You are on page 1of 17

Folia Biologica et Oecologica 8: 117 (2012)

Acta Universitatis Lodziensis

Proteus sp. an opportunistic bacterial pathogen classification, swarming


growth, clinical significance and virulence factors

ANTONI RALSKI 1*, AGNIESZKA TORZEWSKA1, MAGDALENA MORYL1, IWONA KWIL1, AGNIESZKA MASZEWSKA1, KINGA
OSTROWSKA1, DOMINIKA DRZEWIECKA2, AGNIESZKA ZABOTNI2, AGATA PALUSIAK2, MAGORZATA SIWISKA2, PAWE
STCZEK3
1
Department of Immunobiology of Bacteria
2
Department of General Microbiology
3
Department of Genetics of Microorganisms
Institute of Microbiology, Biotechnology and Immunology, University of d, Banacha 12/16, 90-237 d, Poland
*
E-mail: rozala@biol.uni.lodz.pl

ABSTRACT

The genus Proteus belongs to the Enterobacteriaceae family, where it is placed in the
tribe Proteeae, together with the genera Morganella and Providencia. Currently, the
genus Proteus consists of five species: P. mirabilis, P. vulgaris, P. penneri, P. hauseri
and P. myxofaciens, as well as three unnamed Proteus genomospecies. The most
defining characteristic of Proteus bacteria is a swarming phenomenon, a multicellular
differentiation process of short rods to elongated swarmer cells. It allows population of
bacteria to migrate on solid surface. Proteus bacteria inhabit the environment and are
also present in the intestines of humans and animals. These microorganisms under
favorable conditions cause a number of infections including urinary tract infections
(UTIs), wound infections, meningitis in neonates or infants and rheumatoid arthritis.
Therefore, Proteus is known as a bacterial opportunistic pathogen. It causes
complicated UTIs with a higher frequency, compared to other uropathogens. Proteus
infections are accompanied by a formation of urinary stones, containing struvite and
carbonate apatite. The virulence of Proteus rods has been related to several factors
including fimbriae, flagella, enzymes (urease - hydrolyzing urea to CO2 and NH3,
proteases degrading antibodies, tissue matrix proteins and proteins of the complement
system), iron acqusition systems and toxins: hemolysins, Proteus toxin agglutinin
(Pta), as well as an endotoxin - lipopolysaccharide (LPS). Proteus rods form biofilm,
particularly on the surface of urinary catheters, which can lead to serious consequences
for patients. In this review we present factors involved in the regulation of swarming
phenomenon, discuss the role of particular pathogenic features of Proteus spp.,
and characterize biofilm formation by these bacteria.

KEY WORDS: Proteus, pathogenicity, mechanisms of virulence

1. Classification and environmental distribution of Proteus rods


The genus Proteus, which was described Providencia. Proteus rods are distinguishable
for the first time by Hauser in 1885, belongs from most other genera by their ability to
to the Enterobacteriaceae family. In this swarm across agar surfaces of solid media.
family it is placed in the tribe Proteeae, The differentiation of Proteus rods according
together with the genera Morganella and to the results of biochemical tests is shown in

DOI: 10.2478/fobio-2013-0001 1
FOLIA BIOLOGICA ET OECOLOGICA

the table 1. The genus Proteus currently described results of the analysis of partial
consists of five species: P. mirabilis, sequences of the rpoB gene encoding the
P. vulgaris, P. penneri, P. hauseri and -subunit of RNA polymerase, led to the
P. myxofaciens, as well as three unnamed conclusion that a genus-level genetic distance
Proteus genomospecies. Proteus myxofaciens exists between P. myxofaciens and the species
is the only Proteus species without any of the Proteus-Providencia group.
significance in the pathogenicity of humans, it Classification of P. myxofaciens in a new
has been isolated from living and dead larvae genus Cosenzaea gen. nov. has therefore been
of the gypsy moth Porteria dispar (OHARA proposed (GIAMMANCO et al. 2011).
et al. 2000, JANDA & ABBOT 2006). Recently
TABLE 1. Biochemical tests used in differentiation within the genus Proteus (RALSKI & STCZEK 2010, 2011).
+ 90100 % positive, - 0 9.9 % positive; S susceptible, R resistant, V variable.

Test P. mirabilis P. vulgaris P. penneri P. hauseri P. myxofaciens

Salicin fermentation - + - - -

Maltose fermentation - + + + +

D-Xylose fermentation + + + + -

Esculin hydrolysis - + - - -

Ornithine + - - - -
decarboxylase

Indole production - + - + -

Chloramphenicol S V R S S
susceptibility

Proteus microorganisms are widely the decomposing of the organic matter of the
distributed in the natural environment, animal origin. They are also present in the
including polluted water, soil, and manure. intestines of humans and animals. P. mirabilis
Due to their proteolytic activity, the ability to was most frequently isolated from dogs, cows
hydrolyze urea to ammonia and carbon and birds, whereas P. vulgaris was most
dioxide, as well as the oxidative deamination frequently isolated from pigs and cold-
of amino acids, these bacteria are involved in blooded vertebrates (JANDA & ABBOT 2006).

2. Swarming phenomenon
Swarming is a multicellular differentiation dimorphic bacteria, which in liquid media are
phenomenon that allows a population of motile, peritrichously flagellated short rods
bacteria to migrate on a solid surface in a (1.0 to 2.0 m in length with 610 flagella).
coordinate manner. It is important in These bacteria are called swimmer cells.
movement of Proteus rods to new locations However, when transferred onto solid media
and most probably helps them in the these short rods change into elongated (2080
colonization macroorganisms. It involves cell- m in length), hyperflagellated,
to-cell signaling and multicellular interactions multinucleated, nonseptated swarmer cells.
and is connected with the possibility of The latter migrate out from the inoculation
morphological differentiation of bacteria site as long as the population of swarmer cells
depending on growth media. Proteus are is reduced on solid surfaces. Then, the

RALSKI ET AL. 2
FOLIA BIOLOGICA ET OECOLOGICA

consolidation process takes place. In this described. Mutation in the wzz region
period of swarming growth, the long rods encoding O-antigen chain, as well as in genes
disintegrate to short bacteria. The processes of waaD and waaC required for the core region
differentiation and dedifferentiation of of LPS are impaired in swarming growth
Proteus bacteria are cyclic. It results in the (MORGENSTEIN et al. 2010). Swarmer cells
formation of characteristic rings of bacterial translocation is facilitated by an extracellular
growth on the agar plate (fig. 1) acidic polysaccharide designated as Cmf
(VERSTRAETEN et al. 2008, MORGENSTEIN et (colony migration factor), which acts as a
al. 2010, RALSKI & STCZEK 2010). lubricant, reducing surface friction (GYGI et
Differentiation of swimmer cells into al. 1995). This capsular polysaccharide is
swarmer cells is induced by the contact of structurally identical with the O-specific part
bacteria with a solid surface and the inhibition of Proteus LPS serogroup O6 (KNIREL et al.
of flagellar rotation. It was found that the 2011). Most likely, Proteus O-specific
addition of thickening agents to liquid media polysaccharides are important for the
resulted in differentiation of short rods to swarming. It was found that the Re mutant of
swarmer cells. A similar effect was obtained P. mirabilis R45 producing LPS having only
when antiflagellar antibodies were added to the lipid A and Kdo region was unable to
liquid medium (BELAS & SUVANASUTHI swarm. The Ra mutant of P. mirabilis R110
2005). Mutations in gene flaA encoding containing lipid A and a complete core region
flagellin, as well in genes involved in expressed only a limited ability for migration
flagellum assembly led to the inhibition of on the solid medium, whereas most of
Proteus short rods differentiation (BELAS S-forms of P. mirabilis, P. vulgaris and
1994). Lipopolysaccharide (LPS), a surface P. penneri could swarm vigorously (BABICKA
component of these bacteria, seems to be 2001, KWIL 2003).
important in the swarming phenomenon,
however, its exact role has not been

Figure 1. Swarming growth of three strains P. mirabilis strains 34/57 (A), 14/57 (B) and 26/57 (C) on a agar plate
(KWIL data not published).

