You are on page 1of 21

This article was downloaded by: [Glen Ulett]

On: 05 August 2015, At: 20:50


Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: 5 Howick Place,
London, SW1P 1WG

Critical Reviews in Microbiology


Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/imby20

Click for updates

Urinary tract infection of mice to model human


disease: Practicalities, implications and limitations
a

Alison J. Carey , Chee K. Tan , Deepak S. Ipe , Matthew J. Sullivan , Allan W. Cripps , Mark
c

A. Schembri & Glen C. Ulett

Menzies Health Institute Queensland & School of Medical Sciences, Griffith University,
Gold Coast, Australia,
b

Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia, and

School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane,


Australia
Published online: 05 Jun 2015.

To cite this article: Alison J. Carey, Chee K. Tan, Deepak S. Ipe, Matthew J. Sullivan, Allan W. Cripps, Mark A. Schembri &
Glen C. Ulett (2015): Urinary tract infection of mice to model human disease: Practicalities, implications and limitations,
Critical Reviews in Microbiology
To link to this article: http://dx.doi.org/10.3109/1040841X.2015.1028885

PLEASE SCROLL DOWN FOR ARTICLE


Taylor & Francis makes every effort to ensure the accuracy of all the information (the Content) contained
in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no
representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the
Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and
are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and
should be independently verified with primary sources of information. Taylor and Francis shall not be liable for
any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever
or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of
the Content.
This article may be used for research, teaching, and private study purposes. Any substantial or systematic
reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any
form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://
www.tandfonline.com/page/terms-and-conditions

http://informahealthcare.com/mby
ISSN: 1040-841X (print), 1549-7828 (electronic)
Crit Rev Microbiol, Early Online: 120
! 2015 Informa Healthcare USA, Inc. DOI: 10.3109/1040841X.2015.1028885

REVIEW ARTICLE

Urinary tract infection of mice to model human disease: Practicalities,


implications and limitations
Alison J. Carey1, Chee K. Tan1, Deepak S. Ipe1, Matthew J. Sullivan1, Allan W. Cripps2, Mark A. Schembri3, and
Glen C. Ulett1
Menzies Health Institute Queensland & School of Medical Sciences, Griffith University, Gold Coast, Australia, 2Menzies Health Institute Queensland,
Griffith University, Gold Coast, Australia, and 3School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia

Downloaded by [Glen Ulett] at 20:50 05 August 2015

Abstract

Keywords

Urinary tract infections (UTIs) are among the most common bacterial infections in humans.
Murine models of human UTI are vital experimental tools that have helped to elucidate UTI
pathogenesis and advance knowledge of potential treatment and infection prevention
strategies. Fundamentally, several variables are inherent in different murine models, and
understanding the limitations of these variables provides an opportunity to understand how
models may be best applied to research aimed at mimicking human disease. In this review, we
discuss variables inherent in murine UTI model studies and how these affect model usage, data
analysis and data interpretation. We examine recent studies that have elucidated UTI host
pathogen interactions from the perspective of gene expression, and review new studies of
biofilm and UTI preventative approaches. We also consider potential standards for variables
inherent in murine UTI models and discuss how these might expand the utility of models for
mimicking human disease and uncovering new aspects of pathogenesis.

Animal models, bacterial pathogenesis,


Escherichia coli, innate immunity, urinary
tract infection

Introduction
Urinary tract infections (UTIs) are some of the most common
bacterial infections in humans and a major burden to
healthcare systems. UTIs cause an estimated 11 million
doctor visits in the USA annually, and over 150 million doctor
visits globally, with an annual cost upwards of $6 billion
(Bower et al., 2005; Foxman, 2003; Nielubowicz & Mobley,
2010; Stamm & Hooton, 1993; Stamm & Norrby, 2001).
Categories of disease include complicated and uncomplicated
infections, which are further classified according to symptoms
and diagnostic markers as urethritis, acute and chronic
cystitis, prostatitis and pyelonephritis (Hay & Fahey, 2002;
Kaufmann & Modest, 2002; Neild, 2003; Nicolle, 2008).
Complicated UTIs are defined by their association with
structural or functional abnormalities of the urinary tract that
leads to enhanced susceptibility to infection (Johansen et al.,
2011). UTI can progress to life-threatening disease including
urosepsis in the absence of appropriate treatment (Mcbean &
Rajamani, 2001; Siegman-Igra et al., 2002). The UTI spectrum also encompasses asymptomatic bacteriuria (ABU); this
is a carrier state that resembles commensalism and often leads
to the recovery of4105 colony forming units (cfu) per ml of a
single bacterial strain from urine without clinical symptoms.

Address for correspondence: Associate Professor Glen C. Ulett, PhD,


Menzies Health Institute Queensland & School of Medical Sciences,
Griffith University, c/o School of Medical Science, Griffith Health
Centre G40, Gold Coast 4222, Australia. E-mail: g.ulett@griffith.edu.au

History
Received 4 November 2014
Revised 5 March 2015
Accepted 10 March 2015
Published online 26 May 2015

UTIs occur most frequently in women; over half of all


women will experience a UTI in their lifetime (Bower et al.,
2005; Foxman, 2002), and up to 30% of these women will
experience a recurrent infection that may progress to chronic
disease (Brown, 1999; Hooton et al., 1996; Kunin, 1994;
Stamm & Hooton, 1993; Stamm & Norrby, 2001; Stapleton,
1999). The incidence of UTI in women over the age of 65 is
21% compared to only 12% in men of similar age (Lee &
Neild, 2007). The incidence of specific types of UTI is also
disproportionately associated with age and predisposing
factors. For example, ABU is overrepresented in several
populations including elderly institutionalized individuals and
adult women with diabetes mellitus (High et al., 2009; Ipe
et al., 2013; Johansen et al., 2011; Lee & Neild, 2007;
Schneeberger et al., 2014; Van Duin, 2012). A population
with a nearly 100% prevalence of ABU is users of chronic
indwelling urinary catheters (Warren et al., 1982). The major
determinant for development of catheter-associated bacteriuria is the duration of catheterization (Nicolle, 2014).
Symptomatic catheter-associated UTIs [CAUTIs; as advocated by the CDC (Press & Metlay, 2013)] are important
among hospitalized individuals, and in these patients, infection with a broad range of pathogens, including mixed
infections, is common (Foxman, 1990; Jacobsen et al., 2008;
Meddings et al., 2010; Trautner, 2010; Trautner et al., 2005).
CAUTIs as nosocomial infections have been linked to biofilm
formation (Frank et al., 2009; Hola et al., 2012; Maki &
Tambyah, 2001; Ong et al., 2008; Stahlhut et al., 2012), and
are often associated with enhanced antibiotic resistance

A. J. Carey et al.

(Chenoweth & Saint, 2013;


Wagenlehner & Naber, 2004).

Crit Rev Microbiol, Early Online: 120

Johnson

et al.,

1999;

Downloaded by [Glen Ulett] at 20:50 05 August 2015

Causal agents of UTIs


Uropathogenic Escherichia coli (UPEC) cause up to 80% of
all UTIs (Ronald, 2002; Stamm, 2006), particularly among
young women (Salvatore et al., 2011). The major reservoir
for UPEC is the gastrointestinal tract, with spread to the
genital tract and urethral opening providing an opportunity for
bladder colonization (Stamm, 2006). Other noted etiological
agents of UTI include Klebsiella spp., Pseudomonas spp.,
Citrobacter spp. and Proteus spp. (Lee & Neild, 2007;
Ronald, 2003; Tabibian et al., 2008). Several Gram-positive
bacteria including Staphylococcus spp., Enterococcus sp. and
Streptococcus agalactiae (also known as group B streptococcus; GBS) also cause UTI and ABU (Alamuri et al., 2009;
Natarajan, 2008; Raffi et al., 2005; Singh et al., 2007; Tan
et al., 2012b; Tena et al., 2007; Ulett et al., 2009). Some
pathogens are more frequently associated with distinct UTI
clinical conditions and patient groups (Ipe et al., 2013;
Ronald, 2002). For example, Staphylococcus saprophyticus
affects mainly younger women, Morganella morganii,
Providencia
stuartii,
Pseudomonas
aeruginosa,
Enterobacter and Serratia sp. are frequently associated with
severe UTI (Franco, 2005; Johnson et al., 1987; Nielubowicz
& Mobley, 2010; Stamm & Hooton, 1993), and enterococci
are overrepresented among complicated UTI and hospitalacquired CAUTIs (Barnett & Stephens, 1997; Ronald, 2002;
Tabibian et al., 2008; Warren et al., 1982). Nosocomial
CAUTI, which is often associated with mixed bacterial
species, is caused by a more diverse range of Gram-negative
and Gram-positive pathogens, including E. coli, Proteus spp.,
Pseudomonas spp., P. stuartii, Staphylococcus spp. and
Enterococcus spp. (Nielsen et al., 2012; Sillanpaa et al.,
2010). While E. coli causes most ABU, Klebsiella spp.,
enterococci, streptococci and Candida spp. are also relatively
common among certain patient groups (Ipe et al., 2013;
Ronald, 2002, 2003).

Insights into UTI pathogenesis from studies


using mice
In order to successfully colonize the urinary tract, pathogens
must endure a multitude of host factors that mediate localized
defense to prevent UTI (Ragnarsdottir et al., 2011; Schilling
et al., 2001a; Song & Abraham, 2008; Ulett et al., 2013). In
addition to the normal urinary flushing mechanisms and the
(impermeable) urothelial-lining of the bladder, factors
including host antimicrobial peptides, uromodulin (also
known as TammHorsfall protein) and cytokines contribute
to host defense.
Substantial advances in our understanding of the pathogenesis of UTI have come from the development and
characterization of murine models of human infection.
Different versions of the murine UTI model have been used
to dissect the roles of cytokines in innate defense including
CCL2 (Ingersoll et al., 2008; Sivick et al., 2010), CCL4
(Ingersoll et al., 2008), IL-1b (Ingersoll et al., 2008; Ulett
et al., 2010), IL-6 (Hedges et al., 1992; Ingersoll et al.,
2008; Nielubowicz & Mobley, 2010; Sivick & Mobley, 2010;

Wullt, 2003), IL-8 (mouse equivalent CXCL1/2) (Ingersoll


et al., 2008; Nielubowicz & Mobley, 2010; Wullt, 2003;
Wullt et al., 2003), IL-9 (Kline et al., 2011), IL-10 (Duell
et al., 2012a), IL-17A (Ingersoll et al., 2008; Sivick &
Mobley, 2010), IL-12p40 (Ingersoll et al., 2008), G-CSF
(Ingersoll et al., 2008; Sivick & Mobley, 2010) and TNF-a
(Sivick & Mobley, 2010; Sivick et al., 2010). The synthesis
of some of these cytokines begins as early as 1 h postinfection in mice, peaks at 24 h and returns to baseline by
2 weeks. A commonly used model uses the C57BL/6 mouse
strain and has illustrated the early cytokine responses that
correlate with peak bacterial burdens in the bladder and
subsequent UPEC clearance (Sivick & Mobley, 2010). Other
models have shown that production of IL-8 (CXCL1/2 in
mice) triggers an influx of neutrophils (Artifoni et al., 2007;
Hang et al., 2000; Sivick & Mobley, 2010; Sivick et al.,
2010; Smithson et al., 2005) via toll-like receptor (TLR)4dependent and -independent mechanisms (Agace, 1996;
Agace et al., 1993; Hedges et al., 1994; Ragnarsdottir
et al., 2008; Sivick & Mobley, 2010; Schilling et al.,
2001a,b; Wullt et al., 2001). Recently defined elements of
acute UTI pathogenesis have been reviewed elsewhere (Sivick
& Mobley, 2010; Ulett et al., 2013). Pathogens that can
overcome host defenses can exploit the bladder and urine as
unique growth environments. Indeed, growth in urine has
been linked to recurrent cystitis in individuals with voiding
defects (Foxman, 2002; Raz et al., 2000; Scholes et al.,
2000) and is probably related to microbial traits that provide
bacteruric potential, as discussed elsewhere (Ipe et al.,
2013).

How do responses to UTI in mice correlate with


responses in humans?
We can compare key elements of host defense and immune
responses to UTI in mice with what is known about human
infection to gain insight into the degree to which murine
models reflect hostpathogen interactions in patients. TLRencoding genes (Andersen-Nissen et al., 2007; Ragnarsdottir
et al., 2010; Yin et al., 2010) and some chemokine receptors
(Frendeus et al., 2000; Godaly et al., 2000; Svensson et al.,
2011) offer useful paradigms of consistent and divergent
responses between mouse and human UTI. The study of
TLR4 and CXCR1 in the context of mouse genetics and
defense against UTI (Ragnarsdottir et al., 2008; Sivick &
Mobley, 2010; Svanborg et al., 2001; Svanborg Eden et al.,
1985), in particular, provide validation of murine models for
studying human infection.

TLR4 and the host response to LPS


C3H/HeJ mice, which are LPS non-responsive due to
mutation in the tlr4 gene and mount poor inflammatory
responses compared to C3H/HeN LPS-responsive mice, are
highly susceptible to UPEC UTI (Hagberg et al., 1984).
Studies using these mice were critical for establishing the
signaling pathway from TLR4 that leads to secretion of
cytokines such as IL-6 (De Man et al., 1989), chemokines
such as macrophage inflammatory protein 2 (CXCL2), and
recruitment of neutrophils (Yadav et al., 2010) that help to
control UTI. Interestingly, the control of UTI in the bladder

DOI: 10.3109/1040841X.2015.1028885

Downloaded by [Glen Ulett] at 20:50 05 August 2015

depends on TLR4 expression on urothelial and stromal cells,


and this is needed to drive inflammatory responses even in the
presence of hematopoietic cells with intact TLR4 (Schilling
et al., 2003). TLR4 signaling has also been associated with
increased severity of pain due to UPEC UTI in mice (Rudick
et al., 2010), whereas less TLR4 activity has been associated
with ABU in mice (Fischer et al., 2006) and in humans
(Ragnarsdottir et al., 2007, 2010), as noted elsewhere (Ulett
et al., 2013). Analysis of TLR4 expression in humans with
UTI demonstrated that lower TLR4 expression due to
polymorphisms in the promoter attenuates host responses
and promotes asymptomatic carriage of UPEC (Hernandez
et al., 2011; Ragnarsdottir et al., 2007, 2010). Another study
of tlr4 polymorphisms in children associated a TLR4_A896G
polymorphism with development of upper UTI and formation
of renal scarring in pyelonephritis (Akil et al., 2012). Thus,
observations regarding the function of TLR4 in murine UTI
largely parallel findings in human infection and indicate that
carefully regulated innate immune responses are critical to
protect against UTI.

Host responses involving TLR5 and TLR11


TLR5 and TLR11 provide other examples of connect and
disconnect, respectively, when we compare data derived from
murine models and humans. In humans, a polymorphism

Urinary tract infection of mice to model human disease

in the TLR5 gene (TLR5_C1174T) abrogates the flagellin


signaling cascade and has been associated with an increased
susceptibility to recurrent UTI (rUTI) (Hawn et al., 2009).
TLR5 is essential to mediate flagellin-induced inflammatory
responses in mice with cystitis, which aid in control of
infection in both the bladder and kidneys (Andersen-Nissen
et al., 2007). Thus, similar to TLR4, data regarding the
function of TLR5 appear to be comparable between mouse
UTI and human infection. In contrast, TLR11 offers useful
insight into the limitations of murine models of human UTI.
TLR11 is expressed in mice and plays a role in protecting
the kidneys against UPEC (Zhang et al., 2004). The exact
mechanism by which TLR11 is activated and provides
protection in mice is not known. A TLR11 homolog in
humans has yet to be identified, which highlights the
divergence between human clinical UTI and mice, and the
need for caution when extrapolating some findings from
mice to humans.

