Next Article in Journal
Importance of B Lymphocytes and the IgG-Binding Protein Sbi in Staphylococcus aureus Skin Infection
Next Article in Special Issue
Genetic Variations in Vesicoureteral Reflux Sequelae
Previous Article in Journal
Acknowledgement to Reviewers of Pathogens in 2015
Previous Article in Special Issue
The Global Prevalence of Infections in Urology Study: A Long-Term, Worldwide Surveillance Study on Urological Infections
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Strategies in the Prevention and Treatment of Urinary Tract Infections

Department of Microbiology, Tumor and Cell Biology, Division of Clinical Microbiology, Karolinska Institutet and Karolinska University Hospital, Stockholm SE-171 76, Sweden
*
Author to whom correspondence should be addressed.
Current affiliation: Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet and Karolinska University Hospital, Stockholm SE-141 86, Sweden
Pathogens 2016, 5(1), 13; https://doi.org/10.3390/pathogens5010013
Submission received: 16 July 2015 / Revised: 5 August 2015 / Accepted: 21 January 2016 / Published: 27 January 2016
(This article belongs to the Special Issue Molecular Aspects of Urinary Tract Infection)

Abstract

:
Urinary tract infections are one of the most common bacterial infections, especially in women and children, frequently treated with antibiotics. The alarming increase in antibiotic resistance is a global threat to future treatment of infections. Therefore, alternative strategies are urgently needed. The innate immune system plays a fundamental role in protecting the urinary tract from infections. Antimicrobial peptides form an important part of the innate immunity. They are produced by epithelial cells and neutrophils and defend the urinary tract against invading bacteria. Since efficient resistance mechanisms have not evolved among bacterial pathogens, much effort has been put into exploring the role of antimicrobial peptides and possibilities to utilize them in clinical practice. Here, we describe the impact of antimicrobial peptides in the urinary tract and ways to enhance the production by hormones like vitamin D and estrogen. We also discuss the potential of medicinal herbs to be used in the prophylaxis and the treatment of urinary tract infections.

1. Introduction

Urinary tract infections belong to the most common infections worldwide; more than 50% of women will experience at least one UTI during her lifetime [1]. Even though often self-limiting and seldom developing into more severe infections, cystitis causes considerable costs for the patient and the public health system. Moreover, in a substantial number of women, infections will recur several times during the year. While in adults, in the absence of pre-disposing factors, women are several times more often affected than men [2], the gender distribution among children varies with age, with boys being most susceptible as infants below 12 months of age [3,4,5]. The cumulative incidence during the first six years of life however is almost 7% in girls and close to 2% in boys [6]. Most infections have good prognosis, but the risk for renal scarring might be as high as 40%, largely dependent on the diagnostic method used [7,8,9,10]. Similar to UTI in women, there is a risk for recurrence in 25%–40% of children with UTI [11,12].
Acute infections are commonly treated with antibiotics, and antimicrobial prophylaxis might be considered for women suffering from recurrent infections. The most common uropathogen is Escherichia coli (E. coli), accounting for >80% of uncomplicated UTIs. While highly resistant strains are yet not common in countries with relatively strict antibiotic regimes, multi-drug resistance and extended-beta-lactamase (ESBL) resistance is frequently detected in uropathogenic E. coli (UPEC) isolated in countries with less controlled antibiotic usage [13,14].
Even though bacteria have evolved mechanisms to resist the antimicrobial effect of endogenous antibiotics [15,16], resistance to these antimicrobial peptides (AMPs) has not effectively developed. The classical AMPs have common features, as they are small polypeptides of a few amino acids, are cationic and amphipathic. The highly positive charge explains the relatively selective action on bacterial membranes, which are more negatively charged compared to mammalian cell membranes [17]. The interaction with the peptides leads to membrane disruption and eventually lysis of the cell. However, in addition to the direct antibacterial activity, AMPs exhibit alternative functions supporting the immune system [18]. This might be immuno-modulatory properties [19] or actions on cell differentiation and tissue remodeling, which ultimately strengthen mechanical defense of epithelial barriers [20,21,22]. This wide range of actions together with the apparently low risk for resistance development make AMPs interesting mediators for novel treatment strategies. In this review, AMPs in the urinary tract will be presented, and different strategies to enhance innate immunity by AMPs or other epithelial-located mechanisms will be discussed.

2. Recurrent UTI

While the acute infection is self-limiting within a few days and is still easily treated with antibiotics, in a considerable number of women, the infection may recur within weeks or months. Recurrent UTI is defined by two or more infections within six months or at least three infections during a year. The recurring infection is often caused by the same bacterial strain as a previous infection, indicating that the bacterium is persisting in the body despite treatment with a suitable antibiotic [23]. Both the perianal and the vaginal flora serve as reservoirs for infections of the urinary tract [24,25,26]; however, E. coli appears to persist even in the bladder tissue. When entering the bladder, E. coli adheres to the bladder epithelium, and a small proportion of bacteria invades the cells. In this niche, protected from mechanical and cellular immune defense, bacteria multiply rapidly to form so-called intracellular bacterial communities (IBCs) [27,28]. As part of the defense mechanism, infected cells undergo apoptosis, lose contact with the neighboring cells and are shed and excreted with the urine [29,30]. Bacteria released from IBCs can therefore invade cells of deeper layers, which became exposed upon exfoliation of the outer layer of umbrella cells [28]. Within these cells, bacteria do not multiply, but instead form small dormant colonies, referred to as quiescent intracellular reservoirs (QIRs). While the signals inducing re-activation of these bacteria are not fully elucidated yet [31], QIRs are considered the source of recurrent infections [32,33]. Even though this model is based on studies in mice, there is an indication that a similar infection cycle takes place in women [34,35]. IBCs have also been detected in the urine of children with recurrent UTI [36,37]. However, especially in this patient group, other pathomechanisms, in particular vesicoureteral reflux [38,39] and genetic predisposition (reviewed in [40]), have to be considered.
Intravesical bacterial persistence explains the relative ineffectiveness of antimicrobials to prevent recurrence of the infection, since intracellular and metabolically-inactive bacteria make a difficult target for conventional antibiotics and the body’s own defense. Moreover, these intracellular bacteria reside in biofilm-like organizations and express many factors commonly associated with biofilm formation on abiotic surfaces in vitro. The non-fimbrial adhesin antigen 43 (Ag43) has been detected in IBCs in mice [27], and a particular variant of this protein, Ag43aCFT073, was shown to promote persistence in the mouse urinary tract [41]. In a collection of E. coli from children with recurrent UTIs, the presence of agn43 was linked to persisting strains [42]. Infections with strains carrying the gene coding for Ag43aCFT073 in particular caused a lower immune response in these children (as determined by urinary IL-8 levels) compared to strains lacking this gene. This finding might indicate that immune evasion, either during direct contact with the host cell or by more efficient formation of biofilm intracellularly, promotes the persistence of these strains. The major E. coli biofilm components curli and cellulose contribute similarly to the persistence of UPEC in the urinary bladder. In women suffering from recurrent UTIs, persisting strains were more likely to express both factors than strains causing sporadic infections [25]. In a mouse UTI model, reduced immune induction by strains producing cellulose promoted their persistence in the tissue, while curli fimbriae supported early virulence of the strain by mediating adherence [43].
While treatment with conventional antimicrobial agents is effective against acute UTI, it does not prevent the recurrence of the infection, because intracellular, dormant bacteria are not eradicated. Long-term prophylaxis with low doses of antibiotics might efficiently inhibit the emergence of intravesical bacteria during the treatment period [44,45], but infections are likely to re-occur when the prophylactic treatment is terminated [46,47]. Therefore, alternative prophylactic and therapeutic strategies are needed, especially for women suffering from recurrent UTI [48].

