Médecine et maladies infectieuses 36 (2006) 78–91
http://france.elsevier.com/direct/MEDMAL/
General review
Targeting mechanisms of Pseudomonas aeruginosa pathogenesis
Thérapeutiques ciblant les mécanismes
pathogéniques de Pseudomonas aeruginosa
E. Kipnis *, T. Sawa, J. Wiener-Kronish
Department of Anesthesia and Perioperative Care, University of California San Francisco, 513 Parnassus Avenue, Room s-261,
Medical Science Building, Box 0542, San Francisco, CA 94143, USA
Received and accepted 18 October 2005
Available online 19 January 2006
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen responsible for ventilator-acquired pneumonia, acute lower respiratory tract infections
in immunocompromised patients and chronic respiratory infections in cystic fibrosis patients. High incidence, infection severity and increasing
resistance characterize P. aeruginosa infections, highlighting the need for new therapeutic options. One such option is to target the many pathogenic mechanisms conferred to P. aeruginosa by its large genome encoding many different virulence factors. This article reviews the pathogenic
mechanisms and potential therapies targeting these mechanisms in P. aeruginosa respiratory infections.
© 2006 Elsevier SAS. All rights reserved.
Résumé
Pseudomonas aeruginosa est un pathogène opportuniste responsable de pneumonies nosocomiales, infections des voies respiratoires basses
chez les patients immunodéprimés et infections respiratoires chroniques lors de la mucoviscidose. L’incidence élevée, la sévérité et la résistance
croissante sont caractéristiques des infections à P. aeruginosa et soulignent le besoin d’ouvrir de nouvelles voies thérapeutiques. Une voie
thérapeutique possible est de cibler un des nombreux mécanismes pathogéniques dont est doté P. aeruginosa grâce à son génome de grande
taille codant pour de nombreux facteurs de virulence. Nous exposons les différents mécanismes pathogéniques au cours des infections respiratoires à P. aeruginosa ainsi que les voies thérapeutiques potentielles qui les ciblent.
© 2006 Elsevier SAS. All rights reserved.
Keywords: Pseudomonas aeruginosa; Pathogenicity; Virulence factors
Mots clés : Pseudomonas aeruginosa ; Pathogénicité ; Facteurs de virulence
1. Introduction
tance of P. aeruginosa to conventional antimicrobial treatment
has increased over the past decade [5,6].
Pseudomonas aeruginosa is the most common pathogen responsible for both acute respiratory infections in ventilated or
immunocompromised patients and chronic respiratory infections in cystic fibrosis patients [1,2]. P. aeruginosa is also responsible for excessive mortality in VAP [1,3,4]. Adding to the
problems of high incidence and infection severity, the resis-
These three problems of high incidence, severity and resistance persist even though they are now broadly recognized and
various strategies have been proposed addressing them [1,7–12].
*
Corresponding author. Tel.: +1 415 476 1653.
E-mail address: ekipnis@gmail.com (E. Kipnis).
0399-077X/$ - see front matter © 2006 Elsevier SAS. All rights reserved.
doi:10.1016/j.medmal.2005.10.007
Therefore, it is crucial that new therapeutic options for
P. aeruginosa infections be explored. Such new options may
come from specifically targeting the pathogenic mechanisms
of P. aeruginosa. Indeed, P. aeruginosa is a remarkable pathogen in that it is endowed with a uniquely large genome containing genes for many different virulence factors and regulatory mechanisms allowing it to adapt to hostile environments.
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
79
This article reviews the pathogenic mechanisms and potential
therapies targeting these mechanisms in P. aeruginosa respiratory infections.
3. Bacterial cell surface virulence factors (Fig. 1)
2. Schematic overview of P. aeruginosa pathogenesis
in respiratory infections
Flagella are complex proteic structures forming a filamentous polar appendage at the surface of P. aeruginosa. Flagella
are the main motile appendage of gram negative bacteria and
allow the swimming movement of P. aeruginosa through a
propeller or screw-like motion. Flagella have a critical role in
pathogenesis by tethering and adhering to epithelial cells
through binding with a common membrane component, asialoGM1 [13]. They also participate in virulence and elicit an
NFkB dependent inflammatory response through interactions
with Toll-receptors TLR5 and TLR2 and a calcium entry dependent activation of the extracellular regulated kinase pathway leading to IL-8 production [14,15]. However, flagella are
also very immunogenic, rendering their presence a liability for
P. aeruginosa after successful colonization. This is why
P. aeruginosa is capable of adapting by selecting aflagellar mutants to evade host response in chronic infections [16]. It is
therefore not surprising that flagella have been considered interesting targets for immunotherapy. P. aeruginosa pneumonia
was attenuated in rats receiving human antiflagellar monoclonal antibodies [17]. Similar findings led to the development of
a P. aeruginosa flagella vaccine for cystic fibrosis patients that
successfully completed phase I and II trials and is undergoing
phase III evaluation [18,19].
P. aeruginosa is an opportunistic pathogen that, after being
acquired from the environment, colonizes the respiratory
epithelium in patients with predisposing conditions such as
cystic fibrosis, mechanical ventilation, immunodeficiency or
preexisting respiratory disease. Flagella and pili, the motile surface appendages of P. aeruginosa are responsible for bacterial
motility and progression towards epithelial contact. These appendages also act as initial tethers in facilitating bacteria to
epithelial cell contact by binding to the epithelial surface glycolipid asialo-GM1. Additionally, lipopolysaccharide (LPS)
also plays a similar role in bacterial adhesion through asialoGM1 binding. These appendages then play a major role in the
irreversible adhesion to epithelial cells, which is the initial critical step in colonization of the respiratory epithelium. Upon
cell contact, the type III secretion system, a major virulence
determinant, is activated. The type III secretion system allows
P. aeruginosa to inject secreted toxins through a syringe-like
apparatus directly into the eukaryotic cytoplasm. Four effector
proteins are known: ExoY, ExoS, ExoT, and ExoU and all
participate, at varying levels, in the cytotoxicity of
P. aeruginosa leading to invasion and dissemination of
P. aeruginosa. Other virulence factors secreted via type II secretion system into the extracellular space such as elastase, alkaline phosphatase, exotoxin A, and phospholipase C also participate in invasion by destroying the protective glycocalix of
the respiratory epithelium and exposing epithelial ligands to
P. aeruginosa. These secretins also participate in cytotoxicity.
A similar role also exists for pyoverdine and pyocyanin.
In acute infections, invasion, dissemination and extensive
tissue damage predominate. However, in chronic infections,
particularly in cystic fibrosis patients, P. aeruginosa may also
adapt, by losing its most immunogenic features such as pili and
flagella to avoid clearance, and by isolating itself from host
defenses and adhering to the respiratory epithelium by forming
biofilms. In chronic infections, a persistent inflammatory state
is maintained by extracellular secreted virulence factors.
Whether in acute or chronic infections, P. aeruginosa possesses a multiplicity of regulating systems allowing it to adapt
to its environment and notably to host defenses. Among these
systems, quorum-sensing (QS) showcases P. aeruginosa adaptability. Quorum sensing systems are complex bacterial cell-tocell signaling systems that allow the bacteria to sense their own
cell density and to communicate with each other resulting in
coordinated production of virulence factors depending on bacterial density. QS has been shown to be critical to maintaining
airway inflammation through virulence factor production and
to the formation of biofilm in chronic infections.
This schematic overview shows that there are many levels
in the pathogenesis of P. aeruginosa infections that may represent therapeutic targets, which we will now describe in detail.
3.1. Flagella
3.2. Pili
Pili or fimbriae are smaller filamentous surface appendages
of P. aeruginosa. Multiple pili are usually present on the surface. P. aeruginosa pili are among the rare prokaryotic pili involved in bacterial motility. This motility, called twitching is
due to the retractile properties of P. aeruginosa pili and allows
P. aeruginosa to “spread” along hydrated surfaces rather than
“swim” [20]. This feature facilitates the rapid colonization of
the airway [20]. Like flagella, pili are crucial to the adhesion
phase of colonization through binding to asialoGM1 of the
epithelial cell membrane [21,22]. Furthermore, studies have
shown that both pili-mediated adherence and twitching motility
are critical to P. aeruginosa virulence [23,24]. In an infant
mouse model of lung infection, piliated strains of
P. aeruginosa caused more severe and diffuse pneumonia than
corresponding non-piliated mutants [25]. Therefore, pili, like
flagella seem to be legitimate targets in developing antipseudomonal immunotherapy [26–28]. Among such strategies, a chimeric vaccine incorporating both pilin and non-toxic modified
exotoxin A successfully reduced bacterial adherence [29].
However, there are issues linked to the lack of cross reactivity
of antigens targeting pili across different P. aeruginosa strains
that need to be resolved [26,28].
3.3. LPS
Although the inner face of the outer membrane resembles a
typical phospholipid bilayer, the outer face of the outer mem-
80
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
Fig. 1. Overview of P. aeruginosa cell-surface virulence factors.
Fig. 1. Schéma des facteurs de virulence de surface cellulaire de P. aeruginosa.
After reaching the respiratory epithelium through flagellum-dependent “swimming” and spreading along the epithelium through pili-dependent “twitching”
P. aeruginosa is tethered to respiratory epithelial cells through binding of flagellum and pili to asialoGM1. Further adherence is conferred by interactions between
LPS and asialoGM1, CD14 and/or CFTR. Flagellar interaction with TLR5 activates a Calcium entry-dependent MAP-kinase pathway leading to increased IL-8
transcription. Both flagellar interaction with TLR4 and LPS interaction with TLR4/MD2 activate MyD88-dependent NFκB pathways leading to increased IL-8
transcription. In cystic fibrosis patients, alginate increases adhesion and through an unknown mechanism depresses host response.