Cytokine responses in mice and humans


Some of the patterns of cytokine production in mice parallel
the immune responses that have been described in human
patients with UTI. For example, IL-1b, IL-6, IL-8, IL-10,
CCL2, CCL5 and CXC chemokines are produced early during
infection in both mice and humans, as illustrated in Figure 1

Figure 1. Similarities between murine models of UPEC UTI and human infection. Similarities in the host response to UPEC UTI between mouse and
human include involvement of some UPEC virulence factors and gene expression, host cellular responses and cytokine/chemokine signaling.
References are listed in the right column as: 1: (Anderson et al., 2003); 2: (Rosen et al., 2007); 3: (Robino et al., 2013); 4: (Snyder et al., 2004); 5:
(Hagan et al., 2010); 6: (Mulvey, 2002); 7: (Chan et al., 2013); 8: (Chuang & Kuo, 2013); 9: (Kai-Larsen et al., 2010); 10: (Cusumano et al., 2011);
11: (Agace, 1996); 12: (Wu et al., 1996); 13: (Hang et al., 2000); 14: (Smithson et al., 2005); 15: (Nielubowicz & Mobley, 2010); 16: (Schilling et al.,
2001b); 17: (Ko et al., 1993); 18: (Candela et al., 1998); 19: (Wullt et al., 2002); 20: (Roelofs et al., 2006); 21: (Godaly et al., 2007); 22: (Ingersoll
et al., 2008); 23: (Sivick & Mobley, 2010); 24: (Ulett et al., 2010); 25: (Duell et al., 2012a); 26: (Garcia et al., 2013); 27: (Billips et al., 2007); 28:
(Ragnarsdottir et al., 2008); 29: (Duell et al., 2013); 30: (Tan et al., 2012a). Abbreviations: IL, Interleukin; PMN, polymorphonuclear cells; CRAMP,
murine cathelin-related antimicrobial peptide; IBC, intracellular bacterial community; TLR, toll-like receptor.

Downloaded by [Glen Ulett] at 20:50 05 August 2015

A. J. Carey et al.

(Candela et al., 1998; Duell et al., 2012a; Garcia et al.,


2013; Godaly et al., 2007; Ingersoll et al., 2008; Ko et al.,
1993; Roelofs et al., 2006; Sivick & Mobley, 2010; Ulett
et al., 2010; Wullt et al., 2002). Likewise, intracellular
bacterial communities (IBCs) occur in mice (Anderson
et al., 2003; Rosen et al., 2007) and have been observed in
tissues/cells collected from humans with UTI (Robino et al.,
2013, 2014; Rosen et al., 2007). Fimbrial adhesins of UPEC
mediate binding to mouse and human urothelial cells, and
play a major role in colonization of the urinary tract (Connell
et al., 1996; Mulvey, 2002; Nielubowicz & Mobley, 2010).
There have also been recent discoveries regarding the
involvement of mast cells in murine UTI that align with
recent insight into the human response to infection. In mice,
mast cells contribute to the suppression of humoral and cell
mediated immune responses in the bladder via the production
of IL-10, which was shown to contribute to long-term
persistence of UPEC (Chan et al., 2013). Biopsies of
human bladder collected from patients with rUTI also
contained increased mast cells, suggesting that they play a
role in chronic inflammation and contribute to re-occurrence
of infection (Chuang & Kuo, 2013). A summary of the
consistent features in UTI pathogenesis between mice and
humans is presented in Figure 1.

Bacterial manipulation of the host response


UPEC can also pro-actively manipulate key host responses
during UTI. For example, UPEC production of TcpC,
a toll/interleukin-1 receptor (TIR) homology domain-containing protein, results in inhibition of downstream TLR signaling
and enhances UPEC survival in mouse macrophages (Cirl
et al., 2008). The effect of UPEC survival in macrophages
toward the pathogenesis and severity of UTI is unknown.
Interestingly, however, there are differences between the
ability of different E. coli strains to survive in human and
mouse macrophages; UPEC UTI89 and ABU VR50 display
better survival than UPEC CFT073 and ABU 83972 in
murine bone marrow-derived macrophages, whereas in
human monocyte derived macrophages, the intramacrophage
fitness trait of ABU VR50 is absent (Bokil et al., 2011). This
suggests that host species differences may impact on UTI
pathogenesis in the context of macrophage anti-microbial
pathways.
Some UPEC strains can also suppress urothelial cytokine
responses, subduing the secretion of IL-8 (CXCL8) from
human urothelial cells, and inhibiting the transcription of the
mouse equivalents, keratinocyte cytokine (KC/CXCL1) and
macrophage inflammatory protein 2 (CXCL2) in vivo (Billips
et al., 2007). UPEC a-hemolysin promotes shedding of
urothelial cells in vitro for both murine and human cells,
and suppresses the secretion of the inflammatory cytokine,
IL-6, from human bladder urothelial cells (Dhakal & Mulvey,
2012; Hilbert et al., 2012). Curli expression in UPEC also
alters the innate immune response by interacting with and
binding to the human antimicrobial peptide cathelicidin
LL-37, and the murine ortholog, cathelicidin-related
antimicrobial peptide (CRAMP). Binding of antimicrobial
peptide to UPEC curli prevents the former from binding
to the bacterial membrane and mediating cell lysis

Crit Rev Microbiol, Early Online: 120

(Kai-Larsen et al., 2010). Thus, LL-37/CRAMP is secreted


in urine in response to UPEC and provides protection against
UTI in human cell lines in vitro and in mice (Chromek et al.,
2006). The concentration of LL-37 in urine was suggested
to influence susceptibility to UTI (Nielsen et al., 2014).
Other virulence mechanisms used by UPEC to subvert
host responses are reviewed elsewhere (Hannan et al.,
2012; Hunstad & Justice, 2010; Nielubowicz & Mobley,
2010; Sivick & Mobley, 2010; Ulett et al., 2013).

Evaluating fundamental variables in murine


UTI models
Murine models of UTI represent a crucial link in the ability to
study hostpathogen interactions and devise and evaluate new
preventive and treatment strategies to protect against infection
(Hoelzer et al., 2012; Louz et al., 2013; Mastroeni &
Sheppard, 2004, Mylonakis et al., 2007, Silva et al., 2011).
Mice are low cost, easy to handle and can be used with a
wealth of readily available reagents and tools. Genetically
modified strains for studying single gene function also
provide considerable benefits and are increasingly being
used for UTI pathogenesis studies (Anderson et al., 2010;
Duell et al., 2012a; Fischer et al., 2010; Saemann et al.,
2005; Sivick et al., 2010; Yadav et al., 2010). A summary of
murine UTI models used to study UPEC pathogenesis,
treatment, prevention and catheter colonization is provided
in Table 1. This is intended to comprise illustrative examples
of the variables in murine UTI models in detail and is not
intended to represent an exhaustive list of all the studies that
have used murine UTI models to date.
The first murine model of human UTI was described in
1975 (Kalmanson et al., 1975). Refinements to this original
model in the 1980s (Hagberg et al., 1983; Iwahi et al., 1982;
OHanley et al., 1985), and more recently (Hung et al., 2009)
have helped to define how these models reflect human
infection. The fundamental variables inherent in murine UTI
models can by grouped into three categories: (i) mode of
infection, (ii) inoculum properties (infectious dose, volume,
pathogen and strain) and (iii) mouse strain. Below, we discuss
these variables with a focus on carefully defining UTI models
and provide an overview of recent investigations that have
utilized these models to study UTI.

Variable 1: mode of infection


A range of inoculation procedures has been used to establish
infection in the mouse urinary tract. Variable parameters
include the mode of injection, anesthetic conditions, surgical
interventions [e.g. implantation of an indwelling catheter
(Li et al., 2002)], post-inoculation procedures to restrict
voiding [e.g. transient obstruction using collodion film
(Johnson et al., 1993a; Tratselas et al., 2014)], catheter
clamping (Bates et al., 2004; Raffi et al., 2009; Tsuji et al.,
2003) or water restriction (Bates et al., 2004; Chassin et al.,
2006; Li et al., 2010; Raffi et al., 2009; Tsuji et al., 2003;
Vimont et al., 2012; You et al., 2009)], dietary modifications
[e.g. antibiotic supplementation in drinking water (Hannan
et al., 2010; Johnson et al., 2006; Schilling et al., 2002)],
operator skill, and lastly, general animal handling and housing
conditions.

Urinary tract infection of mice to model human disease

DOI: 10.3109/1040841X.2015.1028885

Table 1. Summary of variables in recent and representative murine UTI models used for the experimental modelling of human UTI.
Bacteria

Strain

Downloaded by [Glen Ulett] at 20:50 05 August 2015

Pathogenesis and host response studies


UPEC
CFT073
UPEC
CFT073

Dose (cfu)

Volume (ml)

Mouse strain

Reference

5  107
5  107

50
50

C57BL/6
C57BL/6

Sivick et al. (2010)


Andersen-Nissen et al.
(2007)
Yadav et al. (2010)
Allsopp et al. (2010)
Allsopp et al. (2012)
Ulett et al. (2007)
Tree et al. (2008)
Totsika et al. (2009)
Chromek et al. (2006)
Fischer et al. (2010)
Duell et al. (2012a)y
Jaillon et al. (2014)
Ingersoll et al. (2008)
Danka & Hunstad
(2014)
Wang et al. (2013)
Wang et al. (2014)
Springall et al. (2001)
Roelofs et al. (2006)
Totsika et al. (2011)
Godaly et al. (2000)
Svensson et al. (2011)
Hang et al. (2000)
Frendeus et al. (2000)
Hopkins et al. (1998)

UPEC
UPEC
UPEC
UPEC
UPEC
UPEC
UPEC
UPEC
UPEC
UPEC, ABU E. coli
UPEC
UPEC

CFT073
CFT073
CFT073
CFT073
CFT073
CFT073
CFT073
CFT073
CFT073
CFT073, 83972
UTI89
UTI89

108
5  108
5  108
5  108
5  108
5  108
108
109
3  109
108
107
107

100
20
20
25
25
20
100
100
40
100
50
50

C57BL/6
C57BL/6
C57BL/6
C57BL/6
C57BL/6
C57BL/6
C57BL/6, 129/SvJ
C57BL/6
C57BL/6, CBA
C57BL/6J
C57BL/6
C57BL/6

UPEC
UPEC
UPEC
UPEC
UPEC
UPEC
UPEC
UPEC
UPEC
UPEC

UTI89
UTI89
J96
1677
EC958
1177
1177
1177
1177
1677

107
107
108
109
5  108
5  107
108
12  108
107
108

50
50
50
100
50
100
100
100
100
50

UPEC
UPEC, E. coli
UPEC, E. coli

Clinical
CFT073, K12
CFT073, clinical
strains, MG1655
CFT073
CFT073
CFT073
CFT073
C1, C2b
CFT073,1177, 536,
NU14, 83972
UTI89
UTI89
UTI89, COH1
HT7
CP9
AL511, AL10

109
109
108

50
25
50

C57BL/6
C57BL/6
C57BL/6
C57BL/6
C57BL/6
BALB/c
BALB/c
BALB/c
BALB/c
BALB/c, C3H/HeJ, C57BL/6,
DBA.1, DBA.2, C3H/OuJ,
SJL, SWR, C3H/HeN, AKR
CBA/J
CBA/J
CBA/J

107 or 109
2  109
5  109
108
5  108
109

50
20
50
50
25
25

CBA/J
CBA/J
CBA/J
CBA/J
CBA
CBA

Lane et al. (2007)


Barbieri et al. (2014)
Snyder et al. (2004)y
Hagan & Mobley (2009)
Sidjabat et al. (2009)
Roos et al. (2006)

108
5  1062  107
12  107
108
6.25  105
108

50
50
50
50
27
50

Schilling et al. (2001b)


Schwartz et al. (2015)y
Kline et al. (2014)
Chassin et al. (2007)
Garcia et al. (2013)y
Chassin et al. (2006)

Clinical
UTI89
83972
UTI89, J96, DS17,
AAEC185/pSH2
Various
UIT89,
TOP52

108
107
2.5  108
1  106 or 1010

100
50
20
10,25 or 100

C3H/HeN, C3H/HeJ
C3H/HeN, C57BL/6J
C3H/HeN, C3H/HeJ
C3H/HeN, C3H/HeOuJ
C3H/HeOuJ
C3H/HeOuJ, C3H/HeJ,
C57BL/6J
C3H/HeN
C3H/HeN
C3H/HeJ
129/sv

109
2  1078

50

Nielsen et al. (2014)


Hannan et al. (2010)

K. pneumoniae,
S. saprophyticus
P. stuartii

ATCC33459,
ATCC15305
BE2467

105

25

Albino OF1
C3H/HeN, DBA/2J, BALB/c,
C3HeB/FeJ, 129S1/SvImJ,
C3H/HeOuJ, C57BL/6J,
C3Smn.CB-Prkdcscid/J,
CBA/J, C3H/HeJ,
THP gene KO

107

50

CBA/J

P. stuartii
P. mirabilis
P. mirabilis

NS
2921
HI4320

109
108
107

NS
50
50

CD-1, CBA
CD-1
CBA/J

P. mirabilis
P. mirabilis
P. aeruginosa

HI4320
Clinical
MPAO1

107
105
5  106

50
50
50

CBA/J
129/sv
Swiss-Webster

Armbruster et al.
(2014)*
Johnson et al. (1987)
Umpierrez et al. (2013)
Armbruster et al.
(2014)*
Pearson et al. (2011)y
Raffi et al. (2009)
Bala et al. (2014)

UPEC
UPEC
UPEC
UPEC
UPEC
UPEC, ABU E. coli
UPEC
UPEC
UPEC, GBS
UPEC
UPEC
UPEC
UPEC
UPEC
ABU E. coli
UPEC
UPEC, E. coli
UPEC, K. pneumoniae

Johnson et al. (1998)


Sabri et al. (2009)
Hagan et al. (2010)y

Connell et al. (1996)


Kostakioti et al. (2012)
Watts et al. (2012b)
Bates et al. (2004)

Raffi et al. (2005)

(continued )

A. J. Carey et al.

Crit Rev Microbiol, Early Online: 120

Table 1. Continued

Bacteria
UPEC
UPEC
E. faecalis
GBS
GBS, UPEC
GBS, UPEC

Strain
CP9
CP9
12030
NS
247, CFT073
247, CFT073

CAUTI and biofilm formation studies


UPEC
UTI89
UPEC
UTI80
UPEC
UTI89
E. faecalis
OG1RF
E. faecalis
OG1RF
E. faecalis
OG1RF
E. faecalis
OG1RF

Downloaded by [Glen Ulett] at 20:50 05 August 2015

E. faecalis
P. aeruginosa
P. mirabilis
K. pneumoniae

OG1RF
PA14
296
TOP52

Treatment and preventative studies


UPEC
UTI89
UPEC
UTI89
UPEC
UTI89
UPEC
CFT073, 83972
UPEC
Clinical
UPEC
Clinical
UPEC
EC958
UPEC
UTI89
CT073, HT7
UPEC
UTI89
UPEC
UPEC
P. mirabilis
P. mirabilis
P. aeruginosa

CFT073
9612
Pr2921
Pr2921
SR10396

Dose (cfu)

Volume (ml)

Mouse strain

Reference

64  10
38  107
35  108
103
3  109
109

25 or 50
25 or 50
100
NS
40
20

Swiss-Webster
Swiss-Webster
BALB/c
Cornett
C57BL/6
C57BL/6

Russo et al. (1996b)


Russo et al. (1996a)
Wobser et al. (2014)
Furtado (1976)
Tan et al. (2012a)y
Ulett et al. (2010)

12  107
108
2  107
1056
104
23.5  107
12  107

50
50
50
200
200
50
50

C57BL/6Ncr
C3H/HeN, C3H/HeJ
C3H/HeN
ICR
ICR
C57BL/6Ncr
C57BL/6Ncr

4  107
106
2.5  104
2.33.3  107

50
35
50
50

C57BL/6
CF-1
C57BL/6
C57BL/6NCr

Guiton et al. (2012)


Anderson et al. (2003)
Anderson et al. (2010)
Singh et al. (2007)
Kemp et al. (2007)
Guiton et al. (2010)
Flores-Mireles et al.
(2014)
Nielsen et al. (2012)
Cole et al. (2014)
Janssen et al. (2014)
Murphy et al. (2013)

108
108
12  107
1  106 or 108
8  108
108
2  1078
12  1078
107

50
100 i.p.
50
25
80
50
NS
50
50
50

C57BL/6
C57BL/6Ncr
C57BL/6Ncr
C57BL/6
Ssc-CF1
BALB/c
C3H/HeN
C3H/HeN
C3H/HeN
CBA/J

108
2.2  109
2  108
2  108
4.7  104

50
50
50
50
100

CBA
CD-1 ICR
CD-1
CD-1
SLC/ICR

79

Schilling et al. (2002)


Goller et al. (2014)
Guiton et al. (2012)
Watts et al. (2012a)
Hvidberg et al. (2000)
Tratselas et al. (2014)
Totsika et al. (2013)
Hannan et al. (2014)
Vimont et al. (2012)
Blango & Mulvey
(2010)
Lefort et al. (2014)
You et al. (2009)
Scavone et al. (2014)
Pellegrino et al. (2003)
Tsuji et al. (2003)

NS, not stated. i.p., intraperitoneal challenge for urosepsis model.