3. Antimicrobial Peptides in the Protection of the Urinary Tract

Antimicrobial peptides are produced by immune cells, but also expressed by epithelial cells throughout the body. They confer instant protection by their antimicrobial activity against a wide range of pathogens, but contribute also by immune-modulatory properties or alternative actions on the pathogens. Several AMPs have been described in the lower and upper urinary tract [49,50]. We have previously demonstrated the importance of the human cathelicidin LL-37/hCAP18 for the protection of the urinary tract [51], and recent research has established a major contribution of ribonuclease (RNase) 7 to the urinary defense against uropathogens [52,53]. The expression of human β-defensins (hBDs) in the urogenital tract is well established [54,55], but their role during UTI is only incompletely understood. hBD1 is constitutively expressed in various epithelia throughout the body [56], with high expression levels in the kidney [57] and lower expression in epithelial cells of the urinary bladder [58]. Despite anti-E. coli activity of the AMP in urine [57], hBD1 deficiency in mice was not accompanied with increased bacterial burden in bladder or kidneys [58]. Studies with human patients suggest that hBD1 might play a role during acute pyelonephritis [59], while its role in the defense of the lower urinary tract is less certain [60]. In contrast to the constitutive expression pattern of hBD1, hBD2 is expressed only in response to inflammatory stimuli [61] and has been detected in chronically-infected kidneys [62].
Another group of defensins, the α-defensins, have mainly been investigated in the intestinal tract. Recently, however, the presence and activity of the human α-defensin 5 has been demonstrated also in the urinary tract [63]. Other peptides inhibit bacteria by nutrient depletion, most importantly iron. Hepcidin is a major regulator of iron homeostasis, but may also exhibit direct antibacterial activity against E. coli [64]. Lipocalin 2 and lactoferrin influence bacterial growth by depletion of essential metals, such as iron. Both proteins are stored in neutrophil granules, but are also expressed by epithelia of the urinary tract, primarily in the kidney [65]. Renal lipocalin 2 is upregulated upon infection, secreted with the urine and, thus, contributes to the defense also of the lower urinary tract [66,67]. A similar antibacterial mechanism has been suggested for the S100 protein psoriasin via depletion of zinc [68]. Despite its pronounced activity against E. coli and documented expression in urinary tissues [69,70], the contribution of psoriasin to the urinary defense has not been investigated. Finally, the Tamm-Horsfall protein, or uromodulin, a renal-specific protein, which is secreted with the urine at high concentrations, fends off E. coli and other pathogens by interfering with the adhesion to the host cells of the lower urinary tract [71]. This review will mainly focus on the human cathelicidin LL-37/hCAP18, its role in urinary protection and its interaction with E. coli.

3.1. The Role of Cathelicidin in the Urinary Tract

Similar to other AMPs, the human cathelicidin is produced as an inactive precursor, containing a signal peptide and the propeptide hCAP18 [72,73]. After cleavage, the antimicrobially-active peptide consists of 37 amino acids starting with two lysin residues, therefore referred to as LL-37. Further cleavage products with distinct antimicrobial activities might be generated at different sites of the body [74,75,76]. In mice, the homologues peptide is referred to as CRAMP, cathelicidin-related AMP [77]. Since much of our knowledge is based on experiments in mice and analyses of mouse tissues, the precise role of LL-37/hCAP18 in the human urinary tract can only be estimated.
The sensitivity of E. coli strains isolated from the urinary tract differs. Interestingly, strains isolated from more severe infections, i.e., pyelonephritis, exhibit decreased sensitivity to LL-37 in vitro compared to less invasive strains isolated from cases of cystitis [51]. Several factors expressed by UPEC interfere with the bactericidal action of LL-37. Curli fimbriae (adhesins and biofilm component of E. coli) bind to LL-37 and thus prevent interaction with the bacterial cell membrane ([43], discussed below). Biofilm formation per se might moreover hamper LL-37 penetration and thus impair its action also in a widely unspecific manner. The outer membrane protease OmpT has been shown to contribute to resistance against LL-37 in enteric E. coli pathotypes by degrading the peptide [78]. The UPEC strain CFT073 also expresses OmpT with proteolytic activity against LL-37; the contribution to protection against LL-37 is however less clear [79]. In various Gram-negative bacteria, modification of the outer membrane has been implicated in protection against LL-37 [16]. Changes in the lipid A moiety of the lipopolysaccharide (LPS) can partly neutralize the negative charges and thus renders the surface less susceptible for binding cationic AMPs [80]. As part of the immune-modulatory function of LL-37 is based on its LPS-neutralizing effect [81], a correlating function of LPS in the protection of E. coli against this AMP may be anticipated. However, in E. coli, this has not been studied in detail.
LL-37/hCAP18 is expressed throughout the urinary tract. Epithelial cells of the lower and upper urinary tract express the peptide constitutively at low levels. Higher levels are produced in response to bacteria and by neutrophils recruited during infection, as reflected by increased urinary levels in patients with UTI compared to healthy controls [51,60]. In a mouse model of ascending pyelonephritis, morbidity and mortality were higher in animals deficient for CRAMP compared to wild-type, with higher bacterial titers both in the kidneys and the bladder [51]. Experiments with neutrophil-depleted mice demonstrated that epithelial-derived LL-37 influenced morbidity and bacterial load in the bladder, while neutrophil-derived LL-37 reduced renal bacteria and mortality and inhibited systemic spread of the infection. Neutrophilic LL-37 contributes to intracellular killing of bacteria after phagocytosis, but is also part of neutrophil extracellular traps (NETs), a net of fibers consisting of chromatin and granular proteins released from neutrophils [82]. Rather than killing entrapped pathogens directly, LL-37 appears to promote NET formation and increases NET stability [83,84]. The importance of NETs and the role of LL-37 in this context have not been investigated in the urinary tract so far.
In a mouse strain resistant to vesicoureteral reflux, CRAMP deficiency did not result in higher bacterial titers [85]. Instead, the pro-inflammatory activity of CRAMP resulted in more severe tissue damage and higher bacterial loads in wild-type mice during acute infection. Similar observations were made when LL-37 was directly instilled in the urinary bladder of mice [86,87]. As indicated above, curli fimbriae trap LL-37, preventing it from exerting its bactericidal activity at the bacterial outer membrane [43]. While the non-curliated pyelonephritis strain E. coli CFT073 was used in the first study by Chromek et al. [51], the curliated cystitis strain UTI89 was used in the study by Danka et al. [85]. Due to its curli fimbriae, this strain may be less sensitive to LL-37 (or CRAMP, respectively). Bacterial survival in the bladder might thus not be that much affected by this AMP; instead, the pro-inflammatory activity of CRAMP may be dominating the histopathological picture. In co-infection experiments with isogenic E. coli strains, a curli-expressing wild-type strain was recovered in higher numbers compared to a biofilm-deficient strain lacking curli fimbriae [88]. While this might primarily be due to a deficiency in the formation of IBCs, increased sensitivity of the non-curliated strain towards LL-37 may contribute.
Neutrophil-derived CRAMP was of major importance in the protection of the upper urinary tract and systemic spread of the infection [51]. The role of CRAMP in the acute protection of the urinary bladder is less evident and might to larger extent depend on the infecting strain [51,85]. It can also not be ruled out that CRAMP has an impact on long-term intracellular bacterial persistence and thus recurrence of infections. Lower baseline levels of urinary LL-37 in women who had had a UTI compared to baseline levels from women who never had one indicate that CAMP plays a role in the protection of the lower urinary tract [60].

3.2. Interaction of LL-37 with Bacterial Biofilm

At sub-inhibitory concentrations, LL-37 has been shown to influence biofilm formation of different bacterial species [43,89,90,91,92,93,94], including species with potential relevance in the urinary tract. The direct interaction of LL-37 with microbial surface structures may be responsible in some cases [43,94], including the opportunistic fungal pathogen Candida albicans [95]. In Pseudomonas aeruginosa, LL-37 reduced biofilm formation by impacting quorum sensing systems, leading to decreased bacterial attachment and increased twitching motility [89].
In E. coli, curli-dependent biofilm is inhibited by LL-37 through direct interaction of the AMP with CsgA, the major subunit of curli fibers [43]. By binding of LL-37 to monomeric CsgA, it inhibits the polymerization and thereby the curli formation and ultimately reduces biofilm formation. However, binding of LL-37 to already established curli fimbriae at the bacterial surface neutralizes the bactericidal effect of the peptide.

4. Supporting Endogenous Defense Mechanisms

Since prophylactic antibiotic treatment is unsatisfying and moreover bears the risk for selecting for resistant strains, alternative approaches have been investigated. One attractive strategy is to enhance endogenous defense mechanisms, which will be well tolerated by the host organism while at the same time are less likely to induce resistance in the pathogen.

