DOI] [PubMed] [Google Scholar]

RESOURCES