Swarming growth is characteristic for extracellular signal AI-2 (autoinducer-2) does


population of bacteria, thus it is dependent on not play a role in swarming. Cyclic dipeptides
multicellular interactions and cell to cell were also shown as P. mirabilis signaling
signaling in the phenomenon described as molecules out of importance in swarming
quorum sensing. It was found that (RATHER 2005). Other signal molecules

PROTEUS SP. AN OPPORTUNISTIC BACTERIAL PATHOGEN 3


FOLIA BIOLOGICA ET OECOLOGICA

N-acyl homoserine lactones crucial in quorum DisA most probably inhibits the assembly of
sensing are not produced by P. mirabilis heterotetramer or its binding to DNA
(MORGENSTEIN et al. 2010). Nevertheless, it (MORGENSTEIN et al. 2010).
was shown that N-acyl homoserine lactones Proteus has 16 predicted two-component
added to the media are able to accelerate systems TCS (PEARSON et al. 2008). TCS
migration of some P. mirabilis strains consists of sensor kinase, which activates
(STAKOWSKA et al. 2008). In earlier studies, response regulator DNA a binding protein
glutamine was found to be able to initiate controlling gene expression. Two of TCS -
swarming on minimal media (ALLISON et al. Rcs and Rpp are involved in swarming
1993). Swarming growth is also influenced by regulation (MORGENSTEIN et al. 2010). Rcs
fatty acids. Oleic acid stimulates, whereas system contains RcsC sensor kinase, RcsB
lauric and myristic acids inhibit this response regulator, RcsD intermediate
phenomenon, respectively (LIAW et al. 2004). transferring phosphate to the RcsB and RcsF
The mechanism by which glutamine and fatty which is an outer membrane lipoprotein.
acids play a role in the swarming process as Mutation in the genes coding Rcs system in P.
signals for cell to cell communication is mirabilis lead to the hyperswarming
currently unknown. In swarmer cell phenotype, most probably due to an increased
differentiation putrescine was described as a expression of the flhDC. P. mirabilis Rcs
biochemical signal, however, the mechanism mutant grows in liquid media as elongated
by which this amine regulates swarming is not cells. The Rpp system consists of RppA
clear. Most probably putrescine can form a response regulator and RppB histidine sensor
complex with galacturonic acid which is kinase like protein. Mutation in the Rpp
component of CPS. This suggests a possibility system in P. mirabilis also results in a
that putrescine complexed with cell surface hyperswarming phenotype.
polysaccharide acts as a signal factor or flhDC expression is also regulated by
putrescine regulates expression of CPS several proteins including Umo, MrpJ, WosA,
(RATHER 2005, MORGENSTEIN et al. 2010). Lrp and RsmA. Umo A-D proteins (umo
A number of factors playing a role in upregulated expression of the flhDC master
swarming regulations have been recognized operon) are located in the cell membrane and
(DANIELS et al. 2004, RATHER 2005, periplasm. Mutant defective in production of
VERSTRAETEN et al. 2008, MORGENSTEIN et UmoD protein is not able to swarm, however,
al. 2010). The most important gene involved such effect was not noticed in the case of
in upregulation of flagellin production is UmoA and UmoC (DUFOUR et al. 1998).
flhDC, class 1 gene of flagellar regulon. This mrpJ gene is located in the mrp operon
gene encodes FlhD2C2 complex, encoding MR/P fimbriae, which are required
a heterotetrameric transcriptional regulator, for sessile lifestyle of bacteria (NIELUBOWICZ
which also regulates the expression of & MOBLEY 2010). It is opposite to the
additional genes required for swarmer cells swimming or swarming phenotype of
differentiation. The expression of flhDC bacteria. The existence of bacteria in one of
increases 10-fold during initiation of swarmer two forms is regulated by mrpJ gene product
cells differentiation and it is influenced by MrpJ protein, a transcriptional regulator. It
a variety of environmental factors and binds the flhDC promoter, which causes its
regulating genes. FlhD2C2 activates class 2 repression and results in loss of motility of
genes coding basal body and hook proteins of bacteria (PEARSON & MOBLEY 2008).
flagella, as well as 28 factor, which activates wosA gene coding WosA protein, is
class 3 genes required for synthesis of overexpressed in strains exhibiting
flagellin and flagellum assembly. FlhD2C2 hyperswarming phenotype (wos wild type
activity is regulated by two identified factors onset superswarming). WosA overexpression
DisA (decarboxylase inhibitor of swarming) increases the expression of fhlDC and leads to
and Lon protease (ATP-dependent protease). a differentiation of swarmer cells in liquid

RALSKI ET AL. 4
FOLIA BIOLOGICA ET OECOLOGICA

media. The hyperswarming wosA bacteria of hemolysin, protease, urease and flagellin
move quickly and spend less time in the (LIAW et al. 2003). Overexpressing of rsmA
consolidation phase. wosA expression is led to repression of flhDC (MORGENSTEIN et
growth phase dependent, as well as it is al. 2010).
partially dependent upon the expression of The role of the swarming phenomenon in
flaA gene encoding the flagellar filament the pathogenicity of Proteus bacteria (see
(HATT & RATHER 2008). below) is till now unclear. It was shown that
Leucine responsive protein (Lrp) is a swarmer cells demonstrate higher production
transcriptional global regulator involved in of urease, HpmA hemolysin and IgA
regulating different processes including amino metaloprotease ZapA, as well as flagellin
acids synthesis, peptide transport, and pilin synthesis, as compared to swimmer cells
biogenesis (RATHER 2005, MORGENSTEIN et (RATHER 2005, JACOBSEN et al. 2008). There
al. 2010). In P. mirabilis strains Lrp plays a is some controversy concerning invasiveness
role in the regulation of the swarming of swarmer cells. ALLISON et al. 1992 showed
phenomenon, most probably its action leads in vitro that this morphotype could invade
to the inhibition of hyperflagellation. uroepithelial cells, whereas nonflagellated and
Mutation in lrp results in a decrease in the nonswarming forms were noninvasive. Other
flhDC, flaA and hpm (hemolysin) expression authors (RALSKI et al. 1986, CHIPPENDALE
and, in consequence, a nonswarming et al. 1994) showed the short rods as invasive
phenotype (HAY et al. 1997). Lrp is well forms, rather than swarmer cells. These
known to respond to leucine and to a lesser results were confirmed by JANSEN et al.
extent to alanine. Recently, it was found that (2003) in a mouse model of ascending UTI.
it also responds to methionine and is sensitive These authors have found that the
to isoleucine, histidine and thyrosine (HART & predominant cell type in the urinary tract are
BLUMENTHAL 2010). In additional studies short swimmer cells but not elongated
HART et al. (2011) showed that recognition of swarmer cells. Recently, FUJIHARA et al.
DNA by the helix-turn-helix motif of Lrp is (2011) suggested that P. mirabilis cells
modulated by the N-terminal tail. differentiate into hyperflagellated and
RsmA repressor of secondary multinucleated swarmer cells in acidic pH of
metabolites is a member of regulatory systems the hosts urine and differentiate back into
involved in the expression control of genes swimmer forms, when the urinary pH is
which code factors commited to stationary increased and it is alkaline after urease action
growth processes. This protein stabilizes (see below). In acidic condition swarmer cells
mRNA. In P. mirabilis RsmA is 6.8 kDa exhibit higher cytotoxicity against T24 line.
protein containing 62 amino acids. Increasing In alkaline condition P. mirabilis showed few
its expression results in the inhibition of elongated cells with high number of flagella
swarmer cells differentiation and production and cytotoxic activity.

3. Pathogenicity
Proteus rods are opportunistic bacterial by BLOCH et al. (2010). However, it should be
pathogens which under favorable conditions stressed that Proteus bacteria cause UTIs with
cause urinary tract infections (UTIs), wound higher frequency. This type of infections is
infections, meningitis in neonates or infants classified as uncomplicated or complicated.
and rheumatoid arthritis (OHARA et al. 2000, Uncomplicated infections occur in patients
JANDA & ABBOT 2006). KALRA et al. (2011) who are otherwise considered healthy,
reviewed endocarditis due to Proteus species whereas complicated infections usually take
and OKIMOTO et al. (2010) reported place in patients with an urinary catheter in
P. mirabilis pneumonia. Brain abscesses place or with structural and/or functional
during P. vulgaris bacteremia were described abnormalities in the urinary tract, suffering

PROTEUS SP. AN OPPORTUNISTIC BACTERIAL PATHOGEN 5


FOLIA BIOLOGICA ET OECOLOGICA

from another illness, immunocompromised, as among others mannose-resistant Proteus-like