4.1. Vitamin D

The implication of vitamin D on the immune system is well established and has empirically been employed by the usage of sunlight to fight infections, such as tuberculosis [96]. More recently, a direct linkage of vitamin D and CAMP expression was identified as one underlying mechanism [97]. The promoter region of the CAMP gene contains a vitamin D-responsive element (VDRE), which binds the receptor in a ligand-dependent and -independent manner, but transcription efficacy is greatly enhanced in the presence of vitamin D. This genetic organization is restricted to primates, including humans, and not present in rodents, which hampers animal studies in this field.
The action of vitamin D requires the hormonally-active form 1,25-dihydroxyvitamin D3. Vitamin D precursors obtained by exposure to sunlight or with the diet are converted to the major storage form 25-hydroxyvitamin D3 in the liver [98], while the kidney is the principal organ to convert the inactive form into active 1,25-dihydroxyvitamin D3 by the 1α-hydroxylase (Cyp27B1) [99]. However, a number of other tissues, including epithelial cells, at different body sites express this enzyme and are able to locally activate vitamin D. The Cyp27B1 enzyme could also be detected in bladder epithelial cells [100]. Upregulation of the vitamin D inactivating enzyme 24-hydroxylase (Cyp24A1) upon exposure to 25-hydroxyvitamin D3 indicated furthermore activation of a negative feedback response, indicating the local generation of 1,25-dihydroxyvitamin D3.
Several clinical studies have demonstrated the importance of sufficient vitamin D levels for the protection of the urinary tract against infection. In children with UTI, serum vitamin D was significantly lower compared to healthy controls who never had a UTI [101]; moreover, vitamin D levels correlated with the severity of the infection. Similarly, in premenopausal women, low levels of serum vitamin D were associated with a higher risk for recurrent UTI [102]. In an experimental study, we could show that oral vitamin D supplementation was able to increase CAMP production in the human bladder epithelium in response to E. coli [100]. Together, these studies clearly demonstrate that vitamin D supplementation provides a potent alternative in the protection against UTI in vulnerable patient groups.

4.2. Estrogen

Clinical observations indicate a complex impact of estrogen on UTI pathogenesis. In young women, high estrogen levels are linked to increased receptivity for E. coli infection [103]; in women post-menopause who suffer from recurrent UTI, estrogen supplementation might, on the other hand, improve the condition [104,105,106]. Recently, we were able to provide explanations for the seemingly conflicting findings with respect to the estrogenic action in the urinary tract, based on experimental data from animal and in vitro models and patients’ material [69]. In the presence of high estrogen levels, corresponding to those found in menstruating women, expression of uroplakin (UP) Ia and β1 integrin, major host receptors mediating E. coli adherence and invasion of the uroepithelium [107,108], were elevated, partly explaining the high incidence of UTIs when estrogen levels are high. On the other hand, estrogen caused increased expression of at least three of five tested AMPs in 75% of postmenopausal women after local supplementation with estrogen for two weeks, with the most prominent overall effect on hBD3 [69]. Expression of proteins involved in cell-cell contact was likewise elevated, indicating improved epithelial integrity in the presence of estrogen. Similar to vitamin D, estrogen acts via its receptors and estrogen responsive elements in the gene promoter. The universal effect of estrogen on AMP expression, however, suggests rather an indirect effect linked to estrogen-promoted differentiation of the uroepithelium. In the absence of estrogen, defective differentiation of bladder epithelial cells was noted [109], supporting this hypothesis. The epithelial-protective effect by estrogen might furthermore be supported by dampening the pro-inflammatory response [109], which always coincides with tissue damage. Increased epithelial integrity and higher expression of AMPs as a consequence of estrogenic action on the uroepithelium may together reduce the formation of QIRs as the source of recurrent infections [69,109].
Estrogen levels are high also during pregnancy. While there are no solid data indicating differences in the general receptivity to UTI in pregnant women, an increased risk for ascending infections is well established [110]. Anatomical alterations, such as ureteral dilatation and decreased tone in the bladder and ureter, facilitate ureterovesical reflux and ascendance of bacteria from the bladder to the kidneys. These complex anatomical, physiological and hormonal changes might dominate over-protective estrogenic effects on epithelial defense strategies during pregnancy.

4.3. Medicinal Herbs

Herbal preparations are often part of alternative treatment strategies. The active substance or the way of action has not always been identified; thus, treatment is often empirically and without proven effect. However, efforts are made to understand the mechanisms of these medicines, to evaluate their value as alternatives for conventional antibiotic treatment [111].
Some herbal preparations exhibit direct antibacterial activity against uropathogens, at least in vitro [112,113,114]. A commercial preparation consisting of Armoraciae rusticanae (horseradish) and Tropaeoli majoris (nasturtium) was tested in a clinical trial and found effective for prophylaxis against recurrent UTI [115]. In vitro, the preparation exhibited antibacterial activity against a wide range of bacterial pathogens, including E. coli [112]. A systematic screening of plant extracts traditionally used for UTI treatment showed that several substances did not exhibit direct antimicrobial actions, but interfered with bacterial adhesion to the host cell [116]. Two herbal extracts, Agropyron repens (couch grass) and Zea mays (maize, corn), acted on the bacteria directly. Direct interference with adhesion of P-fimbriated bacteria has also been proposed for the protective mechanism of cranberries [117,118]. Cranberry products have widely been used for the treatment and prevention of (recurrent) UTIs, but the benefit is discussed controversially [119,120].
Three further plant extracts investigated in the study by Rafsanjany et al. [116], i.e., extracts from Betula spp. (birch), Orthosiphon stamineus (Java tea) and Urtica spp. (nettles), acted on the host cell rather than the bacterium to diminish adherence of the pathogen. Such infection-reducing effects have been observed and investigated for other medicinal plants with traditional anti-infective applications. Decoction of Lactuca indica (Indian lettuce), a Vietnamese herbal plant with anti-inflammatory and diuretic activity, reduced bacterial infection by interfering with host cell signaling pathways leading to bacterial uptake [121]. Extracts from the Malaysian flowering plant Labisia pumila var. alata [122] and the seeds from Citrus reticulata Blanco (mandarin) [123] reduced bacterial invasion of bladder epithelial cells by downregulation of the invasion-mediating receptor β1 integrin. In addition to the anti-adhesive effect, Labisia pumila var. alata extracts supported apoptosis-dependent exfoliation of infected cells, but prevented progressive cell death during infection. These actions in combination may reduce the bacterial burden in the upper epithelial cell layer of the urinary bladder while protecting the underlying tissue, prone to developing persisting bacterial reservoirs.
Many medicinal herbs exhibit also anti-inflammatory, analgetic and diuretic effects, which may contribute to their beneficial effect during UTI. A double-blinded, randomized clinical trial comparing symptomatic (ibuprofen) and antibiotic treatments (ciprofloxacin) of women with acute UTI suggested that symptomatic treatment might often be sufficient for the management of most uncomplicated UTI [124]. Thus, anti-inflammatory and analgetic actions might explain the relief by the intake of herbal preparations where direct modes of action are yet unidentified. For example, Gynostemma pentaphyllum, mostly known for its anti-diabetic effect, reduced the bacteria-induced inflammatory response in the urinary bladder of rats receiving the herbal extract [125]. Interestingly, Gynostemma pentaphyllum also modulated expression of several AMPs. In particular, bladder epithelial cells from treated rats responded with a stronger upregulation of psoriasin, an AMP with strong activity against E. coli, compared to control animals.
Even though the action of most traditional herbal medicines is incompletely understood today and placebo-controlled clinical trials are sparse or missing, these studies demonstrate the complex and multifunctional mode of action. Identifying the active substances and establishing their activity will help to develop novel treatment and prophylactic strategies against UTIs.

5. Concluding Remarks

Uncomplicated UTIs are often self-limiting, still a frequent indication for antimicrobial therapy. This conventional treatment is usually effective against the acute infection, but long-term usage of antibiotics is required to reduce recurrences. The imperfect protection against recurrent UTIs and the risk of provoking resistance among the bacterial pathogen are two reasons asking for alternatives in the management of UTIs. A promising possibility is the exploitation of the innate immune defense by supporting endogenous mechanisms. The targeted application of natural substances identified in traditionally-used herbal medicines and the knowledge about their way of action provide another source for the development of alternative strategies.

