You are on page 1of 13

Pediatr Nephrol (2012) 27:2017–2029

DOI 10.1007/s00467-011-2089-1

REVIEW

Susceptibility to acute pyelonephritis or asymptomatic


bacteriuria: Host–pathogen interaction in urinary tract
infections
Bryndís Ragnarsdóttir & Catharina Svanborg

Received: 7 October 2011 / Revised: 30 November 2011 / Accepted: 2 December 2011 / Published online: 12 February 2012
# IPNA 2012

Abstract Our knowledge of the molecular mechanisms of by combining information on bacterial virulence and the
urinary tract infection (UTI) pathogenesis has advanced host response.
greatly in recent years. In this review, we provide a general
background of UTI pathogenesis, followed by an update on Keywords Urinary tract infections . Bacterial virulence .
the mechanisms of UTI susceptibility, with a particular Host resistance . Genetic polymorphisms
focus on genetic variation affecting innate immunity. The
innate immune response of the host is critically important in
the antibacterial defence mechanisms of the urinary tract,
and bacterial clearance normally proceeds without sequelae. Introduction
However, slight dysfunctions in these mechanisms may
result in acute disease and tissue destruction. The symptoms Bacteria often invade the urinary tract, but successful estab-
of acute pyelonephritis are caused by the innate immune lishment of bacteriuria and infection is a much rarer event.
response, and inflammation in the urinary tract decreases Not that urinary tract infections (UTIs) are rare—they are
renal tubular function and may give rise to renal scarring, among the most common bacterial infections in man and
especially in paediatric patients. In contrast, in children with important enough to be taken very seriously by physicians
asymptomatic bacteriuria (ABU), bacteria persist without in any type of practice [1].
causing symptoms or pathology. Pathogenic agents trigger Acute pyelonephritis (APN), a kidney infection with
a response determined by their virulence factors, mediating local and systemic consequences, is the most severe form
adherence to the urinary tract mucosa, signalling through of UTI. Patients experience flank pain, fever >38.5°C and
Toll-like receptors (TLRs) and activating the defence mech- general malaise, and laboratory tests detect elevated levels
anisms. In ABU strains, such virulence factors are mostly of circulating acute phase reactants [C-reactive protein
not expressed. However, the influence of the host on UTI (CRP)] as well as evidence of local inflammation in the
severity cannot be overestimated, and rapid progress is urinary tract, such as pyuria and elevated cytokine levels
being made in clarifying host susceptibility mechanisms. [2]. About 30% of adults with acute pyelonephritis develop
For example, genetic alterations that reduce TLR4 function bacteremia, and urosepsis remains an important cause of
are associated with ABU, while polymorphisms reducing death.
IRF3 or CXCR1 expression are associated with acute py- Asymptomatic bacteriuria (ABU), defined by the pres-
elonephritis and an increased risk for renal scarring. It ence of a large number of bacteria in the urine (>105 cfu/ml
should be plausible to “individualize” diagnosis and therapy urine), often for extended periods of time, without the ac-
companying symptoms of a UTI, is the other extreme of the
B. Ragnarsdóttir : C. Svanborg (*) UTI spectrum [3–5]. ABU is also the most common form of
Institute of Laboratory Medicine, Section of Microbiology, UTI, occurring in about 1% of girls, 2% of pregnant women
Immunology and Glycobiology (MIG), Lund University,
and 20% of both men and women above 70 years of age [3,
Sölvegatan 23,
22362, Lund, Sweden 6–8]. Patients with ABU may develop a mild host response
e-mail: Catharina.Svanborg@med.lu.se that can be quantified in the urine, and after long-term
2018 Pediatr Nephrol (2012) 27:2017–2029

carriage they may develop lymphoid follicles in the bladder and other adaptation strategies and by resistance to the many
mucosa, which resolve after antibiotic therapy [9]. and intricate antibacterial defense mechanisms that guard
Acute cystitis is very common, yet not well defined. At the urinary tract against infection. Escherichia coli (E. coli)
least half of all women experience at least once a sporadic bacteria cause 80–90% of community-acquired acute pyelo-
cystitis episode, accompanied by frequency, dysuria and nephritis episodes and are especially common in children.
suprapubic pain. Treatment consists of antibiotic therapy, Less common infectious agents include Proteus mirabilis
but lower tract symptoms may also precede an upper urinary and Klebsiella as well as Staphylococcus saprophyticus.
tract infection, creating a grey zone between the groups with Nosocomially acquired infections may be caused by many
symptomatic UTI [10]. different organisms, depending on the hospital environment
So why do not all of us develop UTI daily when bacteria and underlying host factors [10].
accidentally gain access to the urinary tract? The answer is The subset of E. coli strains that causes acute pyelone-
to be sought in the properties of the bacteria; virulent uro- phritis in otherwise healthy hosts is designated as “uropa-
pathogenic bacteria that cause acute pyelonephritis are for- thogenic” [12] and defined by packages of chromosomal
tunately not often represented in the perineal flora, except in virulence genes, forming so-called “pathogenicity islands”
highly UTI-prone individuals who carry virulent bacterial (Figs. 1, 2) [13, 14]. The sequential activation of those genes
strains in their fecal/perineal flora for extended periods of facilitates host tissue attack and bacterial survival. For ex-
time. Furthermore, most hosts have an intact urine flow and ample, fimbriae initiate tissue attack through bacterial adhe-
a highly efficient innate immune response that senses the sion to the mucosa [12] and increase tissue exposure to
presence of invaders and destroys them, so that the urinary secreted bacterial virulence factors, such as hemolysin and
tract can regain its sterility [11]. lipopolysaccharide (LPS). Such toxins damage the tissues
In this review, we focus on the determinants of bacterial by interfering with cellular functions or by directly inducing
virulence factors and on host response dysfunction that cell death. The “uropathogenic” strains can be identified by
overrides the normally efficient defence and tilts the balance their surface antigen expression (OKH serotypes) or by
towards inflammation, symptoms and disease. typing for virulence factors like P-fimbriae [12, 15, 16].
In recent years, cellular and molecular mechanisms that
define the antibacterial host defence system and the extent
Pathogenesis—an overview of tissue damage have been extensively studied [17]. Early
studies in the murine UTI model (Fig. 3) as well as clinical
Before invading the urinary tract, bacteria causing UTI often observations (Fig. 4) have clearly shown that resistance to
reside for long periods in the intestinal flora. After spreading infection relies on innate immune mechanisms [18]. Defects
via the perineum to the periurethral area, they ascend against in innate immunity drastically increase UTI susceptibility,
the urine flow and succeed in establishing bacteriuria facil- both in mice carrying single gene deletions and in patients
itated by adhesion mechanisms, flagella-mediated motility prone to acute pyelonephritis or ABU (Table 1) [19–25]. In

Fig. 1 Summary of the


symptoms and signs of urinary tract
infections (UTIs) and of correlating
bacterial virulence factors. Acute
pyelonephritis (APN) is a severe
infection of the kidneys that may
become life threatening if left
untreated. Asymptomatic
bacteriuria (ABU) results in a
symptom-free bacterial coloni-
zation of the bladder. The disease
severity reflects differences in
bacterial virulence, with APN
strains having multiple virulence
factors, such as P-fimbriae,
toxins and proteins such as TcpC
that inhibit an efficient innate
immune response. In contrast,
ABU strains lack many of the
virulence factors seen in APN
strains due to point mutations or
deletions
Pediatr Nephrol (2012) 27:2017–2029 2019