well as after surgical intervention in the fimbriae, urease, and iron uptake systems.
urogenital system. It was found that Genes encoding flagella were downregulated.
Escherichia coli is a common cause of Earlier studies (FLANNERY et al. 200) showed
uncomplicated infections. Complicated UTIs the identification of 94-kb PAI (pathogenicity
might be polymicrobial and are usually island) designated ICEPm1 (integrative and
caused by Gram-negative bacteria Proteus conjugative elements P. mirabilis 1). This
spp., Providencia stuartii, Morganella PAI is common to P. mirabilis, P. stuartii and
morganii, E. coli, Pseudomonas aeruginosa, M. morganii and carries virulence factors
Klebsiella pneumoniae as well as some Gram- including Proteus toxic agglutinin (see below)
positive bacteria (NIELUBOWICZ & MOBLEY and the high pathogenicity island of Yersinia
2010). Proteus rods can cause hematogenous spp.
infections and ascending infections, however, The P. mirabilis genome predicts 17
the latter are more common for these distinct fimbrial operons and 13 additional
microorganisms. The ascending infections orphan fimbrial genes not associated with
comprise colonization subsequently urethra, complete operons (PEARSON et al. 2008,
bladder, ureters and kidneys. P. mirabilis NIELUBOWICZ & MOBLEY 2010). The
causes UTIs with the highest frequency biogenesis and/or the biological role were
among all Proteus species. These bacteria are studied for six types of fimbriae and
a common cause of complicated UTIs with agglutinins: MR/P (mannose resistant Proteus
the frequency of 12 % and catheter associated like fimbriae), MR/K (mannose resistant
bacteriuria (WARREN 1996, JACOBSEN et al. Klebsiella hemagglutinins), PMF (Proteus
2008). They also cause uncomplicated lower mirabilis fimbriae), NAF (nonagglutinating
urinary tract infections, however less fimbriae), ATF (ambient temperature
frequently. CHLABICZ et al. (2011) have found fimbriae) and P. mirabilis P-like fimbriae
in Poland uncomplicated P. mirabilis (fimbriae, which are similar to E. coli P pili)
infections with the frequency of 3.4 %. (COKER et al. 2000, ROCHA et al. 2007b,
The virulence of Proteus rods has been JACOBSEN et al. 2008).
related to several factors including fimbriae; The most important P. mirabilis fimbriae
flagella; enzymes: urease, hydrolyzing urea to seem to be MR/P. This type of fimbriae is
CO2 and NH3; proteases degrading antibodies, encoded by mrp operon containing 10 genes
tissue matrix proteins and proteins of located on bacterial chromosome. The main
complement system; iron acqusition systems structural subunit of these fimbriae is the
and toxins: hemolysins, Proteus toxin MrpA protein. The MrpC protein is an outer
agglutinin (Pta), as well as endotoxin - membrane platform, MrpD plays a role of a
lipopolysaccharide (LPS) (table 2) (COKER et chaperon protein and MrpH is an adhesin
al. 2000, MOBLEY 1996, RALSKI et al. located at the fimbrial tip (BAHRANI et al.
1997, RALSKI 2002, NIELUBOWICZ & 1991, LI et al. 1999, COKER et al. 2000).
MOBLEY 2010). BEVERIDGE (1999) postulated MR/P/ fimbriae undergo phase variation. It
the possible role of outer membrane vesicles was found that the MrpI protein acts as
(OMVs) released from the P. mirabilis P. mirabilis recombinase controlling on/off
surface, as an alternative route of delivery of orientation of the mrp promoter. The fimbrial
some enzymes, as well as some virulence genes are transcribed and MR/P fimbriae are
factors. synthesized when the promoter is in on
PEARSON et al. (2011) using microarrays orientation (LI et al. 2002a, NIELUBOWICZ &
analyzed P. mirabilis gene expression in vivo MOBLEY 2010). In the off orientation the
from experimentally infected mice and production of fimbriae is stopped. Expression
showed 471 gene upregulated and 82 of MR/P fimbriae is increased under oxygen
downregulated in comparison to the in vitro limitation (LANE et al. 2009). These fimbriae
broth culture. Upregulated genes encoded contribute to biofilm formation and facilitate

RALSKI ET AL. 6
FOLIA BIOLOGICA ET OECOLOGICA

colonization of upper urinary tract and were infection. Indeed, it was shown in vivo that
found more often on bacterial strains which flagella-negative mutant of P. mirabilis is less
cause pyelonephritis (JANSEN et al. 2004). virulent, compared to the wild strain, which
MR/P fimbriae are involved in aggregative suggests an important role of these bacterial
adherence to HEp-2 cells (ROCHA et al. surface structures in the pathogenicity
2007a). MR/P fimbriae elicit a strong immune (MOBLEY et al. 1996). However, it must be
response during infection. It is directed stressed, that lack of flagella on P. mirabilis
against the MrpA protein. It was found that human isolate was found (ZUNINO et al.
immunization of mice with the attenuated 1994). Flagella play a role of H antigen and
strain of Salmonella Typhimurium expressing are strongly immunogenic. Bacteria can avoid
MrpA-TetC fusion protein led to an increase immune response of human organism due to
in serum level of IgG antibodies against the antigenic variation process. This
MrpA and significantly reduced P. mirabilis phenomenon is based on flagellin genes
colonization of bladder and kidney. TetC is a rearrangement. Flagellar antigenic variations
non-toxic highly immunogenic fragment of allow bacteria to evade the action of secretory
the tetanus toxin (SCAVONE et al. 2011). The IgA antibodies directed against these
presence of mrp operon was shown not only organelles (BELAS 1994).
in P. mirabilis strains but also in other species P. mirabilis evades the immune system
of the Proteus genus (BABICKA 2001, KWIL, response during infection thanks to the
2002, 2003, KWIL et al. data not published). production of sIgA protease. The best
Other than MR/P fimbriae are known to a characterized is 54-kDa secreted
lesser extent. MR/K hemagglutinins were metalloprotease ZapA, mirabilysin belonging
found more frequently on P. penneri strains to the serralysin family (LOOMES et al. 1990).
than on P. mirabilis strains and most probably ZapA possesses a broad spectrum of
they facilitated the adherence of bacteria to proteolytic specificity, including the activity
the urinary catheters (JACOBSEN et al. 2008). against antibodies of sIgA, IgA2 and IgG,
The exact virulence role of PMF fimbriae is proteins of the complement system, cell
unclear (NIELUBOWICZ & MOBLEY 2010). matrix proteins, cytoskeletal proteins, as well
ZUNINO et al. (2007) by using P. mirabilis as antimicrobial defense components of
pmfA/mrpA-D mutant showed the specific and human innate system peptides hBD1 and
additive roles of MR/P and PMF fimbriae in cathelicidin LL-37 (BELAS et al. 2004,
UTI caused by these bacteria. NAF fimbriae, JACOBSEN et al. 2008). ZapA was found to be
earlier known as UCA fimbriae, allow an important virulence factor in a rat model of
bacteria to colonize uroepithelial cells (WRAY P. mirabilis induced acute and chronic
et al. 1986). It was found that NAF/UCA prostatitis (PHAN et al. 2008). Recently,
binds the following receptors: asialo-GM1, CARSON et al. (2011) described N-alpha
asialo-GM3, lactosyl ceramide and galectin mercaptoamide dipeptides, which can be used
(LEE et al. 2000, ALTMAN et al. 2001). ATF as inhibitors of ZapA. KWIL et al. (2011) have
fimbriae are not important in the shown the presence of the zapA operon in
pathogenicity of human beings (ZUNINO et al. P. vulgaris and P. penneri strains.
2000). PMP fimbriae were found on canine It was shown that amino acid deaminase
UTI isolates (BIJLSMA et al. 1995), however (Aad) produces -keto acids which serves as
genes coding this type of fimbriae were found siderophores involved in iron acquisition
in human clinical isolates (PEARSON et al. (DRECHSEL et al. 1993, MASSAD et al. 1995).
2008). Recently, HIMPSI et al. (2010) described
There are conflicting results concerning the proteobactin and yersiniabactin related
importance of flagella in the infection. Since siderophore function in P. mirabilis. Zinc,
flagella mediate motility of bacteria, they another biologically important element
seem to be required for virulence of needed for bacterial growth, is utilized by
uropathogens, particularly during ascending P. mirabilis by use of ZnuABC high affinity

PROTEUS SP. AN OPPORTUNISTIC BACTERIAL PATHOGEN 7


FOLIA BIOLOGICA ET OECOLOGICA

transport system (NIELUBOWICZ & MOBLEY the precipitation of magnesium and calcium
2010). P. mirabilis also posses high-affinity ions, normally soluble in slightly acidic urine,
phosphate transporter system Pst. It is and the formation of urinary stones,
postulated that this system is important in containing struvite and carbonate apatite
P. mirabilis virulence (JACOBSEN et al. 2008). (RALSKI et al. 1997, JACOBSEN et al. 2008,
Urease is an urea amidohydrolase which NIELUBOWICZ & MOBLEY 2010). The
catalyses the hydrolysis of urea to ammonia carbonate apatite forms an amorphous
and carbamate, then digested to carbone precipitate opposite to struvite, which forms
dioxide and second molecule of ammonia crystals of defined morphology. It was found
(MOBLEY et al. 1995). This process results in that struvite exhibits polar properties
elevation of pH and non-physiological (PRYWER & TORZEWSKA 2010, ROMANOWSKI
alkalization of urine, which in turn induces et al. 2010).

Table 2. Virulence factors (VP), biological features (BF) and types of growth (TG) of Proteus rods in relation to the
pathogenicity.