*Same study.
yStudy that reports transcriptomic analysis based on microarrays.

The most common mode of infection is inoculation by


transurethral (intra-bladder, intravesical) delivery of bacteria
directly into the bladder following insertion of a small catheter
(0.6 mm diameter) through the urethral orifice of female
mice. Here, the catheter is inserted through the urethral orifice,
beneath the pelvic bone and through the external and internal
urethral sphincters (total distance 1.2 cm; the female mouse
urethra is 1 cm long, as measured in 810-week-old mice
(Hopkins et al., 1995)) into the bladder until resistance is felt,
then withdrawn several mm prior to direct inoculation into the
bladder lumen (Hung et al., 2009; Johnson & Brown, 1996).
In humans, bladder/urethral/urinary catheterization is always
transurethral (i.e. into the bladder). Thus, transurethral is the
term that we have used here to define the inoculation method in
mice that most closely parallels catheterization in humans.
Alternatively, insertion of the catheter can be restricted to only
several mm (0.5 cm) through the urethral orifice and through
the external urethral sphincter (but not through the internal
urethral sphincter) to achieve intra-urethral delivery in mice
(Hopkins et al., 1995; Hung et al., 2009). However, there is no
intra-urethral variant of catheter insertion in humans (as in
mice). Transurethral inoculation into the bladder by deeper

catheterization may promote bacterial binding to urothelial


cells better than intra-urethral challenge and therefore may lead
to higher bacterial recovery (Hung et al., 2009). However, no
studies have directly compared these approaches in side-byside trials. A third inoculation approach is also possible;
perurethral inoculation, which can be achieved superficially at
the urethral office. In this approach, the urethra is not
cannulated because the catheter is placed only 0.2 cm
within the urethral orifice and does not pass beneath the
pelvic bone, nor through the external or internal urethral
sphincter (i.e. pelvic diaphragm). There is no perurethral
variant of catheter insertion in humans; but this approach may
provide a parallel to UTI in humans where infection develops
in the absence of transurethral catheterization. The three
inoculation methods, illustrating placement of the catheter tip,
are shown in Figure 2.
The speed of delivery of the inoculum (i.e. challenge force)
may also affect bacterial dispersal following inoculation of
the bladder (Hung et al., 2009; Hvidberg et al., 2000;
Johnson, 1998; Johnson & Brown, 1996; Russo et al.,
1996a). To manage this, electronically or mechanically
regulated injection pumps have been used to control the

DOI: 10.3109/1040841X.2015.1028885

Downloaded by [Glen Ulett] at 20:50 05 August 2015

Figure 2. Three different inoculation techniques for murine UTI. The


procedure can be performed using transurethral (A), intra-urethral (B) or
perurethral (C) inoculation methods. The diagram illustrates placements
of the tip of the catheter for AC in the context of the urethral orifice,
urethral sphincters, urethra and bladder.

speed of delivery of the inoculum (Johnson & Brown, 1996;


Mobley et al., 1993). The most commonly used method
involves the use of a 1-ml syringe directly attached to a
catheter for manual injection of inoculum volumes 50 ll
to prevent vesicoureteral reflux (VUR), as discussed
further below.
Alternative infection procedures that do not use transurethral or intra-urethral delivery have been applied in some
studies. For example, surgical opening of the peritoneal cavity
was used to facilitate injection of the inoculum into the
bladder followed by suturing and recovery in one study
(Nicholson & Glynn, 1975). Another mini-surgical bladder
inoculation technique yielded reliable infection results for the
upper and lower urinary tract without systemic complications
or peritonitis and offers excellent potential for advancing the
study of UTI in male mice (Olson & Hunstad, 2014). It is
noteworthy, however, that these methods probably best
parallel transabdominal surgery through the suprapubic
region. Therefore, it is conceivable that these models could
lead to atypical disease manifestations as a reflection of nonphysiologic means of (natural) bladder infection whereby
bacteria originate from the periurethra and ascend into the
bladder.
Suprapubic catheterization is a common surgical procedure for patients where urethral catheterization is not recommended (Tenke et al., 2014). Complications from this include
urine leakage, hematuria and urine retention, which are
unlikely to be associated with transurethral catheterization
(Healy et al., 2012); more practically, this procedure, independent from transabdominal surgery, has not yet been
reported in mice. Future studies that analyze the incidence
of complications arising from these different infection
procedures in mice would thus be of interest. More importantly, studies on male mice will now be possible and may help
to elucidate the basis of recently reported sex differences
between males and females for the development of more
severe UTI (Olson & Hunstad, 2014).
Finally, bloodstream infection by intravenous infection in a
model of hematogenous spread of streptococci to the urinary

Urinary tract infection of mice to model human disease

tract was used in one study to provide a model of urosepsis


(Furtado, 1976). In humans, bloodstream infection arising
from a primary UTI represents ascending infection with
secondary dissemination; bloodstream challenge murine
models represents hematogenous infection and descending
UTI. Thus, while streptococci do cause pyelonephritis and
urosepsis (Ulett et al., 2009, 2012), murine models of
hematogenous infection and descending UTI are minimally
applicable to the human situation since this infection
approach does not parallel the natural progression to urosepsis
in patients.
A final comment on the limitation of the direct mode of
infection in murine models is also warranted since, as noted,
bacteria typically originate from the periurethra and ascend
into the bladder to cause UTI in humans. Thus, the factors
that determine vaginal and periurethral colonization in
humans, and bacterial entry from the periurethral area into
the bladder, are bypassed by direct instillation of bacteria into
the urinary tract in murine models. The importance of this
disconnect with murine UTI models remains unknown.

Variable 2: inoculum properties


Challenge inoculum is a second key variable in murine UTI
models and is summarized according to pathogen and strain,
infectious dose and challenge volume (Table 1). Typically, the
infectious dose in transurethral models is 107109 cfu;
however, there is considerable variation in this dose and
studies have used as few as 103 cfu, and up to 1010 cfu
(Table 1). An infectious dose of 108 cfu is common, and for
studies that enrich UPEC cultures for type 1 fimbriae
expression by serial static subculture, a dose of 107 cfu is
frequently used. This lower dose is linked to the higher
expression of type 1 fimbriae, and their role in mediating
adherence to uroplakins on superficial bladder epithelial cells
(Cusumano et al., 2011; Nielubowicz & Mobley, 2010;
Wu et al., 1996). The incidence of persistent bacteriuria in
C3H/HeN mice that receive 10-fold more type 1 fimbriaeenriched UPEC to establish chronic infection (108 versus 107
cfu) is increased, but the higher dose does not alter kidneybladder titer ratios after 1 month (Hannan et al., 2010). These
data show that infectious dose can influence model read-outs
in separate compartments of the urinary tract independently.
In interpreting data from different murine models one also
needs to consider that the effects of infectious dose can
change as a function of host background. For example, the
progression to chronic cystitis is dose-dependent in C3H/HeN
mice but not C57BL/6 mice (Hannan et al., 2010). Thus,
adjustments to the standard inoculation approach of a single
infectious dose of 108 cfu (or 107 cfu type 1 fimbriae-enriched
cultures) may be useful for different pathogens or specific
studies; for example, uropathogenic streptococci were administered at a high infectious dose of 109 cfu to achieve
consistent ID90 outcomes (Tan et al., 2012a), and differential
dosing was useful to model the effects of specific virulence
factors including flagella in a study on UPEC (Lane et al.,
2007). A more recent study focused on comparing a single
infectious dose with superinfection in C3H/HeN mice and
C57BL/6 mice (Schwartz et al., 2015). Remarkably, superinfection of C57BL/6 mice with UPEC UTI89 rendered these

A. J. Carey et al.

Crit Rev Microbiol, Early Online: 120

Table 2. Normative standards for murine UTI models and application-based modifications.

Downloaded by [Glen Ulett] at 20:50 05 August 2015

Variable

Standard method

Alternative methods
8

Dose

Single dose of 10 cfu

Volume

2550 ll PBS

Inoculum
Injection

Shaking cultures
Deep transurethral insertion,
manual injection

Post-challenge

No restrictions on inoculum voiding

107 cfu in studies using fimbrial-enriched cultures (i.e. serial static subcultures)
One repeat (2nd) dose for superinfection
Increase volume to 100 ll to model pyelonephritis, induce VUR, or for i.p./i.v.
Carrier alternatives: 1% India ink PBS
For studies of fimbrial adhesion, serial static subcultures ideal
Use of electronic or mechanical injection pumps, mini-surgical via subrapubic
delivery or use of indwelling catheter Intra-urethral, or perurethral challenge
Use of i.p. delivery and 108 cfu for urosepsis
Use of i.v. delivery and 106 cfu for urosepsis
Alternative methods as defined by model
Restrictions on inoculum voiding post-challenge, through diet/water restriction,
collodion film, or catheter clamping

mice (that are normally resistant to chronic cystitis following


a single infection) significantly more susceptible to developing persistent bacteriuria and chronic cystitis; serum levels of
IL-6, KC/CXCL1 and G-CSF predicted the development of
chronic cystitis. Notably, however, the response to infection
differed between C3H/HeN and C57BL/6J mice, especially
for immunoglobulin genes.
Inoculum volumes typically range between 10 and 100 ll
(Table 1) (Furtado, 1976; Hagberg et al., 1983; Hopkins
et al., 1995; Hung et al., 2009; Nicholson & Glynn, 1975;
Thai et al., 2010). Differences in volume affect early events
in bladder colonization. Most notably, the UTI condition
being studied (cystitis, pyelonephritis, urosepsis, VUR)
should guide the choice of volume. A standard of 2550 ll
is typical and is increased to 100150 ll to model pyelonephritis and induce VUR (Table 1) (Hopkins et al., 1998;
Johnson, 1998; Morin & Hopkins, 1998). VUR in murine
models most likely relates to both the (larger) inoculum
volume and the (increased) force of injection/speed that
causes reflux to the kidneys (Hopkins et al., 1995); intraurethral or, alternatively, superficial perurethral challenge (at
the urethral orifice), may not induce VUR as readily as
forceful deeper transurethral injection directly into the
bladder (Hopkins et al., 1998; Hung et al., 2009; Johnson,
1998; Johnson & Brown, 1996). In addition, the potential for
high-pressure VUR to induce kidney trauma (e.g. tearing of
tissue, and gross hematuria) as a result of the use of larger
volumes and increased force has also been documented in the
murine UTI model (Johnson & Manivel, 1991). Hydrostatic
injury to the kidney creates a non-physiologic substrate for
infection that is unrepresentative of uncomplicated pyelonephritis (Johnson & Brown, 1996). Thus, inoculationinduced VUR is not desirable if the goal is to mimic the
natural pathogenesis of ascending pyelonephritis, in which
bacteria must ascend the ureter(s) themselves, unaided by
artificial bladder overpressure. Notably, studies on UPEC and
P. mirabilis have shown true ascending UTI in the murine
model based on transurethral inoculation procedures that do
not induce VUR of the inoculum (i.e. 2550 ll volume
delivered by slow infusion) (Hagberg et al., 1983; Hvidberg
et al., 2000; Johnson et al., 1993b). Routine use of a standard
volume of 2550 ll among non-VUR studies will improve the
ability to compare studies where colonization data is derived
using equivalent inoculation methods. We suggest normative
standards for volume and other variables, as well as

alternative methods for application-based modifications in


Table 2, which should aid standardization procedures for
experimental infection in murine UTI models.
The inoculum properties relating to bacterial strain are a
major variable between studies and in many cases, represent
the actual variable under study. In some cases, the bacterial
strain is simply a way to achieve infection for investigation of
aspects of the infection process other than the characteristics
of the strain. However, for studies in which the characteristics
of the strain are the main focus, the choice of strain is critical.
We have summarized the use of different bacterial strains,
chiefly for UPEC, in Table 1. This lists the commonly used
prototype strains including the urosepsis strain, CFT073, that
was isolated from the blood of a pyelonephritis patient, and
the cystitis strain, UTI89, that was isolated from a patient with
recurrent cystitis. UPEC are a highly heterogeneous population and different strains possess different sets of virulence
factors that are important at different stages of UTI. CFT073
is a highly toxigenic strain that can cause severe damage to
the urothelium and immunopathology (Mobley et al., 1990),
but can also invade epithelial cells and form IBCs (Garofalo
et al., 2007). UTI89 is a more invasive strain that forms IBCs
and survives intracellularly, but also expresses several toxins
that cause urothelial damage (Chen et al., 2009). The mosaic
nature of the UPEC genome means that there are no unique
genetic features that clearly distinguish different pathogenicity lifestyles.
Table 1 also highlights strains for which limited data from
murine models are available, such as the recently emerged
and globally disseminated multidrug resistant UPEC clone
of serotype O25b:H4, sequence type 131 (E. coli ST131)
(Totsika et al., 2011). As discussed, type 1 fimbriae are
essential for UPEC colonization of the mouse urinary tract
and many studies employ UPEC cultures enriched for type 1
fimbriae expression following static growth. Given the
importance of this phase-variable phenotype for bladder
colonization, we suggest that the level of type 1 fimbriae
expression should be carefully monitored, particularly for
experiments comparing wild-type and mutant strains.
Methods such as yeast cell agglutination (Schembri et al.,
2000), fim switch PCR (Gally et al., 1996), hemagglutination
(Hagberg et al., 1981) or western blotting using Fim-specific
antibodies would be appropriate.
The use of co-infection models, where various strains of
bacteria are mixed in the challenge inoculum, has proved

Downloaded by [Glen Ulett] at 20:50 05 August 2015

DOI: 10.3109/1040841X.2015.1028885

highly beneficial in identifying the role that various virulence


factors play in pathogenesis cascades including bladder
colonization, the ascension of infection and nutrient acquisition. Co-infection models can involve either (i) the use of
different wild-type strains, where one is examining interactions between the strains, or comparative fitness, or host
responses; or (ii) the use of a parent strain and its mutant or
transconjugant derivative in an effort to identify the impact of
a specific trait on competitive fitness. Competition studies
using UPEC and mutants deficient in various genes required
for metabolism, for example, showed that the tricarboxylic
acid cycle and gluconeogenesis contributes to virulence
in vivo (Alteri et al., 2009b). Similar studies have been
performed to characterize the role of flagella in UPEC fitness
(Lane et al., 2005), and autotransporter proteins in bladder
colonization (Allsopp et al., 2010, 2012). Mixed infections
using UPEC mutants lacking iron receptors have demonstrated that the uptake of either aerobactin or yersiniabactin is
more important for colonization of the UPEC strain CFT073
than enterobactin or salmochelin utilization (Garcia et al.,
2011). However, a different siderophore dependence profile
for colonization of the urinary tract was reported for the ABU
strain 83972 (Watts et al., 2012b). Co-infection with
P. mirabilis and P. stuartii was recently shown to increase
the incidence of bacteremia in a urease-dependent manner in
a murine model of ascending UTI (Armbruster et al., 2014).
Studies have also shown that mixed infection with different
uropathogens can significantly alter the outcomes of infection. Co-infection of GBS and UPEC, for example, increases
the fitness of UPEC in the bladder during acute infection,
perhaps through modulation of innate immune responses.
This co-infection combination was also shown to increase the
risk of developing latent bacterial reservoirs in mice and
pyelonephritis (Kline et al., 2012), as well as acute high-titer
cystitis in aged multiparous mice (Kline et al., 2014).