Fig. 2 Schematic of the urinary


tract mucosal barrier and
immune cells involved in the
host response. Uropathogenic
Escherichia coli elicit a
complex innate immune
response leading to the
recruitment of inflammatory
cells and to the possibility of a
subsequent specific immune
response. In contrast,
asymptomatic carrier strains do
not elicit a significant innate
immune response and do not
trigger tissue inflammation.
Modified figure from
Ragnarsdóttir et al. [17] with
the permission of Nature
Reviews Urology. PMNs
Polymorphonuclear leukocytes

contrast, the defence against acute UTI does not rely on that they recognize. Type l fimbriae bind to mannosylated
specific immunity defined by antigen-specific lymphocyte epitopes present in the Tamm–Horsfall glycoprotein, in
activation [26]. secretory immunoglobulin A (IgA), in bladder cell uropla-
kins or in integrin molecules [28–32]. S-fimbriae bind sialic
acid epitopes present in renal sialylated proteins and lipids
Bacterial virulence factors [33]. P-fimbriae recognize Galα1–4Galβ-epitopes in the
globoseries of glycolipids [34]. The expression of P-
Bacterial adhesion Adherence to cells in the urinary tract is fimbriae is strongly linked to virulence, being expressed
a critical step to initiate tissue attack [12, 27]. Mediated by by about 80% of strains causing uncomplicated acute py-
bacterial surface fimbriae that recognize specific receptors elonephritis but in only <20% of children with ABU [35,
on host cells, bacteria become associated with the uroepi- 36]. The frequency of P-fimbriated strains is reduced in
thelial lining, and the adherent state facilitates the action of patients with impaired resistance, such as pregnancy, urody-
toxins and invasion factors. Several fimbrial types may be namic defects or genetic abnormalities (see below).
involved in the attachment process, and these are distin-
guished by their structural features as well as by the oligo- Lipopolysaccharide Lipopolysaccharide is an endotoxin of
saccharide sequences in host glycolipids or glycoproteins Gram-negative bacteria, with the lipid A anchored in the

Fig. 3 Innate rather than


adaptive immunity is essential
for bacterial clearance during
urinary tract infections (UTIs).
In the murine model, Tlr4−/− as
well as Myd88−/− and Trif−/−
mice develop asymptomatic
carriage while mCxcr2−/− and
Irf3−/− mice are highly
susceptible and associated with
severe disease and increased
mortality. Defects of the
specific immune response, in
contrast, did not increase
urinary tract infection (UTI)
susceptibility (T, B cell or RAG
mice) [18, 19, 25]
2020 Pediatr Nephrol (2012) 27:2017–2029

Fig. 4 Polymorphisms in three human genes (TLR4, CXCR1 and expression in APN-prone patients is the intronic variant at position
IRF3) have been associated with urinary tract infection (UTI) suscep- G217C. The A/A-CC genotype for IRF3 promoter SNPs is associ-
tibility. Functional studies have confirmed their effect on gene expres- ated with APN, resulting in lower IRF3 promoter activity. Modi-
sion. Single nucleotide polymorphisms (SNPs) that have been shown fied figure from Ragnarsdóttir et al. [17] with the permission of
to result in lower TLR4 promoter activity, in vitro, are -4038, -3612, -2081 Nature Reviews Urology
and -2026. An SNP that has been shown to reduce CXCR1

outer membrane being responsible for the toxic effects. occur more frequently in E. coli causing acute pyelonephri-
Injection of lipid A mimics many of the symptoms associ- tis than in fecal isolates; however, the difference in frequen-
ated with acute pyelonephritis, Gram-negative septicemia or cy is less pronounced than that for P-fimbriae [43].
other serious Gram-negative infections, including fever and
activation of the acute phase response. In addition, LPS Metabolic competition Bacteria compete with the host for
carries an invariable oligosaccharide core and a repeating many nutrients, including iron. Lactoferrins are synthesized
oligosaccharide that determines the O antigen. LPS activates by the mucosa and transferrin is present in the circulation,
Toll-like receptor 4 (TLR4) signalling, after binding to with concentrations increasing in response to infection [44].
soluble or cell surface-associated CD14, in conjunction with Although no single iron uptake system is essential, all
MD2 [37, 38]. This mechanism is essential for innate im- uropathogenic strains express some form of iron uptake.
mune activation during systemic infection but is less impor- Enterobactin is expressed by nearly all E. coli strains, but
tant at mucosal surfaces, where the cell-bound CD14 most E. coli strains causing acute pyelonephritis produce
receptor is often missing [39, 40]. aerobactin, which is a high-affinity iron-binding protein, as
well as other iron-sequestering proteins, such as yersinia-
Capsular polysaccharide Capsular polysaccharides are bactin, ChuA and Iro [45–48].
formed from oligosaccharide polymers. Different capsular
types are defined by their variant antigenic oligosaccharide
epitopes. The capsules surround bacteria in blood and tissues
and have been shown to contribute significantly to bacterial Host response and the antibacterial defense
survival in the presence of a functional host defense by coun-
tering lytic effects of complement and phagocytosis [41]. The innate immune response is activated in a pathogen-
Mutant bacteria with genetically altered capsule expression specific way by P-fimbriae-mediated adhesion. Subsequent
show reduced virulence in experimental UTI models [42]. cleavage of the glycolipid receptors releases ceramide,
which activates TLR4 signalling, and transcription factors
Hemolysin Hemolysins are cytotoxic, pore-forming proteins such as IRF3 trigger cytokine production and neutrophil
that permeate the cell membrane. They are commonly iden- recruitment, which in turn kill the bacteria [25, 49–51].
tified by their ability to lyse erythrocytes. Hemolysin pro- These mechanisms also determine the symptoms and signs
duction was first observed in the 1940s in E. coli causing of acute UTI. The loss of functional TLRs reduces the innate
acute pyelonephritis, and hemolysin have been found to immune response and promotes long-term bacterial
Pediatr Nephrol (2012) 27:2017–2029 2021

26, 52, 94]

TGF-β1, Transforming growth factor-beta1; TLR4, Toll-like receptor 4; SNP, single nucleotide polymorphism; UTR, untranslated region; PBMC, peripheral blood mononuclear cell; ABU,
[21, 22, 62]
[18, 23, 24,
colonization and ABU [18, 24, 52]. In contrast, dysfunc-

References

[95–98]
tional, exaggerated responses result in severe acute infec-

[25]
tions with inflammation (Fig. 5). The resulting defects in
antibacterial effector functions enable the infection to be-
come chronic, and rapid renal scarring occurs in animals
Disease association

with deficiencies in these pathways.

Renal scarring
TLR4 signalling activates the cytokine response in
infected uroepithelial cells, and secreted cytokines orches-
trate mucosal inflammation. Indeed, the presence of a mu-
ABU

APN
APN

APN cosal cytokine response was first observed in UTI. Interleukin


(IL)-6 is secreted by uroepithelial cells and acts as an endog-
enous pyrogen, activates hepatocyte CRP production and

SNPs to RS but no correlation between


PBMC TGF-β1 production was lower in
increased mRNA 3′-processing, reduced

stimulates mucosal B cells to produce IgA antibodies [53,

SNPs and circulating TGF-β1 levels


levels of the long CXCR1 transcript
and low innate response in patients.