Author Contributions

Petra Lüthje performed literature searches and wrote the manuscript. Annelie Brauner supervised the work and reviewed the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Foxman, B.; Barlow, R.; D’Arcy, H.; Gillespie, B.; Sobel, J.D. Urinary tract infection: Self-reported incidence and associated costs. Ann. Epidemiol. 2000, 10, 509–515. [Google Scholar] [CrossRef]
  2. Foxman, B. Urinary tract infection syndromes: Occurrence, recurrence, bacteriology, risk factors, and disease burden. Infect. Dis. Clin. N. Am. 2014, 28, 1–13. [Google Scholar] [CrossRef] [PubMed]
  3. Hoberman, A.; Chao, H.P.; Keller, D.M.; Hickey, R.; Davis, H.W.; Ellis, D. Prevalence of urinary tract infection in febrile infants. J. Pediatr. 1993, 123, 17–23. [Google Scholar] [CrossRef]
  4. Shaw, K.N.; Gorelick, M.; McGowan, K.L.; Yakscoe, N.M.; Schwartz, J.S. Prevalence of urinary tract infection in febrile young children in the emergency department. Pediatrics 1998. [Google Scholar] [CrossRef]
  5. Zorc, J.J.; Levine, D.A.; Platt, S.L.; Dayan, P.S.; Macias, C.G.; Krief, W.; Schor, J.; Bank, D.; Shaw, K.N.; Kuppermann, N.; et al. Clinical and demographic factors associated with urinary tract infection in young febrile infants. Pediatrics 2005, 116, 644–648. [Google Scholar] [CrossRef] [PubMed]
  6. Marild, S.; Jodal, U. Incidence rate of first-time symptomatic urinary tract infection in children under 6 years of age. Acta Paediatr. 1998, 87, 549–552. [Google Scholar] [CrossRef] [PubMed]
  7. Stokland, E.; Hellstrom, M.; Jacobsson, B.; Jodal, U.; Sixt, R. Renal damage one year after first urinary tract infection: Role of dimercaptosuccinic acid scintigraphy. J. Pediatr. 1996, 129, 815–820. [Google Scholar] [CrossRef]
  8. Stokland, E.; Hellstrom, M.; Jacobsson, B.; Jodal, U.; Sixt, R. Evaluation of DMSA scintigraphy and urography in assessing both acute and permanent renal damage in children. Acta Radiol. 1998, 39, 447–452. [Google Scholar] [CrossRef] [PubMed]
  9. Saxton, H.M. Computed tomography or intravenous urography for renal damage in childhood urinary infection? Pediatr. Nephrol. 1995, 9, 256–257. [Google Scholar] [CrossRef] [PubMed]
  10. Kavanagh, E.C.; Ryan, S.; Awan, A.; McCourbrey, S.; O’Connor, R.; Donoghue, V. Can MRI replace DMSA in the detection of renal parenchymal defects in children with urinary tract infections? Pediatr Radiol 2005, 35, 275–281. [Google Scholar] [CrossRef] [PubMed]
  11. Winberg, J.; Andersen, H.J.; Bergstrom, T.; Jacobsson, B.; Larson, H.; Lincoln, K. Epidemiology of symptomatic urinary tract infection in childhood. Acta Paediatr. Scand. Suppl. 1974, 63, 1–20. [Google Scholar] [CrossRef]
  12. Mingin, G.C.; Hinds, A.; Nguyen, H.T.; Baskin, L.S. Children with a febrile urinary tract infection and a negative radiologic workup: Factors predictive of recurrence. Urology 2004, 63, 562–565. [Google Scholar] [CrossRef] [PubMed]
  13. Ramos, N.L.; Dzung, D.T.; Stopsack, K.; Janko, V.; Pourshafie, M.R.; Katouli, M.; Brauner, A. Characterisation of uropathogenic Escherichia coli from children with urinary tract infection in different countries. Eur. J. Clin. Microbiol. Infect. Dis. 2011, 30, 1587–1593. [Google Scholar] [CrossRef] [PubMed]
  14. Ramos, N.L.; Sekikubo, M.; Dzung, D.T.; Kosnopfel, C.; Kironde, F.; Mirembe, F.; Brauner, A. Uropathogenic Escherichia coli isolates from pregnant women in different countries. J. Clin. Microbiol. 2012, 50, 3569–3574. [Google Scholar] [CrossRef] [PubMed]
  15. Joo, H.S.; Otto, M. Mechanisms of resistance to antimicrobial peptides in staphylococci. Biochim. Biophys. Acta 2015. [Google Scholar] [CrossRef] [PubMed]
  16. Gruenheid, S.; Le Moual, H. Resistance to antimicrobial peptides in gram-negative bacteria. FEMS Microbiol. Lett. 2012, 330, 81–89. [Google Scholar] [CrossRef] [PubMed]
  17. Galdiero, S.; Falanga, A.; Berisio, R.; Grieco, P.; Morelli, G.; Galdiero, M. Antimicrobial peptides as an opportunity against bacterial diseases. Curr. Med. Chem. 2015, 22, 1665–1677. [Google Scholar] [CrossRef] [PubMed]
  18. Niyonsaba, F.; Nagaoka, I.; Ogawa, H. Human defensins and cathelicidins in the skin: Beyond direct antimicrobial properties. Crit. Rev. Immunol. 2006, 26, 545–576. [Google Scholar] [CrossRef] [PubMed]
  19. Bowdish, D.M.; Davidson, D.J.; Hancock, R.E. A re-evaluation of the role of host defence peptides in mammalian immunity. Curr. Protein Pept. Sci. 2005, 6, 35–51. [Google Scholar] [CrossRef] [PubMed]
  20. Heilborn, J.D.; Nilsson, M.F.; Weber, G.; Sorensen, O.; Borregaard, N.; Stahle-Backdahl, M.; Kratz, G. The cathelicidin anti-microbial peptide LL-37 is involved in re-epithelialization of human skin wounds and is lacking in chronic ulcer epithelium. J. Investig. Dermatol. 2003, 120, 379–389. [Google Scholar] [CrossRef] [PubMed]
  21. Butmarc, J.; Yufit, T.; Carson, P.; Falanga, V. Human beta-defensin-2 expression is increased in chronic wounds. Wound Repair Regen. 2004, 12, 439–443. [Google Scholar] [CrossRef] [PubMed]
  22. Aberg, K.M.; Man, M.Q.; Gallo, R.L.; Ganz, T.; Crumrine, D.; Brown, B.E.; Choi, E.H.; Kim, D.K.; Schroder, J.M.; Feingold, K.R.; et al. Co-regulation and interdependence of the mammalian epidermal permeability and antimicrobial barriers. J. Investig. Dermatol. 2008, 128, 917–925. [Google Scholar] [CrossRef] [PubMed]
  23. Ejrnaes, K.; Sandvang, D.; Lundgren, B.; Ferry, S.; Holm, S.; Lundholm, R.; Frimodt-Moller, N.; Monsen, T. Pulsed-field gel electrophoresis typing of Escherichia coli strains from samples collected before and after pivmecillinam or placebo treatment of uncomplicated community-acquired urinary tract infection in women. J. Clin. Microbiol. 2006, 44, 1776–1781. [Google Scholar] [CrossRef] [PubMed]
  24. Navas-Nacher, E.L.; Dardick, F.; Venegas, M.F.; Anderson, B.E.; Schaeffer, A.J.; Duncan, J.L. Relatedness of Escherichia coli colonizing women longitudinally. Mol. Urol. 2001, 5, 31–36. [Google Scholar] [CrossRef] [PubMed]
  25. Norinder, B.S.; Lüthje, P.; Yadav, M.; Kadas, L.; Fang, H.; Nord, C.E.; Brauner, A. Cellulose and papg are important for Escherichia coli causing recurrent urinary tract infection in women. Infection 2011, 39, 571–574. [Google Scholar] [CrossRef] [PubMed]
  26. Nielsen, K.L.; Dynesen, P.; Larsen, P.; Frimodt-Moller, N. Faecal Escherichia coli from patients with E. coli urinary tract infection and healthy controls who have never had a urinary tract infection. J. Med. Microbiol. 2014, 63, 582–589. [Google Scholar] [CrossRef] [PubMed]