Variable 3: mouse strain


Different mouse strains respond to UTI distinctly and results
generated in one strain are not always applicable to other
strains. For example, different immune responses in different
mouse strains can result in the development of acute or
chronic UTI. Divergent responses to UTI in mice were first
noted in a comparison of ten inbred strains (Hopkins et al.,
1998). The relative susceptibility and resistance phenotypes
of commonly used mouse strains were more recently
quantitated (Hannan et al., 2010). These data illustrate the
major influence of host genetic background, as reviewed
elsewhere (Ragnarsdottir et al., 2011; Svanborg Eden et al.,
1985). Importantly, dissimilar responses between different
mouse strains reflect the genetic diversity in human populations and provide the opportunity to infer clinical relevance
based on genetic differences (Hopkins et al., 1998;
Ragnarsdottir et al., 2011). In other words, no single mouse
strain is ideally suited to mimic all of the diverse requirements
needed to model different types of human UTI. A summary of
infection and inflammation differences (in terms of severity)
between different mouse strains is shown in Figure 3.
Most studies have used one of three inbred mouse strains:
C57BL/6, CBA/J or C3H/HeN; often with genetically distinct

Urinary tract infection of mice to model human disease

Figure 3. The severity of inflammation and time course of UPEC


bladder infections in different mouse strains. The time course of
infection was derived from comparison of mice transurethrally infected
with 108cfu UPEC (Hannan et al., 2010; Hopkins et al., 1998). The
incidence of persistent bacteriuria in the mouse strains, presented in the
study by Hannan et al. (2010), was used to numerically rank the strains
from lowest to highest incidence. This numerical ranking was also done
for the levels of bladder infections in the same strains of mice (Hannan
et al., 2010). The average of these two rankings was calculated and used
to generate an ordered list of mouse strains that resolve infection, or are
more likely to develop persistent infection. This list was compiled with
the results of the levels of bladder infections reported by Hopkins et al.
(1998) at the day 14 time point. Similarly, the severity of inflammation
based on inflammatory scores reported by Hopkins et al. (1998) at day
14 post-infection, and the mouse strains that were examined for
inflammation by Hannan et al. (2010) were ranked and compiled.
While some of the mouse strains ranked differently between the two
studies, the results from Hannan et al. (2010) were considered to be
actual measures of persistence and were used preferentially.

derivatives or related strains with contrasting susceptibility


phenotypes (e.g. C3H/HeJ) (Svanborg Eden et al., 1985).
C57BL/6 mice are intermediately susceptible to acute UTI
and progressively resolve bacteriuria over 14 days, with
sporadic and low-level kidney infection (Duell et al., 2012a;
Hannan et al., 2010; Hopkins et al., 1998); these mice are
resistant to chronic cystitis but exhibit low-level bladder
colonization (104 cfu/ml) at 14 days after a single infection
(Hannan et al., 2010). Superinfection can lead to increased
rates of persistent bacteriuria and chronic cystitis in the
C57BL/6 strain (Schwartz et al., 2015). In addition, C57BL/6
mice provide the advantage of having many gene-deficient
mutant derivatives available. CBA/J mice are useful to model
ascending UTI and, like DBA/2J and C3H/HeN mice
(Schwartz et al., 2015), are susceptible to chronic cystitis
and inflammation (Hagan & Mobley, 2009; Johnson et al.,
1998; Sabri et al., 2009; Snyder et al., 2004).

Downloaded by [Glen Ulett] at 20:50 05 August 2015

10

A. J. Carey et al.

However, CBA/J mice develop pyelonephritis at a lower


frequency compared to C3H/HeN and C3H/HeN-derived
strains (Hannan et al., 2010). C3H/HeN mice are more
susceptible to persistent bacteriuria compared to C57BL/6
mice, and exhibit two, mutually exclusive, outcomes to acute
infection: either persistent, high-level bacteriuria (4104 cfu),
bladder colonization (4104 cfu) and chronicity over 4 weeks
(Hannan et al., 2010); or resolution of bacteriuria (Schwartz
et al., 2015). C3H/HeN mice have been particularly useful in
studies with the LPS-non-responsive C3H/HeJ strain to
elucidate the role of TLR4 in UTI pathogenesis, as discussed
in the section above.
Approaches to modify innate susceptibility of mice to
model specific forms of human UTI have also been
developed. For example, the use of chemical destruction of
pancreatic islet cells has been used to study diabetes and UTI.
C3H/HeN, C3H/HeJ and C57BL/6 mice that are rendered
diabetic by intraperitoneal injection of streptozocin exhibit
increased susceptibility to UTI following infection with
UPEC, K. pneumoniae and E. faecalis (Rosen et al., 2008).
These data parallel the association between diabetes and
increased susceptibility to UTI in adults (Hoepelman et al.,
2003). UTI in pregnancy has also been modeled using several
strains of mice. In a unique murine model of UTI-induced
pre-term labor in pregnant C3H/HeJ mice, the survival rates
of mothers and pups were influenced by UPEC bearing the Dr
adhesin (Kaul et al., 1999). Another study in pregnant
C57BL/6J mice showed that despite the inability to culture
UPEC from uterine tissue, a pattern of inflammation within
uterine tissue was observed and this correlated with a
decrease in fetal weight (Bolton et al., 2012). These data
suggest that inflammation due to UTI during pregnancy is not
localized to the bladder, and may affect urogenital tract tissue
more generally.
The source of mice may also be important. In one study
using outbred Swiss-Webster mice, for example, the investigators reported a supplier-dependent variation in renal
infection, as well as a divergence in the development of
pyelonephritis (Russo et al., 1996a). It is possible that this
relates to outbred strains and hybrid vigor (i.e. genetic
differences between parents that produce synergistic, favorable effects in offspring), but these findings emphasize the
benefits of single-source suppliers and highlight the potential
problems that may arise from sub-standard animal care,
housing and quality control. Finally, the influence of distinct
anatomy, diet, urinary composition, behavior, and micturition
habits between mice and humans in the context of UTI, and
the potential correlation between these factors and pathogen
(Nesta et al., 2012), is unclear. An entirely unexplored area is
the role of the gut microbiome in influencing susceptibility to
UTI. Recent data related to other mucosal sites (Ichinohe
et al., 2011; Pang & Iwasaki, 2012; Oh et al., 2014) suggest
this will be an exciting area for future murine UTI model
research.
Duration of observation post-inoculation is also an
important variable related to mouse strain (because of
differential susceptibility) (Hannan et al., 2010). There is
some evidence of a time-dependent sequence of events after
bacterial challenge; observation at a given time point will
catch only part of that and the part observed will be

Crit Rev Microbiol, Early Online: 120

determined by the time point chosen. Inoculum properties


also relate to the duration of observation post-inoculation
since differences in doses and virulence may affect the time
course of events after challenge. Particular time points have
highlighted the dynamic nature of cytokine production, for
example, where some responses peak while other subside at a
given time point. Ingersoll et al. (2008) addressed the issue
by performing time course studies of bacterial clearance and
defining how this relates to host responses (Ingersoll et al.,
2008). For data on human UTI, definitive time course analysis
is rarely possible, except in inoculation studies (Hull et al.,
2000; Koves et al., 2014; Sunden et al., 2010; Wullt et al.,
1998). Lane et al. (2007) used an IVIS to monitor infection in
mice in real-time, which provides a valuable approach for
extended time course studies (Lane et al., 2007). Thus, the
murine model provides critical information on the dynamics
of infection and host responses, which is limited by the
duration of observation post-inoculation.

Transcriptomic studies of UPEC and host responses


Analysis of bacterial and host genes that are activated or
inactivated during UTI has led to the identification of several
key elements of pathogenesis. Similar patterns of UPEC gene
expression between in vivo- and in vitro-grown bacteria have
been defined. Analysis of UPEC grown in human urine or
synthetic media in vitro with UPEC recovered ex vivo from
mouse urine or human urine during UTI has also revealed
niche-dependent gene expression. For example, UPEC grown
in human urine in vitro up-regulates genes for iron acquisition
(e.g. sit, fhuA, chuA), capsular polysaccharide and LPS
synthesis (Hagan et al., 2010; Snyder et al., 2004), which
parallels UPEC gene expression patterns in mice (i.e. similar
patterns); however, the expression patterns for these genes
differ in UPEC grown in LB (i.e. divergent patterns) (Snyder
et al., 2004).
Other niche-dependent patterns of gene expression relate
to UPEC genes that encode fimbriae. Enhanced expression of
type 1 fimbriae and down-regulation of P fimbriae and
flagella expression occurs in mice, but not in human urine
cultures in vitro. In the latter, other adhesin and UPEC
motility genes including papA_2 (major P fimbriae subunit)
and fliC are up-regulated (Snyder et al., 2004). In making
such comparisons it is important to consider the experimental
conditions in which gene expression is compared, since, for
example, the mode of in vitro bacterial growth (e.g. shaking
verses static serial subculture) influences the expression of
genes including type 1 fimbriae. Comparisons of gene
expression in UPEC isolated from urine of infected mice,
UPEC isolated from cystitis patients, and UPEC grown in
human urine ex vivo revealed similar gene expression profiles
(Hagan et al., 2010). Comparing patient-recovered UPEC
strains with those grown ex vivo revealed up-regulation of cell
division factors (ftsW), protein secretion components (secD,
yidC), and nucleotide synthesis (guaA, pyrG) in vivo,
implying faster replication of UPEC during UTI in humans.
UPEC isolated from patients also express genes for iron
acquisition (chuA, fhuA) and respiration (cyoABCD, cydAB),
as occurs in mice. Up-regulation of UPEC genes associated
with iron acquisition in IBCs in murine bladder was also

Urinary tract infection of mice to model human disease

DOI: 10.3109/1040841X.2015.1028885

11

Table 3. Future development of murine UTI models for study of human disease.
Model

Whats needed

Possible outcome/advancement

Urosepsis

Characterization of sepsis/systemic infection models


that more closely mimic human route of infection
(bladder > kidney > blood)
Comparison of intra-urethral and/or superficial
perurethral challenge approach for injection
Standardization of challenge dose and volume
Characterization of superinfections models
Study of dynamics of bacterial persistence in urine
in the absence of severe inflammation
Comparison of urinary constituents between mice
and humans utilized by bacteria in ABU
Development of indwelling catheter models for
different uropathogens that cause CAUTI
Characterization of UTI during pregnancy

More physiological studies of human urosepsis


Enable novel comparisons of uropathogens

Transurethral infection

ABU

CAUTI
Pregnancy

Downloaded by [Glen Ulett] at 20:50 05 August 2015

SCID-hu UTI

Development and characterization of novel


humanized murine models of UTI

reported (Reigstad et al., 2007). Iron acquisition is fundamental to microbe survival in the urinary tract, and the
increased transcription of iron acquisition genes by UPEC
during IBC formation contributes to UPEC intracellular
survival. Conserved UPEC transcriptional responses (Figure
1) provide a context for future studies that depend on interspecies convergence of bacterial gene expression patterns.
Divergent gene expression between UPEC in humans and
mice can also provide useful insight into potential disease
mechanisms by revealing inter-species differences. For
example, it was found that genes encoding type 1 fimbriae
(e.g. fimA, fimH) are highly expressed in UPEC recovered
from urine of infected mice but not in UPEC voided in human
urine (Hagan et al., 2010; Snyder et al., 2004). However,
UPEC do express fimA when grown in human urine in vitro
(Hagan et al., 2010). Thus, type 1 fimbriae are required for
(and expressed during) infection in mice but their expression
during distinct stages of human UTI requires more study. It is
likely that adhesin-expressing bacteria may be selectively
retained within the urinary tract and not shed in urine, which
could complicate conclusions of adhesin non-expression in
vivo during UTI. Together, these studies show that conserved
patterns of gene expression in UPEC occur in mice and
human UTI ex vivo (recovered) and in vitro (urine assay), as
do divergent UPEC responses between mice and humans.
Recent studies that profile UPEC gene transcription during
human UTI using RNA-seq have advanced our understanding
of
UPEC
pathogenesis
(Bielecki
et al.,
2014;
Subashchandrabose et al., 2014). Future studies to functionally define the roles of conserved and divergent UPEC gene
expression patterns, including at different stages of infection,
will now be essential.
Recent studies have also probed gene expression in
uropathogens other than UPEC using in vitro and ex vivo
models. In one study, microarray analysis of P. mirabilis
isolated from mice demonstrated that genes encoding mannose-resistant Proteus-like fimbriae (e.g. mrpA) were upregulated along with genes for amino acid and carbohydrate
transporters (e.g. dppA, oppA), energy production and iron

Identify potential differences between methods


Allow a standardized method for more direct comparisons between different studies
Model rUTI after frequent sexual intercourse
More physiological studies of mechanisms underlying human bacteriuria and ABU
Identify bacterial metabolic processes in ABU
Study virulence factors for biofilm formation
Link consequences of untreated UTI to adverse
pregnancy outcomes
More physiological studies of human UTI
Identify human-specific factors and responses
involved in the control of, or susceptibility to, UTI

transport (e.g. exbBD, sitABC) (Pearson et al., 2011). Some


bacteria can grow efficiently in urine and, as a fitness trait,
this can influence the course of UTI, including ABU (Ipe
et al., 2013). The expression of several genes in E. coli
involved in metabolism has been linked to urine growth
(Alteri & Mobley, 2007; Roos & Klemm, 2006; Roos et al.,
2006; Russo et al., 1996b; Watts et al., 2012b). Examination
of E. faecalis in human urine also demonstrated up-regulation
of iron transporter genes (e.g. feoA, feoB) in this organism
(Vebo et al., 2010). Together, these data alongside those for
UPEC confirm the importance of genes involved in iron
transport and metabolism for fitness in the nutrient poor
environment of urine.
Recent transcriptomic studies in mice have also broadened
our knowledge of the host immune response to UTI and
revealed responses that are relevant to co-infection. Initial
bladder responses are complex with 1564-genes and 2507genes altered in the bladder in response to UPEC at 2 and
24 h, respectively (Duell et al., 2012a; Tan et al., 2012a).
Strong activation of IL-10 has implications for immune
responses during UTI because IL-10 is a master regulator of
immunity (Duell et al., 2012b). Direct comparison of gene
expression activated by GBS revealed only 172 genes in the
response to infection; 68 were also triggered by UPEC. Thus,
unique bladder responses are triggered by different uropathogens. Notably, there are few studies on human bladder
responses to UTI and responses between mice and humans
can differ (Seok et al., 2013). A recent study of inflammasomes in the murine bladder urothelium (Hughes et al.,
2014), and their role in inflammation and physiology (i.e.
urodynamics), highlights an area for future research.
Studies on virulence factors that impact pathogen-specific
immune responses are now needed. For example, UPEC
cytotoxic necrotizing factor (CNF1) and hemolysin (HlyA1)
induce the expression of several inflammatory mediators in
the bladder or urine in mice 24 h after infection; including
IL-6, CXCL1, CXCL2 and TNF-a (Garcia et al., 2013),
which is consistent with the transcriptomic studies discussed
above. UPEC infection of the mouse bladder also leads to the

12

A. J. Carey et al.

up-regulation of genes associated with suppression of transforming growth factor-b and the glycoprotein Wnt5a; both
genes are associated with renewal of the bladder epithelium
(Mysorekar et al., 2002). This suppression slows regeneration
of the uroepithelium and may enhance susceptibility to
recurrent infection. It would be of interest to determine
whether UPEC infection of the human bladder triggers
equivalent gene expression responses. Collectively, these
studies have provided important information with regard to
hostpathogen interactions and bacterial subversion of the
immune response to UTI. Future work employing nextgeneration sequencing methods (e.g. RNA-seq) will help to
further profile the genetic signatures that define transcriptomic changes during UTI.

Downloaded by [Glen Ulett] at 20:50 05 August 2015

CAUTI and biofilms


The use of indwelling catheters results in enhanced predisposition to UTI through the introduction of organisms into the
bladder (from the urethra), direct damage to the urothelium,
and the provision of an abiotic surface for biofilm formation.
Biofilms are recalcitrant to antibiotics and immune responses,
a property mediated by multiple factors including the
production of a polymeric matrix that is difficult to penetrate.
Numerous bacterial factors contribute to the establishment of
UPEC biofilms on inert surfaces, including type 1 and some
other chaperone-usher fimbriae, and the autotransporter
proteins Ag43, UpaG and UpaH (Allsopp et al., 2010;
Murphy et al., 2013; Ong et al., 2008; Stahlhut et al.,
2012; Ulett et al., 2007). The production of curli and
cellulose have also been shown to mediate the formation of
biofilms through participation in adherence to human bladder
and kidney epithelial cells in vitro and in the early stages of
murine UTI (Hadjifrangiskou et al., 2012; Kai-Larsen et al.,
2010).
Recently, a murine model of enterococcal CAUTI was used
in which silicone implants were inserted transurethrally,
followed by infection (Guiton et al., 2010; Nielsen et al.,
2012). This model has permitted the identification of new
cell-surface factors that contribute to biofilm formation,
including the enterococcal surface protein, Esp (Shankar
et al., 2001), the pilus-associated sortase A and C (Guiton
et al., 2010; Kemp et al., 2007) and the endocarditis and
biofilm-associated pilus, Ebp (Nielsen et al., 2012). In
addition, this method demonstrated that foreign bodies
elicit major histological changes in the bladder and local
pro-inflammatory cytokine responses. In studies on the
application of the model to E. faecalis-mediated UTIs,
the investigators demonstrated that the bacteria induce the
expression IL-1b and macrophage inflammatory protein 1a
(CCL3). These findings represent a significant advance in our
understanding of CAUTI in the host, and underscore the
importance of urinary catheterization during E. faecalis
uropathogenesis in terms of driving specific pro-inflammatory cytokine responses (Guiton et al., 2010).
Catheter implantation into the bladder permits the study of
bacterial virulence factors that are involved in the formation
of biofilms in vivo. A recent study demonstrated that catheter
implantation induces the release of the extracellular matrix
protein fibrinogen, which can coat catheter surfaces and act as

Crit Rev Microbiol, Early Online: 120

a receptor for E. faecalis Ebp-mediated biofilm formation


(Flores-Mireles et al., 2014). The insertion of a catheter also
triggers sloughing of the urothelium, which can elicit the reactivation of quiescent intracellular bacteria (Guiton et al.,
2012). Urinary catheter implantation in mice that have
resolved bacteriuria can lead to recurrent bacteriuria with
the original infecting strain. This is believed to be associated
with the existence of quiescent intracellular reservoirs of
bacteria that provide a source for recurrent infection (Guiton
et al., 2012). This model could be useful for the evaluation of
new antimicrobial targets for the treatment of CAUTI (Guiton
et al., 2010). Finally, the potential effects of analgesia should
be considered in models that use chronic administration of
analgesics since this has been shown to influence the
development of experimental UTI in rats (Vivaldi, 1968).