Association of promoter and coding 54]. The biological activities of IL-6 thus agree with the
clinical presentation of acute pyelonephritis, and IL-6 is
promoter SNP heterozygotes

detected in the blood and urine of patients with acute pyelo-


Reduced promoter activity,

Reduced promoter activity

Reduced promoter activity


low TLR4 expression

nephritis. Several studies have suggested that urine cytokine


levels directly reflect the degree of renal involvement.
Biological effect

Infected epithelial cells also secrete IL-8, which is che-


motactic for neutrophils. IL-8 forms a gradient across
the mucosa, and neutrophils migrate towards the gradi-
ent and cross the mucosal barrier into the urine. The
concentration of IL-8 correlates with the number of
leucocytes in urine, and the chemotactic activity of
CXCR1 SNP in 3′ UTR region

urine is inhibited by IL-8 antibodies [27, 55]. In addi-


Genetic variants in humans

tion, infection increases the expression of IL-8 receptors,


IRF3 promoter (A/A-C/C)
TLR4 promoter SNPs/GP

which are important targets for the IL-8 family of che-


TGF-β1 promoter and
coding region SNPs
CXCR1 intronic SNP

mokines and support neutrophil migration and activation.


These mechanisms explain the classical phenomenon called
“pyuria”, i.e. the presence of leucocytes in urine, which has
Humans

been used as a sign of UTI since microscopy started to be used


for clinical diagnosis. More recently, a number of additional
chemokines have been found in urine, probably reflecting a
more complex immune response in the mucosa, especially
Acute infection and urosepsis. Extensive

from multifocal, mixed inflammatory

during long-term bacterial carriage [56].


Table 1 Genetic variation and urinary tract infection susceptibility

Mice with acute pyelonephritis have


infection and urosepsis. Extensive
Delayed neutrophil response. Acute

It is not clear if the urine flow, per se, is sufficient to


Uninfected TGF-β1−/− mice suffer
marked kidney cell expression

cell response, tissue necrosis,

maintain sterility. Clearly, the efficiency with which the


abscesses in surviving mice.

abscesses in surviving mice.


delayed bacterial clearance

urinary tract maintains sterility is aided by antibacterial


Low innate response and

organ failure and death

peptides that directly kill the bacteria (for review see [57]).
asymptomatic bacteriuria; APN, acute pyelonephritis
of TGF-β1 mRNA.

In an epidemiologic study of sensitivity to the human cath-


Biological effect

elicidin LL-37, APN strains were found to be more resistant


than lower urinary tract isolates [58]. The importance of
antibacterial peptides is evident in the murine model as well.
Mice have only one urinary tract cathelicidin, CRAMP
(cathelin-related, antimicrobial peptide), and knock-out
mice for this gene were more susceptible to UTI than
Genetically modified mice

wild-type mice [58].


(non-functional Tlr4)
Tlr4−/− or C3H/HeJ

Host factors predispose to UTI


mCxcr2−/−

TGF-β1
Irf3−/−

The frequency of UTI varies with age, gender and socio-


Mice

economic factors. The susceptibility to UTI is higher in


2022 Pediatr Nephrol (2012) 27:2017–2029

Fig. 5 The P-fimbriae on uropathogenic E. coli are essential for the shaded area (Tlr4, Myd88 or Trif) protect against symptomatic infec-
activation of CD14-negative epithelial cells. P-fimbriae use the PapG tions, and infected hosts develop asymptomatic bacteriuria with no
tip adhesin to bind oligosaccharide sequences in the globoseries of evidence of tissue pathology. Gene deletions in the red-shaded area
glycosphingolipids. After binding, the receptors are cleaved, and the (Irf3 or Ifnβ) exacerbate the severity of acute pyelonephritis, with
release of ceramide triggers Toll-like receptor 4 (TLR4) signalling. increased mortality, followed by rapid tissue damage. Modified figure
Defects in the TLR4 signalling pathway greatly affect the host immune from Ragnarsdóttir et al. [17] with permission of Nature Reviews
response and therefore disease outcome. Gene deletions in the green- Urology

women than in men, except during the first year and at the innate immune signalling results in an immunodeficient phe-
end of life. Still more noteworthy are the differences in UTI notype, with increased UTI susceptibility in mice and poly-
susceptibility across these lines, however. Certain individu- morphisms in patients with different forms of UTI.
als develop severe, acute infections, some are likely to suffer Predisposition to acute pyelonephritis is also influenced
recurring UTIs and a subset develops a chronic state of by dysfunctional voiding or vesicoureteric reflux (VUR)
tissue damage, leading to renal scarring. Scientists have [59]. Patients with such dysfunctions have an increased
been working for decades to better understand the factors frequency of UTI and are susceptible to infection with
that determine susceptibility and to develop diagnostic tools bacteria of lower virulence than patients with anatomically
to discriminate these individuals from others who only expe- normal urinary tracts [60].
rience sporadic UTI episodes. A number of genetic risk factors
have now been identified that distinguish pyelonephritis-
prone children from those who consistently develop ABU
(for a review, see [17]). Even if the information is still limited, Genetic variation and ABU susceptibility
it is clear that genetic variation that particularly affects innate
immunity influences UTI susceptibility. These examples The effects of a number of genetic risk factors and suscep-
show that the deletion of single genes with crucial functions in tibility to UTIs are summarized in Table 1.
Pediatr Nephrol (2012) 27:2017–2029 2023

Reduced TLR signalling and ABU As outlined above, the number of genotype patterns and the proportion of specific
activation of TLR signalling and the resulting production of promoter genotypes differed compared to the background
mediators of innate immunity, including cytokines/chemo- population. In contrast, the APN patient group carried the
kines, are crucial for the elimination of bacteria from the eight single nucleotide polymorphisms (SNPs) and did not
urinary tract. Without TLR4, the production of antimicrobial differ in SNP frequency from the paediatric controls.
peptides is abrogated, and neutrophil recruitment is reduced, A few other studies have focused on ABU, albeit with
as is their activation to engage in bacterial phagocytosis and different diagnostic criteria. Decreased TLR4 expression was
to recruit a second wave of inflammatory cells that are detected in patients with a history of recurrent UTIs compared
needed to rid the tissues of debris from dying phagocytic with controls [63]. An increased frequency of three CXCR1
cells. Pharmacological or genetic inhibition of neutrophil polymorphisms was detected in women with Gram-positive
migration/activation has been shown to drastically impair ABU, [64] but their history of childhood APN was not exam-
the antibacterial defense, resulting in poor bacterial clear- ined. A TLR2 polymorphism (2258G > A) was also associated
ance and drastic tissue pathology [21, 61, 62]. Still, genetic with ABU [64].
defects affecting TLR signalling seem to be protective In the murine UTI model, defects in the TLR adaptor
and mainly associated with ABU, suggesting that just molecules Trif/Tram and MyD88 have also been shown to
reducing the efficiency of bacterial clearance does not protect against symptomatic UTI [24], but to date human
create pathology—when the tissue inflammation that causes polymorphisms in the corresponding genes have not been
the pathology is blocked. Mice lacking the Tlr4 gene devel- associated with UTI susceptibility.
op ABU, with >105 cfu/ml of bacteria, but no symptoms or
tissue damage. Lack of murine Tlr4 expression and the result-
ing absence of inflammation thus appear to be protective
rather than destructive [18, 24, 52]. Genetic variation in acute pyelonephritis
It was therefore not surprising to find that children with
ABU express lower levels of the TLR4 than age-matched IRF3 polymorphisms P-fimbriated E. coli activate TLR4
controls or children with acute pyelonephritis [52]. However, signalling. and the IRF3/7 transcription factor enhances
the genetic basis for low TLR4 expression is not explained by the transcription of innate immune response genes, in-
structural gene variation, but rather by mutations in the TLR4 cluding cytokines and chemokine receptors. Mice lacking
promoter that reduce the efficiency of TLR4 expression [23]. the Irf3 gene develop severe acute pyelonephritis with
We examined TLR4 promoter sequences in two groups of urosepsis and renal abscesses (Table 1; Fig. 6) [25] due
highly selected UTI-prone patients with either a consistent to deficient activation of downstream antibacterial effec-
pattern of ABU or APN during at least 5 years of follow-up tor functions, such as those dependent on interferons
[23]. The patients were compared to paediatric and adult alpha and beta (INFα, INFβ).
controls without UTI. The TLR4 promoter sequences differed Based on the predictions from the mouse model, we
between asymptomatic carriers and controls or patients with genotyped pyelonephritis-prone patients for IRF3 promoter
severe disease. The primary ABU patients also had a reduced polymorphisms [25]. The results of that study showed that