  27. Anderson, G.G.; Palermo, J.J.; Schilling, J.D.; Roth, R.; Heuser, J.; Hultgren, S.J. Intracellular bacterial biofilm-like pods in urinary tract infections. Science 2003, 301, 105–107. [Google Scholar] [CrossRef] [PubMed]
  28. Justice, S.S.; Hung, C.; Theriot, J.A.; Fletcher, D.A.; Anderson, G.G.; Footer, M.J.; Hultgren, S.J. Differentiation and developmental pathways of uropathogenic Escherichia coli in urinary tract pathogenesis. Proc. Natl. Acad. Sci. USA 2004, 101, 1333–1338. [Google Scholar] [CrossRef] [PubMed]
  29. Klumpp, D.J.; Rycyk, M.T.; Chen, M.C.; Thumbikat, P.; Sengupta, S.; Schaeffer, A.J. Uropathogenic Escherichia coli induces extrinsic and intrinsic cascades to initiate urothelial apoptosis. Infect. Immun. 2006, 74, 5106–5113. [Google Scholar] [CrossRef] [PubMed]
  30. Thumbikat, P.; Berry, R.E.; Schaeffer, A.J.; Klumpp, D.J. Differentiation-induced uroplakin iii expression promotes urothelial cell death in response to uropathogenic E. coli. Microbes Infect. 2009, 11, 57–65. [Google Scholar] [CrossRef] [PubMed]
  31. Eto, D.S.; Sundsbak, J.L.; Mulvey, M.A. Actin-gated intracellular growth and resurgence of uropathogenic Escherichia coli. Cell. Microbiol. 2006, 8, 704–717. [Google Scholar] [CrossRef] [PubMed]
  32. Mysorekar, I.U.; Mulvey, M.A.; Hultgren, S.J.; Gordon, J.I. Molecular regulation of urothelial renewal and host defenses during infection with uropathogenic Escherichia coli. J. Biol. Chem. 2002, 277, 7412–7419. [Google Scholar] [CrossRef] [PubMed]
  33. Mulvey, M.A.; Schilling, J.D.; Hultgren, S.J. Establishment of a persistent Escherichia coli reservoir during the acute phase of a bladder infection. Infect. Immun. 2001, 69, 4572–4579. [Google Scholar] [CrossRef] [PubMed]
  34. Rosen, D.A.; Hooton, T.M.; Stamm, W.E.; Humphrey, P.A.; Hultgren, S.J. Detection of intracellular bacterial communities in human urinary tract infection. PLoS Med. 2007, 4, e329. [Google Scholar] [CrossRef] [PubMed]
  35. Garofalo, C.K.; Hooton, T.M.; Martin, S.M.; Stamm, W.E.; Palermo, J.J.; Gordon, J.I.; Hultgren, S.J. Escherichia coli from urine of female patients with urinary tract infections is competent for intracellular bacterial community formation. Infect. Immun. 2007, 75, 52–60. [Google Scholar] [CrossRef] [PubMed]
  36. Robino, L.; Scavone, P.; Araujo, L.; Algorta, G.; Zunino, P.; Pirez, M.C.; Vignoli, R. Intracellular bacteria in the pathogenesis of Escherichia coli urinary tract infection in children. Clin. Infect. Dis. 2014, 59, e158–e164. [Google Scholar] [CrossRef] [PubMed]
  37. Robino, L.; Scavone, P.; Araujo, L.; Algorta, G.; Zunino, P.; Vignoli, R. Detection of intracellular bacterial communities in a child with Escherichia coli recurrent urinary tract infections. Pathog. Dis. 2013, 68, 78–81. [Google Scholar] [CrossRef] [PubMed]
  38. Conway, P.H.; Cnaan, A.; Zaoutis, T.; Henry, B.V.; Grundmeier, R.W.; Keren, R. Recurrent urinary tract infections in children: Risk factors and association with prophylactic antimicrobials. JAMA 2007, 298, 179–186. [Google Scholar] [CrossRef] [PubMed]
  39. Nuutinen, M.; Uhari, M. Recurrence and follow-up after urinary tract infection under the age of 1 year. Pediatr. Nephrol. 2001, 16, 69–72. [Google Scholar] [CrossRef] [PubMed]
  40. Godaly, G.; Ambite, I.; Svanborg, C. Innate immunity and genetic determinants of urinary tract infection susceptibility. Curr. Opin. Infect. Dis. 2015, 28, 88–96. [Google Scholar] [CrossRef] [PubMed]
  41. Ulett, G.C.; Valle, J.; Beloin, C.; Sherlock, O.; Ghigo, J.M.; Schembri, M.A. Functional analysis of antigen 43 in uropathogenic Escherichia coli reveals a role in long-term persistence in the urinary tract. Infect. Immun. 2007, 75, 3233–3244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Lüthje, P.; Brauner, A. Ag43 promotes persistence of uropathogenic Escherichia coli isolates in the urinary tract. J. Clin. Microbiol. 2010, 48, 2316–2317. [Google Scholar] [CrossRef] [PubMed]
  43. Kai-Larsen, Y.; Lüthje, P.; Chromek, M.; Peters, V.; Wang, X.; Holm, Å.; Kadas, L.; Hedlund, K.O.; Johansson, J.; Chapman, M.R.; et al. Uropathogenic Escherichia coli modulates immune responses and its curli fimbriae interact with the antimicrobial peptide LL-37. PLoS Pathog. 2010, 6, e1001010. [Google Scholar] [CrossRef] [PubMed]
  44. Albert, X.; Huertas, I.; Pereiro, I.I.; Sanfelix, J.; Gosalbes, V.; Perrota, C. Antibiotics for preventing recurrent urinary tract infection in non-pregnant women. Cochrane Database Syst. Rev. 2004. [Google Scholar] [CrossRef]
  45. Eells, S.J.; Bharadwa, K.; McKinnell, J.A.; Miller, L.G. Recurrent urinary tract infections among women: Comparative effectiveness of 5 prevention and management strategies using a Markov chain Monte Carlo model. Clin. Infect. Dis. 2014, 58, 147–160. [Google Scholar] [CrossRef] [PubMed]
  46. Lorenzo-Gomez, M.F.; Padilla-Fernandez, B.; Garcia-Criado, F.J.; Miron-Canelo, J.A.; Gil-Vicente, A.; Nieto-Huertos, A.; Silva-Abuin, J.M. Evaluation of a therapeutic vaccine for the prevention of recurrent urinary tract infections versus prophylactic treatment with antibiotics. Int. Urogynecol. J. 2013, 24, 127–134. [Google Scholar] [CrossRef] [PubMed]
  47. Beerepoot, M.A.; ter Riet, G.; Nys, S.; van der Wal, W.M.; de Borgie, C.A.; de Reijke, T.M.; Prins, J.M.; Koeijers, J.; Verbon, A.; Stobberingh, E.; et al. Cranberries vs antibiotics to prevent urinary tract infections: A randomized double-blind noninferiority trial in premenopausal women. Arch. Intern. Med. 2011, 171, 1270–1278. [Google Scholar] [CrossRef] [PubMed]
  48. Geerlings, S.E.; Beerepoot, M.A.; Prins, J.M. Prevention of recurrent urinary tract infections in women: Antimicrobial and nonantimicrobial strategies. Infect. Dis. Clin. N. Am. 2014, 28, 135–147. [Google Scholar] [CrossRef] [PubMed]
  49. Spencer, J.D.; Schwaderer, A.L.; Becknell, B.; Watson, J.; Hains, D.S. The innate immune response during urinary tract infection and pyelonephritis. Pediatr. Nephrol. 2014, 29, 1139–1149. [Google Scholar] [CrossRef] [PubMed]
  50. Ali, A.S.; Townes, C.L.; Hall, J.; Pickard, R.S. Maintaining a sterile urinary tract: The role of antimicrobial peptides. J. Urol. 2009, 182, 21–28. [Google Scholar] [CrossRef] [PubMed]