LPS: lipopolysaccharide, asialoGM1: asialoganglioside gangliotetraosylceramide, CD14: cluster of differentiation receptor 14, CFTR: cystic fibrosis transmembrane
conductance regulator, TLR: toll-like receptor, MAP-kinase: mitogen-activated protein kinase, IL-8: interleukin 8, PKC: protein kinase C, c-raf: src/ras-dependent
kinase, MEK1/2: dual specificity MAP kinase/extracellular signal-regulated kinase kinase, ERK 1/2: dual specificity extracellular signal-regulated kinase, PKR:
protein kinase regulated by RNA, IKK: I-kappa-B kinase, NFκB: nuclear factor kappa B, MyD88: myeloid differentiation primary response 88, TRAF6: tumor
necrosis factor receptor-associated factor 6, IRAK: IL-1 receptor-associated kinase.
brane is mainly composed of LPS. LPS associates a hydrophobic domain, Lipid A inserted into the phospholipid bilayer to a
hydrophilic tail composed of the core polysaccharide and the
O-specific polysaccharide.
The variable O-specific polysaccharide chains are the basis
of antigenic identification of P. aeruginosa serotypes. This immunogenicity makes them obvious targets for immunotherapy.
However, the active immunization elicited by O-antigen based
vaccines is lacking in protectiveness even when multiple Oantigens from different serotypes are conjugated [30–32].
Thus, various O-antigen based vaccines have been tested over
decades with limited success [32]. To circumvent this problem
several strategies have been developed. Multiple serotype conjugates can be further conjugated with another target such as
exotoxin A [30]. Such a vaccine has successfully completed
phase I and II trials and is currently undergoing a phase III trial
[30,33]. Equally promising, live attenuated vaccines from deletional mutants of P. aeruginosa or Salmonella enteritica expressing P. aeruginosa O-antigens have been developed and
confer protection in animal models [34–36].
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
Passive immunization therapies against O-specific polysaccharides have been explored for many years [37]. The main
obstacle of administering monoclonal antibodies mainly obtained from mice to humans has been recently overcome with
the advent of transgenic mice producing human monoclonal
antibodies protective against P. aeruginosa [38,39]. Similarly,
human hybridoma derived monoclonal antibodies are also under development against P. aeruginosa [30].
Other strategies have consisted in the use of synthetic antimicrobial peptides known to inhibit LPS such as CAP-18.
Although CAP-18 was a potent in vitro antipseudomonal, in
vivo administration did not improve survival [40].
LPS is a substance biologically active to the extent that it is
used in many models of sepsis or acute lung injury even
though it has recently been shown that its potency in mimicking acute lung injury depends mostly on preexisting damage
[41] and on structure. Indeed, the Lipid A component of
P. aeruginosa LPS activates multiple pro-inflammatory pathways [42–44]. LPS is also critical to virulence [45], mainly
by its role in adhesion through asialoGM1 binding [22],
TLR4/CD14 or TLR4/MD-2 recognition [43,44] or binding to
CFTR [46]. In cystic fibrosis patients, P. aeruginosa adapts by
selecting mutants with specific Lipid A modifications that allow resistance to host antimicrobial peptides and increase
TLR4 activation [44,47,48].
Some immunotherapeutic strategies target Lipid-A,
although their activity is also based on O-antigen reactivity
[49]. However, new strategies specifically target Lipid-A because of its role in pathogenesis and not for recognition by
immune-based methods. These strategies aim to inhibit the first
or second step in synthesis of Lipid-A [50,51]. However, these
inhibitors have not been tested in P. aeruginosa infections.
3.4. Alginate
Alginate is a mucoid exopolysaccharide produced by
P. aeruginosa that is made up of repeating polymers of mannuronic and glucuronic acid. Alginate, like LPS, functions as
an adhesin, anchoring P. aeruginosa to the colonized respiratory epithelium. Surrounding conditions in cystic fibrosis patient’s lungs and host inflammatory response increase alginate
synthesis leading to conversion to an alginate overproducing
mucoid phenotype [52,53]. This mucoid alginate-producing
phenotype is commonly found in cystic fibrosis airways over
the course of P. aeruginosa infections. Overexpressed alginate
protects P. aeruginosa from phagocytosis, antibiotics and even
attenuates the host response [54,55]. Although alginate has
been widely considered to participate in the architecture of
P. aeruginosa biofilm in cystic fibrosis [56] and probably does
have a role in some of the properties of biofilm [54], it has
been recently shown that alginate is not crucial to biofilm development [57].
However, because of this excessive production in cystic fibrosis, alginate has been a recent target in immunotherapy [58,
59]. Human monoclonal antibodies directed against alginate
were effective in a murine model of lethal pneumonia and sur-
81
prisingly even against non-mucoid strains producing undetectable levels of alginate [59]. However, active immunization
against alginate is in the process of being optimized, particularly through conjugation with exoA [58], in order to compensate for the variable efficacy of antibodies produced in response to alginate [60].
4. Secreted virulence factors (Fig. 2)
4.1. Pyocyanin
Pyocyanin is a blue pigment metabolite of P. aeruginosa
that has been shown to have numerous pathogenic effects such
as increasing IL-8 [61,62], depressing host-response [61,63],
and inducing apoptosis in neutrophils [63]. In animal models
of acute and chronic lung infection, pyocyanin was shown to
be essential to P. aeruginosa virulence [64]. The same study
also showed that pyocyanin instillation caused an influx of
neutrophils into the lung. Additionally, due to it’s known oxidoreductive properties, pyocyanin oxidizes glutathione and inactivates catalase in respiratory epithelial cells thus participating in oxidative-stress related damage [65,66]. Furthermore, in
yeast cells, pyocyanin disrupts vacuolar ATPase and mitochondrial electron transport that may impair CFTR chloride channels in cystic fibrosis [67].
Although no therapeutic strategies directly target pyocyanin,
several antioxidant therapies have proved useful in cystic fibrosis [68–70]. Glutathione delivery through aerosolization improved FEV in cystic fibrosis patients [68,70]. Lung function
was improved in cystic fibrosis patients after an 8-week antioxidant supplemented regimen [68].
Recently, authors have suggested inhibiting the synthesis of
pyocyanin as a therapeutic strategy, although such studies have
yet to be conducted [67,71].
4.2. Pyoverdine
Pyoverdine is a siderophore, a small molecule chelating iron
from the environment for use in P. aeruginosa metabolism.
Pyoverdine has also been shown to play a role in
P. aeruginosa virulence [72,73]. One explanation for this role
has recently emerged when it was found that pyoverdine regulates the secretion of other P. aeruginosa virulence factors,
exotoxin A and an endoprotease and its own secretion [74].
Although there are yet no therapeutic strategies aimed at
pyoverdine, this virulence regulating function that doubles as
a cell-to-cell signaling mechanism makes it an attractive target.
4.3. Alkaline protease
Alkaline protease is a fibrin lysing protease secreted by
P. aeruginosa through a type I secretion system [75]. Although
its pathogenic role is only clear in corneal infections as is the
case for most P. aeruginosa proteases [75], it may participate in
the pathogenesis of acute lung injury. Indeed, it has been shown
that there is an early massive intra-alveolar formation of fibrin in
82
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
Fig. 2. Overview of secreted P. aeruginosa virulence factors.
Fig. 2. Schéma des facteurs sécrétoires de virulence de P. aeruginosa.
Pyocyanin, an oxidative reactive pigment increases reactive oxygen species (ROS) within host cells and impairs host cell antioxydant capacities by decreasing
glutathione stock catalase activity. Pyocyanin also increases production of IL-8. Type I secretion system secreted alkaline phosphatase lyses the fibrin matrix limiting
acute lung injury. Elastase disrupts epithelial tight junctions, cleaves surfactant proteins A and D, inactivates proteinase-activated receptor 2 and increases production
of IL-8. Phospholipase C lyses mebrane phospholipids and inactivates surfactant. Exotoxin A (ExoA) inhibits elongation factor 2 (EF2) leading to transcriptional
arrest. Pyoverdin increases the secretion of ExoA and itself.
P. aeruginosa acute lung injury and that inhibition of this initial
fibrinoformation is deleterious in an animal model [76]. Furthermore, the efficacy of passive and active immunization therapies
targeting alkaline protease in a murine model of sepsis shows
that alkaline protease may have a larger role in P. aeruginosa
pathogenesis and is thus a legitimate target [77–79].
P. aeruginosa keratitis [80], it has only recently been established that protease IV is also involved in the pathogenesis of
lung infection through degradation of surfactant proteins A, D
and B [81]. This may indicate that protease IV could be a new
therapeutic target.
4.5. Elastase
4.4. Protease IV
Other proteases secreted by P. aeruginosa such as protease
IV also have a role in pathogenesis. Although, protease IV is
particularly known to participate in the pathogenesis of
Elastase, or lasB, is a metalloproteinase secreted by
P. aeruginosa into the extracellular space through a type II secretion system. Elastase has been shown to have a role in the
pathogenesis of P. aeruginosa respiratory infections by ruptur-
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
ing the respiratory epithelium through tight-junction destruction, thus increasing epithelial permeability and facilitating
neutrophil recruitment [82–84]. Elastase is also pro-inflammatory, increasing IL-8 levels in a rat air pouch inflammation
model [85].
P. aeruginosa elastase can also decrease host immune response through cleavage of respiratory tract surfactant proteins
A and D and Proteinase-activated receptor 2 into inactive
forms [86–88].
Elastase is a candidate for immune based targeting of
P. aeruginosa as attested by the efficacy of immunization with
synthetic epitopes of elastase in decreasing the severity of infection in rats [89]. Likewise, monoclonal antibodies directed
against P. aeruginosa elastase decreased epithelial permeability
in a monolayer of polarized respiratory epithelial cells [90].