Fig. 6 Renal tissue damage in


Irf3−/− mice 7 days after
infection with uropathogenic E.
coli strain 1177. Arrows mark
areas with tissue abscesses.
Virulent strains activate TLR4
signalling and the nuclear
translocation of IRF3. Mice
lacking the Irf3 gene are
therefore highly susceptible,
and surviving mice develop
abscesses and tissue damage
2024 Pediatr Nephrol (2012) 27:2017–2029

the IRF3 genotype present in pyelonephritis-prone children confirmed in patients with APN onset before 15 years of
and adults reduced IRF3 promoter activity. IRF3 promoter age, and reduced CXCR2 levels were found in girls with
sequence variation was detected in two highly UTI-prone recurrent UTI. IL-8 (CXCL8) SNPs were detected in patients
patient populations. One included children in southern Swe- with confirmed APN without VUR, further supporting a role
den who had a consistent UTI pattern over several years: of chemokines and their receptors in the antibacterial de-
either severe recurrent APN or ABU, with no prior symp- fense against UTI and the addition of CXCR2 and CXCL8 to
tomatic infection. These children were identified on the the susceptibility gene repertoire.
basis of a prospective, long-term follow-up of a larger
patient group. The second patient group comprised adults Other polymorphisms A CCL5 variant has been shown to
with a history of childhood febrile UTI who were enrolled in differ between APN-prone children and controls [68]. A
a prospective study in the 1970s and re-evaluated after about TNF variant (-238G/A) showed increased frequency in
30 years. Controls were children without UTI-related mor- UTI-prone patients with early rheumatoid arthritis [69]. In
bidity and adult blood donors from the same geographic Egyptian children with APN, two transforming growth
areas. factor-beta 1 (TGF-β1) variants and two detected vascular
DNA sequencing of IRF3 promoters from these UTI endothelial growth factor (VEGF) promoter variants were
patients revealed variation at the -925 and -776 positions. more frequent in patients with scarring than in healthy
SNPs -925 and -776 were linked in the study population controls, but were not linked to VUR [70]. Both TGF-β1
(r2 01.0), but the IRF3 genotype varied with UTI severity and VEGF promoter variants were associated with renal
and between cases and controls. In the paediatric group, scarring in Irish children [71] but not in a group of Asian
most of the APN patients were homozygous for the two patients with renal scarring, indicating variation between
positions, a finding which was confirmed in the adult patients, ethnic groups [72]. In addition to promoter polymorphisms,
with 75% homozygous and 13% heterozygous SNPs com- genetic variants affecting coding regions of lymphotoxin α
pared to 53 and 37%, respectively, in controls. In addition, the [69] and TLR5 1174C > T [73] have been associated with
minor allele frequency was decreased in APN compared to recurrent UTI. Numerous other SNPs have been examined,
primary ABU and controls. without significant UTI associations.
To examine if IRF3 promoter variation influences tran-
scription efficiency, we cloned promoters from one patient Receptors, P blood group and UTI susceptibility Uropatho-
with APN and one with ABU into a luciferase reporter genic E. coli colonize mucosal surfaces through adhesion;
vector. Transcriptional activity from the vector with the consequently, resistance to infection can be modified by the
APN promoter was about 50% lower than that from the receptor repertoire of the patients and by soluble anti-
vector with the ABU promoter, and this difference was adhesive molecules. Early studies by Stamey et al. [74] led
due to the polymorphic sites. The results show that the to the proposal that women with recurrent UTIs have in-
IRF3 promoter efficiency is reduced by the SNPs occurring creased vaginal carriage of uropathogenic bacteria. Several
in about 80% of APN patients, which is consistent with the research groups subsequently demonstrated that the uroepi-
finding of human SNPs reducing IRF3 expression and in- thelial cells of these women to have an increased tendency
creasing the risk for APN. to bind uropathogenic E. coli, reflecting an increased density
of receptors [60, 75–77].
CXCR1 polymorphisms In 2000, we showed that mice lack- P-fimbriated E. coli bind to the globoseries of glycoli-
ing the IL-8 receptor develop severe acute pyelonephritis pids, which are antigens in the P blood group system.
due to a dysfunctional neutrophil response, with urosepsis Through P blood group determination, it is possible to
and surviving animals showing renal abscess formation and predict the mucosal receptor repertoire and, therefore, the
renal scarring [19]. P blood group can be used as a marker in epidemiologic
Based on the predictions from the mouse model, we also studies. Individuals with blood group P lack both a
investigated whether the IL-8 receptor (CXCR1 and critical enzyme involved in P blood group synthesis
CXCR2) is polymorphic in pyelonephritis-prone patients. and functional receptors for P-fimbriae, leading to the
Reduced expression of the IL-8 receptor, CXCR1, was prediction that they would be more resistant to P-
found in children prone to acute pyelonephritis, and the fimbriated bacterial infection [34]. Children with blood
finding of heterozygous CXCR1 polymorphisms and low group P1 run an increased risk of acute pyelonephritis
mRNA levels suggested that transcription is impaired in this (about 11-fold) and have increased carriage of P-fimbriated
patient group [19, 22, 65]. E. coli in their intestinal flora, which is a UTI risk
Variants of CXCR1 and related genes have subsequently factor [60, 75, 76]. Epithelial receptor expression is also
been identified in patient groups with different diagnostic influenced by the ABO blood group, and individuals express-
criteria [63, 64, 66, 67]. Reduced CXCR1 expression was ing globo-A are preferentially infected by P-fimbriated E. coli
Pediatr Nephrol (2012) 27:2017–2029 2025

that recognize a combined Gala1–4Galb and blood group generated through vaccination studies. Early studies showed
A epitope [77]. that vaccination with formalin-treated whole bacteria mark-
edly increases specific antibody levels. Subsequent studies
Anti-adhesive molecules in urine Urine contains soluble have used purified antigens (LPS, kapsel, fimbriae, outer
receptor molecules that inhibit bacterial adhesion. Soluble membrane proteins) [85–87]. The results clearly show that
receptor oligosaccharides or glycoconjugates bind to fimbri- hyper-immunization can protect against infection, provided
ae and prevent them from mediating adhesion. The Tamm– that the vaccine antigen is expressed by the bacterial strain
Horsfall protein, which is a constituent of normal urine, used for infection and that infectious challenge is given
carries terminal mannose residues that bind type 1- while the immune response remains active. While the anti-
fimbriated E. coli [78], as well as sialic acid residues that genic variation in uropathogenic E. coli is a problem for the
bind S-fimbriae [79, 80]. P-fimbriae, which show the implementation of vaccination in UTI, the development of
strongest disease association, have no known soluble inhib- novel vaccines gives rise to new optimism. Genetic studies
itors in urine. also suggest that genetically susceptible individuals might
be targeted by vaccination, thus identifying a suitable target
population.