  51. Chromek, M.; Slamova, Z.; Bergman, P.; Kovacs, L.; Podracka, L.; Ehren, I.; Hokfelt, T.; Gudmundsson, G.H.; Gallo, R.L.; Agerberth, B.; et al. The antimicrobial peptide cathelicidin protects the urinary tract against invasive bacterial infection. Nat. Med. 2006, 12, 636–641. [Google Scholar] [CrossRef] [PubMed]
  52. Spencer, J.D.; Schwaderer, A.L.; Dirosario, J.D.; McHugh, K.M.; McGillivary, G.; Justice, S.S.; Carpenter, A.R.; Baker, P.B.; Harder, J.; Hains, D.S. Ribonuclease 7 is a potent antimicrobial peptide within the human urinary tract. Kidney Int. 2011, 80, 174–180. [Google Scholar] [CrossRef] [PubMed]
  53. Spencer, J.D.; Schwaderer, A.L.; Wang, H.; Bartz, J.; Kline, J.; Eichler, T.; DeSouza, K.R.; Sims-Lucas, S.; Baker, P.; Hains, D.S. Ribonuclease 7, an antimicrobial peptide upregulated during infection, contributes to microbial defense of the human urinary tract. Kidney Int. 2013, 83, 615–625. [Google Scholar] [CrossRef] [PubMed]
  54. Ganz, T. Defensins in the urinary tract and other tissues. J. Infect. Dis. 2001, 183, S41–S42. [Google Scholar] [CrossRef] [PubMed]
  55. Semple, C.A.; Rolfe, M.; Dorin, J.R. Duplication and selection in the evolution of primate beta-defensin genes. Genome Biol. 2003, 4, R31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Zhao, C.; Wang, I.; Lehrer, R.I. Widespread expression of beta-defensin hBD-1 in human secretory glands and epithelial cells. FEBS Lett. 1996, 396, 319–322. [Google Scholar] [CrossRef]
  57. Valore, E.V.; Park, C.H.; Quayle, A.J.; Wiles, K.R.; McCray, P.B., Jr.; Ganz, T. Human beta-defensin-1: An antimicrobial peptide of urogenital tissues. J. Clin. Investig. 1998, 101, 1633–1642. [Google Scholar] [CrossRef] [PubMed]
  58. Becknell, B.; Spencer, J.D.; Carpenter, A.R.; Chen, X.; Singh, A.; Ploeger, S.; Kline, J.; Ellsworth, P.; Li, B.; Proksch, E.; et al. Expression and antimicrobial function of beta-defensin 1 in the lower urinary tract. PLoS ONE 2013, 8, e77714. [Google Scholar] [CrossRef] [PubMed]
  59. Hiratsuka, T.; Nakazato, M.; Ihi, T.; Minematsu, T.; Chino, N.; Nakanishi, T.; Shimizu, A.; Kangawa, K.; Matsukura, S. Structural analysis of human beta-defensin-1 and its significance in urinary tract infection. Nephron 2000, 85, 34–40. [Google Scholar] [CrossRef] [PubMed]
  60. Nielsen, K.L.; Dynesen, P.; Larsen, P.; Jakobsen, L.; Andersen, P.S.; Frimodt-Moller, N. Role of urinary cathelicidin LL-37 and human beta-defensin 1 in uncomplicated Escherichia coli urinary tract infections. Infect. Immun. 2014, 82, 1572–1578. [Google Scholar] [CrossRef] [PubMed]
  61. Nitschke, M.; Wiehl, S.; Baer, P.C.; Kreft, B. Bactericidal activity of renal tubular cells: The putative role of human beta-defensins. Exp. Nephrol. 2002, 10, 332–337. [Google Scholar] [CrossRef] [PubMed]
  62. Lehmann, J.; Retz, M.; Harder, J.; Krams, M.; Kellner, U.; Hartmann, J.; Hohgrawe, K.; Loch, T.; Raffenberg, U.; Gerber, M.; et al. Expression of human beta-defensins 1 and 2 in kidneys with chronic bacterial infection. BMC Infect. Dis. 2002, 2, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Spencer, J.D.; Hains, D.S.; Porter, E.; Bevins, C.L.; DiRosario, J.; Becknell, B.; Schwaderer, A.L.; Wang, H. Human alpha defensin 5 expression in the human kidney and urinary tract. PLoS ONE 2012, 7, e31712. [Google Scholar] [CrossRef] [PubMed]
  64. Park, C.H.; Valore, E.V.; Waring, A.J.; Ganz, T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J. Biol. Chem. 2001, 276, 7806–7810. [Google Scholar] [CrossRef] [PubMed]
  65. Åbrink, M.; Larsson, E.; Gobl, A.; Hellman, L. Expression of lactoferrin in the kidney: Implications for innate immunity and iron metabolism. Kidney Int. 2000, 57, 2004–2010. [Google Scholar] [CrossRef] [PubMed]
  66. Paragas, N.; Kulkarni, R.; Werth, M.; Schmidt-Ott, K.M.; Forster, C.; Deng, R.; Zhang, Q.; Singer, E.; Klose, A.D.; Shen, T.H.; et al. Alpha-intercalated cells defend the urinary system from bacterial infection. J. Clin. Investig. 2014, 124, 2963–2976. [Google Scholar] [CrossRef] [PubMed]
  67. Steigedal, M.; Marstad, A.; Haug, M.; Damas, J.K.; Strong, R.K.; Roberts, P.L.; Himpsl, S.D.; Stapleton, A.; Hooton, T.M.; Mobley, H.L.; et al. Lipocalin 2 imparts selective pressure on bacterial growth in the bladder and is elevated in women with urinary tract infection. J. Immunol. 2014, 193, 6081–6089. [Google Scholar] [CrossRef] [PubMed]
  68. Gläser, R.; Harder, J.; Lange, H.; Bartels, J.; Christophers, E.; Schröder, J.M. Antimicrobial psoriasin (S100A7) protects human skin from Escherichia coli infection. Nat. Immunol. 2005, 6, 57–64. [Google Scholar] [CrossRef] [PubMed]
  69. Lüthje, P.; Brauner, H.; Ramos, N.L.; Övregaard, A.; Gläser, R.; Hirschberg, A.L.; Aspenström, P.; Brauner, A. Estrogen supports urothelial defense mechanisms. Sci. Transl. Med. 2013, 5, 190ra180. [Google Scholar] [CrossRef] [PubMed]
  70. Østergaard, M.; Wolf, H.; Orntoft, T.F.; Celis, J.E. Psoriasin (S100A7): A putative urinary marker for the follow-up of patients with bladder squamous cell carcinomas. Electrophoresis 1999, 20, 349–354. [Google Scholar] [CrossRef]
  71. Bates, J.M.; Raffi, H.M.; Prasadan, K.; Mascarenhas, R.; Laszik, Z.; Maeda, N.; Hultgren, S.J.; Kumar, S. Tamm-Horsfall protein knockout mice are more prone to urinary tract infection: Rapid communication. Kidney Int. 2004, 65, 791–797. [Google Scholar] [CrossRef] [PubMed]
  72. Lehrer, R.I.; Ganz, T. Cathelicidins: A family of endogenous antimicrobial peptides. Curr. Opin. Hematol. 2002, 9, 18–22. [Google Scholar] [CrossRef] [PubMed]
  73. Zanetti, M. The role of cathelicidins in the innate host defenses of mammals. Curr. Issues Mol. Biol. 2005, 7, 179–196. [Google Scholar] [PubMed]
  74. Murakami, M.; Lopez-Garcia, B.; Braff, M.; Dorschner, R.A.; Gallo, R.L. Postsecretory processing generates multiple cathelicidins for enhanced topical antimicrobial defense. J. Immunol. 2004, 172, 3070–3077. [Google Scholar] [CrossRef] [PubMed]
  75. Murakami, M.; Ohtake, T.; Dorschner, R.A.; Schittek, B.; Garbe, C.; Gallo, R.L. Cathelicidin anti-microbial peptide expression in sweat, an innate defense system for the skin. J. Investig. Dermatol. 2002, 119, 1090–1095. [Google Scholar] [CrossRef] [PubMed]
  76. Sorensen, O.E.; Gram, L.; Johnsen, A.H.; Andersson, E.; Bangsboll, S.; Tjabringa, G.S.; Malm, J.; Hiemstra, P.S.; Egesten, A.; Borregaard, N. Processing of seminal plasma hCAP-18 to ALL-38 by gastricsin: A novel mechanism of generating antimicrobial peptides in vagina. J. Biol. Chem. 2003, 278, 28540–28546. [Google Scholar] [CrossRef] [PubMed]