VP/BF/TG Specific data Contribution to the pathogenicity

Fimbriae P. mirabilis genome predicts 17 Adherence of bacteria to the epithelial tissue


distinct fimbrial operons. Six types Contribution to the biofilm formation
of fimbriae/agglutinins were
identified

Flagella H-antigen Ascension of bacteria in urinary tract. Elicitation


of immunoresponce which can be avoid due to
the antigenic variation
Invassiveness Studied in vitro Penetration and internalization of bacteria into
host cells
Proteases IgA and IgG proteases, which are Diminish or elimination of phagocytosis
also able to degrade other types of
biologically important proteins
Amino acid Production of alpha-keto acids Iron acquisition
deaminase which serve as siderophores
Proteobactin and
yersiniabactin -
related siderophore
ZnuABC High affinity zinc transport system Zinc utilization
Pst High affinity phosphate transporter Phosphate accumulation
Urease Enzyme hydrolyzing urea to NH3 Elevation of urine pH level, resulting in stones
and CO2 formation. Cytotoxicity against urinary epithelial
cells
Protein toxins:
HmpA Cell-bound hemolysin Pore forming toxins mediating cytotoxicity
HlyA Cell free hemolysin, RTX protein
Pta Cytotoxin and agglutinin Cytotoxicity and autoagglutination of bacteria
CPS Most probably structurally identical Formation of biofilm. Importance in formation of
with O-specific part of LPS urinary stones. Facilitation of swarmer cells
translocation
LPS Endotoxin Endotoxicity. Resistance against bactericidal
action of the serum. Production of glycocalyx
important in biofilm formation
OMVs Outer membrane vesicles containing Transport of virulence factors to the host tissues
periplasmic constituents
Swarming growth Differentiation of short rods to Not fully understood
swarmer cells
Biofilm formation Crystalline biofilm, particularly Protection of bacteria against action of
formed on the surface of urinary antibiotics and immune mechanisms
catheters

RALSKI ET AL. 8
FOLIA BIOLOGICA ET OECOLOGICA

The structural and elastic properties of prevention of bacterial adherence to the


struvite crystals were studied by PIECHOTA et uroepithelium (JACOBSEN et al. 2008). In vitro
al. (2011). Formation and morphology of studies performed by TORZEWSKA and
struvite crystals depend on the pH of urine. RALSKI (2009) showed that
PRYWER and TORZEWSKA (2009) have found glycosaminoglycans have no inhibitory effect
that for pH in the range 7.59.0 struvite on infection-induced crystallization.
crystals take single and hemimorphic In a mouse model of ascending UTI it was
morphology. At higher pH many twin and found that the urease negative mutant of P.
dendritic crystals have been shown. mirabilis was less virulent as compared to the
DUMANSKI et al. (1994) has revealed that wild strains, which suggests the important
Proteus capsular polysaccharide structurally role of this enzyme in infection caused by this
identical to O-specific polysaccharide of LPS bacterium (JONES et al. 1990). Therefore,
accelerates crystallization process. investigation of different substances which
TORZEWSKA et al. (2003) confirmed this can be used as urease inhibitors is being
observations and showed that urinary stone performed. Recently, BERLICKI et al. (2011)
formation due to the urease activity, may be described N-substituted amino-methane-
modified by surface O-antigens being a part phosphonic and aminomethane-P-
of lipopolysaccharides (LPS). The sugar methylphosphonic acids as effective inhibitors
composition of these polysaccharides may of P. mirabilis urease.
either enhance or inhibit the crystallization of Proteus rods produce three types of
struvite and carbone apatite depending on cytolysins, HpmA and HlyA hemolysins and
their chemical structure and ability to bind Pta cytotoxic agglutinin. HpmA is the 166
cations. Bacteria present within urinary stones kDa protein, calciumindependent cell-bound
are protected from host defense mechanisms hemolysin secreted by the bacteria and
and antibiotic treatment (LI et al. 2002b). activated in the process mediated by HpmB
Therefore, several phytotherapeutic (UPHOFF & WELCH 1990). This hemolysin
compounds have been investigated to prevent belongs to the pore forming toxins family and
urinary stone formation. For example it is cytotoxic against a number of cells
curcumin an yellow-orange pigment non toxic (SWIHART & WELCH 1990). HlyA hemolysin
to humans or animals extracted from the roots is produced by P. vulgaris, P. penneri and
of turmeric (Curcuma longa) was shown to P. hauseri strains (KORONAKIS et al. 1987,
inhibit urease activity and consequently RALSKI & KOTEKO 1987, RALSKI &
decrease the struvite growth. It was found in STCZEK 2011). It is a strongly cytotoxic,
vitro experiment of struvite formation in calcium dependent 110 kDa extracellular
artificial urine that the addition of curcumin toxin belonging to the RTX family of proteins
increases the induction time and decreases the (RTX - repeat in toxin). Pta (Proteus toxic
efficiency of formation of these crystals agglutinin), an autotransporter with subtilase-
(TORZEWSKA et al. 2010, PRYWER & like serine protease activity exhibits
TORZEWSKA 2010, 2012). Moreover, bifunctional activity. It is both cytotoxin and
curcumin also inhibits biofilm formation and agglutinin, which mediates autoagglutination
block swarming growth (TORZEWSKA et al. of bacteria and cytotoxicity against human
2010). Recently, MALIC et al. (2011) reported bladder and kidney cells. The vaccination of
on the development of an early warning mice with Pta results in protection against
sensor for encrustation of urinary catheters in P. mirabilis UTI (ALAMURI & MOBLEY 2008).
case of Proteus infection. Lipopolysaccharide is composed of three
It was shown that the high level of genetically and structurally distinct regions:
ammonia, a product of urease action also O-specific chain (O-antigen, O-specific
results in damage of the urinary tract polysaccharide), the core oligosaccharide and
epithelium. Toxic ammonia damages bladder lipid A, which anchors the LPS molecule to
surface glycosaminoglycans important in the the bacterial outer membrane (RAETZ &

PROTEUS SP. AN OPPORTUNISTIC BACTERIAL PATHOGEN 9


FOLIA BIOLOGICA ET OECOLOGICA

WHITFIELD 2002). LPS containing all these were found to belong to serogroups O3, O10,
three regions is produced by smooth forms of O11, O13, O23, O27 and O30. Recently,
bacteria. Rough strains synthesize LPS KACA et al. (2011) studying 123 clinical
containing lipid A and the whole core region isolates from Sweden and Poland showed that
or only its part. All three regions of Proteus none of serogroups was predominant.
LPS have been studied (RALSKI 2002, DRZEWIECKA et al. 2010 described seven
2004, 2008, RALSKI et al. 2002). The P. mirabilis strains representing serogroup
differences in the structure of O-antigens O78 which can be classified in one bacterial
serve as a basis for the serological clone widespread in hospitals, most probably
classification of Proteus strains. The due to the nosocomial infection and
serological classification scheme currently autoinfections. The results of detailed studies
consists of 78 serogroups (DRZEWIECKA et al. of Proteus O-specific polysaccharides are
2004, 2008, 2010, DRZEWIECKA & presented in the review by KNIREL et al. 2011.
SIDORCZYK 2005, KNIREL et al. 2011). The WANG et al. (2010) identified five putative
prevalence of particular Proteus O-serogroups Proteus O-antigen gene clusters and Proteus
among clinical isolates has been changing specific genes of strains classified to five
over the time. Previously, clinical isolates serogroups.

Figure 2. Structure of the O-specific repeating unit (A) and core oligosaccharide obtained by mild acid degradation
(B) of lipopolysaccharide of Proteus mirabilis genomic strain HI 4320. Alt - altruronic acid (KNIREL et al. 2011,
VINOGRADOV 2011).

The core region of Proteus E. coli and Salmonella (HOLST 1999,


lipopolysaccharides was studied by use of VINOGRADOV et al. 2002). The Proteus core
rough mutants or smooth forms classified into region is composed of two parts an inner
different serogroups (RADZIEJEWSKA- part, common for several number of strains
LEBRECHT et al. 1989, VINOGRADOV et al. and second, an outer part, which is
1994, VINOGRADOV et al. 2002). The characterized by structural variability from
structural diversity of the core is characteristic strain to strain. The common part is not
for Proteus spp. and makes it different from identical in all Proteus strains and is