Inheritance of UTI susceptibility


Clinical implications
Urinary tract infections have been shown to run in families,
and especially sisters, mothers and grandmothers of the The efficiency of antibiotics in the treatment of symptomatic
patient have a history of UTI. The inheritance of APN UTI has limited interest in new diagnostic and therapeutic
susceptibility has been examined by surveying several gen- approaches to UTI. However, with the increasing frequency
erations of families of APN-prone children [65]. An in- of antibiotic resistance among uropathogens, attitudes are
creased frequency of APN was detected in both male and changing. It is fortunate that a large body of molecular
female family members, and the latter also had lower levels information has been gathered, and the current challenge is
of CXCR1 expression than controls, indicating that CXCR1 to implement the use of such new tools in clinical practice,
expression influences APN susceptibility. However, the fre- not least when hospital administrative structures and cost
quency of cystitis was similar in both groups, emphasizing awareness challenge each new diagnostic test and therapeu-
that APN and cystitis are controlled by different pathoge- tic expense. In view of the frequency of UTI and the con-
netic mechanisms. Scholes et al. detected that susceptibility sequences of long-term sequelae, it should be easy to defend
to acute cystitis susceptibility is inherited [81]. Another a more individualized diagnostic approach and to explore
study showed that 42% of family members of patients with how prophylaxis and therapy may be diversified to better fit
a history of lower UTI were UTI prone compared to 11% of the needs of individual patients.
controls [82]. In individuals with a family history of UTI,
there is also an increased influence of behavioral factors Bacterial virulence The new molecular tools make it possi-
[83]. ble to distinguish virulent bacteria from avirulent strains
based on their virulence factors. Many candidate markers
or combinations of markers are potential tools for identify-
Specific immunity and resistance to UTI ing virulent uropathogenic strains, but to date P-fimbrial
expression shows the strongest association with virulence,
Acute pyelonephritis gives rise to a specific immune re- defined as the ability to cause acute pyelonephritis in a child
sponse after 3–7 days. The mucosal immune response has without abnormalities of the urinary tract or other predis-
a relatively short memory, however, and most patients there- posing conditions. P-fimbriae occur in 70–90% of acute
fore have low levels of specific antibodies in the urine, even pyelonephritis strains and in up to 100% of adults with
when they suffer from recurrent infections. In experimental urosepsis. In human inoculation studies, the expression of
studies, mice lacking a specific immune system have sur- P-fimbriae alone has been shown to increase the ability of a
prisingly not shown a drastic increase in UTI susceptibility non-virulent ABU strain to establish bacteriuria and to trig-
[26, 84], and patients with different specific immuno- ger the innate host response that underlies the development
deficiencies do not primarily develop UTI, suggesting that of symptoms [88–90].
the specific immune system is mainly involved once the P-fimbrial detection in the laboratory is based on agglu-
innate immune response has failed to clear the infection. tination of human P blood group-positive erythrocytes or
The main evidence suggesting that specific immunity receptor-coated particles, and the test is simple to imple-
may protect the urinary tract against infection has been ment. Uropathogenic E. coli strains may be identified by P-
2026 Pediatr Nephrol (2012) 27:2017–2029

fimbrial expression. Detecting a P-fimbriated strain in a predict future risk of acute pyelonephritis. Alternately, pro-
child with a typical case of acute pyelonephritis would be tection from symptomatic UTI, resulting from TLR4 poly-
confirmatory, while infection with a P-fimbriae negative morphisms and low innate immune signalling, might be
strain might suggest host complications that need to be helpful to identify ABU patients who may safely be left
investigated further. Early studies showed, for example, that untreated from those requiring prophylaxis.
children who develop renal scars are infected with P- As family studies have supported the premise that UTI
fimbriae-negative strains more often than children with a susceptibility may be inherited, a thorough family history is
single episode of acute pyelonephritis without scar forma- essential for risk assessment in children with UTI. IRF3
tion [91]. As genetic aberrations have now been shown to promoter polymorphisms may be used to assess the risk
cause dysfunctional inflammation in the mouse model, a for recurrent acute pyelonephritis and renal scarring. Con-
knowledge of the virulence profile of the infecting strain versely, TLR4 promoter polymorphisms associated with re-
might also help in the clinical evaluation of a child with duced TLR4 expression might be used to identify patients
atypical symptoms. who preferentially develop asymptomatic bacteriuria.

Host susceptibility Susceptibility to UTI is usually defined


in childhood, and with long-term follow-up the infection pat-
tern with repeated, febrile infections will reveal who is prone to References
developing acute pyelonephritis. Some UTI-prone children
consistently develop ABU with no symptomatic episodes in 1. Stamm WE, Norrby SR (2001) Urinary tract infections: disease
between (primary ABU), while others develop ABU subse- panorama and challenges. J Infect Dis 183(Suppl 1):S1–S4
quent to a preceding symptomatic episode (secondary ABU). 2. Svanborg C, Bergsten G, Fischer H, Frendeus B, Godaly G,
There has been a lack of tools to distinguish patients at risk of Gustafsson E, Hang L, Hedlund M, Karpman D, Lundstedt AC,
Samuelsson M, Samuelsson P, Svensson M, Wullt B (2001) The
recurrent severe infections and future renal scarring from those
‘innate’ host response protects and damages the infected urinary
who will be protected by long-term ABU and who should be tract. Ann Med 33:563–570
left untreated. There is also a need for suitable parameters that 3. Lindberg U, Claesson I, Hanson LA, Jodal U (1975) Asymptom-
limit the use of invasive procedures to the patient group that atic bacteriuria in schoolgirls. I. Clinical and laboratory findings.
Acta Paediatr Scand 64:425–431
will benefit the most from these procedures.
4. Kass EH (1956) Asymptomatic infection of the urinary tract. Trans
Susceptibility is influenced by many different variables Assoc Am Physicians 69:56–63
and, clearly, patients with malformations or mechanic dys- 5. Wullt B (2003) The role of P fimbriae for Escherichia coli estab-
functions leading to obstructions or VUR should be consid- lishment and mucosal inflammation in the human urinary tract. Int
J Antimicrob Agents 21:605–621
ered as a separate group. However, most children with UTI
6. Stenquist K, Dahlén-Nilsson I, Lidin-Janson G, Lincoln K, Odén
do not have any underlying mechanic defects. Innate im- A, Rignell S, Svanborg-Edén C (1989) Bacteriuria in pregnancy:
mune response biomarkers provide useful diagnostic tools frequency and risk of acquisition. Am J Epidemiol 129:372–379
to evaluate UTI severity and site of infection. It may be 7. Wettergren B, Jodal U, Jonasson G (1985) Epidemiology of bac-
teriuria during the first year of life. Acta Pediatr Scand 74:925–933
difficult to distinguish acute pyelonephritis from meningitis
8. Nordenstam G, Branberg Å, Odén A, Svanborg-Edén C, Svanborg
or pneumonia in infants with unclear symptoms, elevated A (1986) Bacteriuria and mortality in an elderly population. N
CRP and bacteriuria, but cytokines may be quantified in the Engl J Med 314:1152–1156
urine. IL-6 and IL-8 are produced by infected cells in the 9. Marsh FP, Banerjee R, Panchamia P (1974) The relationship be-
tween urinary infection, cystoscopic appearance, and pathology of
urinary tract and elevated levels accompany symptomatic
the bladder in man. J Clin Pathol 27:297–307
infections. In ABU, IL-6 is rarely elevated and IL-8 levels 10. Kunin C (1987) Urinary tract infections. Detection, prevention and
are low. Furthermore, high inflammatory volume from management, 5th edn. Williams & Wilkins, Baltimore
dimercaptosuccinic acid scans in children with acute pyelo- 11. Ragnarsdottir B, Fischer H, Godaly G, Gronberg-Hernandez J,
Gustafsson M, Karpman D, Lundstedt AC, Lutay N, Ramisch S,
nephritis increases the risk of renal scarring [92, 93]. Tools
Svensson ML, Wullt B, Yadav M, Svanborg C (2008) TLR- and
for more extensive cytokine proteomic profiling in urine are CXCR1-dependent innate immunity: insights into the genetics of
also becoming available. urinary tract infections. Eur J Clin Invest 38(Suppl 2):12–20
We propose that the new genetic tools should be applied 12. Svanborg C, Hanson LA, Jodal U, Lindberg U, Akerlund AS
(1976) Variable adherence to normal human urinary-tract epithelial
as widely as possible in children with UTI. As in cystic
cells of Escherichia coli strains associated with various forms of
fibrosis, multiple genetic alterations may be expected to urinary-tract infection. Lancet 1:490–492
converge on the susceptible phenotype in APN-prone 13. Yamamoto S (2007) Molecular epidemiology of uropathogenic
patients. While more extensive, prospective clinical studies Escherichia coli. J Infect Chemother 13:68–73
14. Oelschlaeger TA, Dobrindt U, Hacker J (2002) Virulence factors of
are needed to define the sensitivity and specificity of such
uropathogens. Curr Opin Urol 12:33–38
tests, the known genetic loci that modify the efficiency of 15. Evans DJ Jr, Evans DG (1983) Classification of pathogenic
the innate immune response are strong candidate tools to Escherichia coli according to serotype and the production of
Pediatr Nephrol (2012) 27:2017–2029 2027