The inhibition of elastase secreted by P. aeruginosa with
protease inhibitors could be a potential therapeutic option.
However, current protease inhibitor therapeutic strategies are
aimed at the more overwhelming secretion of elastase by inflammatory neutrophils and have been shown to be effective
in rat models of chronic respiratory infection [91,92].
4.6. Phospholipase C
Phospholipase C, more specifically hemolytic phospholipase C, is a phospholipase secreted by P. aeruginosa into the
extracellular space through a type II secretion system. Hemolytic phospholipase C targets eukaryotic membrane phospholipids and has been shown to participate in the pathogenesis of
P. aeruginosa acute lung injury [93] and in inflammation [94,
95]. Like elastase, part of the pathogenic effect of hemolytic
phospholipase C may be due to surfactant inactivation [96].
Furthermore, hemolytic phospholipase can suppress the host
neutrophil oxidative burst response [97]. However, no therapeutic strategy to date specifically targets phospholipase C
mediated pathogenicity although it is interesting to note that
various antipseudomonal antibiotics can decrease the secretion
of phospholipase C in a clinical isolate of P. aeruginosa [98].
4.7. Exotoxin A
Exotoxin A (ExoA), secreted into the extracellular space
through a type II secretion system is an ADP-ribosyl transferase [99] inhibiting elongation factor-2 (EF-2) thereby inhibiting protein synthesis and leading to cell death [100]. ExoA has
also been shown to depress host response to infection [101,
102]. Among extracellular toxins, ExoA has a major role in
P. aeruginosa virulence [103] although to a lesser extent than
type III secretion system effectors such as ExoS [104]. Indeed,
an ExoA deficient mutant was 20 times less virulent than the
wild strain in mice [103].
Passive immunization targeting ExoA increased survival in
a murine model of P. aeruginosa infection [105] and in a murine model of gut-derived sepsis [77]. Because of these immunogenetic properties, non-toxic altered ExoA has been used
particularly successfully as a target for multivalent immu-
83
notherapy in chimeric vaccine with outer membrane proteins
I and F [106] or pilin protein [29]. An alternative to using altered ExoA to avoid toxicity has been immunization with
ExoA-coding DNA, successfully tested in animals [107,108].
5. Type III secretion system (Fig. 3)
Type III secretion systems are shared among Yersinia, Salmonella, Shigella and Pseudomonas species as a mechanism to
directly inject toxins into the host cells. The type III secretion
system (TTSS) of P. aeruginosa is a complex pilus-like structure allowing the translocation of effector proteins from the
bacteria, across the bacterial membranes and into the eukaryotic cytoplasm through a needle-like appendage forming a pore
in the eukaryotic membrane [109]. There are four known toxins, variably expressed in different strains and isolates, injected
into host cells by P. aeruginosa through the TTSS: ExoY,
ExoS, ExoT and ExoU.
5.1. ExoS
ExoS is a bifunctional cytotoxin with two active domains, a
C-terminal ADP-ribosyltranferase domain and an N-terminal
Rho GTPase-activating protein (GAP) domain. The ADP-ribosyltransferase activity necessitates a eukaryotic cell cofactor:
14-3-3 protein [110]. The pathogenic role of ExoS is mainly
attributable to the ADP-ribosyltranferase activity leading to
disruption of normal cytoskeletal organization [111,112],
although GAP activity also plays a similar role [111,113]. Additionally, it has recently been shown that the C-terminal domain binds to TLR2 and the N-terminal domain binds to
TLR4, showing that ExoS may also modulate the host immune
and inflammatory response [114]. Although it is generally recognized that ExoS is less cytotoxic than ExoU [111,115],
their secretion is mutually exclusive in wild strains of
P. aeruginosa, implying that ExoS is still a major cytotoxin
in certain strains [115,116]. Furthermore, the relative risk of
mortality increased to 8.7 in patients who were infected with
a strain expressing a functional TTSS and secreting either
ExoS or ExoU [117] underlining the importance of this toxin
in the pathogenesis of P. aeruginosa infection. However, there
are no therapeutic strategies that target the toxin in itself.
5.2. ExoT
ExoT is similar to ExoS, with dual ADP-ribosyltranferase
and GAP activities, although the ExoT ADP-ribosyltranferase,
initially thought to be deficient compared to that of ExoS, targets different pathways [118–122]. Although, ExoT has similar
effects on the eukaryotic cytoskeleton as ExoS, and has been
shown to inhibit both Pseudomonas internalization and wound
repair [123,124], it is considered only a minor cytotoxin [111].
Surprisingly, ExoT was even found to diminish the cytotoxicity due to ExoU in a specific strain [115]. Therefore, unless
this protective effect yields a specific therapeutic pathway,
84
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
Fig. 3. Overview of P. aeruginosa type three secretion system.
Fig. 3. Schéma du système sécrétoire de type III de P. aeruginosa.
The type 3 secretion system is constituted of a secretion apparatus(PscC proteins) and a translocator or “needle” (PopD, PopB, PcrV proteins) allowing the
“injection” of Exotoxins (ExoY, ExoS, ExoT and ExoU) into the host cell cytoplasm. ExoY is an adenylate cyclase that causes an increase in cytoplasmic camp
leading to increased epithelial barrier permeability. Exo S and Exo T both have N-terminal GTPase activating protein (GAP) domains that inactivate actin
polymerization disrupting the cytoskeleton. They also have C-terminal ADP-ribosyltransferase (ADPRT) activities on different substrates. ExoS ADPRT, in the
presence of 14-3-3 protein, inhibits the interaction of Ezrin-Radixin-Moesin (E, R and M) and cytoskeletal proteins. ExoT ADPRT inhibits the Rac/integrin
cytoskeletal wound healing pathway. ExoU, a phospholipase/ lysophospholipase destroys epithelial membranes in the presence of an unknown host cell cofactor.
ExoT in itself does not seem to be an interesting candidate in
targeting P. aeruginosa pathogenesis.
[115]. Therefore, ExoY could be a potential, albeit minor, therapeutic target.
5.3. ExoY
5.4. ExoU
ExoY is an adenylate cyclase [125] injected directly into the
host cytosol by the TTSS and increases cytosolic cAMP, enhanced by a eukaryotic cofactor. This increased cytosolic
cAMP leads to increased pulmonary microvascular intercellular gap formation and increased lung permeability [126]. However, this role of ExoY in overall P. aeruginosa pathogenesis
may need to be explored further as most in vitro and in vivo
models of cytotoxicity only show a minor effect of ExoY
ExoU was recently found to have a phospholipase/lysophospholipase activity disrupting eukaryotic cell membranes after
translocation into the cell by the TTSS and activation by a yet
unknown eukaryotic cofactor [127–129]. ExoU is widely recognized as being the major cytotoxin secreted via the TTSS,
even though a recent study has shown that the cumulative
ExoU-independent/TTSS-mediated pathogenicity is far from
minor [130]. ExoU is 100 times more cytotoxic than ExoS
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
[115] and ExoU secretion alone by the TTSS confers cytotoxicity in animal models of lung injury leading to sepsis [111,131,
132]. Furthermore, ExoU is responsible for decompartmentalization of the inflammatory response in a model of acute lung
injury leading to sepsis [133]. In patients infected with a strain
expressing ExoU the relative risk of mortality increased to 2.3
[117]. In P. aeruginosa-induced ventilator associated pneumonia, ExoU was secreted by isolates from more severely ill patients [134]. Because ExoU plays such a major role in the
pathogenesis of P. aeruginosa infection, it represents a very
attractive therapeutic target. Indeed, anti-ExoU immunotherapy
has been explored but deemed disappointing. Immunization of
mice with recombinant ExoU only conferred 80% survival
against PA103 intratracheal challenge compared to 100% survival with recombinant PcrV [135]. Likewise, passive immunization with Anti-ExoU IgG only allowed 50% survival.
Although we are just beginning to understand the mechanism
of ExoU cytotoxicity, phospholipase A2 inhibitors have already successfully been used in vitro [136]. Additionally, elucidating the mechanism of activation of ExoU by unknown
eukaryotic cofactors could present new therapeutic options.
As we have seen, the TTSS is involved in pathogenesis
through the translocation of multiple toxins into host cells,
however, it also appears that it may participate in
P. aeruginosa virulence in itself. Indeed, mutants expressing
the TTSS but not the toxins are cytotoxic [115]. This cytotoxicity imputable to the TTSS apparatus itself may explain, in
part, why ExoU-independent pathogenicity as a whole is quite
major [130] although ExoS is considered moderately cytotoxic
and ExoT and ExoY are considered minor cytotoxins. Therefore, the TTSS is a major cancdidate for pathogenesis-targeting
therapeutics both because it translocates cytotoxins into the eukaryotic cell and because the apparatus itself may play a part in
virulence.
Several proteins composing this system have been studied,
among which PcrV is a probable structural protein of the translocation structure, or “needle”, of the TTSS. PcrV plays a role
in the assembly of the translocation pore in the eukaryotic cell
membrane [137]. Early studies showed 100% survival in mice
receiving either active or passive anti-PcrV immunotherapy before an intratracheal instillation of PA103 [135]. Subsequent
studies have continued to prove the efficacy of anti-PcrV immunotherapy in animal models of P. aeruginosa infection and
monoclonal antibodies have been successfully generated [138–
141].
85
sion in an entire bacterial population. This coordinated gene
expression concerns survival genes and more importantly,
genes coding for virulence factors and biofilm formation. The
discovery of these systems has created an entire field of research and several in-depth reviews are available [142,143].