Intracellular bacterial communities


UPEC form IBCs within superficial bladder epithelial cells of
mice during the early stages of acute UTI (Anderson et al.,
2003; Mulvey et al., 2001). IBCs share several characteristics
with biofilms; for example UPEC utilize type 1 pili and Ag43
for the formation of IBCs and produce a matrix comprised
primarily of polysaccharide material. Bacteria within an IBC
also undergo characteristic morphological changes as seen in
biofilm communities on abiotic surfaces (Anderson et al.,
2010). IBCs have been identified in cells obtained from the
urine of children with recurrent E. coli UTIs; staining of cells
isolated from the urine revealed long filamentous bacteria and
disrupted cells with bacteria within and protruding from the
cell membrane (Robino et al., 2013, 2014). Similar to this,
cells isolated from women with acute UTI or ABU
demonstrated the presence of IBCs and characteristic filamentous bacteria (Rosen et al., 2007). Data from murine
models suggest that IBCs possess similar properties to
biofilms; they protect UPEC from host immune responses
and antibiotic treatment. The use of murine models to
examine this aspect of UTI has been invaluable and has
provided novel insight into UPEC pathogenesis.

Treatment and prevention studies in mice


Blango & Mulvey (2010) tested 17 different antibiotics for
effectiveness in killing UPEC UTI89 both in vitro and in vivo
in relation to IBCs. This study showed that most antibiotics
were able to limit UTI89 growth in vitro. However, in their
murine UTI model, the authors reported a significantly higher
minimum inhibitory concentration for biofilm cultures
compared to those observed in vitro, which were associated
with IBC formation in the bladder. This led to a conclusion
that UPEC persistence in the bladder is related to the entry of
UPEC into a quiescent or semi-quiescent state within host
cells, and the resilient permeability barrier function associated with the bladder urothelium (Blango & Mulvey, 2010).
Other murine UTI models have also been used to examine the
efficacy of various treatments. For example, Hvidberg et al.
(2000) used eleven clinical isolates of UPEC and one nonpathogenic laboratory E. coli strain to test the therapeutic
efficacy of cefuroxime and gentamicin and measured the
concentrations of the antibiotics in serum, urine, kidney and
bladder tissues. These studies concluded that while the levels

Downloaded by [Glen Ulett] at 20:50 05 August 2015

DOI: 10.3109/1040841X.2015.1028885

of antibiotics were high in urine, this had little effect on the


number of infecting bacteria in the bladder (Hvidberg et al.,
2000). Ultimately, the limitations and advantages of murine
models for evaluating new therapeutics in such studies depend
on the degree to which these models can mimic human
pharmacology and UTI features.
With the increasing rates of antibiotic resistance, there is
an emerging need to identify alternative approaches to prevent
UPEC infection of the urinary tract. The use of mannosides,
which function as anti-adhesive compounds by preventing
FimH-mediated binding to the bladder epithelium, has proven
to be effective in the prevention of acute UTI and the
treatment of chronic UTI in murine models (Cusumano et al.,
2011; Guiton et al., 2012; Han et al., 2010; Totsika et al.,
2013). The treatment of mice with mannosides reduces IBC
formation in animals with catheter implants and potentiates
the effect of antibiotics (Guiton et al., 2012). This approach
may provide an alternative to antibiotic therapy that is often
ineffective in humans. Importantly, the prophylactic use of the
lead mannoside derivative ZFH-04269 prevented the establishment of acute UTI by the multidrug resistant E. coli
ST131 strain EC958, and significantly reduced chronic
bladder infection in mice following one round of treatment
(Totsika et al., 2013). These studies provide strong evidence
for the utilization of mannosides in the treatment of UTI
caused by multidrug resistant UPEC strains. In a more recent
study reported by Hannan et al. (2014), the authors showed
that non-steroidal anti-inflammatory drugs such as cyclooxygenase-2 inhibitors may represent an effective treatment
and/or preventive strategy as an alternative to antibiotics in
certain patients who are susceptible to rUTI (Hannan et al.,
2014). Another conceptually new class of therapeutic molecules that target capsule biogenesis was recently used to
successfully treat systemic UPEC infection in mice (Goller
et al., 2014).
Murine models have also been used for testing the efficacy
of UPEC vaccine antigens. Multiple animal studies have
shown immunogenicity and the presence of vaccine-specific
antibodies after immunization for several different vaccine
approaches, as reviewed elsewhere (Brumbaugh & Mobley,
2012; Moriel & Schembri, 2013). Systemic immunization
with FimH afforded long lasting protection against UPEC
UTI in one study, leading to a 99% reduction in mouse
bladder colonization and kidney infection and high IgG titers
in serum and urine (Langermann et al., 1997); similarly
FimH fused to FliC, a major flagellin subunit, afforded
protection in another murine model (Asadi Karam et al.,
2013). Pyelonephritis was prevented by subcutaneous immunization with synthetically produced peptide fragments of the
UPEC P fimbrial major subunit protein (Schmidt et al.,
1988). In another study, recombinant P fimbriae were
delivered subcutaneously and afforded protection against
homologous and heterologous piliated UPEC strains (Pecha
et al., 1989).
Subcutaneous immunization with IroN, a siderophore
receptor on E. coli, prevented pyelonephritis but did not
provide protection against cystitis (Russo et al., 2003).
A large-scale screening and selection process paired with
in vivo experiments identified three conserved vaccine
candidates, Hma, IutA and IreA (Alteri et al., 2009a).

Urinary tract infection of mice to model human disease

13

These surface-expressed proteins, all involved in iron acquisition by UPEC, provided protection against challenge with
the UPEC strain CFT073. Intranasal immunization with IutA
and IreA stimulated antigen-specific IgA secretion into the
urine, which was directly correlated to protection in the
bladder. Despite antigen-specific IgA responses in the urinary
tract of animals immunized with Hma, only the kidneys were
protected from infection (Alteri et al., 2009a). Subsequent to
this, FyuA, another UPEC iron receptor, was identified as a
potential vaccine candidate, protecting from kidney infection
through the production of IgG (Brumbaugh et al., 2013).
A combination of the four UPEC surface-expressed iron
receptors may prove efficacious as a multivalent vaccine
(Wieser et al., 2010). Overall, however, the efficacy of
experimental vaccines to prevent rUTI in humans has been
difficult to establish in the absence of well-structured clinical
trials. Still, there is promise; for example, clinical trials of
Uro-Vaxom that have been underway for 30-years have
demonstrated efficacy for reducing the incidence of rUTI, as
reviewed elsewhere (Brumbaugh & Mobley, 2012). Data
derived from murine UTI models (Baier et al., 1997; Bessler
et al., 2009, 2010; Bosch et al., 1988; Huber et al., 2000)
provide the opportunity for complementary insight into such
clinical trials. Finally, a recent study reported successful
blocking of the binding of E. faecalis to fibrinogen to prevent
catheter-associated bladder infection in mice as a novel
vaccine approach for this type of UTI (Flores-Mireles et al.,
2014).

Are human-mouse chimeras the model of the future?


Finally, future studies aimed at the development of UTI
models utilizing humanmouse chimeras could serve as a
valuable tool to provide mouse models that more closely
parallel human UTI. Severe combined immunodeficient
(SCID)-hu mice have been used to model hostpathogen
interactions in various forms (Davis & Stanley, 2003). In one
study, SCID mice were more susceptible to UTI than nude
mice (Hopkins et al., 1993). While this indicates a lack of
thymus-derived T cells does not predispose to UTI (Hopkins
et al., 1993; Jones-Carson et al., 1999), the differences in
lymphocyte responses between mice and humans (Deknuydt
et al., 2009; Ness-Schwickerath & Morita, 2011) imply a need
to study SCID mice further in UTI chimera models to explore
these findings. There is also the possibility of utilizing
xenograft models in the future if human tissues or cell samples
can be acquired. An intestinal model using SCID mice has
been successfully developed where human intestinal xenografts were placed into the subcapsular regions of the mice and
allowed to develop (Savidge et al., 1995). This was used to
examine responses to various infections, including Shigella
(Zhang et al., 2001). In theory, such an approach could be
adapted for the urinary tract using similar methods; the
limiting factor being the availability of human bladder tissue.
Murine models of this type could help to verify observations
from other standard murine models of UTI (Table 3).

Conclusions
Murine UTI models represent a powerful method to study
human UTI. It is vital to consider the variables encompassed

Downloaded by [Glen Ulett] at 20:50 05 August 2015

14

A. J. Carey et al.

in these models and the manner in which they relate to human


UTI. In this review, we provide a framework that should
aid better standardization of procedures to study UTI. In
particular, normative standards for the variables discussed
herein including dose, volume, inoculum, injection and
procedures post-challenge should be applied where possible
to aid standardization of methods in studies. This includes
application-based modifications to the standard method (i.e.
single 108 cfu dose in 2550 ml PBS, using deep transurethral
insertion and manual injection with no restrictions on
inoculum voiding) in the context of alternative strategies,
such as those aimed at modeling VUR, super-infection and
urosepsis. Some alternatives to this standard method may
include a lower infectious dose, repeat dose(s), a higher
inoculum volume (e.g. to model pyelonephritis), the use of
an indwelling catheter, alternate challenge method (e.g. i.p.
delivery to model urosepsis), or restrictions on inoculum
voiding post-challenge. Consideration of these methods and
alternatives will help to bring the central variables in murine
UTI models to the forefront of studies so that we may improve
data interpretation and study design. All of the murine models
described in this review will continue to advance the UTI
field by offering data to inform and guide new pathogenesis,
treatment and preventative studies for these very common
infections.

Declaration of interest
This study was supported with funding from the National
Health and Medical Research Council of Australia
(APP1084889). A.J.C. is supported by a National Health
and Medical Research Council of Australia Peter Doherty
Australian Biomedical Fellowship (APP1052464); C.K.T. is a
Prime Ministers Endeavour Asia Fellow; M.A.S. and G.C.U.
are supported by Future Fellowships from the Australian
Research Council (FT100100662 and FT110101048, respectively). There are no other declarations of interest.

References
Agace WW. (1996). The role of the epithelial cell in Escherichia coli
induced neutrophil migration into the urinary tract. Eur Respir J
9:171328.
Agace WW, Hedges SR, Ceska M, Svanborg C. (1993). Interleukin-8
and the neutrophil response to mucosal gram-negative infection. J Clin
Invest 92:7805.
Akil I, Ozkinay F, Onay H, et al. (2012). Assessment of toll-like
receptor-4 gene polymorphism on pyelonephritis and renal scar. Int J
Immunogenet 39:3037.
Alamuri P, Eaton KA, Himpsl SD, et al. (2009). Vaccination with proteus
toxic agglutinin, a hemolysin-independent cytotoxin in vivo, protects
against Proteus mirabilis urinary tract infection. Infect Immun
77:63241.
Allsopp LP, Beloin C, Ulett GC, et al. (2012). Molecular characterization
of UpaB and UpaC two new autotransporter proteins of uropathogenic
Escherichia coli CFT073. Infect Immun 80:32132.
Allsopp LP, Totsika M, Tree JJ, et al. (2010). UpaH is a newly identified
autotransporter protein that contributes to biofilm formation and
bladder colonization by uropathogenic Escherichia coli CFT073.
Infect Immun 78:165969.
Alteri CJ, Hagan EC, Sivick KE, et al. (2009a). Mucosal immunization
with iron receptor antigens protects against urinary tract infection.
PLoS Pathog 5:e1000586.
Alteri CJ, Mobley HL. (2007). Quantitative profile of the uropathogenic
Escherichia coli outer membrane proteome during growth in human
urine. Infect Immun 75:267988.

Crit Rev Microbiol, Early Online: 120

Alteri CJ, Smith SN, Mobley HL. (2009b). Fitness of Escherichia coli
during urinary tract infection requires gluconeogenesis and the TCA
cycle. PLoS Pathog 5:e1000448.
Andersen-Nissen E, Hawn TR, Smith KD, et al. (2007). Cutting edge:
Tlr5/ mice are more susceptible to Escherichia coli urinary tract
infection. J Immunol 178:471720.
Anderson GG, Goller CC, Justice S, et al. (2010). Polysaccharide
capsule and sialic acid-mediated regulation promote biofilm-like
intracellular bacterial communities during cystitis. Infect Immun 78:
96375.
Anderson GG, Palermo JJ, Schilling JD, et al. (2003). Intracellular
bacterial biofilm-like pods in urinary tract infections. Science 301:
1057.
Armbruster CE, Smith SN, Yep A, Mobley HL. (2014). Increased
incidence of urolithiasis and bacteremia during Proteus mirabilis and
Providencia stuartii coinfection due to synergistic induction of urease
activity. J Infect Dis 209:152432.
Artifoni L, Negrisolo S, Montini G, et al. (2007). Interleukin-8 and
CXCR1 receptor functional polymorphisms and susceptibility to acute
pyelonephritis. J Urol 177:11026.
Asadi Karam MR, Oloomi M, Mahdavi M, et al. (2013). Vaccination
with recombinant FimH fused with flagellin enhances cellular and
humoral immunity against urinary tract infection in mice. Vaccine 31:
121016.
Baier W, Sedelmeier EA, Bessler WG. (1997). Studies on the immunogenicity of an Escherichia coli extract after oral application in mice.
Arzneim-Forsch 47:9805.
Bala A, Chhibber S, Harjai K. (2014). Pseudomonas quinolone
signalling system: a component of quorum sensing cascade is a
crucial player in the acute urinary tract infection caused by
Pseudomonas aeruginosa. Int J Med Microbiol 304:1199208.
Barbieri NL, Nicholson B, Hussein A, et al. (2014). FNR regulates
expression of important virulence factors contributing to pathogenicity of uropathogenic Escherichia coli. Infect Immun 82:508698.
Barnett BJ, Stephens DS. (1997). Urinary tract infection: an overview.
Am J Med Sci 314:2459.
Bates JM, Raffi HM, Prasadan K, et al. (2004). TammHorsfall protein
knockout mice are more prone to urinary tract infection: rapid
communication. Kidney Int 65:7917.
Bessler WG, Puce K, Vor Dem Esche U, et al. (2009).
Immunomodulating effects of OM-89, a bacterial extract from
Escherichia coli, in murine and human leukocytes. Arzneim-Forsch
59:5717.
Bessler WG, Vor Dem Esche U, Zgaga-Griesz A, Ataullakhanov R.
(2010). Immunostimulatory properties of the bacterial extract OM-89
in vitro and in vivo. Arzneim-Forsch 60:3249.
Bielecki P, Muthukumarasamy U, Eckweiler D, et al. (2014). In vivo
mRNA profiling of uropathogenic Escherichia coli from diverse
phylogroups reveals common and group-specific gene expression
profiles. MBio 5:e0107514.
Billips BK, Forrestal SG, Rycyk MT, et al. (2007). Modulation of host
innate immune response in the bladder by uropathogenic Escherichia
coli. Infect Immun 75:535360.
Blango MG, Mulvey MA. (2010). Persistence of uropathogenic
Escherichia coli in the face of multiple antibiotics. Antimicrob
Agents Chemother 54:185563.
Bokil NJ, Totsika M, Carey AJ, et al. (2011). Intramacrophage survival
of uropathogenic Escherichia coli: differences between diverse
clinical isolates and between mouse and human macrophages.
Immunobiology 216:116471.
Bolton M, Horvath Jr DJ, Li B, et al. (2012). Intrauterine growth
restriction is a direct consequence of localized maternal uropathogenic Escherichia coli cystitis. PLoS One 7:e33897.
Bosch A, Benedi VJ, Pares R, Jofre J. (1988). Enhancement of the
humoral immune response and resistance to bacterial infection in mice
by the oral administration of a bacterial immunomodulator (OM-89).
Immunopharm Immunotoxicol 10:33343.
Bower JM, Eto DS, Mulvey MA. (2005). Covert operations of
uropathogenic Escherichia coli within the urinary tract. Traffic 6:
1831.
Brown PD. (1999). Antibiotic selection for urinary tract infection: new
microbiologic considerations. Cur Infect Dis Rep 1:3848.
Brumbaugh AR, Mobley HL. (2012). Preventing urinary tract infection:
progress toward an effective Escherichia coli vaccine. Expert Rev
Vaccines 11:66376.