  77. Pestonjamasp, V.K.; Huttner, K.H.; Gallo, R.L. Processing site and gene structure for the murine antimicrobial peptide CRAMP. Peptides 2001, 22, 1643–1650. [Google Scholar] [CrossRef]
  78. Thomassin, J.L.; Brannon, J.R.; Gibbs, B.F.; Gruenheid, S.; le Moual, H. OmpT outer membrane proteases of enterohemorrhagic and enteropathogenic Escherichia coli contribute differently to the degradation of human LL-37. Infect. Immun. 2012, 80, 483–492. [Google Scholar] [CrossRef] [PubMed]
  79. Brannon, J.R.; Thomassin, J.L.; 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, 64–71. [Google Scholar] [CrossRef] [PubMed]
  80. Richards, S.M.; Strandberg, K.L.; Gunn, J.S. Salmonella-regulated lipopolysaccharide modifications. Subcell. Biochem. 2010, 53, 101–122. [Google Scholar] [PubMed]
  81. Rosenfeld, Y.; Papo, N.; Shai, Y. Endotoxin (lipopolysaccharide) neutralization by innate immunity host-defense peptides. Peptide properties and plausible modes of action. J. Biol. Chem. 2006, 281, 1636–1643. [Google Scholar] [CrossRef] [PubMed]
  82. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  83. Neumann, A.; Berends, E.T.; Nerlich, A.; Molhoek, E.M.; Gallo, R.L.; Meerloo, T.; Nizet, V.; Naim, H.Y.; von Köckritz-Blickwede, M. The antimicrobial peptide LL-37 facilitates the formation of neutrophil extracellular traps. Biochem. J. 2014, 464, 3–11. [Google Scholar] [CrossRef] [PubMed]
  84. Neumann, A.; Völlger, L.; Berends, E.T.; Molhoek, E.M.; Stapels, D.A.; Midon, M.; Friaes, A.; Pingoud, A.; Rooijakkers, S.H.; Gallo, R.L.; et al. Novel role of the antimicrobial peptide LL-37 in the protection of neutrophil extracellular traps against degradation by bacterial nucleases. J. Innate Immun. 2014, 6, 860–868. [Google Scholar] [CrossRef] [PubMed]
  85. Danka, E.S.; Hunstad, D.A. Cathelicidin augments epithelial receptivity and pathogenesis in experimental Escherichia coli cystitis. J. Infect. Dis. 2015, 211, 1164–1173. [Google Scholar] [CrossRef] [PubMed]
  86. Oottamasathien, S.; Jia, W.; McCoard, L.; Slack, S.; Zhang, J.; Skardal, A.; Job, K.; Kennedy, T.P.; Dull, R.O.; Prestwich, G.D. A murine model of inflammatory bladder disease: Cathelicidin peptide induced bladder inflammation and treatment with sulfated polysaccharides. J. Urol. 2011, 186, 1684–1692. [Google Scholar] [CrossRef] [PubMed]
  87. Oottamasathien, S.; Jia, W.; Roundy, L.M.; Zhang, J.; Wang, L.; Ye, X.; Hill, A.C.; Savage, J.; Lee, W.Y.; Hannon, A.M.; et al. Physiological relevance of LL-37 induced bladder inflammation and mast cells. J. Urol. 2013, 190, 1596–1602. [Google Scholar] [CrossRef] [PubMed]
  88. Lüthje, P.; Chromek, M.; Brauner, A. Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden. Unpublished work. 2007. [Google Scholar]
  89. Overhage, J.; Campisano, A.; Bains, M.; Torfs, E.C.; Rehm, B.H.; Hancock, R.E. Human host defense peptide LL-37 prevents bacterial biofilm formation. Infect. Immun. 2008, 76, 4176–4182. [Google Scholar] [CrossRef] [PubMed]
  90. Dean, S.N.; Bishop, B.M.; van Hoek, M.L. Natural and synthetic cathelicidin peptides with anti-microbial and anti-biofilm activity against Staphylococcus aureus. BMC Microbiol. 2011, 11, 114. [Google Scholar] [CrossRef] [PubMed]
  91. Haisma, E.M.; de Breij, A.; van Dissel, J.T.; Drijfhout, J.W.; Hiemstra, P.S.; El Ghalbzouri, A.; Chan, H.; Nibbering, P.H. Ll-37-derived peptides eradicate multidrug-resistant Staphylococcus aureus from thermally wounded human skin equivalents. Antimicrob. Agents Chemother. 2014, 58, 4411–4419. [Google Scholar] [CrossRef] [PubMed]
  92. Amer, L.S.; Bishop, B.M.; van Hoek, M.L. Antimicrobial and antibiofilm activity of cathelicidins and short, synthetic peptides against Francisella. Biochem. Biophys. Res. Commun. 2010, 396, 246–251. [Google Scholar] [CrossRef] [PubMed]
  93. Feng, X.; Sambanthamoorthy, K.; Palys, T.; Paranavitana, C. The human antimicrobial peptide LL-37 and its fragments possess both antimicrobial and antibiofilm activities against multidrug-resistant Acinetobacter baumannii. Peptides 2013, 49, 131–137. [Google Scholar] [CrossRef] [PubMed]
  94. Sol, A.; Ginesin, O.; Chaushu, S.; Karra, L.; Coppenhagen-Glazer, S.; Ginsburg, I.; Bachrach, G. Ll-37 opsonizes and inhibits biofilm formation of Aggregatibacter actinomycetemcomitans at subbactericidal concentrations. Infect. Immun. 2013, 81, 3577–3585. [Google Scholar] [CrossRef] [PubMed]
  95. Tsai, P.W.; Yang, C.Y.; Chang, H.T.; Lan, C.Y. Human antimicrobial peptide LL-37 inhibits adhesion of Candida albicans by interacting with yeast cell-wall carbohydrates. PLoS ONE 2011, 6, e17755. [Google Scholar] [CrossRef] [PubMed]
  96. Schwalfenberg, G.K. A review of the critical role of vitamin D in the functioning of the immune system and the clinical implications of vitamin D deficiency. Mol. Nutr. Food. Res. 2011, 55, 96–108. [Google Scholar] [CrossRef] [PubMed]
  97. Gombart, A.F.; Borregaard, N.; Koeffler, H.P. Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3. FASEB J. 2005, 19, 1067–1077. [Google Scholar] [CrossRef] [PubMed]
  98. Ponchon, G.; Kennan, A.L.; DeLuca, H.F. “Activation” of vitamin D by the liver. J. Clin. Investig. 1969, 48, 2032–2037. [Google Scholar] [CrossRef] [PubMed]
  99. Zehnder, D.; Bland, R.; Walker, E.A.; Bradwell, A.R.; Howie, A.J.; Hewison, M.; Stewart, P.M. Expression of 25-hydroxyvitamin D3-1alpha-hydroxylase in the human kidney. J. Am. Soc. Nephrol. 1999, 10, 2465–2473. [Google Scholar] [PubMed]
  100. Hertting, O.; Lüthje, P.; Brauner, H.; Dyrdak, R.; Jonasson, A.F.; Wiklund, P.; Chromek, M.; Holm, Å.; Brauner, A. Vitamin D induction of the human antimicrobial peptide cathelicidin in the urinary bladder. PLoS ONE 2010, 5, e15580. [Google Scholar] [CrossRef] [PubMed]
  101. Tekin, M.; Konca, C.; Celik, V.; Almis, H.; Kahramaner, Z.; Erdemir, A.; Gulyuz, A.; Uckardes, F.; Turgut, M. The association between vitamin D levels and urinary tract infection in children. Horm Res Paediatr 2015, 83, 198–203. [Google Scholar] [CrossRef] [PubMed]
  102. Nseir, W.; Taha, M.; Nemarny, H.; Mograbi, J. The association between serum levels of vitamin D and recurrent urinary tract infections in premenopausal women. Int. J. Infect. Dis. 2013, 17, e1121–e1124. [Google Scholar] [CrossRef] [PubMed]
  103. Sonnex, C. Influence of ovarian hormones on urogenital infection. Sex. Transm. Infect. 1998, 74, 11–19. [Google Scholar] [CrossRef] [PubMed]