RALSKI ET AL. 10
FOLIA BIOLOGICA ET OECOLOGICA

subdivided into three forms known as mechanisms (DONLAN & COSTERTON 2002,
glycoforms I-III. The outer part of the core STICKLER & FENLEY 2010). Proteus bacteria
region (outer core) contains an form a crystalline biofilm particularly on the
oligosaccharide characteristic for particular surface of catheters (STICKLER 2008). It
Proteus strains (VINOGRADOV et. al 2002, contains calcium phosphate and magnesium
RALSKI 2008). The structure of the core phosphate crystals formed due to the action of
part of P. mirabilis genomic strain HI4320 urease. JONES et al. (2007) reported that the
(formerly classified into serogroup O10) was structure of P. mirabilis biofilm depends on
shown by VINOGRADOV (2011) (fig. 2). The the environment (artificial urine, laboratory
epitope specificity of the core region of media) in which it occurs. Biofilms in Luria-
P. penneri strains were studied by PALUSIAK Bertani (LB) broth formed a mushroom-like
and SIDORCZYK (2010). They have identified architecture, while biofilms grown in urine
seven epitopes responsible for cross reactivity formed a flat layer. MORYL et al. (2008)
of LPS of these bacteria with appropriate studied the effect of environmental factors
antisera. The identification of genes required such as glucose, acidic pH, albumin and some
for the biosynthesis of the core region of antimicrobial agents: ciprofloxacin and
P. mirabilis were described by AQUILINI et al. methylene blue on the formation of
(2010). P. mirabilis biofilm. The most inhibiting
LPS, released from pathogenic bacteria effect was observed after incubation of
during infection induces a spectrum of bacteria with albumin and in acidic growth
biological activities. Therefore, LPS is also media. KWIECINSKA-PIRG (2010) studied the
known as an endotoxin. The mechanism of formation of biofilm on different surfaces,
biological action of the endotoxin is common and has found silicone latex surface to be
to most bacteria. Its biological domain is better than polychloride vinyl and
lipid A (UKASIEWICZ & UGOWSKI 2003). polypropylene surfaces for P. mirabilis
Proteus lipid A contains glucosamine growth in biofilm. Octenisept R an antiseptic
disaccharide substituted with phosphate products occured to be active against these
residues and fatty acids. It also contains bacteria growing in biofilm. Prevention and
4-amino-4-deoxy-L-arabinopyranose eradication of P. mirabilis biofilm by use of
(L-Arap4N), which quantitatively substitutes lytic bacteriophages was shown by CARSON et
the ester-linked phosphate residue of the al. (2010). It was also found that two quorum
glucosamine backbone (SIDORCZYK et al. sensing antagonists p-nitrophenyl glycerol
1983). (PNPG) and tannic acid inhibit the formation
Proteus LPS is associated with a broad of P. mirabilis biofilm in artificial urine
spectrum of biological activities, and with (JONES et al. 2009). P. mirabilis biofilm
interactions with bacterial or eukaryotic cells formation on glass and polystyrene can also
(RALSKI 2008). LPS of S-forms of bacteria be inhibited by extract of Ibucella lutea, a
contributes to their resistance against native plant of America (SOSA & ZUNINO
bactericidal action of the serum (KACA & 2009). MORYL (2010) observed inhibition of
UJAZDA 1998, MIELNIK et al. 2004, BABICKA biofilm formation by P. mirabilis growing
2001, KWIL 2003, KACA et al. 2009). The O- together with Escherichia coli or Providencia
specific polysaccharide chain exposed outside stuartii. STAKOWSKA et al. (2011) showed a
bacteria is involved in glycocalyx formation. significant influence of N-butanoyl
Glycocalyx enables bacteria to grow in a form homoserine lactone (BHL) on biofilm
of biofilm on a solid surface. formation by P. mirabilis strain belonging to
Bacterial biofilm is defined as a matrix- serogroup O18. Recently, SCHLAPP et al.
enclosed bacterial population adhering to the (2011) excluded the contribution of swarmer
surfaces. Bacteria enclosed in a glycocalyx cells in biofilm formation during the 7-day
capsule are protected against the action of incubation of bacteria in LB broth on glass
antibodies, as well as against other immune coverslips. These authors showed that

PROTEUS SP. AN OPPORTUNISTIC BACTERIAL PATHOGEN 11


FOLIA BIOLOGICA ET OECOLOGICA

P. mirabilis biofilm formation is a five-stage extracellular materials and decreasing of


process: i) reversible adhesion to the solid elongation, iii) acceleration of bacterial
surface with slow growth of bacteria, growth, further decrease in elongation and
presence of elongated cells and absence of halting production of extracellular material,
extracellular matrix, ii) irreversible bacterial iv) maturation of biofilm, v) detachment of
adhesion, beginning of production of bacterial cells and dispersion of biofilm.

4. Summary
Proteus bacteria inhabit the environment population of bacteria to migrate in a
and they are also present in the intestines of coordinate manner on a solid surface. The role
humans and animals. These microorganisms of short rods and elongated swarmer cells in
under favorable conditions cause a number of the pathogenicity of Proteus bacteria is still
infections including urinary tract infections not established. The elongated morphotype
(UTIs), wound infections, meningitis in seems to be in the minority during infection.
neonates or infants and rheumatoid arthritis. Proteus bacteria form a crystalline biofilm,
Proteus opportunistic pathogens express particularly on the surface of catheters present
virulence factors associated with adherence, in the urinary tract of patients. It contains
motility, immunoavoidance, nutrient struvite and hydroxyapatite crystals formed
acquisition, host damage, biofilm formation due to the action of urease. The results of the
and endotoxicity. The most defining study on Proteus virulence and fitness factors
characteristics of Proteus bacteria is the will lead to a better understanding of
swarming phenomenon, a multicellular infectious processes and will subsequently
differentiation process of short rods to allow developing new methods of prevention
elongated swarmer cells. It allows the and clinical treatment.

5. References
ALAMURI, P., MOBLEY, H.L.T. 2008. A novel this species. Ph.D. thesis. University of Lodz. (In
autotransporter of uropathogenic Proteus mirabilis Polish).
is both a cytotoxin and an agglutinin. Mol.
BAHRANI, F.K., JOHNSON, D.E., ROBBINS, D. M.,
Microbiol. 68: 9971017.
MOBLEY, H.L.T. 1991. Proteus mirabilis flagella
ALLISON, C., COLEMAN, N., JONES, P.L., HUGES, C. and MR/P fimbriae: isolation, purification, N-
1992. Ability of Proteus mirabilis to invade terminal analysis, and serum antibody response
urothelial cells is coupled to motility and swarming following urinary tract infection. Infect. Immun. 59:
differentiation. Infect. Immun. 60: 47404746. 35743480.
ALLISON, C., LAI, H.C., HUGHES, C. 1993. Cell BELAS, R. 1994. Expression of multiple flagellin-
differentiation of Proteus mirabilis is initiated by encoding genes of Proteus mirabilis. J. Bacteriol.,
glutamine, a specific chemoattractant for swarming 176: 71697181.
cells. Mol. Microbiol. 8: 5360.
BELAS, R., MANOS, J., SUVANASUTHI, R. 2004. Proteus
ALTMAN, E., HARRISON, B.A., LATTA, R.K., LEE, K.K., mirabilis ZapA metalloprotease degrades a broad
KELLY, J.F., TIMBAULT, P. 2001. Galectin-3- spectrum of substrates, including antimicrobial
mediated adherence of Proteus mirabilis to Madin- peptides. Infect. Immun.72: 51595167.
Darby canine kidney cells. Biochem. Cell. Biol. 79:
BELAS, R., SUVANASUTHI, R. 2005. The ability of
783788.
Proteus mirabilis to sense surfaces and regulate
AQULIINI, E., AZEVEDO, J., JIMENEZ, N., BOUAMAMA, virulence gene expression involves FliL, a flagellar
L., TOMAS, J.M., REGUE, M. 2010. Functional basal body protein. J. Bacteriol. 187: 67896803.
identification of the Proteus mirabilis core
BERLICKI, L., BOCHNO, M., GRABOWIECKA, A.,
lipopolysaccharide biosynthetic genes. J. Bacteriol.
BIAAS, A., KOSIKOWSKA, P., KAFARSKI, P. 2011.
192: 44134424.
N-substituted aminomethanephosphonic and
BABICKA, D. 2001. Investigations of serological aminomethane-P-methylphosphinic acids as
specificity of O-antigens of Proteus vulgaris O1, inhibitors of ureases. Amino Acids. DOI:
O2, O4 and O9 and selected biological features of 10.1007/s00726-011-0920-4.