For our purposes, it is sufficient to know that there are two
such QS systems in P. aeruginosa, las and rhl that interact in
a hierarchical manner, the first activating the second and both
following similar overall pathways (Fig. 4). Briefly, a gene encoding an autoinducer synthase (“I” genes, lasI or rhlI) is activated, the synthesized autoinducer (Oxododecanyl or oxohexanoyl-homoserine lactone) diffuses into the environment,
reaches a threshold concentration allowing it to bind to transcriptional activators forming a complex that activates, among
others, genes coding virulence factors such as elastase, ExoA,
type II secretion system apparatus proteins, alkaline protease,
alginate, pyocyanin and pyoverdine.
This ability of P. aeruginosa to coordinate the upregulation
of virulence genes in a whole population translates into increased pathogenicity of QS capable strains compared to QS
deficient mutants throughout a variety of animal models [45,
144–146]. Furthermore, several studies have shown that the
autoinducer N-(3-oxododecanoyl)-L-homoserine-lactone is immunomodulatory and can depress host responses [147–150].
These roles of QS in virulence and immunomodulation make
it a highly interesting candidate for pathogenesis targeting therapeutics to the extent that potential strategies involving each
6. Quorum-sensing (Fig. 4)
QS is a mechanism shared by Gram-negative bacteria that
allows bacteria-to-bacteria cell signaling through small molecules, acyl homoserine lactones (AHL), also known as autoinducers that diffuse freely across bacterial membranes. When a
certain bacterial density or “quorum” is obtained, these molecules reach a threshold concentration at which, as cofactors of
transcriptional regulators, they allow coordinated gene expres-
Fig. 4. Overview of P. aeruginosa QS.
Fig. 4. Schéma du QS de P. aeruginosa.
“I” genes (lasI or rhlI), encoding an autoinducer synthase are activated, the
synthesized autoinducer (acy-homoserine lactone, AHL) diffuses into the
environment. An increase in bacterial density during infection leads to an
increase in autoinducer concentration past a threshold allowing it to bind to
transcriptional activators (LasR, RhlR) forming a complex that activates genes
coding virulence factors.
86
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
step of QS have been mapped out in an in-depth review [151].
Among such strategies, several have been explored experimentally. QS can be successfully inhibited by competitive antagonist analogs of acyl-homoserine-lactones (AHLs) leading to decreased elastase production and biofilm formation [152]. The
signaling AHLs are degraded by AHL-lactonases. Plants naturally expressing these lactonases have increased resistance to
QS capable pathogens [153]. Insertion of an AHL-lactonase
gene into PAO1 led to decreased production of virulence factors [154]. In this search for QS inhibitors, following the discovery that antibacterial furanones produced by marine macroalga were capable of inhibiting QS, synthetic furanones were
developed [155]. Synthetic furanones successfully inhibit QS
and reduce virulence factor expression, increase bacterial clearance and survival in murine models of P. aeruginosa lung infection [155–157]. Pursuing the quest for QS inhibitors has led
to the development of screening strategies that have successfully shown QS inhibiting properties of novel compounds from
garlic extracts [158]. Closer to clinical practice, it has been
found that azithromycin inhibits QS, decreasing virulence factor expression and biofilm production [159,160]. This effect
may account, in part, for the antipseudomonal effects of subinhibitory concentrations of macrolides [161,162].
7. Genetics
The source of the pathogenic versatility of P. aeruginosa is
undoubtedly it’s unique genome. Sequencing of this genome
was achieved in 2000 and revealed a uniquely large genome
of 6.3 million base pairs in 5570 predicted genes(PAO1 strain).
Among these, 8.4% are involved in gene regulation, making
P. aeruginosa the pathogen with the greatest number of genes
devoted to regulation [163]. This in itself could explain the
adaptability of P. aeruginosa to so many hostile environments
and its impressive array of pathogenic mechanisms. Additionally, although the genome of P. aeruginosa is remarkably conserved in strains among different isolates [164] and even
among different strains [163–165], 10% of the genome is variable and organized in blocks, or islands in the conserved core
[164]. These islands have been successively shown to comprise
material coding for known virulence factors such as ExoU
[165], and up to 19 genes involved in virulence in a model of
mouse thermal injury [166]. These islands, thus dubbed
“pathogenicity islands” provide P. aeruginosa with even greater pathogenic versatility.
Is pathogenesis accessible to therapeutic targeting on a genetic level in P. aeruginosa? Given the above information it
would be highly desirable. In fact, antisense oligonucleotides
or antisense peptide nucleic acids inhibit specific gene expression in Escherichia coli [167] and in Staphylococcus aureus
[168]. In an in vivo animal study in which anti-NF-kappaB
antisense therapy increased survival in endotoxic shock and
peritonitis [169]. However, this approach has yet to be developed in P. aeruginosa infections.
8. Conclusions
Although we have limited the scope of this review to the
therapeutic targeting of direct bacterial mechanisms of pathogenesis, P. aeruginosa also interacts with host cells in triggering or modulating a multitude of pathways leading to further
impairment of host defenses or injury [170]. These interactions
may also be accessible to specific therapies as well, as the success of activated protein C in modulating the inflammation/
coagulation disorders of sepsis has shown [171].
Among the therapeutic strategies we have reviewed, many,
including the most promising, are either active or passive immunotherapy-based. The advantages of immune-based therapeutics are notably the high specificity that allows targeting
of these very specific pathogenic mechanisms and thus, few
adverse effects. Additionally, active immunization strategies
against P. aeruginosa are being pursued with the development
of an anti-PcrV vaccine that could potentially transform the
evolution of patients highly susceptible to P. aeruginosa such
as cystic fibrosis patients. A current review addressing the entire range of vaccine development strategies against
P. aeruginosa has recently been published [172].
The diversity of pathogenic mechanisms of P. aeruginosa
combined with the extreme adaptability conferred to
P. aeruginosa by it’s large genome highly devoted to regulation of gene expression make P. aeruginosa very much the
“Superbug”…and it is high time we find the “Kryptonite”.
Acknowledgements
This research was supported by NIH Grants GM08440 - HL
74005 - HL69809, and funding from the Société de Réanimation de Langue Française (SRLF) and the Association pour la
Recherche en Pathologie Infectieuse (ARREPI).
References
[1]
[2]
[3]
[4]
[5]
[6]
[7]
Chastre J, Fagon JY. Ventilator-associated pneumonia. Am J Respir
Crit Care Med 2002;165:867–903.
Chastre J, Trouillet JL. Problem pathogens (Pseudomonas aeruginosa
and acinetobacter). Semin Respir Infect 2000;15:287–98.
Rello J, Rue M, Jubert P, Muses G, Sonora R, Valles J, et al. Survival
in patients with nosocomial pneumonia: Impact of the severity of illness
and the etiologic agent. Crit Care Med 1997;25:1862–7.
Crouch Brewer S, Wunderink RG, Jones CB, Leeper Jr. KV. Ventilatorassociated pneumonia due to Pseudomonas aeruginosa. Chest 1996;
109:1019–29.
Obritsch MD, Fish DN, MacLaren R, Jung R. National surveillance of
antimicrobial resistance in Pseudomonas aeruginosa isolates obtained
from intensive care unit patients from 1993 to 2002. Antimicrob Agents
Chemother 2004;48:4606–10.
Friedland I, Gallagher G, King T, Woods GL. Antimicrobial susceptibility patterns in Pseudomonas aeruginosa: Data from a multicenter intensive care unit surveillance study (iss) in the United States. J Chemother 2004;16:437–41.
Kollef MH. Prevention of hospital-associated pneumonia and ventilatorassociated pneumonia. Crit Care Med 2004;32:1396–405.
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
[8]
[9]
[10]
[11]
[12]
[13]
[14]
[15]
[16]
[17]
[18]
[19]
[20]
[21]
[22]
[23]
[24]
[25]
[26]
[27]
Hoffken G, Niederman MS. Nosocomial pneumonia: The importance of
a de-escalating strategy for antibiotic treatment of pneumonia in the icu.
Chest 2002;122:2183–96.
Warren DK, Hill HA, Merz LR, Kollef MH, Hayden MK, Fraser VJ,
et al. Cycling empirical antimicrobial agents to prevent emergence of
antimicrobial-resistant gram-negative bacteria among intensive care unit
patients. Crit Care Med 2004;32:2450–6.
Burgess DS. Use of pharmacokinetics and pharmacodynamics to optimize antimicrobial treatment of Pseudomonas aeruginosa infections.
Clin Infect Dis 2005;40(Suppl 2):S99–S104.
Iregui M, Ward S, Sherman G, Fraser VJ, Kollef MH. Clinical importance of delays in the initiation of appropriate antibiotic treatment for
ventilator-associated pneumonia. Chest 2002;122:262–8.
Klepser ME. Role of nebulized antibiotics for the treatment of respiratory infections. Curr Opin Infect Dis 2004;17:109–12.
Feldman M, Bryan R, Rajan S, Scheffler L, Brunnert S, Tang H, et al.
Role of flagella in pathogenesis of Pseudomonas aeruginosa pulmonary
infection. Infect Immun 1998;66:43–51.
Adamo R, Sokol S, Soong G, Gomez MI. Pseudomonas aeruginosa flagella activate airway epithelial cells through asialogm1 and toll-like receptor 2 as well as toll-like receptor 5. Am J Respir Cell Mol Biol 2004;
30:627–34.
DiMango E, Zar HJ, Bryan R, Prince A. Diverse Pseudomonas aeruginosa gene products stimulate respiratory epithelial cells to produce interleukin-8. J Clin Invest 1995;96:2204–10.
Mahenthiralingam E, Campbell ME, Speert DP. Nonmotility and phagocytic resistance of Pseudomonas aeruginosa isolates from chronically
colonized patients with cystic fibrosis. Infect Immun 1994;62:596–605.
Landsperger WJ, Kelly-Wintenberg KD, Montie TC, Knight LS,
Hansen MB, Huntenburg CC, et al. Inhibition of bacterial motility with
human antiflagellar monoclonal antibodies attenuates Pseudomonas
aeruginosa-induced pneumonia in the immunocompetent rat. Infect Immun 1994;62:4825–30.