virulence factors, with special reference to colonization-factor 32. Eto DS, Jones TA, Sundsbak JL, Mulvey MA (2007) Integrin-
antigens. Rev Infect Dis 5(Suppl 4):S692–S701 mediated host cell invasion by type 1-piliated uropathogenic
16. Orskov I, Orskov F, Jann B, Jann K (1977) Serology, chemistry, and Escherichia coli. PLoS Pathogens 3:e100
genetics of O and K antigens of Escherichia coli. Bacteriol Rev 33. Parkkinen J, Virkola R, Korhonen TK (1983) Escherichia coli
41:667–710 strains binding neuraminyl α2-3 galactosides. Biochem Biophys
17. Ragnarsdottir B, Lutay N, Gronberg-Hernandez J, Koves B, Res Commun 11:456–461
Svanborg C (2011) Genetics of innate immunity and UTI 34. Leffler H, Svanborg-Edén C (1980) Chemical identification of a
susceptibility. Nat Rev Urol 8:449–468 glycosphingolipid receptor for Escherichia coli attaching to human
18. Hagberg L, Hull R, Hull S, McGhee JR, Michalek SM, Svanborg- urinary tract epithelial cells and agglutinating human erythrocytes.
Edén C (1984) Difference in susceptibility to Gram-negative uri- FEMS Microbiol Lett 8:127–134
nary tract infection between C3H/HeJ and C3H/HeN mice. Infect 35. Leffler H, Svanborg-Edén C (1981) Glycolipid receptors for uro-
Immun 46:839–844 pathogenic Escherichia coli on human erythrocytes and uroepithe-
19. Frendeus B, Godaly G, Hang L, Karpman D, Lundstedt AC, lial cells. Infect Immun 34:920–929
Svanborg C (2000) Interleukin 8 receptor deficiency confers sus- 36. Plos K, Connell H, Jodal U, Marklund B, Mårild S, Wettergren B,
ceptibility to acute experimental pyelonephritis and may have a Svanborg C (1995) Intestinal carriage of P fimbriated Escherichia
human counterpart. J Exp Med 192:881–890 coli and the susceptibility to urinary tract infection in young
20. Hang L, Frendeus B, Godaly G, Svanborg C (2000) Interleukin- children. J Infect Dis 171:625–631
8 receptor knockout mice have subepithelial neutrophil entrapment 37. Palsson-McDermott EM, O'Neill LA (2004) Signal transduction
and renal scarring following acute pyelonephritis. J Infect Dis by the lipopolysaccharide receptor, Toll-like receptor-4. Immunol-
182:1738–1748 ogy 113:153–162
21. Svensson M, Irjala H, Alm P, Holmqvist B, Lundstedt AC, 38. Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X,
Svanborg C (2005) Natural history of renal scarring in sus- Birdwell D, Alejos E, Silva M, Galanos C, Freudenberg M,
ceptible mIL-8Rh-/- mice. Kidney Int 67:103–110 Ricciardi-Castagnoli P, Layton B, Beutler B (1998) Defective
22. Lundstedt AC, McCarthy S, Gustafsson MC, Godaly G, Jodal U, LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations
Karpman D, Leijonhufvud I, Linden C, Martinell J, Ragnarsdottir B, in Tlr4 gene. Science 282:2085–2088
Samuelsson M, Truedsson L, Andersson B, Svanborg C (2007) A 39. Hedlund M, Wachtler C, Johansson E, Hang L, Somerville
genetic basis of susceptibility to acute pyelonephritis. PLoS One 2: JE, Darveau RP, Svanborg C (1999) P fimbriae-dependent,
e825 lipopolysaccharide-independent activation of epithelial cytokine
23. Ragnarsdottir B, Jonsson K, Urbano A, Gronberg-Hernandez J, responses. Mol Microbiol 33:693–703
Lutay N, Tammi M, Gustafsson M, Lundstedt AC, Leijonhufvud I, 40. Samuelsson P, Hang L, Wullt B, Irjala H, Svanborg C (2004) Toll-
Karpman D, Wullt B, Truedsson L, Jodal U, Andersson B, Svanborg like receptor 4 expression and cytokine responses in the human
C (2010) Toll-like receptor 4 promoter polymorphisms: common urinary tract mucosa. Infect Immun 72:3179–3186
TLR4 variants may protect against severe urinary tract infection. 41. Horwitz MA, Silverstein SC (1980) Influence of the Escherichia
PLoS One 5:e10734 coli capsule on complement fixation and on phagocytosis and
24. Fischer H, Yamamoto M, Akira S, Beutler B, Svanborg C (2006) killing by human phagocytes. J Clin Invest 65:82–94
Mechanism of pathogen-specific TLR4 activation in the mucosa: 42. Svanborg-Eden C, Hagberg L, Hull R, Hull S, Magnusson KE,
fimbriae, recognition receptors and adaptor protein selection. Eur J Ohman L (1987) Bacterial virulence versus host resistance in the
Immunol 36:267–277 urinary tracts of mice. Infect Immun 55:1224–1232
25. Fischer H, Lutay N, Ragnarsdottir B, Yadav M, Jonsson K, Urbano 43. O'Hanley P, Low D, Romero I, Lark D, Vosti K, Falkow S,
A, Al Hadad A, Ramisch S, Storm P, Dobrindt U, Salvador E, Schoolnik G (1985) Gal-Gal binding and hemolysin phenotypes
Karpman D, Jodal U, Svanborg C (2010) Pathogen specific, IRF3- and genotypes associated with uropathogenic Escherichia coli. N
dependent signaling and innate resistance to human kidney infec- Engl J Med 313:414–420
tion. PLoS Pathogens 6:e1001109 44. Kontoghiorghes GJ, Weinberg ED (1995) Iron: mammalian de-
26. Frendeus B, Godaly G, Hang L, Karpman D, Svanborg C (2001) fense systems, mechanisms of disease, and chelation therapy
Interleukin-8 receptor deficiency confers susceptibility to acute approaches. Blood Rev 9:33–45
pyelonephritis. J Infect Dis 183(Suppl 1):S56–S60 45. Carbonetti MH, Williams PH (1985) Detection of aerobactin pro-
27. Agace WW, Hedges SR, Ceska M, Svanborg C (1993) Interleukin- duction. In: Sussman M (ed) The virulence of Escherichia coli.
8 and the neutrophil response to mucosal Gram-negative infection. Special Publications of the Society of General Microbiology.
J Clin Invest 92:780–785 Academic Press, London, pp 419–424
28. Pak J, Pu Y, Zhang ZT, Hasty DL, Wu XR (2001) Tamm-Horsfall 46. Johnson JR, Stell AL (2000) Extended virulence genotypes of
protein binds to type 1 fimbriated Escherichia coli and prevents E. Escherichia coli strains from patients with urosepsis in relation
coli from binding to uroplakin Ia and Ib receptors. J Biol Chem to phylogeny and host compromise. J Infect Dis 181:261–272
276:9924–9930 47. Kanamaru S, Kurazono H, Ishitoya S, Terai A, Habuchi T, Nakano
29. Wold A, Mestecky J, Tomana M, Kobata A, Ohbayashi H, Endo T, M, Ogawa O, Yamamoto S (2003) Distribution and genetic asso-
Svanborg-Edén C (1990) Secretory immunoglobulin-A carries ol- ciation of putative uropathogenic virulence factors iroN, iha,
igosaccharide receptors for Escherichia coli type 1 fimbrial lectin. kpsMT, ompT and usp in Escherichia coli isolated from urinary
Infect Immun 58:3073–3077 tract infections in Japan. J Urol 170:2490–2493
30. Xie B, Zhou G, Chan SY, Shapiro E, Kong XP, Wu XR, Sun TT, 48. Garcia EC, Brumbaugh AR, Mobley HL (2011) Redundancy and
Costello CE (2006) Distinct glycan structures of uroplakins Ia and specificity of Escherichia coli iron acquisition systems during
Ib: structural basis for the selective binding of FimH adhesin to urinary tract infection. Infect Immun 79:1225–1235
uroplakin Ia. J Biol Chem 281:14644–14653 49. Hedlund M, Nilsson Å, Duan RD, Svanborg C (1998) Sphingomye-
31. Malaviya R, Gao Z, Thankavel K, van der Merwe PA, Abraham lin, glycosphingolipids and ceramide signalling in cells exposed to P
SN (1999) The mast cell tumor necrosis factor alpha response fimbriated Escherichia coli. Mol Microbiol 29:1297–1306
to FimH-expressing Escherichia coli is mediated by the 50. Hedlund M, Svensson M, Nilsson A, Duan RD, Svanborg C (1996)
glycosylphosphatidylinositol-anchored molecule CD48. Proc Role of the ceramide-signaling pathway in cytokine responses to P-
Natl Acad Sci USA 96:8110–8115 fimbriated Escherichia coli. J Exp Med 183:1037–1044
2028 Pediatr Nephrol (2012) 27:2017–2029

51. Fischer H, Ellstrom P, Ekstrom K, Gustafsson L, Gustafsson M, Upper urinary tract infections are associated with RANTES pro-
Svanborg C (2007) Ceramide as a TLR4 agonist; a putative sig- moter polymorphism. J Pediatr 157:1038–1040.e1
nalling intermediate between sphingolipid receptors for microbial 69. Hughes LB, Criswell LA, Beasley TM, Edberg JC, Kimberly RP,
ligands and TLR4. Cell Microbiol 9:1239–1251 Moreland LW, Seldin MF, Bridges SL (2004) Genetic risk factors
52. Ragnarsdottir B, Samuelsson M, Gustafsson MC, Leijonhufvud I, for infection in patients with early rheumatoid arthritis. Genes
Karpman D, Svanborg C (2007) Reduced toll-like receptor 4 Immun 5:641–647
expression in children with asymptomatic bacteriuria. J Infect 70. Hussein A, Askar E, Elsaeid M, Schaefer F (2010) Functional
Dis 196:475–484 polymorphisms in transforming growth factor-beta-1 (TGFbeta-1)
53. Hedges S, Anderson P, Lidin JG, de Man P, Svanborg C (1991) and vascular endothelial growth factor (VEGF) genes modify risk
Interleukin-6 response to deliberate colonization of the human of renal parenchymal scarring following childhood urinary tract
urinary tract with Gram-negative bacteria. Infect Immun 59:421– infection. Nephrol Dial Transplant 25:779–785
427 71. Kuroda S, Solari V, Puri P (2007) Association of transforming
54. Hedges S, Svensson M, Svanborg C (1992) Interleukin-6 response growth factor-beta1 gene polymorphism with familial vesicoure-
of epithelial cell lines to bacterial stimulation in vitro. Infect teral reflux. J Urol 178:1650–1653
Immun 60:1295–1301 72. Lee-Chen GJ, Liu KP, Lai YC, Juang HS, Huang SY, Lin CY
55. Godaly G, Proudfoot AE, Offord RE, Svanborg C, Agace WW (2004) Significance of the tissue kallikrein promoter and trans-
(1997) Role of epithelial interleukin-8 (IL-8) and neutrophil IL- forming growth factor-beta1 polymorphisms with renal progres-
8 receptor A in Escherichia coli-induced transuroepithelial neutro- sion in children with vesicoureteral reflux. Kidney Int 65:1467–
phil migration. Infect Immun 65:3451–3456 1472
56. Godaly G, Otto G, Burdick MD, Strieter RM, Svanborg C (2007) 73. Hawn TR, Scholes D, Li SS, Wang H, Yang Y, Roberts PL,
Fimbrial lectins influence the chemokine repertoire in the urinary Stapleton AE, Janer M, Aderem A, Stamm WE, Zhao LP, Hooton
tract mucosa. Kidney Int 71:778–786 TM (2009) Toll-like receptor polymorphisms and susceptibility to
57. Ali AS, Townes CL, Hall J, Pickard RS (2009) Maintaining a sterile urinary tract infections in adult women. PLoS One 4:e5990
urinary tract: the role of antimicrobial peptides. J Urol 182:21–28 74. Stamey TA (1987) Recurrent urinary tract infections in female
58. Chromek M, Slamova Z, Bergman P, Kovacs L, Podracka L, Ehren patients: an overview of management and treatment. Rev Infect
I, Hokfelt T, Gudmundsson GH, Gallo RL, Agerberth B, Brauner Dis 9(Suppl 2):S195–S210
A (2006) The antimicrobial peptide cathelicidin protects the uri- 75. Lomberg H, Jodal U, Svanborg-Edén C, Leffler H, Samuelsson B
nary tract against invasive bacterial infection. Nat Med 12:636– (1981) P1 blood group and urinary tract infection. Lancet i:551–
641 552
59. Montini G, Tullus K, Hewitt I (2011) Febrile urinary tract infec- 76. Stapleton A, Nudelman E, Clausen H, Hakomori S, Stamm WE
tions in children. N Engl J Med 365:239–250 (1992) Binding of uropathogenic Escherichia coli R45 to glyco-
60. Lomberg H, Hanson L, Jacobsson B, Jodal U, Leffler H, lipids extracted from vaginal epithelial cells is dependent on histo-
Svanborg-Edén C (1983) Correlation of P blood group phenotype, blood group secretor status. J Clin Invest 90:965–972
vesicoureteral reflux and bacterial attachment in patients with 77. Lindstedt R, Larson G, Falk P, Jodal U, Leffler H, Svanborg-Edén
recurrent pyelonephritis. N Engl J Med 308:1189–1192 C (1991) The receptor repertoire defines the host range for attach-
61. Svensson M, Yadav M, Holmqvist B, Lutay N, Svanborg C, ing Escherichia coli recognizing globo-A. Infect Immun 59:1086–
Godaly G (2011) Acute pyelonephritis and renal scarring are 1092
caused by dysfunctional innate immunity in mCxcr2 heterozygous 78. Orskov I, Ferencz A, Orskov F (1980) Tamm-Horsfall protein or
mice. Kidney Int 80:1064–1072 uromucoid is the normal urinary slime that traps type 1 fimbriated
62. Svensson M, Irjala H, Svanborg C, Godaly G (2008) Effects of Escherichia coli. Lancet 1:887
epithelial and neutrophil CXCR2 on innate immunity and resis- 79. Korhonen TK, Parkkinen J, Hacker J, Finne J, Pere A, Rhen M,
tance to kidney infection. Kidney Int 74:81–90 Holthofer H (1986) Binding of Escherichia coli S fimbriae to
63. Yin X, Hou T, Liu Y, Chen J, Yao Z, Ma C, Yang L, Wei L (2010) human kidney epithelium. Infect Immun 54:322–327
Association of Toll-like receptor 4 gene polymorphism and expres- 80. Korhonen TK, Väisänen-Rhen V, Rhen M, Pere A, Parkkinen J,
sion with urinary tract infection types in adults. PLoS One 5: Finne J (1984) Escherichia coli fimbriae recognize sialyl galacto-
e14223 sides. J Bacteriol 159:762–766
64. Hawn TR, Scholes D, Wang H, Li SS, Stapleton AE, Janer M, 81. Scholes D, Hooton TM, Roberts PL, Stapleton AE, Gupta K,
Aderem A, Stamm WE, Zhao LP, Hooton TM (2009) Genetic Stamm WE (2000) Risk factors for recurrent urinary tract infection
variation of the human urinary tract innate immune response and in young women. J Infect Dis 182:1177–1182
asymptomatic bacteriuria in women. PLoS One 4:e8300 82. Stauffer CM, van der Weg B, Donadini R, Ramelli GP, Marchand
65. Lundstedt AC, Leijonhufvud I, Ragnarsdottir B, Karpman D, S, Bianchetti MG (2004) Family history and behavioral abnormal-
Andersson B, Svanborg C (2007) Inherited susceptibility to acute ities in girls with recurrent urinary tract infections: a controlled
pyelonephritis: a family study of urinary tract infection. J Infect study. J Urol 171:1663–1665
Dis 195:1227–1234 83. Scholes D, Hawn TR, Roberts PL, Li SS, Stapleton AE, Zhao LP,
66. Artifoni L, Negrisolo S, Montini G, Zucchetta P, Molinari PP, Stamm WE, Hooton TM (2010) Family history and risk of recur-
Cassar W, Destro R, Anglani F, Rigamonti W, Zacchello G, Murer rent cystitis and pyelonephritis in women. J Urol 184:564–569
L (2007) Interleukin-8 and CXCR1 receptor functional polymor- 84. Svanborg Eden C, Briles D, Hagberg L, McGhee J, Michalec S
phisms and susceptibility to acute pyelonephritis. J Urol 177:1102– (1984) Genetic factors in host resistance to urinary tract infection.
1106 Infection 12:118–123
67. Smithson A, Sarrias MR, Barcelo J, Suarez B, Horcajada JP, Soto 85. Pecha B, Low D, O'Hanley P (1989) Gal-Gal pili vaccines prevent
SM, Soriano A, Vila J, Martinez JA, Vives J, Mensa J, Lozano F pyelonephritis by piliated Escherichia coli in a murine model.
(2005) Expression of interleukin-8 receptors (CXCR1 and Single-component Gal-Gal pili vaccines prevent pyelonephritis
CXCR2) in premenopausal women with recurrent urinary tract by homologous and heterologous piliated E. coli strains. J Clin
infections. Clin Diagn Lab Immunol 12:1358–1363 Invest 83:2102–2108
68. Centi S, Negrisolo S, Stefanic A, Benetti E, Cassar W, Da Dalt L, 86. Langermann S, Palaszynski S, Barnhart M, Auguste G, Pinkner JS,
Rigamonti W, Zucchetta P, Montini G, Murer L, Artifoni L (2010) Burlein J, Barren P, Koenig S, Leath S, Jones CH, Hultgren SJ
Pediatr Nephrol (2012) 27:2017–2029 2029

(1997) Prevention of mucosal Escherichia coli infection by FimH- 93. Wang YT, Chiu NT, Chen MJ, Huang JJ, Chou HH, Chiou YY
adhesin-based systemic vaccination. Science 276:607–611 (2005) Correlation of renal ultrasonographic findings with inflam-
87. Langermann S, Mollby R, Burlein JE, Palaszynski SR, Auguste matory volume from dimercaptosuccinic acid renal scans in chil-
CG, DeFusco A, Strouse R, Schenerman MA, Hultgren SJ, Pinkner dren with acute pyelonephritis. J Urol 173:190–194, discussion
JS, Winberg J, Guldevall L, Soderhall M, Ishikawa K, Normark S, 194
Koenig S (2000) Vaccination with FimH adhesin protects cynomol- 94. Hopkins W, Gendron-Fitzpatrick A, McCarthy DO, Haine JE,
gus monkeys from colonization and infection by uropathogenic Uehling DT (1996) Lipopolysaccharide-responder and nonre-
Escherichia coli. J Infect Dis 181:774–778 sponder C3H mouse strains are equally susceptible to an induced
88. Wullt B, Bergsten G, Connell H, Rollano P, Gebretsadik N, Hull R, Escherichia coli urinary tract infection. Infect Immun 64:1369–
Svanborg C (2000) P fimbriae enhance the early establishment of 1372
Escherichia coli in the human urinary tract. Mol Microbiol 38:456– 95. Cotton SA, Gbadegesin RA, Williams S, Brenchley PE, Webb NJ
464 (2002) Role of TGF-beta1 in renal parenchymal scarring following
89. Wullt B, Bergsten G, Samuelsson M, Gebretsadik N, Hull R, childhood urinary tract infection. Kidney Int 61:61–67
Svanborg C (2001) The role of P fimbriae for colonization and 96. Khalil A, Brauner A, Bakhiet M, Burman LG, Jaremko G,
host response induction in the human urinary tract. J Infect Dis 183 Wretlind B, Tullus K (1997) Cytokine gene expression during
(Suppl 1):S43–S46 experimental Escherichia coli pyelonephritis in mice. J Urol
90. Bergsten G, Samuelsson M, Wullt B, Leijonhufvud I, Fischer H, 158:1576–1580
Svanborg C (2004) PapG-dependent adherence breaks mucosal inertia 97. Kulkarni AB, Karlsson S (1993) Transforming growth factor-beta
and triggers the innate host response. J Infect Dis 189:1734–1742 1 knockout mice. A mutation in one cytokine gene causes a
91. P, Jodal U, Lincoln K, Eden CS (1988) Bacterial attachment and dramatic inflammatory disease. Am J Pathol 143:3–9
inflammation in the urinary tract. J Infect Dis 158:29–35 98. Shull MM, Ormsby I, Kier AB, Pawlowski S, Diebold RJ, Yin M,
92. Huang YY, Chen MJ, Chiu NT, Chou HH, Lin KY, Chiou YY Allen R, Sidman C, Proetzel G, Calvin D, Annunziata N, Doetschman
(2011) Adjunctive oral methylprednisolone in pediatric acute py- T (1992) Targeted disruption of the mouse transforming growth factor-
elonephritis alleviates renal scarring. Pediatrics doi:10.1542/ beta 1 gene results in multifocal inflammatory disease. Nature
peds.2010-0297 359:693–699

You might also like