RALSKI ET AL. 12
FOLIA BIOLOGICA ET OECOLOGICA

BEVERIDGE, T.J. 1999. Structures of Gram-negative DRZEWIECKA, D., ARBATSKY, N.P., SHASHKOV, A.S.,
cell walls and their derived membrane vesicles. J. STCZEK, P., KNIREL, Y.A., SIDORCZYK, Z. 2008.
Bacteriol. 181: 47254733. Structure and serological properties of the O-
antigen of two clinical Proteus mirabilis strains
BIJLSMA, I.G., VAN DIJK, L., KUSTERS, J.G., GAASTRA,
classified into a new Proteus O77 serogroup. FEMS
W. 1995. Nucleotide sequence of two fimbrial
Immunol. Med. Microbiol. 54: 185194.
subunit genes, pmpA and ucaA from canine -
uropathogenic Proteus mirabilis strains. DRZEWIECKA, D., ARBATSKY, N.P., STCZEK, P.,
Microbiology. 141: 13491357. SHASHKOV, A.S., KNIREL, Y.A., SIDORCZYK, Z.
2010. Structure and serological studies of the O-
BLOCH, J., LEMAIRE, X., LEGOUT, L., FERRIBY, D.,
polysaccharide of strains from a newly created
YAZDANPANAH, Y., SENNEVILLE, E. 2010. Brain
Proteus O78 serogroup prevalent in Polish patients.
abscesses during Proteus vulgaris bacteremia.
FEMS Immunol. Med. Microbiol. 58: 269276.
Neurol. Sci. 32: 661663.
DRZEWIECKA, D., SIDORCZYK, Z. 2005.
CARSON, L., CATHCART, G.R., SCOTT, C.J.,
Characterization of Proteus penneri species
HOLLENBERG, M.D., WALKER, B., CERI, H.,
human opportunistic pathogens. Post. Mikrobiol.
GILMORE B.F. 2011 Comprehensive inhibitory
44: 113126. (In Polish).
profiling of the Proteus mirabilis metalloprotease
virulence factor ZapA (mirabilysin). Biochemie. DRZEWIECKA, D., ZYCH, K. SIDORCZYK, Z. 2004.
93:18241827. Characterization and serological classification of a
collection of Proteus penneri clinical strains. Arch.
CARSON, L., GORMAN, S.P., GILMORE, BF. 2010. The
Immunol. Therap. Exp. 52:121128.
use of lytic bacteriophages in the prevention and
eradication of biofilms of Proteus mirabilis and DUMANSKI, A.J., HEDELIN, H., EDIN-LILJEGREN, A.,
Escherichia coli. FEMS Immunol. Med. Microbiol. BEACHEMIN, D., MCLEAN, R.J.C. 1994. Unique
59: 447455. ability of Proteus mirabilis capsule to enhance
mineral growth in infectious urinary calculi. Infect.
CHIPPENDALE, G.R., WARREN, J.W., TRIFILLIS, A.L.,
Immun. 62: 29983003.
MOBLEY, H.L.T. 1994. Internalization of Proteus
mirabilis by human renal epithelial cells. Infect. FLANNERY, E., MODY, L., MOBLEY, H.L.T. 2009.
Immun. 62: 31153121. Identification of a modular pathogenicity island that
is widespread among urease-producing
CHLABICZ, S., LESZCZYSKA, K., LUKAS, W., GUALCO,
uropathogens and shares features with diverse
L., SCHITO, G., NABER, K.G. 2011. Uncomplictaed
group of mobile elements. Infect. Immun. 77:
lower urinary tract infections in females-clinical
48874894.
aspects, aetiology and antimicrobial resistance
epidemiology. Results of the ARESC FUJIHARA, M., OBARA, H., WATANABE, Y., ONO, H.K.,
(Antimicrobial Resistance Epidemiological Survey SASAKI J., GORYO, M., HARASAWA, R. 2011. Acidic
on Cystitis) study in Poland on their implications environments induce differentiation of Proteus
for empiric therapy. Przegl. Epidemiol. 65: 345 mirabilis into swarmer morphotypes. Microbiol.
351. (In Polish). Immunol. 55: 489493.
COKER, C., POORE A., LI, X., MOBLEY, H.L.T. 2000. GIAMMANCO, G.M., GRIMONT, P.A., GRIMONT, F.,
Pathogenesis of Proteus mirabilis urinary tract LEFEVRE, M., GIAMMANCO, G., PIGNATO, S. 2011.
infections. Microb. Infect. 2: 14971505. Phylogenic analysis of the genera Proteus,
Morganella, Providencia by comparison of rpoB
DANIELS, R., VANDERLEYDEN, J., MICHIELS, J. 2004.
gene sequence of type and clinical strains suggests
Quorum sensing and swarming migration in
reclassification of Proteus myxofaciens in a new
bacteria. FEMS Microbiol. Rev. 28: 261289.
genus, Cosenzaea gen. nov., as Cosenzaea
DONLAN, R.M., COSTERTON, J.W. 2002. Biofilms: myxofaciens comb. nov. Int. J. Syst. Evol.
survival mechanism of clinically relevant Microbiol. 61:16381644.
microorganisms. Clin. Microbiol. Rev. 15: 167
GYGI, D., RAHMAN, M.M., LAI, H.C., CARLSON, R.,
193.
GUARD-PETTER, J., HUGHES, C. 1995. A cell
DOUFOUR, A., FURNESS, R.B., HUGHES, C. 1998. Novel surface polysaccharide that facilitates rapid
genes that upregulate the Proteus mirabilis flhDC population migration by differentiated swarm cells
master operon controlling flagellar biogenesis and of Proteus mirabilis. Mol. Microbiol. 17: 1167
swarming. Mol. Microbiol. 29: 741751. 1175.
DRECHSEL, H.A., THIECKEN, A., REISSBRODT, R., JUNG, HART, B.R., BLUMENTHAL, R.M. 2010. Unexpected
G., WINKELMANN, G. 1993. -keto acids are novel coregulator range in the global regulator Lrp of
siderophores in the genera Proteus, Providencia Escherichia coli and Proteus mirabilis. J. Bacteriol.
and Morganella and are produced by amino acid 193: 10541064.
deaminases. J. Bacteriol. 175: 27272733.

PROTEUS SP. AN OPPORTUNISTIC BACTERIAL PATHOGEN 13


FOLIA BIOLOGICA ET OECOLOGICA

HART, B.R., MISHRA, P.K., LINTER, R.E., HINERMAN, of crystalline Proteus mirabilis biofilm. Int. J.
J.M., HERR, A.B., BLUMENTHAL, R.M. 2011. Antimicrob. Agen. 34: 360364.
Recognition of DNA by the helix-turn-helix global
KACA, W., ARABSKI, M., FUDAA, R., HOLMSTROM, E.,
regulatory protein Lrp is modulated by the amino
SJOHOLM, A., WEINTRAUB, A., FUTOMA-KOOCH.,
terminus. J. Bacteriol. 193:37943803.
B., BUGLA-POSKOSKA, G., DOROSZKIEWICZ, W.
HATT, J.K., RATHER, P.N. 2008. Characterization of 2009. Human complement activation by smooth
novel gene, wosA, regulating FlhDC expression in and rough Proteus mirabilis lipopolysaccharides.
Proteus mirabilis. J. Bacteriol. 190: 19461955. Arch. Immunol. Ther. Exp. 57: 383391.
HAY, N.A., TIPPER, D., GYGI, D., HUGHES, C. 1997. A KACA, W., GLENSKA, J., LACHOWICZ, L., GRABOWSKI,
nonswarming mutant of Proteus mirabilis lacks the S., BRAUNER, A., KWINKOWSKI, M. 2011.
Lrp global transcriptional regulator. J. Bacteriol. Serotyping of Proteus mirabilis clinical strains
179: 47414746. based on lipopolysaccharide, O-polysaccharide and
core oligosaccharide structures. Biochemistry
HIMPSI, S.D., PEARSON, M.M., AREWANG, C.J., NUSCA
(Moscow). 76: 851861.
T.D., SHERMAN D.H., MOBLEY, H.L.T. 2010.
Proteobactin and a yersiniabactin-related KACA, W., UJAZDA, E. 1998. Complement activation
siderophore mediate iron acquisition in Proteus by bacterial endotoxin. Post. Mikrobiol. 37: 421
mirabilis. Molec. Microbiol. 78: 138157. 427. (In Polish).
HOLST, O. 1999. Chemical structure of the core region KALRA, A., COOLEY, C., TSIGRELIS, C. 2011. Treatment
of lipopolysaccharides. In: H. BRADE, S.M. OPAL, of endocarditis due to Proteus species: a literature
S.N. VOGEL, D.C. MORRISON (eds). Endotoxin in review. Int. J. Infect. Dis. 15: 222225.
health and disease. Marcel Dekker, Inc. New York,
KNIREL, Y.A., PEREPELOV, A.V., KONDAKOVA, A.,
Basel, pp. 115154.
SENCHEKOVA, S.N., SIDORCZYK, Z., RALSKI, A.,
JACOBSEN, S.M., STICKLER D.J., MOBLEY, H.L.T., KACA, W. 2011. Structure and serology of O-
SHIRTLIFF, M.E. 2008. Complicated catheter- antigens as the basis for classification of Proteus
associated urinary tract infections due to strains. Inn. Immun. 17: 7096.
Escherichia coli and Proteus mirabilis. Clin.
KORONAKIS, V., CROSS, M., SENIOR, B., KORONAKIS,
Microbiol. Rev. 21: 2659.
E., HUGHES, C. 1987. The secreted hemolysins of
JANDA, J.M., ABBOT, S.L. 2006. The Proteus mirabilis, Proteus vulgaris, and
Enterobacteriaceae. ASM Press. Washington. 233 Morganella morganii are genetically related to each
259. other and to the alpha-hemolysin of Escherichia
coli. J. Bacteriol. 169: 15091515.
JANSEN, A.M., LOCKATELL, C.V., JOHNSON, D.E.,
MOBLEY, H.L.T. 2004. Mannose-resistant Proteus- KWIECISKA-PIRG, J. 2010. Estimation of production
like fimbriae are produced by most Proteus of biofilm by Proteus mirabilis bacilli. Ph.D. thesis.
mirabilis strains infecting the urinary tract, dictate Nicolas Copernicus University of Toru. Collegium
the in vivo localization of bacteria, and contribute to Medicum in Bydgoszcz. (In Polish).
biofilm formation. Infect. Immun. 72: 72927305.
KWIL, I., BABICKA, D., STCZEK, P., RALSKI, A.
JANSEN, B.V., LOCKATELL, C.V., JOHNSON, D.E., 2002. Applying the PCR metod for searching genes
MOBLEY, H.L.T. 2003. Visualisation of Proteus encoding main structural fimbrial proteins. In: 5 th
mirabilis morphotypes in the urinary tract: the Conference on Molecular biology in diagnostics of
elongated swarmer cell is rarely observed in infectious disease and biotechnology. Warsaw,
ascending urinary tract infection. Infect. Immun. SGGW Publisher. 8891.
71: 36073613.
KWIL, I. 2003. Investigations of selected virulence
JONES, B.D., LOCKATELL, C.V., JOHNSON, D.E., factors of Proteus penneri strains. Ph.D. thesis.
WARREN, J.W., MOBLEY, H.L.T. 1990. Construction University of Lodz. (In Polish).
of urease-negative mutant of Proteus mirabilis:
KWIL, I., BIEGASKA, H., OBAREK, K., RALSKI, A.
analysis of virulence in mouse model of ascending
2006. Expression of selected virulence factors
urinary tract infection. Infect. Immun. 58: 1120
among new bacterial species from the genus
1123.
Proteus: P. hauseri and Proteus genomospecies 4,
JONES, S.M., YERLY, J., HU, Y., CERI, H., MARTINUZZI, 5, 6. In: 9th Conference on molecular biology in
R. 2007. Structure of Proteus mirabilis biofilm diagnostics of infectious disease and biotechnology.
grown in artificial urine and standard laboratory Warsaw, SGGW Publisher. 9396.
media. FEMS Microbiol. Lett. 268:16021.
LANE, M.C., PEARSON, M.M., SIMMS, A.N., MOBLEY,
JONES, S.M., DANG, T., MARTINUZZI, R. 2009. Use of H.L.T. 2009. Oxygen-limiting conditions enrich for
quorum sensing antagonists to deter the formation fimbriate cells of uropathogenic Proteus mirabilis
and Escherichia coli. J. Bacteriol. 191: 13821392.

RALSKI ET AL. 14
FOLIA BIOLOGICA ET OECOLOGICA

LEE, K.K., HARRISON, B.A., LATTA, R., ALTMAN, E. management. ASM Press, Washington DC, pp.
2000. The binding of Proteus mirabilis 245269.
nonagglutinating fimbriae to ganglio-series
MOBLEY, H.L.T, BELAS, R., LOCKATELL, V.,
asialoglycolipids and lactosyl ceramide. Can. J.
CHIPPENADALE, G., TRIFILLIS A.L., JOHNSON, D.E.,
Microbiol. 46: 961966.
WARREN, J.W. 1996. Construction of a flagellum-
LI, X., JOHNSON, D.E., MOBLEY, H.L.T. 1999. negative mutant of Proteus mirabilis: effect on
Requirement of MrpH for mannose-resistant internalization by human renal epithelial cells and
Proteus-like fimbria- mediated hemagglutination by virulence in a mouse model of ascending urinary
Proteus mirabilis. Infect. Immun. 67: 28222833. tract infection. Infect. Immun. 64: 53325340.
LI, X., LOCKATELL, C.V., JOHNSON, D.E., MOBLEY, MOBLEY, H.L.T., ISLAND, M.D., HAUSINGER, R.P.
H.L.T. 2002a. Identification of MrpI as the sole 1995. Molecular biology of microbial ureases.
recombinase that regulates the phase variation of Microbiol. Rev. 59: 451480.
MR/P fimbria, a bladder colonization factor of
MORGENSTEIN, R.M., SZOSTEK, B., RATHER, P.N. 2010.
uropathogenic Proteus mirabilis. Mol. Microbiol.
Regulation of gene expression during swarmer cell
45: 865874.
differentiation in Proteus mirabilis. FEMS
LI, X., ZHAO, H., LOCKATELL, C.V., DRACHENBERG, Microbiol. Rev. 34: 753763.
C.B., JOHNSON, D.E., MOBLEY, H.L.T. 2002b.
MORYL, M. 2010. Investigations of biofilm formation
Visualization of Proteus mirabilis within the matrix
by Proteus mirabilis and its sensitivity to
of urease-induced bladder stones during
antibacterial agents. Ph.D. Thesis. University of
experimental urinary tract infection. Infect. Immun.
Lodz. (In Polish).
70: 389394.
MORYL, M., TORZEWSKA, A., RALSKI, A. 2008.
LIAW., S.J., LAI, H.C., HO, S.W., LUH, K.T., WANG,
Substances affecting Proteus mirabilis biofilm. In:
W.B. 2003. Role of RsmA in the regulation of
Conference Vaccines: Advances in plant and
swarming motility and virulence factor expression
microbial biotechnology, infectious immunity and
in Proteus mirabilis. J. Med. Microbiol. 52: 1928.
cancer therapy. Warsaw, SGGW Publisher. 179
LIAW, S.J., LAI, H.C., WANG., W.B. 2004. Modulation 181.
of swarming and virulence by fatty acids trough the
NIELUBOWICZ, G., MOBLEY, H.L.T. 2010. Host-
RsbA protein in Proteus mirabilis. Infect. Immun.
pathogen interactions in urinary tract infections.
72: 68366845.
Nature Rev. Urology 7: 430441.
LOOMES, L.M., SENIOR, B.W., KERR, M.A. 1990. A
O' HARA, C., BRENNER, F.W., MILLER, J.M. 2000.
proteolytic enzyme secreted by Proteus mirabilis
Classification, identification, and clinical
degrades immunoglobulins of the immunoglobulin
significance of Proteus, Providencia, and
A1 (IgA1), IgA2, and IgG isotypes. Infect. Immun.
Morganella. Clin. Microbiol. Rev. 13: 534546.
58: 19701985.
OKIMOTO, N., HAYASHI, T., ISHIGA, M., NANBA, F.,
UKASIEWICZ, J., UGOWSKI, C. 2003. Biological
KISHIMOTO, M., YAGI, S., KURIHARA, T., ASOKA,
activities of lipopolysaccharide. Post. Hig. Med.
N., TAMADA, S. 2010. Clinical features of Proteus
Dowiad. 57: 3353. (In Polish).
mirabilis pneumonia. J. Infect. Chemother. 16:
MALIC, W., WATTERS, M,G., BASIL, L., STICKLER, D.J., 364366.
WILLIAMS, D.M. 2011. Development of an early
PALUSIAK, A., SIDORCZYK, Z. 2009. Serological
warning sensor for encrustation of urinary
characterization of the core region of
catheters following Proteus infection. J. Biomed.
lipopolysaccarides of rough Proteus sp. strains.
Mater Res. B Appl. Biomater. DOI:
Arch. Immunol. Ther. Exp. 57: 303310.
10.1002/jbm.b.31930.
PALUSIAK, A., SIDORCZYK, Z. 2010. Characterization of
MASSAD, G., ZHAO, H., MOBLEY, H.L.T. 1995. Proteus
epitope specificity of Proteus penneri 7. ABP. 57:
mirabilis aminoacid deaminase: cloning, nucleotide
529532.
sequence, and characterization of aad. J. Bacteriol.
177: 58785883. PEARSON, M.M., MOBLEY, H.L.T. 2008. Repression
motility during fimbrial expression; identification
MIELNIK, G., DOROSZKIEWICZ, W., KORZENIOWSKA
of 14 mrpJ gene paralogues in Proteus mirabilis.
KOWAL, A. 2004. External structures of Gram-
Mol. Microbiol. 69: 548558.
negative bacteria and bactericidal activity of
complement. Post. Mikrobiol. 43: 3957. (In PERSON, M.M. et al. 2008. Complete genome sequence
Polish). of uropathogenic Proteus mirabilis, a master of
both adherence and motility. J. Bacteriol. 190:
MOBLEY, H.L.T. 1996. Virulence of Proteus mirabilis.
40274037.
In: H.L.T. MOBLEY, J.W. WARREN (eds.) Urinary
tract infections, molecular pathogenesis and clinical

PROTEUS SP. AN OPPORTUNISTIC BACTERIAL PATHOGEN 15


FOLIA BIOLOGICA ET OECOLOGICA

PHAN, V., BELAS, R., GILMORE, B.F., CERI, H. 2008. RALSKI, A., DUGOSKA, H., KOTEKO, K. 1986.
ZapA, a virulence factor in a rat model of Proteus Cell invasiveness of Proteus mirabilis and Proteus
mirabilis-induced acute and chronic prostatitis. vulgaris strains. Arch. Immunol. Ther. Exp. 34:
Infect. Immun. 76: 48594864. 505511.
PIECHOTA, J., PRYWER, J., TORZEWSKA, A. 2012. Ab RALSKI, A., KOTEKO, K. 1987. Hemolytic activity
initio prediction of structural and elastic properties and invasiveness in strains of Proteus penneri. J.
of struvite: contribution to urinary stone research. Clin. Microbiol. 25: 10941096.
Comput. Meth. Biochem. Engin. 15: 13291336.
RALSKI, A., SIDORCZYK, Z., KOTEKO, K. 1997.
PRYWER, J., TORZEWSKA, A. 2009. Bacterially induced Potential virulence factors of Proteus bacilli.
struvite growth from synthetic urine: experimental Microbiol. Mol. Biol. Rev. 61: 6589.
and theoretical characterization of crystal
RALSKI, A., STCZEK, P. 2010. Proteus. In: D. LIU,
morphology. Cryst. Growth Design. 9: 35383543.
(ed.) Molecular detection of foodborne pathogens.
PRYWER, J., TORZEWSKA, A. 2010. Biomineralization CRS Press, Taylor and Francis Group. Boca Raton,
of struvite crystals by Proteus mirabilis from p. 417430.
artificial urine and their mesoscopic structure.
RALSKI, A., STCZEK, P. 2011. Proteus. In:
Cryst. Res. Technol. 45: 12831289.
Molecular detection of human bacterial pathogens.
PRYWER, J., TORZEWSKA, A. 2012. Effect of curcumin In: D. Liu (ed.), CRC Press, Taylor and Francis
against Proteus mirabilis during crystallization of Group. Boca Raton, p. 981996.
struvite form artificial urine. Evidence Base
RALSKI, A., TORZEWSKA, A., BARTODZIEJSKA B.,
Complementary and Alternative Medicin. 862794:
BABICKA D., KWIL, I., PEREPELOV, A.V.,
17.
KONDAKOVA, A.N., SENCHENKOVA, S.N., KNIREL,
RADZIEJEWSKA-LEBRECHT, J., MAYER, H. 1989. The Y.A., VINOGRADOV, E.V. 2002. Chemical structure,
core region of Proteus mirabilis R110/1959 antigenic specificity, the role in the pathogenicity of
lipopolysaccharide. Eur. J. Biochem. 183: 573581. lipopolysaccharide (LPS, endotoxin) one the
Proteus vulgaris bacterias example. Wiadomoci
RAETZ, C.R., WHITFIELD, C. 2002.
Chemiczne. 56: 585604. (In Polish).
LIPOPOLYSACCHARIDE ENDOTOXIN. ANN. REV.
BIOCHEM. 71: 635700. SCAVONE, P., UMPIERREZ, A., MASKELL, D.J., ZUNINO,
P. 2011. Nasal immunization with attenuated
RATHER, P.N. 2005. Swarmer cell differentiation in
Salmonella Thyphimurium expressing and MrpA-
Proteus mirabilis. Environm. Microbiol. 7: 1065
TetC fusion protein significantly reduces Proteus
1073.
mirabilis colonization in mouse urinary tract. J.
ROCHA, S.P.D., ELIAS, W.P., CIANCIARULLO, A.M., Med. Microbiol. 60: 899904.
MENEZES M.A., NARA, J.M., PIAZZA, R.F.M.,
SCHLAPP, G., SCAVONE, P., ZUNINO, P., HARTEL, S.
SILVA, M.R.L., MOREIRA, C.G., PELAYO, J.S.
2011. Development of 3D architecture of
2007a. Aggregative adherence of uropathogenic
uropathogenic Proteus mirabilis bath culture
Proteus mirabilis to cultured epithelial cells. FEMS
biofilms A quantitative confocal microscopy
Immunol. Med. Microbiol. 51: 319326.
approach. J. Microbiol. Meth. 87: 234240.
ROCHA, S.P.D., ELIAS, W.P., PELAYO, J.S. 2007b.
SIDORCZYK, Z., ZAHRINGER, U., RIETSCHEL, E.T. 1983.
Fimbriae of uropathogenic Proteus mirabilis.
Chemical structure of the lipid A component of the
FEMS Immunol. Med. Microbiol. 51: 17.
lipopolysacharide of Proteus mirabilis Re mutant.
ROMANOWSKI, Z., KEMPISTY, P., PRYWER, J., Eur. J. Biochem. 137: 1522.
KRUKOWSKI, S., TORZEWSKA A. 2010. Density
SOSA, V., ZUNINO, P. 2009. Effect of Ibicella lutea on
functional theory determination of structural and
uropathogenic Proteus mirabilis growth, virulence
electronic properties of struvite: J. Phys. Chem.
and biofilm formation. J. Infect. Dev. Ctries. 3:
114: 78007808.
762770.
RALSKI, A. 2002. Molecular basis of the
STAKOWSKA, D., CZERWONKA, G., RALSKA, S.,
pathogenicity of Proteus bacteria. Adv. Clin. Exp.
GROSICKA, M., DZIADEK, J., KACA, W. 2012.
Med. 11: 318.
Influence of quorum sensing signal molecules on
RALSKI, A. 2004. Lipopolysaccharide and others biofilm formation in Proteus mirabilis O18. Folia
virulence factors of Proteus bacteria. Post. Microbiol. 57: 5360.
Mikrobiol. 43: 409431. (In Polish).
STAKOWSKA, D., KWINKOWSKI, M., KACA, W. 2008.
RALSKI, A. 2008. Lipopolysaccharide (LPS, Quantification of Proteus mirabilis virulence
endotoxin) of Proteus bacteria chemical structure, factors and modulation by acylated homoserine
serological specificity and the role in pathogenicity. lactones. J. Microbiol. Immunol. Infect. 41: 243
Folia Biolog. Oecolog. 4: 524. 253.

RALSKI ET AL. 16
FOLIA BIOLOGICA ET OECOLOGICA

STICKLER, D.J., 2008. Bacterial biofilms in patients MOBLEY, J.W. WARREN (eds.) Urinary tract
with indwelling urinary catheters. Nature Clin. infections, molecular pathogenesis and clinical
Pract. Urology. 5: 598607. management. ASM Press, Washington DC, p. 228.
STICKLER, D.J., FENELEY, R.C.L. 2010. The WRAY, S.K., HULL, S.I., COOK, R.G., BARRISH, J.,
encrustation and blockage of long-term indwelling HULL, R.A. 1986. Identification and
bladder catheters: a way forward in prevention and characterization of a uroepithelial cell adhesion
control. Spinal Cord. 48: 784790. from a uropathogenic isolate of Proteus mirabilis.
Infect. Immun. 54: 4349.
SWIHART, K.G., WELCH, R.A. 1990. Cytotoxic activity
of Proteus hemolysin HpmA. Infect. Immun. 58: ZUNINO, P., GEYMONT, L., ALLEN, A.G., LEGNANI-
18611869. FAJARDO, C., MASKELL, D.J. 2000. Virulence of
Proteus mirabilis atf isogenic mutant is not
TORZEWSKA, A., RALSKI, A. 2009. Studies on
impaired in a mouse model ascending urinary tract
infection-induced urinary calculi formation in the
infection. FEMS Immunol. Med. Microbiol. 29:
presence of glycosaminoglycans. X Jubilee
137143.
Conference "Molecular biology in diagnostic of
infectious diseases and biotechnology, Warsaw ZUNINO, P., PICCINI, C., LEGNIANI-FAJARDO, C. 1994.
University of Life Sciences - SGGW, 2009. Flagellate and non flagellate Proteus mirabilis in
Scientific materials, 179182. the development of experimental urinary tract
infection. Microb. Path. 16: 379385.
TORZEWSKA, A., STCZEK, P., RALSKI, A. 2003. The
crystallization of urine mineral components may ZUNINO, P., SOSA, V., SHLAPP, G., ALLEN, A.G.,
depend on the chemical nature of Proteus PRESTON A., MASKELL, D.J. 2007. Mannose-
endotoxin polysaccharides. J. Medical. Microbiol. resistant Proteus-like and Proteus mirabilis
52: 471477. fimbriae have specific and additive roles in
P. mirabilis urinary tract infections. FEMS
TORZEWSKA, A., ZIELISKA, E., WIDER, S., RALSKI,
Immunol. Med. Microbiol. 51: 125133.
A. 2010. Influence of curcumin on Proteus
mirabilis pathogenicity. Sepsis. 4: 276277.
UPHOFF, T.S., WELCH, R.A. 1990. Nucleotide
sequencing of the Proteus mirabilis calcium
independent hemolysin genes (hpmA and hpmB)
reveals sequence similarity with Serratia
marcescens hemolysin genes (shlA and shlB). J.
Bacteriol. 172: 12061216.
VERSTRAETEN, N., BREKANEN, K., DEBKUMARI, B.,
FAUVART, M., FRANSAER, J., VERMANT J.,
MICHIELS, J. 2008. Living on a surface: swarming
and biofilm formation. Trends Microbiol. 16: 496
506.
VINOGRADOV, E. 2011. Structure of the core part of the
lipopolysaccharide form Proteus mirabilis genomic
strain HI4320. Biochemistry (Moscow). 76: 803
807.
VINOGRADOV, E.V., SIDORCZYK, Z., KNIREL, Y.A.
2002. Structure of lipopolysaccharide core region of
the genus Proteus. Aust. J. Chem. 55: 6167.
VINOGRADOV, E.V., THOMAS-OATES, E., BRADE, H.,
HOLST, O. 1994. Structural investigations of the
lipopolysaccharide from Proteus mirabilis R45
(Re-chemotype). J. End. Res. 1: 199206.
WANG, Q., TORZEWSKA, A., RUAN, X., WANG, X.,
RALSKI, A., SHAO, Z., GUO, X., FENG, L., WANG,
L. 2010. Molecular and genetic analyses of the
putative Proteus O-antigen gene locus. App. Env.
Microbiol. 76: 54715480.
WARREN, J.W. 1996. Clinical presentations and
epidemiology of urinary tract infections. In: H.L.T.

PROTEUS SP. AN OPPORTUNISTIC BACTERIAL PATHOGEN 17

You might also like