Doring G, Pfeiffer C, Weber U, Mohr-Pennert A, Dorner F. Parenteral
application of a Pseudomonas aeruginosa flagella vaccine elicits specific anti-flagella antibodies in the airways of healthy individuals. Am J
Respir Crit Care Med 1995;151:983–5.
Doring G, Dorner F. A multicenter vaccine trial using the Pseudomonas
aeruginosa flagella vaccine immuno in patients with cystic fibrosis.
Behring Inst Mitt 1997:338–44.
Mattick JS. Type iv pili and twitching motility. Annu Rev Microbiol
2002;56:289–314.
Hahn HP. The type-4 pilus is the major virulence-associated adhesin of
Pseudomonas aeruginosa—a review. Gene 1997;192:99–108.
Gupta SK, Berk RS, Masinick S, Hazlett LD. Pili and lipopolysaccharide of Pseudomonas aeruginosa bind to the glycolipid asialo gm1. Infect Immun 1994;62:4572–9.
Farinha MA, Conway BD, Glasier LM, Ellert NW, Sherburne R,
Irvin RT, et al. Alteration of the pilin adhesin of Pseudomonas aeruginosa pao results in normal pilus biogenesis but a loss of adherence to
human pneumocyte cells and decreased virulence in mice. Infect Immun
1994;62:4118–23.
Comolli JC, Hauser AR, Waite L, Whitchurch CB, Mattick JS,
Engel JN. Pseudomonas aeruginosa gene products pilt and pilu are required for cytotoxicity in vitro and virulence in a mouse model of acute
pneumonia. Infect Immun 1999;67:3625–30.
Tang H, Kays M, Prince A. Role of Pseudomonas aeruginosa pili in
acute pulmonary infection. Infect Immun 1995;63:1278–85.
Hahn H, Lane-Bell PM, Glasier LM, Nomellini JF, Bingle WH, Paranchych W, et al. Pilin-based anti-Pseudomonas vaccines: Latest developments and perspectives. Behring Inst Mitt 1997:315–25.
Sheth HB, Glasier LM, Ellert NW, Cachia P, Kohn W, Lee KK, et al.
Development of an anti-adhesive vaccine for Pseudomonas aeruginosa
targeting the c-terminal region of the pilin structural protein. Biomed
Pept Proteins Nucleic Acids 1995;1:141–8.
[28]
[29]
[30]
[31]
[32]
[33]
[34]
[35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
87
Cachia PJ, Kao DJ, Hodges RS. Synthetic peptide vaccine development: Measurement of polyclonal antibody affinity and cross-reactivity
using a new peptide capture and release system for surface plasmon resonance spectroscopy. J Mol Recognit 2004;17:540–57.
Hertle R, Mrsny R, Fitzgerald DJ. Dual-function vaccine for Pseudomonas aeruginosa: Characterization of chimeric exotoxin a-pilin protein.
Infect Immun 2001;69:6962–9.
Lang AB, Horn MP, Imboden MA, Zuercher AW. Prophylaxis and
therapy of Pseudomonas aeruginosa infection in cystic fibrosis and immunocompromised patients. Vaccine 2004;22(Suppl. 1):S44–8.
Hatano K, Pier GB. Complex serology and immune response of mice to
variant high-molecular-weight o polysaccharides isolated from Pseudomonas aeruginosa serogroup o2 strains. Infect Immun 1998;66:3719–
26.
Pier GB. Promises and pitfalls of Pseudomonas aeruginosa lipopolysaccharide as a vaccine antigen. Carbohydr Res 2003;338:2549–56.
Lang AB, Rudeberg A, Schoni MH, Que JU, Furer E, Schaad UB. Vaccination of cystic fibrosis patients against Pseudomonas aeruginosa reduces the proportion of patients infected and delays time to infection.
Pediatr Infect Dis J 2004;23:504–10.
Priebe GP, Brinig MM, Hatano K, Grout M, Coleman FT, Pier GB,
et al. Construction and characterization of a live, attenuated aroa deletion mutant of Pseudomonas aeruginosa as a candidate intranasal vaccine. Infect Immun 2002;70:1507–17.
Priebe GP, Meluleni GJ, Coleman FT, Goldberg JB, Pier GB. Protection against fatal Pseudomonas aeruginosa pneumonia in mice after nasal immunization with a live, attenuated aroa deletion mutant. Infect Immun 2003;71:1453–61.
DiGiandomenico A, Rao J, Goldberg JB. Oral vaccination of balb/c
mice with salmonella enterica serovar typhimurium expressing Pseudomonas aeruginosa o antigen promotes increased survival in an acute
fatal pneumonia model. Infect Immun 2004;72:7012–21.
Holder IA. Pseudomonas immunotherapy: A historical overview. Vaccine 2004;22:831–9.
Lai Z, Kimmel R, Petersen S, Thomas S, Pier G, Bezabeh B, et al. Multi-valent human monoclonal antibody preparation against Pseudomonas
aeruginosa derived from transgenic mice containing human immunoglobulin loci is protective against fatal Pseudomonas sepsis caused by
multiple serotypes. Vaccine 2005;23:3264–71.
Hemachandra S, Kamboj K, Copfer J, Pier G, Green LL, Schreiber JR.
Human monoclonal antibodies against Pseudomonas aeruginosa lipopolysaccharide derived from transgenic mice containing megabase human immunoglobulin loci are opsonic and protective against fatal Pseudomonas sepsis. Infect Immun 2001;69:2223–9.
Sawa T, Kurahashi K, Ohara M, Gropper MA, Doshi V, Larrick JW,
et al. Evaluation of antimicrobial and lipopolysaccharide-neutralizing
effects of a synthetic cap18 fragment against Pseudomonas aeruginosa
in a mouse model. Antimicrob Agents Chemother 1998;42:3269–75.
Zhou Z, Kozlowski J, Schuster DP. Physiologic, biochemical, and imaging characterization of acute lung injury in mice. Am J Respir Crit
Care Med 2005.
Wieland CW, Siegmund B, Senaldi G, Vasil ML, Dinarello CA, Fantuzzi G. Pulmonary inflammation induced by Pseudomonas aeruginosa
lipopolysaccharide, phospholipase c, and exotoxin a: Role of interferon
regulatory factor 1. Infect Immun 2002;70:1352–8.
Backhed F, Normark S, Schweda EK, Oscarson S, Richter-Dahlfors A.
Structural requirements for tlr4-mediated lps signalling: A biological
role for lps modifications. Microbes Infect 2003;5:1057–63.
Hajjar AM, Ernst RK, Tsai JH, Wilson CB, Miller SI. Human toll-like
receptor 4 recognizes host-specific lps modifications. Nat Immunol
2002;3:354–9.
Tang HB, DiMango E, Bryan R, Gambello M, Iglewski BH,
Goldberg JB, et al. Contribution of specific Pseudomonas aeruginosa
virulence factors to pathogenesis of pneumonia in a neonatal mouse
model of infection. Infect Immun 1996;64:37–43.
88
[46]
[47]
[48]
[49]
[50]
[51]
[52]
[53]
[54]
[55]
[56]
[57]
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
Pier GB, Grout M, Zaidi TS. Cystic fibrosis transmembrane conductance regulator is an epithelial cell receptor for clearance of Pseudomonas aeruginosa from the lung. Proc Natl Acad Sci USA 1997;94:
12088–93.
Ernst RK, Hajjar AM, Tsai JH, Moskowitz SM, Wilson CB, Miller SI.
Pseudomonas aeruginosa lipid a diversity and its recognition by tolllike receptor 4. J Endotoxin Res 2003;9:395–400.
Ernst RK, Yi EC, Guo L, Lim KB, Burns JL, Hackett M, et al. Specific
lipopolysaccharide found in cystic fibrosis airway Pseudomonas aeruginosa. Science 1999;286:1561–5.
Yokota S, Ohtsuka H, Kohzuki T, Noguchi H. A polyreactive human
anti-lipid a monoclonal antibody having cross reactivity to polysaccharide portions of Pseudomonas aeruginosa lipopolysaccharides. FEMS
Immunol Med Microbiol 1996;14:31–8.
Kline T, Andersen NH, Harwood EA, Bowman J, Malanda A, Endsley
S, et al. Potent, novel in vitro inhibitors of the Pseudomonas aeruginosa
deacetylase lpxc. J Med Chem 2002;45:3112–29.
Onishi HR, Pelak BA, Gerckens LS, Silver LL, Kahan FM, Chen MH,
et al. Antibacterial agents that inhibit lipid a biosynthesis. Science 1996;
274:980–2.
Davies JC. Pseudomonas aeruginosa in cystic fibrosis: Pathogenesis
and persistence. Paediatr Respir Rev 2002;3:128–34.
Mathee K, Ciofu O, Sternberg C, Lindum PW, Campbell JI, Jensen P,
et al. Mucoid conversion of Pseudomonas aeruginosa by hydrogen peroxide: A mechanism for virulence activation in the cystic fibrosis lung.
Microbiology 1999;145(Pt 6):1349–57.
Hentzer M, Teitzel GM, Balzer GJ, Heydorn A, Molin S, Givskov M,
et al. Alginate overproduction affects Pseudomonas aeruginosa biofilm
structure and function. J Bacteriol 2001;183:5395–401.
Cobb LM, Mychaleckyj JC, Wozniak DJ, Lopez-Boado YS. Pseudomonas aeruginosa flagellin and alginate elicit very distinct gene expression
patterns in airway epithelial cells: Implications for cystic fibrosis disease. J Immunol 2004;173:5659–70.
Boyd A, Chakrabarty AM. Pseudomonas aeruginosa biofilms: Role of
the alginate exopolysaccharide. J Ind Microbiol 1995;15:162–8.
Wozniak DJ, Wyckoff TJ, Starkey M, Keyser R, Azadi P, O’Toole GA,
et al. Alginate is not a significant component of the extracellular polysaccharide matrix of pa14 and pao1 Pseudomonas aeruginosa biofilms.
Proc Natl Acad Sci USA 2003;100:7907–12.
Theilacker C, Coleman FT, Mueschenborn S, Llosa N, Grout M,
Pier GB. Construction and characterization of a Pseudomonas aeruginosa mucoid exopolysaccharide-alginate conjugate vaccine. Infect Immun
2003;71:3875–84.
Pier GB, Boyer D, Preston M, Coleman FT, Llosa N, MueschenbornKoglin S, et al. Human monoclonal antibodies to Pseudomonas aeruginosa alginate that protect against infection by both mucoid and nonmucoid strains. J Immunol 2004;173:5671–8.
Pier GB, DesJardin D, Grout M, Garner C, Bennett SE, Pekoe G, et al.
Human immune response to Pseudomonas aeruginosa mucoid exopolysaccharide (alginate) vaccine. Infect Immun 1994;62:3972–9.
Leidal KG, Munson KL, Denning GM. Small molecular weight secretory factors from Pseudomonas aeruginosa have opposite effects on il-8
and rantes expression by human airway epithelial cells. Am J Respir
Cell Mol Biol 2001;25:186–95.
Denning GM, Wollenweber LA, Railsback MA, Cox CD, Stoll LL,
Britigan BE. Pseudomonas pyocyanin increases interleukin-8 expression by human airway epithelial cells. Infect Immun 1998;66:5777–84.
Allen L, Dockrell DH, Pattery T, Lee DG, Cornelis P, Hellewell PG,
et al. Pyocyanin production by Pseudomonas aeruginosa induces neutrophil apoptosis and impairs neutrophil-mediated host defenses in vivo.
J Immunol 2005;174:3643–9.
Lau GW, Ran H, Kong F, Hassett DJ, Mavrodi D. Pseudomonas aeruginosa pyocyanin is critical for lung infection in mice. Infect Immun
2004;72:4275–8.
O’Malley YQ, Reszka KJ, Spitz DR, Denning GM, Britigan BE. Pseudomonas aeruginosa pyocyanin directly oxidizes glutathione and de-
[66]
[67]
[68]
[69]
[70]
[71]
[72]
[73]
[74]
[75]
[76]
[77]
[78]
[79]
[80]
[81]
[82]
[83]
[84]
[85]
creases its levels in airway epithelial cells. Am J Physiol Lung Cell
Mol Physiol 2004;287:L94–L103.
O’Malley YQ, Reszka KJ, Rasmussen GT, Abdalla MY, Denning GM,
Britigan BE. The Pseudomonas secretory product pyocyanin inhibits
catalase activity in human lung epithelial cells. Am J Physiol Lung Cell
Mol Physiol 2003;285:L1077–L1086.
Lau GW, Hassett DJ, Ran H, Kong F. The role of pyocyanin in Pseudomonas aeruginosa infection. Trends Mol Med 2004;10:599–606.
Wood LG, Fitzgerald DA, Lee AK, Garg ML. Improved antioxidant
and fatty acid status of patients with cystic fibrosis after antioxidant
supplementation is linked to improved lung function. Am J Clin Nutr
2003;77:150–9.
Rancourt RC, Tai S, King M, Heltshe SL, Penvari C, Accurso FJ, et al.
Thioredoxin liquefies and decreases the viscoelasticity of cystic fibrosis
sputum. Am J Physiol Lung Cell Mol Physiol 2004;286:L931–L938.
Griese M, Ramakers J, Krasselt A, Starosta V, Van Koningsbruggen S,
Fischer R, et al. Improvement of alveolar glutathione and lung function
but not oxidative state in cystic fibrosis. Am J Respir Crit Care Med
2004;169:822–8.
Smith RS, Iglewski BHP. Aeruginosa quorum-sensing systems and
virulence. Curr Opin Microbiol 2003;6:56–60.
Meyer JM, Neely A, Stintzi A, Georges C, Holder IA. Pyoverdin is essential for virulence of Pseudomonas aeruginosa. Infect Immun 1996;
64:518–23.
Takase H, Nitanai H, Hoshino K, Otani T. Impact of siderophore production on Pseudomonas aeruginosa infections in immunosuppressed
mice. Infect Immun 2000;68:1834–9.
Lamont IL, Beare PA, Ochsner U, Vasil AI, Vasil ML. Siderophoremediated signaling regulates virulence factor production in Pseudomonas aeruginosa. Proc Natl Acad Sci USA 2002;99:7072–7.
Guzzo J, Pages JM, Duong F, Lazdunski A, Murgier M. Pseudomonas
aeruginosa alkaline protease: Evidence for secretion genes and study of
secretion mechanism. J Bacteriol 1991;173:5290–7.
Kipnis E, Guery BP, Tournoys A, Leroyt X, Robrique L, Fialdes P,
et al. Massive alveolar thrombin activation in Pseudomonas aeruginosa-induced acute lung injury. Shock 2004;21:444–51.
Matsumoto T, Furuya N, Tateda K, Miyazaki S, Ohno A, Ishii Y, et al.
Effect of passive immunotherapy on murine gut-derived sepsis caused
by Pseudomonas aeruginosa. J Med Microbiol 1999;48:765–70.
Matsumoto T, Tateda K, Furuya N, Miyazaki S, Ohno A, Ishii Y, et al.
Efficacies of alkaline protease, elastase and exotoxin a toxoid vaccines
against gut-derived Pseudomonas aeruginosa sepsis in mice. J Med Microbiol 1998;47:303–8.
Matsumoto T, Tateda K, Miyazaki S, Furuya N, Ohno A, Ishii Y, et al.
Effect of immunisation with Pseudomonas aeruginosa on gut-derived
sepsis in mice. J Med Microbiol 1998;47:295–301.
Matsumoto K. Role of bacterial proteases in pseudomonal and serratial
keratitis. Biol Chem 2004;385:1007–16.
Malloy JL, Veldhuizen RA, Thibodeaux BA, O’Callaghan RJ,
Wright JR. Pseudomonas aeruginosa protease iv degrades surfactant
proteins and inhibits surfactant host defense and biophysical functions.
Am J Physiol Lung Cell Mol Physiol 2005;288:L409–L418.
Azghani AO. Pseudomonas aeruginosa and epithelial permeability:
Role of virulence factors elastase and exotoxin a. Am J Respir Cell
Mol Biol 1996;15:132–40.
Azghani AO, Gray LD, Johnson AR. A bacterial protease perturbs the
paracellular barrier function of transporting epithelial monolayers in culture. Infect Immun 1993;61:2681–6.
Azghani AO, Miller EJ, Peterson BT. Virulence factors from Pseudomonas aeruginosa increase lung epithelial permeability. Lung 2000;
178:261–9.
Kon Y, Tsukada H, Hasegawa T, Igarashi K, Wada K, Suzuki E, et al.
The role of Pseudomonas aeruginosa elastase as a potent inflammatory
factor in a rat air pouch inflammation model. FEMS Immunol Med Microbiol 1999;25:313–21.
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
[86]
Alcorn JF, Wright JR. Degradation of pulmonary surfactant protein d
by Pseudomonas aeruginosa elastase abrogates innate immune function. J Biol Chem 2004;279:30871–9.
[87]
Mariencheck WI, Alcorn JF, Palmer SM, Wright JR. Pseudomonas aeruginosa elastase degrades surfactant proteins a and d. Am J Respir Cell
Mol Biol 2003;28:528–37.
[88]
Dulon S, Leduc D, Cottrell GS, D’Alayer J, Hansen KK, Bunnett NW,
et al. Pseudomonas aeruginosa elastase disables proteinase-activated receptor 2 in respiratory epithelial cells. Am J Respir Cell Mol Biol 2005;
32:411–9.
[89]
Sokol PA, Kooi C, Hodges RS, Cachia P, Woods DE. Immunization
with a Pseudomonas aeruginosa elastase peptide reduces severity of experimental lung infections due to P. aeruginosa or Burkholderia cepacia. J Infect Dis 2000;181:1682–92.
[90]
Azghani AO, Bedinghaus T, Klein R. Detection of elastase from Pseudomonas aeruginosa in sputum and its potential role in epithelial cell
permeability. Lung 2000;178:181–9.
[91]
Woods DE, Cantin A, Cooley J, Kenney DM, Remold-O’Donnell E.
Aerosol treatment with mnei suppresses bacterial proliferation in a model of chronic Pseudomonas aeruginosa lung infection. Pediatr Pulmonol
2005;39:141–9.
[92]
Cantin AM, Woods DE. Aerosolized prolastin suppresses bacterial proliferation in a model of chronic Pseudomonas aeruginosa lung infection. Am J Respir Crit Care Med 1999;160:1130–5.
[93]
Wiener-Kronish JP, Sakuma T, Kudoh I, Pittet JF, Frank D, Dobbs L,
et al. Alveolar epithelial injury and pleural empyema in acute
P. aeruginosa pneumonia in anesthetized rabbits. J Appl Physiol 1993;
75:1661–9.
[94]
Konig B, Vasil ML, Konig W. Role of hemolytic and nonhemolytic
phospholipase c from Pseudomonas aeruginosa for inflammatory mediator release from human granulocytes. Int Arch Allergy Immunol 1997;
112:115–24.
[95]
Konig B, Jaeger KE, Sage AE, Vasil ML, Konig W. Role of Pseudomonas aeruginosa lipase in inflammatory mediator release from human
inflammatory effector cells (platelets, granulocytes, and monocytes. Infect Immun 1996;64:3252–8.
[96]
Holm BA, Keicher L, Liu MY, Sokolowski J, Enhorning G. Inhibition
of pulmonary surfactant function by phospholipases. J Appl Physiol
1991;71:317–21.
[97]
Terada LS, Johansen KA, Nowbar S, Vasil AI, Vasil ML. Pseudomonas
aeruginosa hemolytic phospholipase c suppresses neutrophil respiratory
burst activity. Infect Immun 1999;67:2371–6.
[98]
Hybenova D, Majtan V. The influence of postantibiotic effects and
postantibiotic effects of sub-inhibitory concentrations of quinolones
and aminoglycosides on phospholipase c of Pseudomonas aeruginosa.
Pharmazie 1997;52:157–9.
[99]
Wick MJ, Hamood AN, Iglewski BH. Analysis of the structure–function relationship of Pseudomonas aeruginosa exotoxin a. Mol Microbiol 1990;4:527–35.
[100] Pavlovskis OR, Iglewski BH, Pollack M. Mechanism of action of Pseudomonas aeruginosa exotoxin a in experimental mouse infections: Adenosine diphosphate ribosylation of elongation factor 2. Infect Immun
1978;19:29–33.
[101] Schultz MJ, Speelman P, Zaat SA, Hack CE, van Deventer SJ, van der
Poll T. The effect of Pseudomonas exotoxin a on cytokine production in
whole blood exposed to Pseudomonas aeruginosa. FEMS Immunol
Med Microbiol 2000;29:227–32.
[102] Schultz MJ, Rijneveld AW, Florquin S, Speelman P, van Deventer SJ,
van der Poll T, et al. Impairment of host defence by exotoxin a in Pseudomonas aeruginosa pneumonia in mice. J Med Microbiol 2001;50:
822–7.
[103] Miyazaki S, Matsumoto T, Tateda K, Ohno A, Yamaguchi K. Role of
exotoxin a in inducing severe Pseudomonas aeruginosa infections in
mice. J Med Microbiol 1995;43:169–75.
89
[104] Kudoh I, Wiener-Kronish JP, Hashimoto S, Pittet JF, Frank D. Exoproduct secretions of Pseudomonas aeruginosa strains influence severity of
alveolar epithelial injury. Am J Physiol 1994;267:L551–L556.
[105] El-Zaim HS, Chopra AK, Peterson JW, Vasil ML, Heggers JP. Protection against exotoxin a (eta) and Pseudomonas aeruginosa infection in
mice with eta-specific antipeptide antibodies. Infect Immun 1998;66:
5551–4.
[106] Chen TY, Shang HF, Chen TL, Lin CP, Hui CF, Hwang J. Recombinant protein composed of Pseudomonas exotoxin a, outer membrane
proteins i and f as vaccine against P. aeruginosa infection. Appl Microbiol Biotechnol 1999;52:524–33.
[107] Shiau JW, Tang TK, Shih YL, Tai C, Sung YY, Huang JL, et al. Mice
immunized with DNA encoding a modified Pseudomonas aeruginosa
exotoxin a develop protective immunity against exotoxin intoxication.
Vaccine 2000;19:1106–12.
[108] Denis-Mize KS, Price BM, Baker NR, Galloway DR. Analysis of immunization with DNA encoding Pseudomonas aeruginosa exotoxin a.
FEMS Immunol Med Microbiol 2000;27:147–54.
[109] Sawa T, Wiener-Kronish JP. A therapeutic strategy against the shared
virulence mechanism utilized by both yersinia pestis and Pseudomonas
aeruginosa. Anesthesiol Clin North America 2004;22:591–606 viii–ix.
[110] Fu H, Coburn J, Collier RJ. The eukaryotic host factor that activates
exoenzyme s of Pseudomonas aeruginosa is a member of the 14-3-3
protein family. Proc Natl Acad Sci USA 1993;90:2320–4.
[111] Shaver CM, Hauser AR. Relative contributions of Pseudomonas aeruginosa exou, exos, and exot to virulence in the lung. Infect Immun
2004;72:6969–77.
[112] Maresso AW, Baldwin MR, Barbieri JT. Ezrin/radixin/moesin proteins
are high affinity targets for adp-ribosylation by Pseudomonas aeruginosa exos. J Biol Chem 2004;279:38402–8.
[113] Krall R, Sun J, Pederson KJ, Barbieri JT. In vivo rho gtpase-activating
protein activity of Pseudomonas aeruginosa cytotoxin exos. Infect Immun 2002;70:360–7.
[114] Epelman S, Stack D, Bell C, Wong E, Neely GG, Krutzik S, et al. Different domains of Pseudomonas aeruginosa exoenzyme s activate distinct tlrs. J Immunol 2004;173:2031–40.
[115] Lee VT, Smith RS, Tummler B, Aktories K, Barbieri JT. Activities of
Pseudomonas aeruginosa effectors secreted by the type iii secretion
system in vitro and during infection. Infect Immun 2005;73:1695–705.
[116] Goehring UM, Schmidt G, Pederson KJ, Aktories K, Barbieri JT. The
n-terminal domain of Pseudomonas aeruginosa exoenzyme s is a
gtpase-activating protein for rho gtpases. J Biol Chem 1999;274:
36369–72.
[117] Roy-Burman A, Savel RH, Racine S, Swanson BL, Revadigar NS, Fujimoto J, et al. Type iii protein secretion is associated with death in lower respiratory and systemic Pseudomonas aeruginosa infections. J Infect
Dis 2001;183:1767–74.
[118] Yahr TL, Barbieri JT, Frank DW. Genetic relationship between the 53and 49-kilodalton forms of exoenzyme s from Pseudomonas aeruginosa. J Bacteriol 1996;178:1412–9.
[119] Aktories K, Barbieri JT. Bacterial cytotoxins: Targeting eukaryotic
switches. Nat Rev Microbiol 2005;3:397–410.
[120] Henriksson ML, Sundin C, Jansson AL, Forsberg A, Palmer RH, Hallberg B. Exoenzyme s shows selective adp-ribosylation and gtpase-activating protein (gap) activities towards small gtpases in vivo. Biochem J
2002;367:617–28.
[121] Kazmierczak BI, Engel JN. Pseudomonas aeruginosa exot acts in vivo
as a gtpase-activating protein for rhoa, rac1, and cdc42. Infect Immun
2002;70:2198–205.
[122] Sundin C, Hallberg B, Forsberg A. Adp-ribosylation by exoenzyme t of
Pseudomonas aeruginosa induces an irreversible effect on the host cell
cytoskeleton in vivo. FEMS Microbiol Lett 2004;234:87–91.
[123] Garrity-Ryan L, Kazmierczak B, Kowal R, Comolli J, Engel JN, Hauser
A. The arginine finger domain of exot contributes to actin cytoskeleton
disruption and inhibition of internalization of Pseudomonas aeruginosa
by epithelial cells and macrophages. Infect Immun 2000;68:7100–13.
90
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
[124] Geiser TK, Kazmierczak BI, Garrity-Ryan LK, Garrity-Ryan LK,
Matthay MA, Engel JN. Pseudomonas aeruginosa exot inhibits in vitro
lung epithelial wound repair. Cell Microbiol 2001;3:223–36.
[125] Yahr TL, Vallis AJ, Hancock MK, Frank DW, Barbieri JT. Exoy, an
adenylate cyclase secreted by the Pseudomonas aeruginosa type iii system. Proc Natl Acad Sci USA 1998;95:13899–904.
[126] Sayner SL, Frank DW, King J, Chen H, VandeWaa J, Stevens T. Paradoxical camp-induced lung endothelial hyperpermeability revealed by
Pseudomonas aeruginosa exoy. Circ Res 2004;95:196–203.
[127] Sato H, Frank DW, Hillard CJ, Feix JB, Pankhaniya RR, Moriyama K,
et al. The mechanism of action of the Pseudomonas aeruginosa-encoded type III cytotoxin, exou. EMBO J 2003;22:2959–69.
[128] Tamura M, Ajayi T, Allmond LR, Moriyama K, Wiener-Kronish JP,
Sawa T. Lysophospholipase a activity of Pseudomonas aeruginosa type
III secretory toxin exou. Biochem Biophys Res Commun 2004;316:
323–31.
[129] Pankhaniya RR, Tamura M, Allmond LR, Moriyama K, Ajayi T, Wiener-Kronish JP, et al. Pseudomonas aeruginosa causes acute lung injury via the catalytic activity of the patatin-like phospholipase domain
of exou. Crit Care Med 2004;32:2293–9.
[130] Ader F, Le Berre R, Faure K, Gosset P, Epaulard O, Toussaint B, et al.
Alveolar response to Pseudomonas aeruginosa: Role of the type iii secretion system. Infect Immun 2005;73:4263–71.
[131] Finck-Barbancon V, Goranson J, Zhu L, Sawa T, Wiener-Kronish JP,
Fleiszig SM, et al. Exou expression by Pseudomonas aeruginosa correlates with acute cytotoxicity and epithelial injury. Mol Microbiol 1997;
25:547–57.
[132] Allewelt M, Coleman FT, Grout M, Priebe GP, Pier GB. Acquisition of
expression of the Pseudomonas aeruginosa exou cytotoxin leads to increased bacterial virulence in a murine model of acute pneumonia and
systemic spread. Infect Immun 2000;68:3998–4004.
[133] Kurahashi K, Kajikawa O, Sawa T, Ohara M, Gropper MA, Frank DW,
et al. Pathogenesis of septic shock in Pseudomonas aeruginosa pneumonia. J Clin Invest 1999;104:743–50.
[134] Hauser AR, Cobb E, Bodi M, Mariscal D, Valles J, Engel JN, et al.
Type iii protein secretion is associated with poor clinical outcomes in
patients with ventilator-associated pneumonia caused by Pseudomonas
aeruginosa. Crit Care Med 2002;30:521–8.
[135] Sawa T, Yahr TL, Ohara M, Kurahashi K, Gropper MA, WienerKronish JP, et al. Active and passive immunization with the Pseudomonas v antigen protects against type iii intoxication and lung injury. Nat
Med 1999;5:392–8.
[136] Phillips RM, Six DA, Dennis EA, Ghosh P. In vivo phospholipase activity of the Pseudomonas aeruginosa cytotoxin exou and protection of
mammalian cells with phospholipase a2 inhibitors. J Biol Chem 2003;
278:41326–32.
[137] Goure J, Pastor A, Faudry E, Chabert J, Dessen A, Attree I. The v antigen of Pseudomonas aeruginosa is required for assembly of the functional popb/popd translocation pore in host cell membranes. Infect Immun 2004;72:4741–50.
[138] Neely AN, Holder IA, Wiener-Kronish JP, Sawa T. Passive anti-pcrv
treatment protects burned mice against Pseudomonas aeruginosa challenge. Burns 2005;31:153–8.
[139] Faure K, Fujimoto J, Shimabukuro DW, Ajayi T, Shime N, Moriyama
K, et al. Effects of monoclonal anti-pcrv antibody on Pseudomonas aeruginosa-induced acute lung injury in a rat model. J Immune Based Ther
Vaccines 2003;1:2.
[140] Frank DW, Vallis A, Wiener-Kronish JP, Roy-Burman A, Spack EG,
Mullaney BP, et al. Generation and characterization of a protective
monoclonal antibody to Pseudomonas aeruginosa pcrv. J Infect Dis
2002;186:64–73.
[141] Shime N, Sawa T, Fujimoto J, Faure K, Allmond LR, Karaca T, et al.
Therapeutic administration of anti-pcrv f(ab’)(2) in sepsis associated
with Pseudomonas aeruginosa. J Immunol 2001;167:5880–6.
[142] Finch RG, Pritchard DI, Bycroft BW, Williams P, Stewart GS. Quorum
sensing: A novel target for anti-infective therapy. J Antimicrob Chemother 1998;42:569–71.
[143] Van Delden C, Iglewski BH. Cell-to-cell signaling and Pseudomonas
aeruginosa infections. Emerg Infect Dis 1998;4:551–60.
[144] Pearson JP, Feldman M, Iglewski BH, Prince A. Pseudomonas aeruginosa cell-to-cell signaling is required for virulence in a model of acute
pulmonary infection. Infect Immun 2000;68:4331–4.
[145] Wu H, Song Z, Givskov M, Doring G, Worlitzsch D, Mathee K, et al.
Pseudomonas aeruginosa mutations in lasi and rhli quorum sensing
systems result in milder chronic lung infection. Microbiol 2001;147:
1105–13.
[146] Lesprit P, Faurisson F, Join-Lambert O, Roudot-Thoraval F, Foglino M,
Vissuzaine C, et al. Role of the quorum-sensing system in experimental
pneumonia due to Pseudomonas aeruginosa in rats. Am J Respir Crit
Care Med 2003;167:1478–82.
[147] Telford G, Wheeler D, Williams P, Tomkins PT, Appleby P, Sewell H,
et al. The Pseudomonas aeruginosa quorum-sensing signal molecule n(3-oxododecanoyl)-L-homoserine lactone has immunomodulatory activity. Infect Immun 1998;66:36–42.
[148] Smith RS, Harris SG, Phipps R, Iglewski B. The Pseudomonas aeruginosa quorum-sensing molecule n-(3-oxododecanoyl)homoserine lactone
contributes to virulence and induces inflammation in vivo. J Bacteriol
2002;184:1132–9.
[149] Smith RS, Fedyk ER, Springer TA, Mukaida N, Iglewski BH,
Phipps RP. Il-8 production in human lung fibroblasts and epithelial cells
activated by the Pseudomonas autoinducer n-3-oxododecanoyl homoserine lactone is transcriptionally regulated by nf-kappa b and activator
protein-2. J Immunol 2001;167:366–74.
[150] Smith RS, Kelly R, Iglewski BH, Phipps RP. The Pseudomonas autoinducer n-(3-oxododecanoyl) homoserine lactone induces cyclooxygenase-2 and prostaglandin e2 production in human lung fibroblasts: Implications for inflammation. J Immunol 2002;169:2636–42.
[151] Smith RS, Iglewski BH. Pseudomonas aeruginosa quorum sensing as a
potential antimicrobial target. J Clin Invest 2003;112:1460–5.
[152] Smith KM, Bu Y, Suga H. Library screening for synthetic agonists and
antagonists of a Pseudomonas aeruginosa autoinducer. Chem Biol
2003;10:563–71.
[153] Dong YH, Wang LH, Xu JL, Zhang HB, Zhang XF, Zhang LH.
Quenching quorum-sensing-dependent bacterial infection by an n-acyl
homoserine lactonase. Nature 2001;411:813–7.
[154] Reimmann C, Ginet N, Michel L, Keel C, Michaux P, Krishnapillai V,
et al. Genetically programmed autoinducer destruction reduces virulence
gene expression and swarming motility in Pseudomonas aeruginosa
pao1. Microbiol 2002;148:923–32.
[155] Hentzer M, Riedel K, Rasmussen TB, Heydorn A, Andersen JB,
Parsek MR, et al. Inhibition of quorum sensing in Pseudomonas aeruginosa biofilm bacteria by a halogenated furanone compound. Microbiol 2002;148:87–102.
[156] Hentzer M, Wu H, Andersen JB, Riedel K, Rasmussen TB, Bagge N,
et al. Attenuation of Pseudomonas aeruginosa virulence by quorum
sensing inhibitors. EMBO J 2003;22:3803–15.
[157] Wu H, Song Z, Hentzer M, Andersen JB, Molin S, Givskov M, et al.
Synthetic furanones inhibit quorum-sensing and enhance bacterial clearance in Pseudomonas aeruginosa lung infection in mice. J Antimicrob
Chemother 2004;53:1054–61.
[158] Rasmussen TB, Bjarnsholt T, Skindersoe ME, Hentzer M, Kristoffersen
P, Kote M, et al. Screening for quorum-sensing inhibitors (qsi) by use
of a novel genetic system, the qsi selector. J Bacteriol 2005;187:1799–
814.
[159] Tateda K, Comte R, Pechere JC, Kohler T, Yamaguchi K, Van Delden
C, et al. Azithromycin inhibits quorum sensing in Pseudomonas aeruginosa. Antimicrob Agents Chemother 2001;45:1930–3.
[160] Imamura Y, Yanagihara K, Mizuta Y, Seki M, Ohno H, Higashiyama
Y, et al. Azithromycin inhibits muc5ac production induced by the Pseudomonas aeruginosa autoinducer n-(3-oxododecanoyl) homoserine lac-
E. Kipnis et al. / Médecine et maladies infectieuses 36 (2006) 78–91
[161]
[162]
[163]
[164]
[165]
[166]
tone in nci-h292 cells. Antimicrob Agents Chemother 2004;48:3457–
61.
Tateda K, Standiford TJ, Pechere JC, Yamaguchi K. Regulatory effects
of macrolides on bacterial virulence: Potential role as quorum-sensing
inhibitors. Curr Pharm Des 2004;10:3055–65.
Wozniak DJ, Keyser R. Effects of subinhibitory concentrations of
macrolide antibiotics on Pseudomonas aeruginosa. Chest 2004;125:
62S–69S (quiz 9S).
Stover CK, Pham XQ, Erwin AL, Mizoguchi SD, Warrener P,
Hickey MJ, et al. Complete genome sequence of Pseudomonas aeruginosa pa01, an opportunistic pathogen. Nature 2000;406:959–64.
Spencer DH, Kas A, Smith EE, Raymond CK, Sims EH, Hastings M,
et al. Whole-genome sequence variation among multiple isolates of
Pseudomonas aeruginosa. J Bacteriol 2003;185:1316–25.
Wolfgang MC, Kulasekara BR, Liang X, Boyd D, Wu K, Yang Q, et al.
Conservation of genome content and virulence determinants among
clinical and environmental isolates of Pseudomonas aeruginosa. Proc
Natl Acad Sci USA 2003;100:8484–9.
He J, Baldini RL, Deziel E, Saucier M, Zhang Q, Liberati NT, et al.
The broad host range pathogen Pseudomonas aeruginosa strain pa14
[167]
[168]
[169]
[170]
[171]
[172]
91
carries two pathogenicity islands harboring plant and animal virulence
genes. Proc Natl Acad Sci USA 2004;101:2530–5.
Good L, Awasthi SK, Dryselius R, Dryselius R, Larsson O, Nielsen PE.
Bactericidal antisense effects of peptide-pna conjugates. Nat Biotechnol
2001;19:360–4.
Nekhotiaeva N, Awasthi SK, Nielsen PE, Good L. Inhibition of Staphylococcus aureus gene expression and growth using antisense peptide
nucleic acids. Mol Ther 2004;10:652–9.
D’Souza MJ, Jin Z, Oettinger CW. Treatment of experimental septic
shock with microencapsulated antisense oligomers to nf-kappab. J Interferon Cytokine Res 2005;25:311–20.
Sadikot RT, Blackwell TS, Christman JW, Prince AS. Pathogen–host
interactions in Pseudomonas aeruginosa pneumonia. Am J Respir Crit
Care Med 2005;171:1209–23.
Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, LopezRodriguez A, et al. Efficacy and safety of recombinant human activated
protein c for severe sepsis. N Engl J Med 2001;344:699–709.
Sedlak-Weinstein E, Cripps AW, Kyd JM, Foxwell AR. Pseudomonas
aeruginosa: The potential to immunise against infection. Expert Opin
Biol Ther 2005;5:967–82.