Downloaded by [Glen Ulett] at 20:50 05 August 2015

DOI: 10.3109/1040841X.2015.1028885

Brumbaugh AR, Smith SN, Mobley HL. (2013). Immunization with the
yersiniabactin receptor, FyuA protects against pyelonephritis in a
murine model of urinary tract infection. Infect Immun 81:330916.
Candela JV, Park E, Gerspach JM, et al. (1998). Evaluation of urinary
IL-1alpha and IL-1beta in gravid females and patients with bacterial
cystitis and microscopic hematuria. Urol Res 26:17580.
Chan CY, St John AL, Abraham SN. (2013). Mast cell interleukin-10
drives localized tolerance in chronic bladder infection. Immunity 38:
34959.
Chassin C, Goujon JM, Darche S, et al. (2006). Renal collecting duct
epithelial cells react to pyelonephritis-associated Escherichia coli by
activating distinct TLR4-dependent and -independent inflammatory
pathways. J Immunol 177:477384.
Chassin C, Hornefm W, Bens M, et al. (2007). Hormonal control of the
renal immune response and antibacterial host defense by arginine
vasopressin. J Exp Med 204:283752.
Chen SL, Hung CS, Pinkner JS, et al. (2009). Positive selection identifies
an in vivo role for FimH during urinary tract infection in addition to
mannose binding. Proc Natl Acad Sci USA 106:2243944.
Chenoweth C, Saint S. (2013). Preventing catheter-associated
urinary tract infections in the intensive care unit. Crit Care Clin 29:
1932.
Chromek M, Slamova Z, Bergman P, et al. (2006). The antimicrobial
peptide cathelicidin protects the urinary tract against invasive
bacterial infection. Nat Med 12:63641.
Chuang FC, Kuo HC. (2013). Increased urothelial cell apoptosis and
chronic inflammation are associated with recurrent urinary tract
infection in women. PLoS One 8:e63760.
Cirl C, Wieser A, Yadav M, et al. (2008). Subversion of toll-like receptor
signaling by a unique family of bacterial Toll/interleukin-1 receptor
domain-containing proteins. Nat Med 14:399406.
Cole SJ, Records AR, Orr MW, et al. (2014). Catheter-associated urinary
tract infection by Pseudomonas aeruginosa is mediated by exopolysaccharide-independent biofilms. Infect Immun 82:204858.
Connell I, Agace W, Klemm P, et al. (1996). Type 1 fimbrial expression
enhances Escherichia coli virulence for the urinary tract. Proc Natl
Acad Sci USA 93:982732.
Cusumano CK, Pinkner JS, Han Z, et al. (2011). Treatment and
prevention of urinary tract infection with orally active FimH
inhibitors. Sci Transl Med 3:109ra115.
Danka ES, Hunstad DA. (2015). Cathelicidin augments epithelial
receptivity and pathogenesis in experimental Escherichia coli cystitis.
J Infect Dis 211:116473.
Davis PH, Stanley Jr SL. (2003). Breaking the species barrier: use of
SCID mousehuman chimeras for the study of human infectious
diseases. Cell Microbiol 5:84960.
De Man P, Van Kooten C, Aarden L, et al. (1989). Interleukin-6 induced
at mucosal surfaces by gram-negative bacterial infection. Infect
Immun 57:33838.
Deknuydt F, Scotet E, Bonneville M. (2009). Modulation of inflammation through IL-17 production by gammadelta T cells: mandatory in
the mouse, dispensable in humans? Immunol Lett 127:812.
Dhakal BK, Mulvey MA. (2012). The UPEC pore-forming toxin alphahemolysin triggers proteolysis of host proteins to disrupt cell
adhesion, inflammatory, and survival pathways. Cell Host Microbe
11:5869.
Duell BL, Carey AJ, Dando SJ, et al. (2013). Human bladder
uroepithelial cells synergize with monocytes to promote IL-10
synthesis and other cytokine responses to uropathogenic Escherichia
coli. PLoS One 8:e78013.
Duell BL, Carey AJ, Tan CK, et al. (2012a). Innate transcriptional
networks activated in bladder in response to uropathogenic
Escherichia coli drive diverse biological pathways and rapid synthesis
of IL-10 for defense against bacterial urinary tract infection.
J Immunol 188:78192.
Duell BL, Tan CK, Carey AJ, et al. (2012b). Recent insights into
microbial triggers of interleukin-10 production in the host and the
impact on infectious disease pathogenesis. FEMS Immunol Med
Microbiol 64:295313.
Fischer H, Lutay N, Ragnarsdottir B, et al. (2010). Pathogen specific,
IRF3-dependent signaling and innate resistance to human kidney
infection. PLoS Pathog 6:e1001109.
Fischer H, Yamamoto M, Akira S, et al. (2006). Mechanism of pathogenspecific TLR4 activation in the mucosa: fimbriae, recognition
receptors and adaptor protein selection. Eur J Immunol 36:26777.

Urinary tract infection of mice to model human disease

15

Flores-Mireles AL, Pinkner JS, Caparon MG, Hultgren SJ. (2014). EbpA
vaccine antibodies block binding of Enterococcus faecalis to fibrinogen to prevent catheter-associated bladder infection in mice. Sci
Transl Med 6:254ra127.
Foxman B. (1990). Recurring urinary tract infection: incidence and risk
factors. Am J Public Health 80:3313.
Foxman B. (2002). Epidemiology of urinary tract infections: incidence,
morbidity, and economic costs. Am J Med 113:5S13S.
Foxman B. (2003). Epidemiology of urinary tract infections: incidence,
morbidity, and economic costs. Disease-a-Month 49:5370.
Franco AV. (2005). Recurrent urinary tract infections. Best Pract Res
Clin Obstet Gynaecol 19:86173.
Frank DN, Wilson SS, St Amand AL, Pace NR. (2009). Cultureindependent microbiological analysis of foley urinary catheter
biofilms. PLoS One 4:e7811.
Frendeus B, Godaly G, Hang L, et al. (2000). Interleukin 8 receptor
deficiency confers susceptibility to acute experimental pyelonephritis
and may have a human counterpart. J Exp Med 192:88190.
Furtado D. (1976). Experimental group B streptococcal infections in
mice: hematogenous virulence and mucosal colonization. Infect
Immun 13:131520.
Gally DL, Leathart J, Blomfield IC. (1996). Interaction of FimB and
FimE with the fim switch that controls the phase variation of type 1
fimbriae in Escherichia coli K-12. Mol Microbiol 21:72538.
Garcia EC, Brumbaugh AR, Mobley HL. (2011). Redundancy and
specificity of Escherichia coli iron acquisition systems during urinary
tract infection. Infect Immun 79:122535.
Garcia TA, Ventura CL, Smith MA, et al. (2013). Cytotoxic necrotizing
factor 1 and hemolysin from uropathogenic Escherichia coli elicit
different host responses in the murine bladder. Infect Immun 81:
99109.
Garofalo CK, Hooton TM, Martin SM, et al. (2007). Escherichia coli
from urine of female patients with urinary tract infections is
competent for intracellular bacterial community formation. Infect
Immun 75:5260.
Godaly G, Hang L, Frendeus B, Svanborg C. (2000). Transepithelial
neutrophil migration is CXCR1 dependent in vitro and is defective in
IL-8 receptor knockout mice. J Immunol 165:528794.
Godaly G, Otto G, Burdick MD, et al. (2007). Fimbrial lectins influence
the chemokine repertoire in the urinary tract mucosa. Kidney Int 71:
77886.
Goller CC, Arshad M, Noah JW, et al. (2014). Lifting the mask:
identification of new small molecule inhibitors of uropathogenic
Escherichia coli group 2 capsule biogenesis. PLoS One 9:e96054.
Guiton PS, Cusumano CK, Kline KA, et al. (2012). Combinatorial smallmolecule therapy prevents uropathogenic Escherichia coli catheterassociated urinary tract infections in mice. Antimicrob Agents
Chemother 56:473845.
Guiton PS, Hung CS, Hancock LE, et al. (2010). Enterococcal biofilm
formation and virulence in an optimized murine model of foreign body-associated urinary tract infections. Infect Immun 78:
416675.
Hadjifrangiskou M, Gu AP, Pinkner JS, et al. (2012). Transposon
mutagenesis identifies uropathogenic Escherichia coli biofilm factors.
J Bacteriol 194:6195205.
Hagan EC, Lloyd AL, Rasko DA, et al. (2010). Escherichia coli global
gene expression in urine from women with urinary tract infection.
PLoS Pathog 6:e1001187.
Hagan EC, Mobley HL. (2009). Haem acquisition is facilitated by a
novel receptor Hma and required by uropathogenic Escherichia coli
for kidney infection. Mol Microbiol 71:7991.
Hagberg L, Engberg I, Freter R, et al. (1983). Ascending, unobstructed
urinary tract infection in mice caused by pyelonephritogenic
Escherichia coli of human origin. Infect Immun 40:27383.
Hagberg L, Hull R, Hull S, et al. (1984). Difference in susceptibility to
gram-negative urinary tract infection between C3H/HeJ and C3H/HeN
mice. Infect Immun 46:83944.
Hagberg L, Jodal U, Korhonen TK. (1981). Adhesion, hemagglutination,
and virulence of Escherichia coli causing urinary tract infections.
Infect Immun 31:56470.
Han Z, Pinkner JS, Ford B, et al. (2010). Structure-based drug design and
optimization of mannoside bacterial FimH antagonists. J Med Chem
53:477992.
Hang L, Frendeus B, Godaly G, Svanborg C. (2000). Interleukin-8
receptor knockout mice have subepithelial neutrophil entrapment and

Downloaded by [Glen Ulett] at 20:50 05 August 2015

16

A. J. Carey et al.

renal scarring following acute pyelonephritis. J Infect Dis 182:


173848.
Hannan TJ, Mysorekar IU, Hung CS, et al. (2010). Early severe
inflammatory responses to uropathogenic E. coli predispose to chronic
and recurrent urinary tract infection. PLoS Pathog 6:e1001042.
Hannan TJ, Roberts PL, Riehl TE, et al. (2014). Inhibition of
cyclooxygenase-2 prevents chronic and recurrent cystitis.
EBioMedicine 1:4657.
Hannan TJ, Totsika M, Mansfield KJ, et al. (2012). Hostpathogen
checkpoints and population bottlenecks in persistent and intracellular
uropathogenic Escherichia coli bladder infection. FEMS Microbiol
Rev 36:61648.
Hawn TR, Scholes D, Li SS, et al. (2009). Toll-like receptor
polymorphisms and susceptibility to urinary tract infections in adult
women. PLoS ONE 4:e5990.
Hay AD, Fahey T. (2002). Clinical diagnosis of urinary tract infection.
J Am Med Assoc 288:1229; author reply 12301.
Healy EF, Walsh CA, Cotter AM, Walsh SR. (2012). Suprapubic
compared with transurethral bladder catheterization for gynecologic
surgery: a systematic review and meta-analysis. Obstet Gynecol 120:
67887.
Hedges S, Agace W, Svensson M, et al. (1994). Uroepithelial cells are
part of a mucosal cytokine network. Infect Immun 62:231521.
Hedges S, Svensson M, Svanborg C. (1992). Interleukin-6 response
of epithelial cell lines to bacterial stimulation in vitro. Infect Immun
60:1295301.
Hernandez JG, Sunden F, Connolly J, et al. (2011). Genetic control of the
variable innate immune response to asymptomatic bacteriuria. PLoS
One 6:e28289.
High KP, Bradley SF, Gravenstein S, et al. (2009). Clinical practice
guideline for the evaluation of fever and infection in older adult
residents of long-term care facilities: 2008 update by the Infectious
Diseases Society of America. Clin Infect Dis 48:14971.
Hilbert DW, Paulish-Miller TE, Tan CK, et al. (2012). Clinical
Escherichia coli isolates utilize alpha-hemolysin to inhibit in vitro
epithelial cytokine production. Microbes Infect 14:62838.
Hoelzer K, Pouillot R, Dennis S. (2012). Animal models of listeriosis:
a comparative review of the current state of the art and lessons
learned. Vet Res 43:18.
Hoepelman AI, Meiland R, Geerlings SE. (2003). Pathogenesis and
management of bacterial urinary tract infections in adult patients with
diabetes mellitus. Int J Antimicrob Agents 22:3543.
Hola V, Peroutkova T, Ruzicka F. (2012). Virulence factors in Proteus
bacteria from biofilm communities of catheter-associated urinary tract
infections. FEMS Immunol Med Microbiol 65:3439.
Hooton TM, Scholes D, Hughes JP, et al. (1996). A prospective study of
risk factors for symptomatic urinary tract infection in young women.
New Engl J Med 335:46874.
Hopkins WJ, Gendron-Fitzpatrick A, Balish E, Uehling DT. (1998).
Time course and host responses to Escherichia coli urinary tract
infection in genetically distinct mouse strains. Infect Immun 66:
2798802.
Hopkins WJ, Hall JA, Conway BP, Uehling DT. (1995). Induction of
urinary tract infection by intraurethral inoculation with Escherichia
coli: refining the murine model. J Infect Dis 171:4625.
Hopkins WJ, James LJ, Balish E, Uehling DT. (1993). Congenital
immunodeficiencies in mice increase susceptibility to urinary tract
infection. J Urol 149:9225.
Huber M, Krauter K, Winkelmann G, et al. (2000). Immunostimulation
by bacterial components: II. Efficacy studies and meta-analysis of the
bacterial extract OM-89. Int J Immunopharmacol 22:110311.
Hughes Jr FM, Vivar NP, Kennis JG, et al. (2014). Inflammasomes are
important mediators of cyclophosphamide-induced bladder inflammation. Am J Physiol Renal Physiol 306: F299308.
Hull R, Rudy D, Donovan W, et al. (2000). Urinary tract infection
prophylaxis using Escherichia coli 83972 in spinal cord injured
patients. J Urol 163:8727.
Hung CS, Dodson KW, Hultgren SJ. (2009). A murine model of urinary
tract infection. Nat Protocols 4:123043.
Hunstad DA, Justice SS. (2010). Intracellular lifestyles and immune
evasion strategies of uropathogenic Escherichia coli. Annu Rev
Microbiol 64:20321.
Hvidberg H, Struve C, Krogfelt KA, et al. (2000). Development of a
long-term ascending urinary tract infection mouse model for antibiotic
treatment studies. Antimicrob Agents Chemother 44:15663.

Crit Rev Microbiol, Early Online: 120

Ichinohe T, Pang IK, Kumamoto Y, et al. (2011). Microbiota regulates


immune defense against respiratory tract influenza A virus infection.
Proc Natl Acad Sci USA 108:53549.
Ingersoll MA, Kline KA, Nielsen HV, Hultgren SJ. (2008). G-CSF
induction early in uropathogenic Escherichia coli infection of the
urinary tract modulates host immunity. Cell Microbiol 10:256878.
Ipe DS, Sundac L, Benjamin Jr WH, et al. (2013). Asymptomatic
bacteriuria: prevalence rates of causal microorganisms, etiology of
infection in different patient populations, and recent advances in
molecular detection. FEMS Microbiol Lett 346:110.
Iwahi T, Abe Y, Tsuchiya K. (1982). Virulence of Escherichia coli
in ascending urinary-tract infection in mice. J Med Microbiol 15:
30316.
Jacobsen SM, Stickler DJ, Mobley HL, Shirtliff ME. (2008).
Complicated catheter-associated urinary tract infections due to
Escherichia coli and Proteus mirabilis. Clin Microbiol Rev 21:2659.
Jaillon S, Moalli F, Ragnarsdottir B, et al. (2014). The humoral pattern
recognition molecule PTX3 is a key component of innate immunity
against urinary tract infection. Immunity 40:62132.
Janssen C, Lo J, Jager W, et al. (2014). A high throughput, minimally
invasive, ultrasound guided model for the study of catheter associated
urinary tract infections and device encrustation in mice. J Urol 192:
185663.
Johansen TE, Botto H, Cek M, et al. (2011). Critical review of current
definitions of urinary tract infections and proposal of an EAU/ESIU
classification system. Int J Antimicrob Agents 38:6470.
Johnson JR. (1998). Reflux in the mouse model of urinary tract infection.
Infect Immun 66:60634.
Johnson JR, Brown JJ. (1996). Defining inoculation conditions for the
mouse model of ascending urinary tract infection that avoid immediate
vesicoureteral reflux yet produce renal and bladder infection. J Infect
Dis 173:7469.
Johnson JR, Clabots C, Rosen H. (2006). Effect of inactivation of the
global oxidative stress regulator oxyR on the colonization ability of
Escherichia coli O1:K1:H7 in a mouse model of ascending urinary
tract infection. Infect Immun 74:4618.
Johnson JR, Delavari P, Azar M. (1999). Activities of a nitrofurazonecontaining urinary catheter and a silver hydrogel catheter against
multidrug-resistant bacteria characteristic of catheter-associated urinary tract infection. Antimicrob Agents Chemother 43:29905.
Johnson JR, Manivel JC. (1991). Vesicoureteral reflux induces renal
trauma in a mouse model of ascending, unobstructed pyelonephritis.
J Urol 145:130611.
Johnson DE, Lockatell CV, Hall-Craigs M, et al. (1987).
Uropathogenicity in rats and mice of Providencia stuartii from
long-term catheterized patients. J Urol 138:6325.
Johnson DE, Lockatell CV, Russell RG, et al. (1998). Comparison of
Escherichia coli strains recovered from human cystitis and pyelonephritis infections in transurethrally challenged mice. Infect Immun
66:305965.
Johnson DE, Russell RG, Lockatell CV, et al. (1993a). Urethral
obstruction of 6 hours or less causes bacteriuria, bacteremia, and
pyelonephritis in mice challenged with nonuropathogenic
Escherichia coli. Infect Immun 61:34228.
Johnson DE, Russell RG, Lockatell CV, et al. (1993b). Contribution of
Proteus mirabilis urease to persistence, urolithiasis, and acute
pyelonephritis in a mouse model of ascending urinary tract infection.
Infect Immun 61:274854.
Jones-Carson J, Balish E, Uehling DT. (1999). Susceptibility of
immunodeficient gene-knockout mice to urinary tract infection.
J Urol 161:33841.
Kai-Larsen Y, Luthje P, Chromek M, et al. (2010). Uropathogenic
Escherichia coli modulates immune responses and its curli fimbriae
interact with the antimicrobial peptide LL-37. PLoS Pathog
6:e1001010.
Kalmanson GM, Harwick HJ, Turck M, Guze LB. (1975). Urinary-tract
infection: localisation and virulence of Escherichia coli. Lancet 1:
1346.
Kaufmann J, Modest GA. (2002). Clinical diagnosis of urinary tract
infection. J Am Med Assoc 288:122930; author reply 12301.
Kaul AK, Khan S, Martens MG, et al. (1999). Experimental gestational
pyelonephritis induces preterm births and low birth weights in
C3H/HeJ mice. Infect Immun 67:595866.
Kemp KD, Singh KV, Nallapareddy SR, Murray BE. (2007). Relative
contributions of Enterococcus faecalis OG1RF sortase-encoding

Downloaded by [Glen Ulett] at 20:50 05 August 2015

DOI: 10.3109/1040841X.2015.1028885

genes, srtA and bps (srtC), to biofilm formation and a murine model
of urinary tract infection. Infect Immun 75:5399404.
Kline KA, Schwartz DJ, Gilbert NM, et al. (2012). Immune modulation
by group B streptococcus influences host susceptibility to urinary
tract infection by uropathogenic Escherichia coli. Infect Immun 80:
418694.
Kline KA, Schwartz DJ, Gilbert NM, Lewis AL. (2014). Impact of host
age and parity on susceptibility to severe urinary tract infection in a
murine model. PLoS One 9:e97798.
Kline KA, Schwartz DJ, Lewis WG, et al. (2011). Immune activation and
suppression by group B streptococcus in a murine model of urinary
tract infection. Infect Immun 79:358895.
Ko YC, Mukaida N, Ishiyama S, et al. (1993). Elevated interleukin-8
levels in the urine of patients with urinary tract infections. Infect
Immun 61:130714.
Kostakioti M, Hadjifrangiskou M, Cusumano CK, et al. (2012).
Distinguishing the contribution of type 1 pili from that of other
QseB-misregulated factors when QseC is absent during urinary tract
infection. Infect Immun 80:282634.
Koves B, Salvador E, Gronberg-Hernandez J, et al. (2014). Rare
emergence of symptoms during long-term asymptomatic Escherichia
coli 83972 carriage without an altered virulence factor repertoire.
J Urol 191:51928.
Kunin CM. (1994). Urinary tract infections in females. Clin Infect Dis
18:110; quiz 1112.
Lane MC, Alteri CJ, Smith SN, Mobley HL. (2007). Expression of
flagella is coincident with uropathogenic Escherichia coli ascension to the upper urinary tract. Proc Natil Acad Sci USA 104:
1666974.
Lane MC, Lockatell V, Monterosso G, et al. (2005). Role of motility in
the colonization of uropathogenic Escherichia coli in the urinary tract.
Infect Immun 73:764456.
Langermann S, Palaszynski S, Barnhart M, et al. (1997). Prevention of
mucosal Escherichia coli infection by FimH-adhesin-based systemic
vaccination. Science 276:60711.
Lee JBL, Neild GH. (2007). Urinary tract infection. Medicine 35:4238.
Lefort A, Chau F, Lepeule R, et al. (2014). Activity of fosfomycin alone
or combined with cefoxitin in vitro and in vivo in a murine model of
urinary tract infection due to Escherichia coli harbouring CTX-M-15type extended-spectrum beta-lactamase. Int J Antimicrob Agents 43:
3669.
Li CR, Li Y, Li GQ, et al. (2010). In vivo antibacterial activity of
nemonoxacin, a novel non-fluorinated quinolone. J Antimicrob
Chemother 65:241115.
Li X, Zhao H, Lockatell CV, et al. (2002). Visualization of Proteus
mirabilis within the matrix of urease-induced bladder stones during
experimental urinary tract infection. Infect Immun 70:38994.
Louz D, Bergmans HE, Loos BP, Hoeben RC. (2013). Animal models
in virus research: their utility and limitations. Crit Rev Microbiol
39:32561.
Maki DG, Tambyah PA. (2001). Engineering out the risk for infection
with urinary catheters. Emerg Infect Dis 7:3427.
Mastroeni P, Sheppard M. (2004). Salmonella infections in the mouse
model: host resistance factors and in vivo dynamics of bacterial spread
and distribution in the tissues. Microbes Infect 6:398405.
Mcbean M, Rajamani S. (2001). Increasing rates of hospitalization due
to septicemia in the US elderly population, 19861997. J Infect Dis
183:596603.
Meddings J, Rogers MA, Macy M, Saint S. (2010). Systematic review
and meta-analysis: reminder systems to reduce catheter-associated
urinary tract infections and urinary catheter use in hospitalized
patients. Clin Infect Dis 51:55060.
Mobley HL, Green DM, Trifillis AL, et al. (1990). Pyelonephritogenic
Escherichia coli and killing of cultured human renal proximal tubular
epithelial cells: role of hemolysin in some strains. Infect Immun 58:
12819.
Mobley HL, Jarvis KG, Elwood JP, et al. (1993). Isogenic P-fimbrial
deletion mutants of pyelonephritogenic Escherichia coli: the role of
alpha Gal(14) beta Gal binding in virulence of a wild-type strain.
Mol Microbiol 10:14355.
Moriel DG, Schembri MA. (2013). Vaccination approaches for the
prevention of urinary tract infection. Curr Pharm Biotechnol 14:
96774.
Morin MD, Hopkins WJ. (1998). Treatment of mice with staphylococcal
enterotoxin B enhances resolution of an induced Escherichia coli

Urinary tract infection of mice to model human disease

17

urinary tract infection and stimulates production of proinflammatory


cytokines. Infect Immun 66:246670.
Mulvey MA. (2002). Adhesion and entry of uropathogenic Escherichia
coli. Cell Microbiol 4:25771.
Mulvey MA, Schilling JD, Hultgren SJ. (2001). Establishment of a
persistent Escherichia coli reservoir during the acute phase of a
bladder infection. Infect Immun 69:45729.
Murphy CN, Mortensen MS, Krogfelt KA, Clegg S. (2013). Role of
Klebsiella pneumoniae type 1 and type 3 fimbriae in colonizing
silicone tubes implanted into the bladders of mice as a model
of catheter-associated urinary tract infections. Infect Immun 81:
300917.
Mylonakis E, Casadevall A, Ausubel FM. (2007). Exploiting amoeboid
and non-vertebrate animal model systems to study the virulence of
human pathogenic fungi. PLoS Pathog 3:e101.
Mysorekar IU, Mulvey MA, Hultgren SJ, Gordon JI. (2002). Molecular
regulation of urothelial renewal and host defenses during infection
with uropathogenic Escherichia coli. J Biol Chem 277:741219.
Natarajan V. (2008). Urinary tract infection. Surgery (Oxford) 26:1936.
Neild GH. (2003). Urinary tract infection. Medicine (Abingdon) 31:
8590.
Ness-Schwickerath KJ, Morita CT. (2011). Regulation and function of
IL-17A- and IL-22-producing gammadelta T cells. Cell Mol Life Sci
68:237190.
Nesta B, Spraggon G, Alteri C, et al. (2012). FdeC, a novel broadly
conserved Escherichia coli adhesin eliciting protection against urinary
tract infections. MBio 3:e0001012.
Nicholson AM, Glynn AA. (1975). Investigation of the effect of K
antigen in Escherichia coli urinary tract infections by use of a mouse
model. Br J Exp Pathol 56:54953.
Nicolle LE. (2008). Uncomplicated urinary tract infection in adults
including uncomplicated pyelonephritis. Urol Clin North Am 35:
112, v.
Nicolle LE. (2014). Catheter associated urinary tract infections.
Antimicrob Resist Infect Control 3:2330.
Nielsen HV, Guiton PS, Kline KA, et al. (2012). The metal iondependent adhesion site motif of the Enterococcus faecalis EbpA pilin
mediates pilus function in catheter-associated urinary tract infection.
MBio 3:e0017712.
Nielsen KL, Dynesen P, Larsen P, et al. (2014). Role of urinary
cathelicidin LL-37 and human beta-defensin 1 in uncomplicated
Escherichia coli urinary tract infections. Infect Immun 82:15728.
Nielubowicz GR, Mobley HL. (2010). Hostpathogen interactions in
urinary tract infection. Nat Rev Urol 7:43041.
OHanley P, Lark D, Falkow S, Schoolnik G. (1985). Molecular basis of
Escherichia coli colonization of the upper urinary tract in BALB/c
mice. GalGal pili immunization prevents Escherichia coli pyelonephritis in the BALB/c mouse model of human pyelonephritis. J Clin
Invest 75:34760.
Oh JZ, Ravindran R, Chassaing B, et al. (2014). TLR5-mediated sensing
of gut microbiota is necessary for antibody responses to seasonal
influenza vaccination. Immunity 41:47892.
Olson PD, Hunstad DA. (2014). A new model of urinary tract infection
reveals male susceptibility to chronic cystitis and severe pyelonephritis. American Society for Microbiology, 114th General Meeting.
Boston, USA.
Ong CL, Ulett GC, Mabbett AN, et al. (2008). Identification of type 3
fimbriae in uropathogenic Escherichia coli reveals a role in biofilm
formation. J Bacteriol 190:105463.
Pang IK, Iwasaki A. (2012). Control of antiviral immunity by pattern
recognition and the microbiome. Immunol Rev 245:20926.
Pearson MM, Yep A, Smith SN, Mobley HL. (2011). Transcriptome of
Proteus mirabilis in the murine urinary tract: virulence and nitrogen
assimilation gene expression. Infect Immun 79:261931.
Pecha B, Low D, OHanley P. (1989). GalGal pili vaccines prevent
pyelonephritis by piliated Escherichia coli in a murine model.
Single-component GalGal pili vaccines prevent pyelonephritis by
homologous and heterologous piliated E. coli strains. J Clin Invest
83:21028.
Pellegrino R, Galvalisi U, Scavone P, et al. (2003). Evaluation of Proteus
mirabilis structural fimbrial proteins as antigens against urinary tract
infections. FEMS Immunol Med Microbiol 36:10310.
Press MJ, Metlay JP. (2013). Catheter-associated urinary tract infection:
does changing the definition change quality? Infect Control Hosp
Epidemiol 34:31315.

Downloaded by [Glen Ulett] at 20:50 05 August 2015

18

A. J. Carey et al.

Raffi HS, Bates Jr JM, Laszik Z, Kumar S. (2005). TammHorsfall


protein acts as a general host-defense factor against bacterial cystitis.
Am J Nephrol 25:5708.
Raffi HS, Bates Jr JM, Laszik Z, Kumar S. (2009). TammHorsfall
protein protects against urinary tract infection by Proteus mirabilis.
J Urol 181:23328.
Ragnarsdottir B, Fischer H, Godaly G, et al. (2008). TLR- and CXCR1dependent innate immunity: insights into the genetics of urinary tract
infections. Eur J Clin Invest 38:1220.
Ragnarsdottir B, Jonsson K, Urbano A, et al. (2010). Toll-like receptor 4
promoter polymorphisms: common TLR4 variants may protect against
severe urinary tract infection. PLoS One 5:e10734.
Ragnarsdottir B, Lutay N, Gronberg-Hernandez J, et al. (2011). Genetics
of innate immunity and UTI susceptibility. Nat Rev Urol 8:44968.
Ragnarsdottir B, Samuelsson M, Gustafsson MC, et al. (2007). Reduced
toll-like receptor 4 expression in children with asymptomatic bacteriuria. J Infect Dis 196:47584.
Raz R, Gennesin Y, Wasser J, et al. (2000). Recurrent urinary tract
infections in postmenopausal women. Clin Infect Dis 30:1526.
Reigstad CS, Hultgren SJ, Gordon JI. (2007). Functional genomic studies
of uropathogenic Escherichia coli and host urothelial cells when
intracellular bacterial communities are assembled. J Biol Chem 282:
2125967.
Robino L, Scavone P, Araujo L, et al. (2013). Detection of intracellular
bacterial communities in a child with Escherichia coli recurrent
urinary tract infections. Pathog Dis 68:7881.
Robino L, Scavone P, Araujo L, et al. (2014). Intracellular bacteria in the
pathogenesis of Escherichia coli urinary tract infection in children.
Clin Infect Dis 59:e15864.
Roelofs JJ, Rouschop KM, Teske GJ, et al. (2006). The urokinase
plasminogen activator receptor is crucially involved in host defense
during acute pyelonephritis. Kidney Int 70:19427.
Ronald A. (2002). The etiology of urinary tract infection: traditional and
emerging pathogens. Am J Med 113:14S19S.
Ronald A. (2003). The etiology of urinary tract infection: traditional and
emerging pathogens. Disease-a-Month 49:7182.
Roos V, Klemm P. (2006). Global gene expression profiling of the
asymptomatic bacteriuria Escherichia coli strain 83972 in the human
urinary tract. Infect Immun 74:356575.
Roos V, Ulett GC, Schembri MA, Klemm P. (2006). The asymptomatic
bacteriuria Escherichia coli strain 83972 outcompetes uropathogenic
E. coli strains in human urine. Infect Immun 74:61524.
Rosen DA, Hooton TM, Stamm WE, et al. (2007). Detection of
intracellular bacterial communities in human urinary tract infection.
PLoS Med 4:e329.
Rosen DA, Hung CS, Kline KA, Hultgren SJ. (2008). Streptozocininduced diabetic mouse model of urinary tract infection. Infect Immun
76:42908.
Rudick CN, Billips BK, Pavlov VI, et al. (2010). Hostpathogen
interactions mediating pain of urinary tract infection. J Infect Dis 201:
12409.
Russo T, Brown JJ, Jodush ST, Johnson JR. (1996a). The O4 specific
antigen moiety of lipopolysaccharide but not the K54 group 2 capsule
is important for urovirulence of an extraintestinal isolate of
Escherichia coli. Infect Immun 64:23438.
Russo TA, Jodush ST, Brown JJ, Johnson JR. (1996b). Identification of
two previously unrecognized genes (guaA and argC) important for
uropathogenesis. Mol Microbiol 22:21729.
Russo TA, Mcfadden CD, Carlino-Macdonald UB, et al. (2003). The
Siderophore receptor IroN of extraintestinal pathogenic Escherichia
coli is a potential vaccine candidate. Infect Immun 71:71649.
Sabri M, Houle S, Dozois CM. (2009). Roles of the extraintestinal
pathogenic Escherichia coli ZnuACB and ZupT zinc transporters
during urinary tract infection. Infect Immun 77:115564.
Saemann MD, Weichhart T, Zeyda M, et al. (2005). TammHorsfall
glycoprotein links innate immune cell activation with adaptive
immunity via a toll-like receptor-4-dependent mechanism. J Clin
Invest 115:46875.
Salvatore S, Salvatore S, Cattoni E, et al. (2011). Urinary tract infections
in women. Eur J Obstet Gynecol Reproduct Biol 156:1316.
Savidge TC, Morey AL, Ferguson DJ, et al. (1995). Human intestinal
development in a severe-combined immunodeficient xenograft model.
Differentiation 58:36171.
Scavone P, Umpierrez A, Rial A, et al. (2014). Native flagellin does not
protect mice against an experimental Proteus mirabilis ascending

Crit Rev Microbiol, Early Online: 120

urinary tract infection and neutralizes the protective effect of MrpA


fimbrial protein. Anton van Leeuw 105:113948.
Schembri MA, Sokurenko EV, Klemm P. (2000). Functional flexibility
of the FimH adhesin: insights from a random mutant library. Infect
Immun 68:263846.
Schilling JD, Lorenz RG, Hultgren SJ. (2002). Effect of trimethoprimsulfamethoxazole on recurrent bacteriuria and bacterial persistence
in mice infected with uropathogenic Escherichia coli. Infect Immun
70:70429.
Schilling JD, Martin SM, Hung CS, et al. (2003). Toll-like receptor 4
on stromal and hematopoietic cells mediates innate resistance to
uropathogenic Escherichia coli. Proc Natl Acad Sci USA 100:42038.
Schilling JD, Mulvey MA, Hultgren SJ. (2001a). Dynamic interactions
between host and pathogen during acute urinary tract infections.
Urology 57:5661.
Schilling JD, Mulvey MA, Vincent CD, et al. (2001b). Bacterial invasion
augments epithelial cytokine responses to Escherichia coli through a
lipopolysaccharide-dependent mechanism. J Immunol 166:114855.
Schmidt MA, OHanley P, Lark D, Schoolnik GK. (1988). Synthetic
peptides corresponding to protective epitopes of Escherichia coli
digalactoside-binding pilin prevent infection in a murine pyelonephritis model. Proc Natl Acad Sci USA 85:124751.
Schneeberger C, Kazemier BM, Geerlings SE. (2014). Asymptomatic
bacteriuria and urinary tract infections in special patient groups:
women with diabetes mellitus and pregnant women. Curr Opin Infect
Dis 27:10814.
Scholes D, Hooton TM, Roberts PL, et al. E. (2000). Risk factors
for recurrent urinary tract infection in young women. J Infect Dis 182:
117782.
Schwartz DJ, Conover MS, Hannan TJ, Hultgren SJ. (2015).
Uropathogenic Escherichia coli superinfection enhances the severity
of mouse bladder infection. PLoS Pathog 11:e1004599.
Seok J, Warren HS, Cuenca AG, et al. (2013). Genomic responses in
mouse models poorly mimic human inflammatory diseases. Proc Natl
Acad Sci USA 110:350712.
Shankar N, Lockatell CV, Baghdayan AS, et al. (2001). Role of
Enterococcus faecalis surface protein Esp in the pathogenesis of
ascending urinary tract infection. Infect Immun 69:436672.
Sidjabat HE, Chin JJ, Chapman T, et al. (2009). Colonisation dynamics
and virulence of two clonal groups of multidrug-resistant Escherichia
coli isolated from dogs. Microbes Infect 11:1007.
Siegman-Igra Y, Fourer B, Orni-Wasserlaufr R, et al. (2002). Reappraisal
of community-acquired bacteremia: a proposal of a new classification
for the spectrum of acquisition of bacteremia. Clin Infect Dis 34:
14319.
Sillanpaa J, Nallapareddy SR, Singh KV, et al. (2010). Characterization
of the ebp(fm) pilus-encoding operon of Enterococcus faecium and its
role in biofilm formation and virulence in a murine model of urinary
tract infection. Virulence 1:23646.
Silva TM, Costa EA, Paixao TA, et al. (2011). Laboratory animal models
for brucellosis research. J Biomed Biotechnol 2011:518323.
Singh KV, Nallapareddy SR, Murray BE. (2007). Importance of the ebp
(endocarditis- and biofilm-associated pilus) locus in the pathogenesis
of Enterococcus faecalis ascending urinary tract infection. J Infect Dis
195:16717.
Sivick KE, Mobley HL. (2010). Waging war against uropathogenic
Escherichia coli: winning back the urinary tract. Infect Immun 78:
56885.
Sivick KE, Schaller MA, Smith SN, Mobley HL. (2010). The innate
immune response to uropathogenic Escherichia coli involves IL-17A
in a murine model of urinary tract infection. J Immunol 184:206575.
Smithson A, Sarrias MR, Barcelo J, et al. (2005). Expression of
interleukin-8 receptors (CXCR1 and CXCR2) in premenopausal
women with recurrent urinary tract infections. Clin Diagn Lab
Immunol 12:135863.
Snyder JA, Haugen BJ, Buckles EL, et al. (2004). Transcriptome of
uropathogenic Escherichia coli during urinary tract infection. Infect
Immun 72:637381.
Song J, Abraham SN. (2008). TLR-mediated immune responses in the
urinary tract. Curr Opin Microbiol 11:6673.
Springall T, Sheerin NS, Abe K, et al. (2001). Epithelial secretion of C3
promotes colonization of the upper urinary tract by Escherichia coli.
Nat Med 7:8016.
Stahlhut SG, Struve C, Krogfelt KA, Reisner A. (2012). Biofilm
formation of Klebsiella pneumoniae on urethral catheters requires

Downloaded by [Glen Ulett] at 20:50 05 August 2015

DOI: 10.3109/1040841X.2015.1028885

either type 1 or type 3 fimbriae. FEMS Immunol Med Microbiol 65:


3509.
Stamm WE. (2006). Theodore E. Woodward Award: hostpathogen
interactions in community-acquired urinary tract infections. Trans Am
Clin Climatol Assoc 117:7583; discussion 834.
Stamm WE, Hooton TM. (1993). Management of urinary tract infections
in adults. New Engl J Med 329:132834.
Stamm WE, Norrby SR. (2001). Urinary tract infections: disease
panorama and challenges. J Infect Dis 183:S14.
Stapleton A. (1999). Prevention of recurrent urinary-tract infections in
women. Lancet 353:78.
Subashchandrabose S, Hazen TH, Brumbaugh AR, et al. (2014). Hostspecific induction of Escherichia coli fitness genes during human
urinary tract infection. Proc Natl Acad Sci USA 111:1832732.
Sunden F, Hakansson L, Ljunggren E, Wullt B. (2010). Escherichia coli
83972 bacteriuria protects against recurrent lower urinary tract
infections in patients with incomplete bladder emptying. J Urol 184:
17985.
Svanborg C, Bergsten G, Fischer H, et al. (2001). The innate host
response protects and damages the infected urinary tract. Ann Med
33:56370.
Svanborg Eden C, Briles D, Hagberg L, et al. (1985). Genetic factors in
host resistance to urinary tract infection. Infection 13:S1716.
Svensson M, Yadav M, Holmqvist B, et al. (2011). Acute pyelonephritis
and renal scarring are caused by dysfunctional innate immunity in
mCxcr2 heterozygous mice. Kidney Int 80:106472.
Tabibian JH, Gornbein J, Heidari A, et al. (2008). Uropathogens and host
characteristics. J Clin Microbiol 46:39806.
Tan CK, Carey AJ, Cui X, et al. (2012a). Genome-wide mapping of
cystitis due to Streptococcus agalactiae and Escherichia coli in mice
identifies a unique bladder transcriptome that signifies pathogenspecific antimicrobial defense against urinary tract infection. Infect
Immun 80:314560.
Tan CK, Ulett KB, Steele M, et al. (2012b). Prognostic value of semiquantitative bacteruria counts in the diagnosis of group B streptococcus urinary tract infection: a 4-year retrospective study in adult
patients. BMC Infect Dis 12:273.
Tena D, Gonzalez-Praetorius A, Bisquert J. (2007). Urinary tract
infection due to non-typhoidal Salmonella: report of 19 cases. J Infect
54:2459.
Tenke P, Koves B, Johansen TE. (2014). An update on prevention and
treatment of catheter-associated urinary tract infections. Curr Opin
Infect Dis 27:1027.
Thai KH, Thathireddy A, Hsieh MH. (2010). Transurethral induction of
mouse urinary tract infection. J Vis Exp 42:pii2070.
Totsika M, Beatson SA, Sarkar S, et al. (2011). Insights into a multidrug
resistant Escherichia coli pathogen of the globally disseminated
ST131 lineage: genome analysis and virulence mechanisms. PLoS
One 6:e26578.
Totsika M, Heras B, Wurpel DJ, Schembri MA. (2009). Characterization
of two homologous disulfide bond systems involved in virulence
factor biogenesis in uropathogenic Escherichia coli CFT073.
J Bacteriol 191:39018.
Totsika M, Kostakioti M, Hannan TJ, et al. (2013). A FimH inhibitor
prevents acute bladder infection and treats chronic cystitis caused by
multidrug-resistant uropathogenic Escherichia coli ST131. J Infect
Dis 208:9218.
Tratselas A, Simitsopoulou M, Giannakopoulou A, et al. (2014). Effect
of ceftriaxone on the outcome of murine pyelonephritis caused
by extended-spectrum-beta-lactamase-producing Escherichia coli.
Antimicrob Agents Chemother 58:710211.
Trautner BW. (2010). Management of catheter-associated urinary tract
infection. Curr Opin Infect Dis 23:7682.
Trautner BW, Hull RA, Darouiche RO. (2005). Prevention of
catheter-associated urinary tract infection. Curr Opin Infect Dis 18:
3741.
Tree JJ, Ulett GC, Ong CL, et al. (2008). Trade-off between iron uptake
and protection against oxidative stress: deletion of cueO promotes
uropathogenic Escherichia coli virulence in a mouse model of urinary
tract infection. J Bacteriol 190:690912.
Tsuji M, Takema M, Miwa H, et al. (2003). In vivo antibacterial activity
of S-3578, a new broad-spectrum cephalosporin: methicillinresistant Staphylococcus aureus and Pseudomonas aeruginosa
experimental infection models. Antimicrob Agents Chemother 47:
250712.

Urinary tract infection of mice to model human disease

19

Ulett KB, Benjamin Jr WH, Zhuo F, et al. (2009). Diversity of group B


streptococcus serotypes causing urinary tract infection in adults. J Clin
Microbiol 47:205560.
Ulett KB, Shuemaker JH, Benjamin Jr WH, et al. (2012). Group B
streptococcus cystitis presenting in a diabetic patient with a massive
abdominopelvic abscess: a case report. J Med Case Rep 6:23740.
Ulett GC, Totsika M, Schaale K, et al. (2013). Uropathogenic
Escherichia coli virulence and innate immune responses during
urinary tract infection. Curr Opin Microbiol 16:1007.
Ulett GC, Valle J, Beloin C, et al. (2007). Functional analysis of antigen
43 in uropathogenic Escherichia coli reveals a role in long-term
persistence in the urinary tract. Infect Immun 75:323344.
Ulett GC, Webb RI, Ulett KB, et al. (2010). Group B Streptococcus
(GBS) urinary tract infection involves binding of GBS to bladder
uroepithelium and potent but GBS-specific induction of interleukin
1alpha. J Infect Dis 201:86670.
Umpierrez A, Scavone P, Romanin D, et al. (2013). Innate immune
responses to Proteus mirabilis flagellin in the urinary tract. Microbes
Infect 15:68896.
Van Duin D. (2012). Diagnostic challenges and opportunities in older
adults with infectious diseases. Clin Infect Dis 54:9738.
Vebo HC, Solheim M, Snipen L, et al. (2010). Comparative genomic
analysis of pathogenic and probiotic Enterococcus faecalis isolates,
and their transcriptional responses to growth in human urine. PLoS
One 5:e12489.
Vimont S, Boyd A, Bleibtreu A, et al. (2012). The CTX-M-15-producing
Escherichia coli clone O25b: H4-ST131 has high intestine colonization and urinary tract infection abilities. PLoS One 7:e46547.
Vivaldi E. (1968). Effect of chronic administration of analgesics on
resistance to experimental infection of the urinary tract in rats.
Nephron 5:2029.
Wagenlehner FM, Naber KG. (2004). Emergence of antibiotic resistance
and prudent use of antibiotic therapy in nosocomially acquired urinary
tract infections. Int J Antimicrob Agents 23:S249.
Wang C, Symington JW, Ma E, et al. (2013). Estrogenic modulation of
uropathogenic Escherichia coli infection pathogenesis in a murine
menopause model. Infect Immun 81:7339.
Wang C, Yuan X, Ma E, et al. (2014). NOD2 is dispensable for
ATG16L1 deficiency-mediated resistance to urinary tract infection.
Autophagy 10:3318.
Warren JW, Tenney JH, Hoopes JM, et al. (1982). A prospective
microbiologic study of bacteriuria in patients with chronic indwelling
urethral catheters. J Infect Dis 146:71923.
Watts RE, Tan CK, Ulett GC, et al. (2012a). Escherichia coli 83972
expressing a P fimbriae oligosaccharide receptor mimic impairs
adhesion of uropathogenic E. coli. J Infect Dis 206:12429.
Watts RE, Totsika M, Challinor VL, et al. (2012b). Contribution of
siderophore systems to growth and urinary tract colonization of
asymptomatic bacteriuria Escherichia coli. Infect Immun 80:33344.
Wieser A, Romann E, Magistro G, et al. (2010). A multiepitope subunit
vaccine conveys protection against extraintestinal pathogenic
Escherichia coli in mice. Infect Immun 78:343242.
Wobser D, Ali L, Grohmann E, et al. (2014). A novel role for
D-alanylation of lipoteichoic acid of Enterococcus faecalis in urinary
tract infection. PLoS One 9: e107827.
Wu XR, Sun TT, Medina JJ. (1996). In vitro binding of type 1-fimbriated
Escherichia coli to uroplakins Ia and Ib: relation to urinary tract
infections. Proc Natl Acad Sci USA 93:96305.
Wullt B. (2003). The role of P fimbriae for Escherichia coli establishment and mucosal inflammation in the human urinary tract. Int J
Antimicrob Agents 21:60521.
Wullt B, Bergsten G, Connell H, et al. (2001). P-fimbriae trigger
mucosal responses to Escherichia coli in the human urinary tract.
Cell Microbiol 3:25564.
Wullt B, Bergsten G, Fischer H, et al. (2003). The host response to
urinary tract infection. Infect Dis Clin North Am 17:279301.
Wullt B, Bergsten G, Samuelsson M, Svanborg C. (2002). The role of P
fimbriae for Escherichia coli establishment and mucosal inflammation
in the human urinary tract. Int J Antimicrob Agents 19:52238.
Wullt B, Connell H, Rollano P, et al. (1998). Urodynamic factors
influence the duration of Escherichia coli bacteriuria in deliberately
colonized cases. J Urol 159:205762.
Yadav M, Zhang J, Fischer H, et al. (2010). Inhibition of TIR domain
signaling by TcpC: MyD88-dependent and independent effects on
Escherichia coli virulence. PLoS Pathog 6:e1001120.

20

A. J. Carey et al.

Downloaded by [Glen Ulett] at 20:50 05 August 2015

Yin X, Hou T, Liu Y, et al. (2010). Association of toll-like receptor 4


gene polymorphism and expression with urinary tract infection types
in adults. PLoS One 5:e14223.
You XF, Li CR, Yang XY, et al. (2009). In vivo antibacterial activity of
vertilmicin, a new aminoglycoside antibiotic. Antimicrob Agents
Chemother 53:45258.

Crit Rev Microbiol, Early Online: 120

Zhang Z, Jin L, Champion G, et al. (2001). Shigella infection in a SCID


mousehuman intestinal xenograft model: role for neutrophils in
containing bacterial dissemination in human intestine. Infect Immun
69:32407.
Zhang D, Zhang G, Hayden MS, et al. (2004). A toll-like receptor that
prevents infection by uropathogenic bacteria. Science 303:15226.

You might also like