  104. Epp, A.; Larochelle, A.; Lovatsis, D.; Walter, J.E.; Easton, W.; Farrell, S.A.; Girouard, L.; Gupta, C.; Harvey, M.A.; Robert, M.; et al. Recurrent urinary tract infection. J. Obstet. Gynaecol. Can. 2010, 32, 1082–1101. [Google Scholar] [PubMed]
  105. Perrotta, C.; Aznar, M.; Mejia, R.; Albert, X.; Ng, C.W. Oestrogens for preventing recurrent urinary tract infection in postmenopausal women. Cochrane Database Syst. Rev. 2008. [Google Scholar] [CrossRef]
  106. Salvatore, S.; Salvatore, S.; Cattoni, E.; Siesto, G.; Serati, M.; Sorice, P.; Torella, M. Urinary tract infections in women. Eur. J. Obstet. Gynecol. Reprod. Biol. 2011, 156, 131–136. [Google Scholar] [CrossRef] [PubMed]
  107. Eto, D.S.; Jones, T.A.; Sundsbak, J.L.; Mulvey, M.A. Integrin-mediated host cell invasion by type 1-piliated uropathogenic Escherichia coli. PLoS Pathog. 2007, 3, e100. [Google Scholar] [CrossRef] [PubMed]
  108. Zhou, G.; Mo, W.J.; Sebbel, P.; Min, G.; Neubert, T.A.; Glockshuber, R.; Wu, X.R.; Sun, T.T.; Kong, X.P. Uroplakin Ia is the urothelial receptor for uropathogenic Escherichia coli: Evidence from in vitro fimH binding. J. Cell. Sci. 2001, 114, 4095–4103. [Google Scholar] [PubMed]
  109. Wang, C.; Symington, J.W.; Ma, E.; Cao, B.; Mysorekar, I.U. Estrogenic modulation of uropathogenic Escherichia coli infection pathogenesis in a murine menopause model. Infect. Immun. 2013, 81, 733–739. [Google Scholar] [CrossRef] [PubMed]
  110. Schnarr, J.; Smaill, F. Asymptomatic bacteriuria and symptomatic urinary tract infections in pregnancy. Eur. J. Clin. Investig. 2008, 38, 50–57. [Google Scholar] [CrossRef] [PubMed]
  111. Foxman, B.; Buxton, M. Alternative approaches to conventional treatment of acute uncomplicated urinary tract infection in women. Curr. Infect. Dis. Rep. 2013, 15, 124–129. [Google Scholar] [CrossRef] [PubMed]
  112. Conrad, A.; Biehler, D.; Nobis, T.; Richter, H.; Engels, I.; Biehler, K.; Frank, U. Broad spectrum antibacterial activity of a mixture of isothiocyanates from nasturtium (Tropaeoli majoris herba) and horseradish (Armoraciae rusticanae radix). Drug Res. Stuttg 2013, 63, 65–68. [Google Scholar] [CrossRef] [PubMed]
  113. Liao, S.-G.; Zhang, L.-J.; Sun, F.; Zhang, J.-J.; Chen, A.Y.; Lan, Y.-Y.; Li, Y.-J.; Wang, A.-M.; He, X.; Xiong, Y.; et al. Antibacterial and anti-inflammatory effects of extracts and fractions from Polygonum capitatum. J. Ethnopharmacol. 2011, 134, 1006–1009. [Google Scholar] [CrossRef] [PubMed]
  114. Peng, M.-M.; Fang, Y.; Hu, W.; Huang, Q. The pharmacological activities of compound Salvia plebeia granules on treating urinary tract infection. J. Ethnopharmacol. 2010, 129, 59–63. [Google Scholar] [CrossRef] [PubMed]
  115. Albrecht, U.; Goos, K.H.; Schneider, B. A randomised, double-blind, placebo-controlled trial of a herbal medicinal product containing Tropaeoli majoris herba (nasturtium) and Armoraciae rusticanae radix (horseradish) for the prophylactic treatment of patients with chronically recurrent lower urinary tract infections. Curr. Med. Res. Opin. 2007, 23, 2415–2422. [Google Scholar] [PubMed]
  116. Rafsanjany, N.; Lechtenberg, M.; Petereit, F.; Hensel, A. Antiadhesion as a functional concept for protection against uropathogenic Escherichia coli: In vitro studies with traditionally used plants with antiadhesive activity against uropathognic Escherichia coli. J. Ethnopharmacol. 2013, 145, 591–597. [Google Scholar] [CrossRef] [PubMed]
  117. Chen, C.S.; Ho, D.R.; Chang, P.J.; Lin, W.Y.; Huang, Y.C. Urine post equivalent daily cranberry juice consumption may opsonize uropathogenicity of Escherichia coli. J. Infect. Chemother. 2013, 19, 812–817. [Google Scholar] [CrossRef] [PubMed]
  118. Gupta, K.; Chou, M.Y.; Howell, A.; Wobbe, C.; Grady, R.; Stapleton, A.E. Cranberry products inhibit adherence of P-fimbriated Escherichia coli to primary cultured bladder and vaginal epithelial cells. J. Urol. 2007, 177, 2357–2360. [Google Scholar] [CrossRef] [PubMed]
  119. Bosmans, J.E.; Beerepoot, M.A.; Prins, J.M.; ter Riet, G.; Geerlings, S.E. Cost-effectiveness of cranberries vs. antibiotics to prevent urinary tract infections in premenopausal women: A randomized clinical trial. PLoS ONE 2014, 9, e91939. [Google Scholar] [CrossRef] [PubMed]
  120. Jepson, R.G.; Williams, G.; Craig, J.C. Cranberries for preventing urinary tract infections. Cochrane Database Syst. Rev. 2012, 10, CD001321. [Google Scholar] [PubMed]
  121. Lüthje, P.; Dzung, D.N.; Brauner, A. Lactuca indica extract interferes with uroepithelial infection by Escherichia coli. J. Ethnopharmacol. 2011, 135, 672–677. [Google Scholar] [CrossRef] [PubMed]
  122. Fazliana, M.; Ramos, N.L.; Lüthje, P.; Sekikubo, M.; Holm, Å.; Wan Nazaimoon, W.M.; Brauner, A. Labisia pumila var. alata reduces bacterial load by inducing uroepithelial cell apoptosis. J. Ethnopharmacol. 2011, 136, 111–116. [Google Scholar] [PubMed]
  123. Vollmerhausen, T.L.; Ramos, N.L.; Dzung, D.T.; Brauner, A. Decoctions from Citrus reticulata blanco seeds protect the uroepithelium against Escherichia coli invasion. J. Ethnopharmacol. 2013, 150, 770–774. [Google Scholar] [CrossRef] [PubMed]
  124. Bleidorn, J.; Gagyor, I.; Kochen, M.M.; Wegscheider, K.; Hummers-Pradier, E. Symptomatic treatment (ibuprofen) or antibiotics (ciprofloxacin) for uncomplicated urinary tract infection?—Results of a randomized controlled pilot trial. BMC Med. 2010, 8, 30. [Google Scholar] [CrossRef] [PubMed]
  125. Lüthje, P.; Lokman, E.F.; Sanström, C.; Östenson, C.-G.; Brauner, A. Gynostemma pentaphyllum exhibits anti-inflammatory properties and modulates antimicrobial peptide expression in the urinary bladder. J. Funct. Foods 2015, 17, 283–292. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Lüthje, P.; Brauner, A. Novel Strategies in the Prevention and Treatment of Urinary Tract Infections. Pathogens 2016, 5, 13. https://doi.org/10.3390/pathogens5010013

AMA Style

Lüthje P, Brauner A. Novel Strategies in the Prevention and Treatment of Urinary Tract Infections. Pathogens. 2016; 5(1):13. https://doi.org/10.3390/pathogens5010013

Chicago/Turabian Style

Lüthje, Petra, and Annelie Brauner. 2016. "Novel Strategies in the Prevention and Treatment of Urinary Tract Infections" Pathogens 5, no. 1: 13. https://doi.org/10.3390/pathogens5010013

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop