Sunteți pe pagina 1din 24

REVIEW ARTICLE

Di¡erential adaptation of microbial pathogens to airways


of patients with cystic ¢brosis and chronic obstructive
pulmonary disease
Gerd Döring1, Iyer G. Parameswaran2 & Timothy F. Murphy2
1
Institute of Medical Microbiology and Hygiene, Universitätsklinikum Tübingen, Tübingen, Germany; and 2Department of Medicine and New York
State Center of Excellence in Bioinformatics and Health Sciences, Division of Infectious Diseases, University at Buffalo, State University of New York,
Buffalo, NY, USA

Correspondence: Gerd Döring, Institute of Abstract


Medical Microbiology and Hygiene,
Universitätsklinikum Tübingen,
Cystic fibrosis (CF), the most common autosomal recessive disorder in Cauca-
Wilhelmstrasse 31, D-72074 Tübingen, sians, and chronic obstructive pulmonary disease (COPD), a disease of adults, are
Germany. Tel.: 149 7071 298 2069; characterized by chronic lung inflammation, airflow obstruction and extensive
fax: 149 7071 293 011; e-mail: tissue remodelling, which have a major impact on patients’ morbidity and
gerd.doering@med.uni-tuebingen.de mortality. Airway inflammation is stimulated in CF by chronic bacterial infections
and in COPD by environmental stimuli, particularly from smoking. Pseudomonas
Received 11 February 2010; revised 21 May aeruginosa is the major bacterial pathogen in CF, while in COPD, Haemophilus
2010; accepted 25 May 2010.
influenzae is most frequently observed. Molecular studies indicate that during
Final version published online 24 June 2010.
chronic pulmonary infection, P. aeruginosa clones genotypically and phenotypi-
DOI:10.1111/j.1574-6976.2010.00237.x
cally adapt to the CF niche, resulting in a highly diverse bacterial community that
is difficult to eradicate therapeutically. Pseudomonas aeruginosa clones from COPD
Editor: Dieter Haas patients remain within the airways only for limited time periods, do not adapt and
are easily eradicated. However, in a subgroup of severely ill COPD patients,
P. aeruginosa clones similar to those in CF persist. In this review, we will discuss the
MICROBIOLOGY REVIEWS

Keywords
Pseudomonas aeruginosa; Haemophilus pathophysiology of lung disease in CF and COPD, the complex genotypic and
influenzae; Staphylococcus aureus ; phenotypic adaptation processes of the opportunistic bacterial pathogens and
Burkholderia ssp.; mutator; adaptive radiation. novel treatment options.

domain. Over 1500 mutations and sequence variants have


Introduction been described to date and reported to the Cystic Fibrosis
Cystic fibrosis (CF) is the most common autosomal Genetic Analysis Consortium. Most of these mutations are
recessive disorder in Caucasians, affecting 1 : 2500 chil- rare and only four mutations occur at a frequency of
dren, with a carrier frequency of 1 : 25. In the CF lung, 4 1%. The most frequent mutation, F508del, occurs in
disease develops as a consequence of mutations in the CF the DNA sequence that codes for the NBD1 (Döring &
transmembrane conductance regulator (CFTR) gene, Ratjen, 2005). Abnormal transport of chloride and sodium
which encodes a membrane-bound cAMP-regulated chlor- ions caused by CFTR mutations affects water movement
ide channel (Kerem et al., 1989; Riordan et al., 1989; across epithelia, leading to pathophysiological conse-
Rommens et al., 1989). The primary structure of CFTR quences in various organs including the respiratory, gas-
indicates that it belongs to a family of transmembrane trointestinal and reproductive tract, the pancreas and the
proteins called ATP-binding cassette transporters, which liver. The CF phenotype is rather heterogeneous due to
form a large family of proteins responsible for the translo- many different mutations in CFTR and the influence of
cation of a variety of compounds across membranes of modifier genes (Davies, 2007). Chronic respiratory infec-
both prokaryotes and eukaryotes. CFTR is composed of tions caused by several opportunistic bacterial pathogens
five domains: two membrane-spanning domains, two are recognized to have the largest impact on morbidity and
nucleotide-binding domains (NBDs) and a regulatory mortality in CF patients (Høiby & Frederiksen, 2000).


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 125

Bacterial infections stimulate inflammatory defence me- Pathophysiology of CF lung disease


chanisms, leading to tissue destruction and extensive tissue
remodelling (Döring & Ratjen, 2007). The resulting A variety of innate immune functions have been reported to
emphysema and fibrosis mainly determine the reduced life be dysregulated in CF, and a number of mechanisms have
expectancy in individuals with CF. Once established, these been put forward to explain the fact that practically every CF
infections are difficult to treat with antibiotics and the patient suffers from chronic bacterial lung infections. The
pathogens are rarely eradicated (Döring et al., 2004). inability of mutated CFTR to effectively secrete chloride
Unlike CF, chronic obstructive pulmonary disease from respiratory epithelial cells into the airway surface
(COPD), a chronic, progressive disorder of lung parenchy- liquid causes excessive water absorption from the airway
ma and airways, is exclusively a disease of adults, especially surface liquid, leading to impaired mucociliary clearance in
of individuals over the age of 60 years. COPD is the fourth individuals with CF (Matsui et al., 1998; Boucher, 2007)
leading cause of death globally, accounting for approxi- (Fig. 1). This in turn facilitates the colonization of the
mately 2.7 million deaths in 2003 (Lopez et al., 2006). viscous mucus layer on the respiratory epithelium with
COPD is defined by airflow obstruction that is not fully bacteria, rather than the colonization of the epithelial sur-
reversible, in contrast to asthma, which is associated with face per se (Ulrich et al., 1998; Worlitzsch et al., 2002).
reversible airflow obstruction (Celli & MacNee, 2004). The Furthermore, an abnormal accumulation of ceramide in
degree of reduction in forced expiratory volume in 1 s the lungs of CF mice and in epithelial cells from CF patients
(FEV1) is used to classify COPD into four stages (mild, has been shown to result in an increased death rate of
moderate, severe and very severe). Although genetic factors respiratory epithelial cells and DNA deposits on the respira-
influence COPD (Wood & Stockley, 2006; Silverman et al., tory epithelium, which facilitates bacterial adherence
2009; Smolonska et al., 2009), these influences are complex (Teichgräber et al., 2008). In addition to these mechanisms,
and mostly unknown. COPD results from responses to which explain the high incidence of chronic bacterial lung
environmental stimuli, particularly from smoking, in ge- infections in CF, other investigators have demonstrated
netically susceptible individuals (Celli & MacNee, 2004; that alveolar macrophages from CF mice exhibit defective
Wood & Stockley, 2006). Bacterial pathogens also play an killing of internalized P. aeruginosa (Di et al., 2006). After
important role in the course and pathogenesis of COPD phagocytosis by alveolar macrophages, bacteria enter the
(Sethi & Murphy, 2008). phagosome and lysosomes, exhibiting a low pH due to
Many similarities exist between CF and COPD. In both CFTR and vacuolar ATPase, and fuse with the phagosome,
diseases, exuberant inflammation causes airflow obstruc- forming the phagolysosome. The mature organelle exhibits
tion, tissue destruction and extensive tissue remodelling, an acid pH that ensures the final destruction of the ingested
leading to emphysema and fibrosis. Thus, inflammation bacteria by a variety of proteolytic enzymes. Defective
plays an important role in the morbidity and mortality of CFTR-dependent acidification of lysosomal pH (Barasch
patients. However, CF and COPD differ with regard to et al., 1991) has been linked to significant survival of
infection. While the role of chronic bacterial infections, P. aeruginosa (Di et al., 2006). The contribution of CFTR to
particularly caused by Pseudomonas aeruginosa, is well the regulation of pH in intracellular organelles has been
established in CF (Høiby & Frederiksen, 2000), the impact discussed controversially (Haggie & Verkman, 2007, 2009)
of these infections in patients with COPD is less clear due to experimental difficulties to ensure targeting of
(Murphy et al., 2008). Furthermore, pathogens such as identical vesicle populations by pH-sensitive fluorescent
Moraxella catarrhalis and Streptococcus pneumoniae are dyes. Pseudomonas aeruginosa infection may also be facili-
frequently observed in patients with COPD (Sethi & Mur- tated in CF airways directly by defective CFTR, which, in its
phy, 2001), and yet are seldom present in the airways of CF functional state, binds the pathogen within lipid rafts and
patients. Because of improved symptomatic treatment removes it from the epithelial surface via internalization
strategies and antibiotic therapy, the prognosis of CF (Pier, 2000).
individuals has improved considerably and many children The viscosity of the secretions has several further negative
now reach adulthood. While antibiotic therapy is beneficial consequences for the pathophysiology of CF lung disease.
in treating acute exacerbations of COPD (Anthonisen, 2002; These secretions are present in submucosal gland ducts of
Ram et al., 2006), this therapy has not been demonstrated to CF patients as a result of mutated CFTR (Engelhardt et al.,
affect the course of the disease. In this review, we will discuss 1992), and may impair the transport of antimicrobial
the pathophysiology of lung disease in CF and COPD, oligopeptides (Lehrer & Ganz, 2002; Selsted & Quellette,
bacterial colonization of airways and the complex genotypic 2003) onto the epithelium (Verkman et al., 2003; Joo et al.,
and phenotypic adaptation processes of the opportunistic 2004) (Fig. 1). They may also negatively affect the migration
bacterial pathogens in these diseases and novel treatment of neutrophils towards the pathogens within the mucus
options. overlaying the respiratory epithelium (Matsui et al., 2005)

FEMS Microbiol Rev 35 (2011) 124–146 


c2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
126 G. Döring et al.

Fig. 1. Bacterial killing mechanism of innate


immunity in the respiratory tract of CF patients
and healthy individuals. (a) In healthy individuals,
bacteria entering the mucus layer overlaying the
respiratory epithelium are effectively removed
from the airways by a functional mucociliary
clearance system and killed by CAMPs derived
from submucosal glands or epithelial cells, by
functional neutrophils (PMN) or macrophages
(MF) or within epithelial cells after uptake via
functional CFTR. (b) In individuals with CF,
defective CFTR leads to a highly viscous mucus
layer that impairs mucociliary clearance, the
migration of CAMPs, neutrophils and
macrophages towards the bacterial targets, the
uptake of bacteria by CFTR itself, and induces a
pH shift to alkaline values in epithelial lysosomes.
Because of ceramide accumulation, this results in
DNA deposits, which in turn serve as adhesion
matrices for bacteria. A similar pH shift in the
phagolysosomes of macrophages impairs
bacterial killing. Ceramide accumulation also
triggers cytokine release, which induces further
neutrophil influx (adapted from Döring &
Gulbins, 2009).

(Fig. 1). This may allow bacterial multiplication and pro- tory response dominated by neutrophils not only facilitates
found changes in the bacterial phenotypes that facilitate tissue destruction but also induces tissue remodelling
chronic infection. Furthermore, within the highly viscous (Döring & Gulbins, 2009; Ulrich et al., 2010). An essential
mucus, a microaerobic/anaerobic milieu prevails due to role in these pathogenic reactions is related to the imbalance
oxygen consumption by bacterial pathogens such as P. of serine and metalloproteases to their respective inhibitors
aeruginosa (Worlitzsch et al., 2002; Hassett et al., 2009) or (Döring, 1999; Ratjen et al., 2002). Particularly, neutrophil
invading neutrophils (Kolpen et al., 2010). Consequently, elastase has been implicated in the pathophysiology of CF
the generation of reactive oxygen species (ROS) by neutro- lung disease. The serine protease has been shown to cleave
phils and other cells is abolished and bacterial killing is the endogenous serine protease inhibitor a1-antitrypsin,
impaired (Fig. 2a and b), particularly with regard to patho- immunoglobulins, surfactant proteins, complement com-
gens, which are intrinsically resistant to nonoxidative kill- ponents and a variety of cell receptors (Döring & Gulbins,
ing, or capable of changing their phenotype towards 2009). Cleavage of the complement receptor 1 on neutro-
resistance to nonoxidative killing. phils and the activated complement component C3bi on
Lung inflammation is a key finding in patients with CF opsonized bacteria creates an ‘opsonin-receptor mismatch’
(Konstan et al., 1994). While ceramide accumulation has (Berger et al., 1989; Tosi et al, 1990), which significantly
been shown to induce a proinflammatory status in the impairs P. aeruginosa killing.
respiratory tissue of CF mice, which precedes bacterial The vicious cycle of persisting bacterial pathogens, en-
infection, including an increased production of cytokines cased in biofilms, and decaying neutrophils leads to increas-
and recruitment of macrophages (Teichgräber et al., 2008), ing endobronchial DNA deposition and large mucus plugs,
inflammation clearly increases considerably in response to which further obstruct the airways of the patients (Whit-
persistent bacterial infections. The exaggerated inflamma- church et al., 2002; Worlitzsch et al., 2002).


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 127

of airway inflammation – increased neutrophils, IL-8, TNF-


a and neutrophil elastase – are all increased compared with
healthy controls (Keatings et al., 1996; Hill et al., 1999a b;
Barnes et al., 2003). Chronic colonization of the airways
contributes to this airway inflammation (Hill et al., 2000;
Parameswaran et al., 2009). However, the degree of airway
inflammation does not reach the threshold of patient-
perceived symptoms. When airway inflammation crosses
this threshold for short episodes, these are called exacerba-
tions (Hill et al., 1999a, b). The course of COPD is punc-
tuated by episodes of acute exacerbations, which are
associated with increased airway inflammation compared
with baseline levels (Hill et al., 1999a, b; Sethi et al., 2008).
Goblet cell hyperplasia and mucus hypersecretion are pre-
sent in the airways of most COPD patients, especially in
those with clinical features of chronic bronchitis (Barnes,
2000). Although mucus viscosity is considerably lower in
COPD compared with CF, mucociliary clearance of bacterial
pathogens may be impaired by the fact that respiratory
epithelial cells lose cilia and undergo squamous metaplasia
(Hogg & Timens, 2009). Lung inflammation leads to the
activation of innate immune cells, particularly macro-
phages, which have a profound effect on immune cell
recruitment into the airways and tissue remodelling (Barnes,
2004). In the small airway walls, infiltrating neutrophils and
lymphocytes cause airway thickening and remodelling,
resulting in reduced airway diameter and airflow (Hogg
et al., 2004). Inflammatory cellular infiltration of alveolar
walls is associated with the destruction of alveoli and
enlargement of air spaces, i.e. emphysema (Cosio et al.,
2009). Infiltration especially by CD81 lymphocytes occurs
in pulmonary vessel walls that are thickened due to in-
creased collagen deposition (Cosio et al., 2009). As in CF,
chronic lung inflammation is accompanied by an imbalance
between proteases and antiproteases in COPD (Stockley,
1999). Furthermore, oxidative stress is persistently enhanced
(Rahman & MacNee, 1996) as well as a number of other
Fig. 2. Bacterial sensitivity and resistance to neutrophil-mediated killing
innate immune functions that lead to tissue remodelling
in aerobic and anaerobic environments. (a) On the respiratory epithelium
of healthy individuals, neutrophils effectively kill the CF-related bacterial (Barnes, 2000; Cosio et al., 2009; Hogg & Timens, 2009).
pathogens Pseudomonas aeruginosa (red), Staphylococcus aureus (blue) Alveolar macrophages from COPD patients have decreased
and Burkholderia cepacia (green) by ROS and CAMPs. (b) Neutrophils in sensitivity to nontypeable Haemophilus influenzae (NTHI)
the anaerobic milieu in the mucus layer overlaying the respiratory antigens and decreased ability to phagocytose NTHI, com-
epithelium in individuals with CF are devoid of ROS, leading to intraneu- pared with normal controls (Berenson et al., 2006a, b). The
trophil survival of S. aureus (blue), which produces a protective exopoly-
structural abnormalities in airways, alveoli and pulmonary
saccharide coat (red), and B. cepacia (green), which is intrinsically
vasculature vary among COPD patients (Hogg & Timens,
resistant to nonoxidative killing (adapted from Döring & Gulbins, 2009).
2009).

Bacterial colonization of the CF lung


Pathophysiology of COPD Chronic bacterial pulmonary infection leading to an irre-
The pathophysiology of COPD is characterized by chronic versible decline in lung function is the main cause of
inflammation of airways as a result of active and passive mortality and morbidity in CF patients, with 4 95% of
smoking (Barnes, 2000; Celli & MacNee, 2004). The indices deaths due to respiratory failure (Dodge et al., 2007).

FEMS Microbiol Rev 35 (2011) 124–146 


c 2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
128 G. Döring et al.

Staphylococcus aureus is the most prevalent opportunistic fascinating view is that ‘cheater’ cells that refrain from
bacterial pathogen, which chronically infects CF airways in producing ‘expensive’ metabolites benefit in such commu-
the age group of 0–9 years (55%), while P. aeruginosa is the nities from those produced by the rest of the population
most prevalent pathogen thereafter (81%) (Høiby & Freder- (Keller & Surette, 2006; Duan et al., 2009). The chemical and
iksen, 2000). At a much lower prevalence, H. influenzae, S. physical nature of the mucus plugs in CF airways, in which
pneumoniae, Escherichia coli, strains of the Burkholderia the bacterial pathogens multiply, favours such a microevo-
cepacia complex (Bcc), comprising 10 genomovars, and lution during the course of chronic infection. Interestingly,
fast-growing mycobacteria can be present in airway speci- it has been found that the number of taxa and the phyloge-
mens of CF patients (Høiby & Frederiksen, 2000). netic diversity and evenness within the microbiota in the
In addition, a large number of strict anaerobes have been airways of CF patients decrease with age, which has been
detected, which may be surprising for an air-filled organ such attributed to either repeated antibiotic therapy courses or
as the lung. However, the viscous mucus plugs, in which all invasion of P. aeruginosa into the CF airways (Klepac-Ceraj
these bacteria thrive, have been shown to create microaerobic/ et al., 2010). Microevolution processes have been studied
anaerobic conditions (Worlitzsch et al., 2002), which favour most widely in P. aeruginosa.
the growth of anaerobes. Anaerobic bacteria have been
detected in significant numbers (4105 CFU mL1) in the
lungs of CF patients using classic culturing methods, in-
Pseudomonas aeruginosa is the most prevalent
pathogen
dicating that a highly diverse bacterial community is present
within the CF lung (Brook & Fink, 1983; Jewes & Spencer, The capacity of P. aeruginosa to cause infections in CF
1990; Tunney et al., 2008; Worlitzsch et al., 2009). These patients and other hosts can be traced back to its consider-
include diverse species belonging to the genera Prevotella, able adaptability to specific environmental conditions and
Bifidobacterium, Veillonella, Peptostreptococcus and Fusobac- the production of numerous virulence factors (Lee et al.,
terium. Single obligate anaerobic species persisted for up to 2006) (see Box 1). This adaptability is linked to a large
11 months in sputum plugs in vivo (Worlitzsch et al., 2009). number of regulatory genes. For example, in the 6.3 Mb
Whether these bacteria contribute to the pathophysiology of genome of P. aeruginosa PAO1, which contains 5570 pre-
CF lung disease is still an open question. CF patients with dicted ORFs, 521 known or putative regulatory genes have
and without obligate anaerobes in sputum specimens did been identified, demonstrating a highly complex gene
not differ in lung function (Worlitzsch et al., 2009). regulation (Stover et al., 2000). The P. aeruginosa chromo-
Culture-independent profiling methodologies (Nocker some contains a core genome consisting of the sequences
et al., 2007) such as terminal restriction fragment length that are common to all strains of the taxon and variable
polymorphism have considerably advanced our knowledge amounts of accessory DNA segments. The accessory genome
of the polymicrobial nature of CF lung disease (Rogers et al., that can be horizontally transferred among strains repre-
2003, 2004, 2006, 2009; Harris et al., 2007; Bittar et al., 2008; sents the flexible gene pool that frequently undergoes
Sibley et al., 2008, 2009; Klepac-Ceraj et al., 2010). For acquisition and loss of genetic information and hence plays
instance, it has been shown by culture-independent meth- an important role in the adaptive evolution of bacteria. The
ods that the Streptococcus milleri group of viridans strepto- flexible gene pool is made up of elements such as bacter-
cocci, comprising Streptococcus constellatus, Streptococcus iophages, plasmids, insertion elements, transposons, con-
intermedius and Streptococcus anginosus, is present in CF jugative transposons, integrons and genomic islands.
sputum specimens and related to the pathophysiology of CF Isolates from CF patients and environment are endowed
lung disease (Sibley et al., 2008). with a variable repertoire of genomic islands (Klockgether
During acute infections, which may resolve after several et al., 2007; Wiehlmann et al., 2007). The genes required for
days, microorganisms reversibly regulate gene expression by pathogenicity in one strain of P. aeruginosa are neither
responding to environmental signals, and thereby adapt required for nor predictive of virulence in other strains
their phenotypes accordingly (Green & Paget, 2004; Gilles- (Lee et al., 2006; Bragonzi et al., 2009), indicating a broad
Gonzalez & Gonzalez, 2005). During the course of chronic intraclonal diversity of pathogenicity of this organism, yet
infections, which may last 4 10 years, however, mutants no difference in virulence. Despite the high overall genome
can be selected that differ genotypically and phenotypically similarity of P. aeruginosa of o 0.5% sequence diversity in
from the originally infecting strain. Given the large micro- the core genome, comprising 80% of all genes, differences in
biota in the airways of infected CF patients, complex phenotypes can be striking even at the intraclonal level of
interactions not only occur between microorganisms and genetically highly related strains (Wehmhöner et al., 2003;
the environment but also among members of the commu- Salunkhe et al., 2005a, b).
nity that use primary or secondary metabolites in a multi- Various morphotypes that are not normally present in
tude of chemical interactions (Duan et al., 2009). A environmental P. aeruginosa strains have been described in


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 129

Box 1. Virulence factors of Pseudomonas aeruginosa and their regulation by quorum sensing
Pseudomonas aeruginosa produces many virulence factors, which can be divided into cell-associated and extracellular virulence factors. Many
virulence factors are regulated by the quorum-sensing circuit and other regulatory genes, which have been demonstrated in vitro and in vivo. During
the chronic course of lung infections in patients with CF, many mutants have been characterized that are defective in virulence factor expression and
quorum sensing (D’Argenio, 2004; Hogardt et al., 2004, 2007; van Delden, 2004; Ramsey & Wozniak, 2005; Lee et al., 2006; Williams & Camara,
2009; Winstanley & Fothergill, 2009).
Cell-associated virulence factors
Flagella, type IV pili, lipopolysaccharide and three different exopolysaccharides, encoded by the alg, psl and pel clusters, contribute to adhesion of the
microorganism to various surfaces. On epithelial cells, flagellar proteins bind to toll-like receptor (TLR)-5 and lipopolysaccharide to TLR-4, leading to
the stimulation of innate immune functions and inflammation. The exopolysaccharides have been implicated in biofilm formation. Flagella,
lipopolysaccharide and alginate antibodies have been detected in the sera of infected CF patients.
Extracellular virulence factors
Type I, II and III secretion systems secrete protein toxins and provide contact with eukaryotic cells. Alkaline protease is secreted by the type I secretion
system, while the LasB and LasA elastases, protease IV, exotoxin A, phospholipase C and lipase are secreted by the type II secretion system. The type III
secretion system injects the exoenzymes S, T, Y and U into eukaryotic cells. Other extracellular virulence factors include the secondary metabolites
pyocyanin and other phenazine pigments, siderophores, hydrogen cyanide and rhamnolipids. Most exoenzymes and secondary metabolites have been
shown to be cytotoxic in in vitro and in animal models, and antibodies to many of these factors have been detected in the sera of infected CF patients.
Quorum-sensing circuit
Several virulence factors are regulated in a cell-density-dependent manner by the quorum sensing circuit composed of lasRI, rhlRI and pqs systems.
Pseudomonas aeruginosa produces several diffusible signal molecules with autoinducer properties such as N-3-oxo-dodecanoyl homoserine lactone
(produced by the LasI enzyme) and N-butanoyl homoserine lactone (produced by theRhlI enzyme), which bind at critical concentrations to the
corresponding regulators LasR and RhlR. The lasRI system mainly regulates lasB, lasA, exoA, xcpP and xcpR expression. The rhlRI system regulates the
expression of rpoS, rhamnolipid, LasB and LasA elastase, hydrogen cyanide, pyocyanin, lipase and alkaline protease. The Pseudomonas quinolone signal
system potentiates the effects of the rhlRI system. Quorum sensing controls not only virulence in P. aeruginosa but also many other genes involved in
cellular processes, including chemotaxis and biofilm formation, comprising altogether about 10% of the P. aeruginosa genome. In addition, quorum-
sensing molecules also modulate host responses.

P. aeruginosa strains, isolated from chronically infected CF functions but also against opsonophagocytosis involving
patients, including strains that are mucoid, smooth, rough, specific antibodies, produced by the adaptive immune
dwarf, colourless or present as small colony variants (SCV) system. A key element in alginate regulation is the alter-
(size: 1–3 mm). The latter may contribute to the progressive native sigma factor AlgT, also known as AlgU, RpoE or s22,
pulmonary damage in CF and persistence in CF airways which induces the expression of algD and increases the
(Häussler et al., 1999, 2003; Lory et al., 2009; Starkey et al., expression of regulatory proteins that enhance algD tran-
2009). Additionally, ‘rugose SCVs’ have been detected that scription (Ramsey & Wozniak, 2005). The anti-sigma factor
have characteristics of enhanced biofilm formation and MucA inhibits AlgT; thus, inactivation of mucA leads to
display increased expression of the pel and psl polysacchar- upregulation of AlgT and conversion to mucoidy. Over-
ide gene clusters, decreased expression of motility functions production of alginate has been linked to biofilm formation
and a defect in growth on some amino acid and tricarboxylic in P. aeruginosa – a phenotype that favours chronic persis-
acid cycle intermediates as sole carbon sources (Starkey tence of the pathogen in murine and CF lungs (Hoffmann
et al., 2009). The psl (polysaccharide synthesis locus) genes et al., 2005). However, both mucoid and nonmucoid
are responsible for the production of mannose-rich exopo- P. aeruginosa clonal strains occur in the airways of CF
lysaccharides and the pel locus for pellicle formation. patients simultaneously (Bragonzi et al., 2009). Further-
A consistent finding in CF airways is the presence of more, the finding that a high number of nonmucoid strains
mucoid P. aeruginosa strains that overproduce the exo- from CF patients carry a mucA mutation (Bragonzi et al.,
polysaccharide alginate, a negatively charged polymer of 2006) suggests that a mucoid morphotype, intermittently
b-1-4-linked D-mannuronate, and its C5 epimer, L-gulur- selected in the CF lungs, can be suppressed at a later stage
onate (Deretic et al., 1995; Boucher et al., 1997). Alginate due to other, secondary site mutations (Boles et al., 2004),
expression is upregulated by microaerobic environmental including mutations in algU (Ciofu et al., 2008). Interest-
conditions that are present in the viscous mucus of CF ingly, P. aeruginosa rpoS mutants reveal a significantly
patients (Worlitzsch et al., 2002). Alginate expression is reduced production of alginate (Suh et al., 1999). Mucoid
considered to be an important factor for the persistence of P. aeruginosa strains from CF patients also contain increased
P. aeruginosa in the airways of CF patients as alginate levels of G6PDH activity and expression of the correspond-
protects the pathogen not only against innate immune ing zwf gene. This allows the bacterium to supply sufficient

FEMS Microbiol Rev 35 (2011) 124–146 


c2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
130 G. Döring et al.

amounts of precursor for the alginate pathway (Silo-Suh The genetic analysis of clonal isolates, obtained from the
et al., 2005). CF lung of single patients over several years, revealed the
Other phenotypic changes of P. aeruginosa strains occur- presence of mutations in many genes (Mahenthiralingam
ring during the course of chronic CF lung infection include et al., 1994; Anthony et al., 2002; Nguyen & Singh, 2006;
loss of motility (Luzar et al., 1985; Rau et al., 2010), Smith et al., 2006; Tümmler, 2006; Hogardt et al., 2007;
decreased exotoxin A expression (Suh et al., 1999), loss of Hoboth et al., 2009; Mandsberg et al., 2009; Ciofu et al.,
O antigen in lipopolysaccharide (Hancock et al., 1983) and 2010; Rau et al., 2010). When a P. aeruginosa isolate was
altered lipid moieties (Ernst et al., 1999; Sabra et al., 2003). compared with its clonal variants, isolated 90 months later
The latter change confers enhanced resistance to aminogly- from a single CF patient, revealing 68 mutations, the
coside antibiotics, cationic antimicrobials such as polymyx- analysis suggested a reduced virulence of the latter strains
in E (colistin) and potentially to cationic peptides produced with regard to their ability to induce acute infections, based
by the host (defensins). Mutations in nfxB, the repressor of on mutations in many virulence genes including type III
the MexCD-OprJ operon, results in increased resistance to secretion, quorum sensing and motility (Smith et al., 2006)
fluoroquinolones (Rau et al., 2010). Furthermore, increased (Table 1). The progressive microevolution of P. aeruginosa in
auxotrophy (Taylor et al., 1993; Thomas et al., 2000), defects CF patients, known as adaptive radiation, has been inter-
in type II (Woods et al., 1986) and III secretion (Jain et al., preted as an in vivo selection process, resulting in less
2004; Lee et al., 2005; Hogardt et al., 2007; Rau et al., 2010), virulent variants that consequently do less harm to their
reduced production of proteases and phospholipase C, loss host than the original colonizing strain (Smith et al., 2006).
of pyoverdine, pyocins and elastase expression (Hogardt Although this notion may actually be correct, the defini-
et al., 2007), altered metabolic activities (Silo-Suh et al., tion of virulence also includes the capacity of the pathogen
2005) and antibiotic resistance (Thomassen et al., 1979; to persist in a given host, causing chronic infection. To test
Oliver et al., 2000; Smith et al., 2006; Hogardt et al., 2007) this hypothesis, Bragonzi et al. (2009) isolated P. aeruginosa
have been described in P. aeruginosa strains from CF clones from airways of six CF patients during a period of up
patients. Particularly during acute exacerbations, methio- to 16.3 years and assessed the virulence of clonal variants in
nine-dependent auxotrophic mutants of P. aeruginosa have mice by monitoring acute and chronic lung infection.
been isolated from CF patients (Taylor et al., 1993). In Because the definition of virulence is also dependent on a
conclusion, chronic exposure to the CF environment selects specific host, they tested the clonal variants in mice with
for a variety of mutations in P. aeruginosa strains. different genetic backgrounds, including CF mice. The data
The frequency of mutations is increased in hypermutable or demonstrate that early and intermediate P. aeruginosa CF
mutator strains that reveal defects in genes involved in the isolates are more virulent to cause acute infections (e.g.
DNA repair or error avoidance systems, particularly in the bacteraemia) in mouse lungs than late isolates; however,
methyl-directed mismatch repair system (Oliver et al., 2002; early, intermediate and late isolates do not differ in their
Ciofu et al., 2005, 2010; Hogardt et al., 2007; Montanari et al., capacity to cause chronic infection (Fig. 3). These findings
2007). The percentage of CF patients (60%) carrying at least suggest that clonal expansion of P. aeruginosa strains during
one hypermutable strain is remarkably high (Montanari et al., microevolution within CF lungs leads to populations with
2007). Analysis of the genetic background of the P. aeruginosa altered, but not reduced virulence in mouse lung models.
mutator phenotypes from CF airways showed that mutS was Thus, it may be questionable to define late isolates as less
the most commonly affected gene, followed by mutL (Ciofu virulent than early isolates or to describe the pathogenicity
et al., 2010). A sequential analysis of CF isolates revealed that of CF lung disease as a process leading to an increasingly
mutations in mucA, leading to the mucoid phenotype and benign situation. However, the animal experiments used to
lasR, leading to loss of quorum sensing (Box 1), occurred determine virulence do not reflect a natural colonization
earlier than mutations in the mut genes, showing that hyper- process with initially small numbers of bacteria.
mutability is not a prerequisite for the acquisition of mucoidy Pseudomonas aeruginosa proteins are upregulated in the
and loss of quorum sensing (Ciofu et al., 2010). Accumulation CF lung, including the outer membrane protein OprF and
of mutators in the CF lung is associated especially with enzymes of the arginine deiminase pathway (Hoboth et al.,
multidrug resistance development, thus suggesting that the 2009) (Table 2). These changes indicate an adaptive shift
intensive antibiotic treatment is one of the main selective towards constitutive expression of genes required for growth
pressures for the maintenance and amplification of mutators under the nutritional and microaerobic conditions present
(Oliver et al., 2000; Ciofu et al., 2010). The acquisition of a in the sputum of CF patients. The high viscosity of the CF
stable mutator phenotype may confer a selective advantage for mucus also leads to a limited diffusion of siderophore
bacteria, particularly in stressful and/or fluctuating environ- molecules and therefore to an increased fitness of side-
ments because this may increase the probability of generation rophore-producing cells (Cornelis et al., 2009; Kummerli
of adaptive variants (Taddei et al., 1997). et al., 2009).


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 131

Table 1. Gene mutations present in Pseudomonas aeruginosa cystic fibrosis isolates


Name Annotation Function References
mexZ w
PA2020 Transcriptional regulator of multidrug efflux 1
lasRw PA1430 Transcriptional regulator of quorum sensing 1
PA0313 Probable permease of the ABC transporter 1
mexAw PA0425 RND multidrug efflux membrane fusion protein 1
accCw PA4848 Biotin carboxylase 1
vfrw PA0652 Transcriptional regulator of lasR 1
mexSw PA2491 Probable oxidoreductase 1
exsAw PA1713 Transcriptional regulator of type III secretion 1
PA0506 Probable acyl-CoA dehydrogenase 1
wspF PA3703 Probable methylesterase involved in chemosensory signal 1
transduction
rpoN PA4462 RNA polymerase s-54 factor 1
fleQ PA1097 Transcriptional regulator of flagella synthesis 1
mexT PA2492 Transcriptional regulator of multidrug efflux 1
nalD PA3574 Probable transcriptional regulator 1
ampD PA4522 b-Lactamase expression regulator 1
PA0366 Probable aldehyde dehydrogenase 1
cyaB PA3217 Adenylate cyclase 1
pilB PA4526 Type 4 fimbrial biogenesis protein 1
PA1333 Hypothetical protein 1
PA3565 Probable transcriptional regulator 1
PA2312 Probable transcriptional regulator 1
anr PA1544 Transcriptional regulator of anaerobic metabolism 1
rhlR PA3477 Transcriptional regulator of quorum sensing 1
phoP PA1179 Two-component response regulator 1
rhlI PA3476 Autoinducer synthesis protein 1
PA4796 Hypothetical protein 1
pqsB PA0997 Homologous to b-keto-acyl-acyl carrier 1
protein synthase 1
toxR PA0707 Transcriptional regulator 1
PA2435 Probable cation-transporting P-type ATPase 1
PA4420 Hypothetical protein 1
PA2121 Hypothetical protein 1
mutS PA3620 DNA mismatch repair protein MutS 2–4
mutL PA4946 DNA mismatch repair protein MutL 3, 4
mutY PA5147 A/G specific adenine glycosylase 4, 5
uvrD PA5443 DNA helicase II 3, 4
mucA PA0763 Anti-sigma factor MucA 4, 6
nfxB PA4600 Transcriptional regulator 7
Numbers relate to the genome of PAO1. References 1, Smith et al. (2006); 2, Hogardt et al. (2007); 3, Montanari et al. (2007); 4, Ciofu et al. (2010); 5,
Mandsberg et al. (2009); 6, Anthony et al. (2002); 7, Rau et al. (2010).
w
Genes that are mutated in 4 5% of isolates.

Other bacterial pathogens in CF glucosamine (PNAG), differs structurally from the S. aureus
capsule polysaccharides, which are lost during growth in the
Staphylococcus aureus also changes its phenotype under CF airways due to elevated pCO2 (4%) (Herbert et al.,
microaerobic/anaerobic environmental conditions in vitro 1997). The PNAG-encased cells confer resistance to nonox-
and in the CF mucus, forming biofilm-like aggregates idative killing (Ulrich et al., 2007). Phage mobilization,
(McKenney et al., 1999; Cramton et al., 2001; Ulrich et al., possibly via transduction, contributes significantly to gen-
2007), which differ genotypically and phenotypically from ome alteration in S. aureus during infection in CF patients
environmental strains and strains from the nasal habitat (Goerke et al., 2004), and agr mutants are frequently isolated
(Goerke & Wolz, 2004). The exopolysaccharide present in from these specimens (Goerke et al., 2000). Agr (for
S. aureus biofilms (Cramton et al., 1999), initially named accessory gene regulator) is a well-characterized global
polysaccharide intracellular adhesin and later poly-N-acetyl regulatory locus that is expressed from two promoters: P2

FEMS Microbiol Rev 35 (2011) 124–146 


c 2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
132 G. Döring et al.

Clinical strains 2008). Treatment of CF lung infections using the amino-


100 glycoside tobramycin may result in local subinhibitory
concentrations that favour the emergence of aminoglyco-
80
side-resistant SCVs (Mitchell et al., 2010), involving the
60 global regulator SigB (Biswas et al., 2009). Besides antibio-
% Mortality, survival, chronic

tics, coinfecting P. aeruginosa can also promote the forma-


infection and clearance

40
tion of S. aureus SCVs by producing respiration inhibitors
20 such as 4-hydroxy-2-heptylquinoline-N-oxide (Hoffman
0 et al., 2006; Biswas et al., 2009). Molecular characterization
100
of SCVs from CF patients also revealed that they may be
80
defective in the expression of the agr system (Novick et al.,
60 1993). Taken together, the adaptation of S. aureus to the CF
40
lung leads to biofilm formation and SCVs, both of which
favour chronic infection states as a result of increased
20 resistance to innate immune defence mechanisms and anti-
0 biotic treatment. Without optimized anti-staphylococcal
treatment strategies that reduce the bacterial load in the
Fig. 3. Virulence of Pseudomonas aeruginosa strains in a murine
model of airways infection. C57Bl/6NCrlBR mice were infected with airways (Döring et al., 2004), chronic S. aureus lung infec-
1–2  106 CFU per lung of 32 P. aeruginosa different strains embedded in tions may be considerably harmful for the CF patient.
agar beads. Mortality induced by bacteraemia (red) and survival (grey) Indeed, before the introduction of anti-staphylococcal
were evaluated on challenged mice. Clearance (white) and capacity to agents, virtually all CF patients were infected with this
establish chronic airways infection (green) after 14 days from challenge opportunistic pathogen (Andersen, 1938), which was held
were determined on surviving mice. The data show the percentage of
responsible for the widespread pulmonary changes in CF
mice infected with P. aeruginosa reference strains (PAO1, n = 66; PA14,
(di Sant’Agnese & Talamo, 1967).
n = 14), a pool of environmental strains (n = 65) and a pool of clinical
strains (n = 274), isolated at the onset (n = 91), during the intermediate The Bcc is a group of genetically distinct, but phenotypi-
phase of colonization (n = 65) or later during chronic infection (n = 118) cally similar bacteria that are divided into at least nine
(adapted from Bragonzi et al., 2009). species (Coenye et al., 2001). All group members can cause
infections in CF patients, albeit with a low prevalence
(2–3%). Bcc strains are extremely sensitive towards ROS;
and P3 (Novick et al., 1993, 1995). Agr regulates staphylo- this is reflected by the minimal mortality of neutropenic
coccal virulence and other accessory gene functions, includ- mice when infected with this opportunistic pathogen (Chu
ing the expression of haemolysins. While the P2 RNAII et al., 2002). However, in sputum plugs in airways of CF
transcript codes for the quorum-sensing elements AgrA, patients where ROS are essentially absent due to the
AgrC, AgrB and AgrD, P3 activation, during the late anaerobic/microaerobic environment and in patients with
exponential growth phase, leads to the transcription of the hereditary disorder chronic granulomatous disease
RNAIII, resulting in the repression of surface proteins such (CGD) who suffer from mutations in NADPH oxidase, this
as adhesins or protein A and in the enhanced expression of opportunistic pathogen is highly virulent due to its intrinsic
exoproteins such as haemolysins (Novick, 2003). Agr mu- resistance to nonoxidative killing during phagocytosis by
tants show enhanced biofilm formation in S. aureus, as Agr professional phagocytes (Speert et al., 1994; Pollock et al.,
activation results in the expression of extracellular proteases 1995; Baird et al., 1999; Sahly et al., 2003; Segal et al., 2003;
(Boles & Horswill, 2008) and membrane-active molecules Sousa et al., 2007). Resistance to cationic antimicrobial
(Kong et al., 2006), such as the d-toxin, which contribute to peptides (CAMPs) has been linked to the presence of 4-
the dispersal of biofilms. amino-4-deoxyarabinose moieties, which are attached to the
Often SCVs of S. aureus, which may persist in respiratory phosphate residues in the lipid A backbone of Bcc lipopoly-
epithelial cells, are detected in CF lung specimens (Sadowska saccharides (Cox & Wilkinson, 1991; Vinion-Dubiel &
et al., 2002; von Eiff, 2008). SCVs are mostly nonpigmented Goldberg, 2003).
and nonhaemolytic and their colonies are 10 times smaller While some strains may cause long-term asymptomatic
than those of the parental strain. In general, SCVs are airway colonization, others cause a rapid decline of lung
auxotrophic for haemin and menadione, compounds in- function in CF patients and are highly transmissible, result-
volved in the biosynthesis of electron transport chain ing in patient-to-patient spread within and between CF
components. Most SCVs of S. aureus, isolated from CF centres (Govan et al., 1993; Mahenthiralingam et al., 2005).
patients, are thymidine-auxotrophic due to long-term tri- For instance, Burkholderia cenocepacia strains of the multi-
methoprim-sulphomethoxazol treatment (Schneider et al., locus enzyme electrophoresis type 12 lineage can cause a


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 133

Table 2. Metabolic adaptation of Pseudomonas aeruginosa strains during chronic lung infection in cystic fibrosis patients based on protein expression
Name Annotation Function
rbsB PA1946 Binding protein RbsB of ABC ribose transporter
aotJ PA0888 Arginine-ornithine-binding protein
PA5153 Probable lysine-arginine-ornithine-binding periplasmic protein
PA1260 Probable lysine-arginine-ornithine-binding periplasmic protein
braC PA1074 Periplasmic branched-chain amino acid transport protein
spuD PA0300 Polyamine transport protein
oprFw PA1777 Outer membrane porin OprF
oprD PA0958 Outer membrane porin OprD
fda PA0555 Fuctose-1,6-bisphosphate aldolase
adk PA3686 Adenylate kinase
azuw PA4922 Azurin precursor
ccpRw PA4587 Cytochrome c551 peroxidase precursor
leuD PA3120 3-Isopropylmalate dehydratase small subunit
hisF1 PA5140 Imidazoleglycerol-phosphate synthase, cyclase subunit
arcAw PA5171 Arginine deiminase
arcCw PA5173 Carbamate kinase
PA2888 Probable biotin-dependent carboxylase
acpP PA2966 Acyl carrier protein
accB PA4847 Acetyl-CoA carboxylase
ppaw PA4031 Inorganic pyrophosphatase
trxA PA5240 Thioredoxin
sucC PA1588 Succinyl-CoA synthetase (b-chain)
ansB PA1337 Glutaminase-asparaginase
lpd3 PA4829 Dihydrolipoamid dehydrogenase 3
aceBw PA0482 Malate synthase G (glyoxylate shunt)
atpF PA5558 ATP synthase B chain
atpH PA5557 ATP synthase H chain
pdxH PA1049 Pyridoxamin 5-phosphate oxidase
glnK PA5288 Nitrogen-regulatory protein P-II 2
atoD PA1999 Probable CoA-transferase, subunit A
atoB PA2001 Acetyl-CoA acetyltransferase
PA0318 Hypothetical protein
PA2575 Hypothetical protein
PA0388 Hypothetical protein
PA3440 Hypothetical protein
PA1677 Hypothetical protein
PA5178 Hypothetical protein
PA3309 Hypothetical protein
PA5339 Hypothetical protein
btuEw PA0838 Glutathione peroxidase
sodB PA4366 Superoxide dismutase
kynB PA2081 Kynurenine formamidase
bfrA PA4235 Bacterioferritin A
katA PA4236 Katalase
ahpC PA0139 Alkyl hydroperoxide reductase subunit C
tsaA PA3529 Probable peroxidase
ibpA PA3126 Heat-shock protein IbpA
ppiB PA1793 Peptidyl-prolyl cis-trans isomerase B
groEL PA4385 GroEL
rpsF PA4935 30S ribosomal protein S6
PA4671 Probable ribosomal protein L25
greA PA4755 Transcription elongation factor GreA
rplL PA4271 50S ribosomal protein L7/L12
tsf PA3655 Elongation factor
tufA PA4265 Elongation factor Tu
guaB PA3770 Inosine-5 0 -monophosphate dehydrogenase
pilH PA0409 Twitching motility protein PilH

FEMS Microbiol Rev 35 (2011) 124–146 


c 2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
134 G. Döring et al.

Table 2. Continued.
Name Annotation Function
fliC PA1092 Flagellin type B
murF PA4416 UDP-N-acetylmuramoylalanyl-D-glutamyl-2,
6-diaminopimelate-D-alanyl-D-alanyl ligase
ftsZ PA4407 Cell division protein FtsZ
Adapted from Hoboth et al. (2009).
w
Genes whose expression tends to be strongly upregulated during late chronic infection.

fulminate, necrotizing pneumonia, which may occasionally aeruginosa are detectable in airways of approximately
be complicated by rapidly fatal septicaemia termed the 25–50% of clinically stable COPD patients (Sethi & Murphy,
cepacia syndrome in CF patients (Corey & Farewell, 1996; 2008). Pathogenic bacteria in the lower airways induce
Jones et al., 2001). increased airway inflammation, a hallmark of COPD, even
As with other opportunistic pathogens, Bcc virulence in in the absence of symptoms of acute exacerbation (Soler
CF and CGD patients is multifactorial (for a comprehensive et al., 1999; Hill et al., 2000), supporting the ‘vicious circle’
review of Bcc virulence factors, see Mahenthiralingam et al., hypothesis that proposes that persistent bacterial infections
2005). Besides lipopolysaccharides, which cause significantly cause progressive lung damage in COPD. This notion is
more inflammation than does P. aeruginosa lipooligosac- further substantiated by the finding that the frequency of
charides (Mahenthiralingam et al., 2005), biofilm formation positive cultures increases in patients with more severe
has been implicated to play a role in the pathophysiology of COPD (Eller et al., 1998). Thus, chronic bacterial and/or
CF lung disease (Mahenthiralingam et al., 2005). Many Bcc viral infections may play a role in the development and
strains can produce biofilms in vitro (Ferreira et al., 2010), progression of COPD (Sethi & Murphy, 2008). However,
associated with the ability to produce N-acyl-homoserine conclusive evidence for this hypothesis is lacking at present.
lactones (Conway et al., 2002), which may protect single A key question on which research effort should be focused is
cells from antibiotic killing (Desai et al., 1998). Also, the whether or not chronic infection of the airways in COPD
ability of Bcc strains to form mixed biofilms with accelerates the progressive loss of lung function. Under-
P. aeruginosa has been demonstrated (Riedel et al., 2001). standing the role of chronic infection in the course of the
In a CGD mouse model of Bcc lung infection (Sousa et al., disease would serve as an important guide in directing
2007), survival and death of the infected animals was research in the field. For example, if chronic infection were
correlated to the growth rate of the different Bcc strains. shown to cause increased progression of the disease, then the
The highest growth rate was found for the epidemic strain priority should be to develop methods such as vaccines or
J2315, which multiplied from 103 CFU–108 CFU within 3 immunomodulators to eradicate chronic colonization.
days, whereas for example a Burkholderia vietnamensis strain The causative role of pathogenic microorganisms in acute
never killed any mouse and reached only 104 CFU after 12 exacerbations of COPD, however, is well established (Sethi
days of infection. Thus, genes active in metabolism may be & Murphy, 2008). Opportunistic pathogenic bacteria are
important for Bcc virulence in CF and CGD. Whether Bcc responsible for approximately 50% of exacerbations, while
strains adapt genotypically or phenotypically to the CF virus-induced exacerbations caused by rhinovirus, respira-
niche is not known. tory syncytial virus, adenovirus, influenza virus and human
As in P. aeruginosa, hypermutable phenotypes have been metapneumovirus (Greenberg et al., 2000; Seemungal et al.,
observed repeatedly in S. aureus (Prunier et al., 2003), H. 2001) are approximately 30% (Sethi & Murphy, 2008).
influenzae (Roman et al., 2004; Watson et al., 2004) and Bcc Coinfections are frequent. Table 3 shows the relative dis-
strains from CF patients (Burns, 2005), suggesting that this tribution of bacterial pathogens that caused exacerbations in
mechanism may play a crucial role in the pathogenesis of a 10-year prospective study (Murphy & Parameswaran,
chronic lung infection. 2009). Studies of COPD patients using bronchoscopy and
protected specimen brush sampling of distal airways, avoid-
ing upper airway contamination, have consistently shown
Bacteria colonization in COPD pathogenic bacteria in quantities indicative of infection
In contrast to CF, the role of pathogenic bacteria in the (Monso et al., 1995; Soler et al., 1999). Moreover, a
progression of COPD is an area of uncertainty and the comparison of exacerbation and stable state patients, using
subject of active research. Opportunistic pathogenic bacteria the same technique, showed significantly higher quantities
including NTHI, M. catarrhalis, S. pneumoniae and P. of these bacteria during exacerbation (Fagon et al., 1990;


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 135

Table 3. Relative distribution of bacterial pathogens in COPD indicating that selected strains persist in the airways for
Bacterial species Positive culture Exacerbation w months to years. A 7-year prospective study (Murphy et al.,
2004) including 104 COPD patients observed 17 episodes of
Haemophilus influenzae 707 84
Moraxella catarrhalis 310 68 prolonged ( 4 6 months) periods of negative sputum cul-
Streptococcus pneumoniae 219 11 tures preceded and followed by genotypically identical
Pseudomonas aeruginosa 230 15 strains of NTHI. Clearing and reacquisition of the identical
Number of positive cultures for the pathogen, isolated from the sputum strain by the patients was ruled out, because NTHI strains
of adults with COPD, followed in a 10-year prospective study in Buffalo,
are genetically highly heterogeneous, making the reacquisi-
NY. tion of the same strain circulating in the community a highly
w
Number of exacerbations in the same 10-year prospective study as unlikely event. Furthermore, H. influenzae does not remain
defined by the acquisition of a new bacterial strain (based on molecular viable on inanimate objects in the environment even for
typing of cultured isolates) simultaneous with symptoms of exacerbation minutes. Finally, analysis of sputum samples yielding nega-
(adapted from Murphy & Parameswaran, 2009). tive cultures revealed the presence of strain-specific NTHI
genes, further establishing persistent colonization in COPD
patients. Newly acquired NTHI strains isolated from COPD
Soler et al., 1998). Longitudinal molecular typing of patho- patients during exacerbations, when compared with persist-
genic bacteria from sputum cultures showed that acquisition ing strains, induce more airway inflammation, including
of a new strain was associated with a twofold increase in the neutrophil influx and IL-8 secretion from respiratory
incidence of exacerbations (Sethi et al., 2002). Purulent epithelial cells, and adhere more efficiently to the epithelial
sputum, a marker of exacerbation, is strongly associated cells in vitro (Chin et al., 2005). These data suggest that, as
with the presence of pathogenic bacteria in sputum (White with P. aeruginosa in chronic CF lung infection, adaptive
et al., 2003; Soler et al., 2007). processes may occur that modulate the virulence of this
pathogen. However, similar molecular studies as mentioned
above for P. aeruginosa (Smith et al., 2006; Bragonzi et al.,
NTHI and M. catarrhalis are prevalent pathogens
2009) have not yet been carried out for sequential NTHI
Haemophilus influenzae strains may produce antigenically isolates from single COPD patients to substantiate this
distinct polysaccharide capsules, which distinguishes strain notion. Analysis of the genome content of strains of NTHI
types ‘a’ through ‘f’. In contrast, strains of H. influenzae that that persist in the airways of COPD patients in comparison
colonize and infect the airways in COPD are nonencapsu- with those that are cleared quickly will help elucidate the
lated as demonstrated by a lack of reactivity with typing sera relative roles of the host and pathogen in the dynamics of
for each of the six known capsular serotypes. Thus, these infection in COPD airways.
strains are referred to as nontypeable, abbreviated NTHI. Also, M. catarrhalis is regarded as an important human
Haemophilus influenzae strains use several genetic mechan- mucosal pathogen in COPD patients (de Vries et al., 2009;
isms that alter either the gene expression or the gene content Murphy & Parameswaran, 2009). Virulence factors include a
that allow rapid adaptation and improved fitness to chan- family of ubiquitous surface proteins, including the ubiqui-
ging environments including phase variation based on tous surface protein A1 (UspA1) and UspA2, which can bind
slipped-strand mispairing, mediated by short DNA repeats a1-antichymotrypsin, thereby potentially increasing the
in either the coding regions or the upstream promoter protease activity in vivo, which may cause excessive inflam-
regions of virulence genes (Gilsdorf et al., 2004; Srikhanta mation in COPD patients (Perez Vidakovics & Riesbeck,
et al., 2010). Phase variation results in a large genetic 2009). In contrast, UspA1 has been shown to bind to the
diversity among H. influenzae strains present in different human-specific extracellular immunoglobulin V (IgV)-like
environments, which complicates the interpretation of viru- amino-terminal domain of carcinoembryonic antigen-re-
lence-related phenotypes. The multifactorial virulence of H. lated cell adhesion molecule 1, resulting in reduced toll-like
influenzae strains includes factors such as lipooligosacchar- receptor 2-initiated transcription factor nuclear factor-kB-
ides (Schweda et al., 2007), fimbriae encoded by the hif dependent inflammation (Slevogt et al., 2008). Which of
locus, IgA1 protease(s) (Erwin & Smith, 2007) and the these mechanisms dominate during M. catarrhalis infection
ability to form biofilms (Starner et al., 2006). in COPD patients is unclear.
NTHI strains show a highly variable pattern of carriage COPD patients who acquire new strains of M. catarrhalis
among COPD patients. Some strains of NTHI, causing usually clear this species efficiently from their airways in
exacerbations, can also persist for longer periods of time approximately 1 month. In a study including 104 COPD
than is apparent from sputum cultures. Molecular analysis patients who experienced 560 exacerbations, molecular
shows that a strain is present in COPD airways during typing identified 120 episodes of acquisition and clearance
periods when it cannot be recovered by routine culture, of M. catarrhalis in 50 patients (Murphy et al., 2005).

FEMS Microbiol Rev 35 (2011) 124–146 


c2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
136 G. Döring et al.

Reacquisition of the same strain was rare. The rapid estab- aeruginosa in COPD. This epidemiological signature is
lishment of protective antibody responses against the patho- divergent from that of the chronic P. aeruginosa infections
gen in these patients suggested that the clearance of in patients with CF, the latter being characterized by no or
M. catarrhalis is a function of a functional immune system slow turnover of clones (Rakhimova et al., 2009).
(Murphy et al., 2005). The findings that 57 (47.5%) of Circumstantial evidence suggests that chronic infection of
M. catarrhalis acquisitions were associated with clinical COPD airways by P. aeruginosa occurs in those with more
exacerbations (Murphy et al., 2005), and that increased severe COPD, particularly among patients who require
inflammation and tilting of the protease–antiprotease bal- mechanical ventilation for severe exacerbations (Miravitlles
ance in favour of protease activity occurred in airways of et al., 1999; Ferrer et al., 2005; Rosell et al., 2005; Nseir et al.,
COPD patients during exacerbation as well as colonization 2006), or in other words, patients with more severe reduc-
with M. catarrhalis (Parameswaran et al., 2009), suggest that tion in FEV1, when compared with those with milder
this pathogen contributes to the long-term decrease of lung disease, are more likely to harbour P. aeruginosa in their
function in COPD patients. Calculations revealed that M. lower airways (Eller et al., 1998). Patients with more severe
catarrhalis likely causes 10% of exacerbations of COPD. COPD also experience a higher number of acute exacerba-
tions and there is increasing recognition that P aeruginosa is
responsible for such exacerbations (Murphy et al., 2008).
Pseudomonas aeruginosa does not commonly Whether chronic P aeruginosa infection has a negative
show persistence in COPD
impact on lung function and the patients’ prognosis requires
In a chronic lung disease such as COPD, which is character- further study.
ized by chronic inflammation, progressive tissue destruction The role of P. aeruginosa in the clinical evolution of
and increased mucus production, one would expect persist- COPD is not well understood. Unlike in CF, no clear
ing P. aeruginosa clones undergoing a similar microevolu- evidence of worsening of lung infection and increased
tion as described above for the P. aeruginosa strains in CF mortality is correlated to chronic P. aeruginosa infection in
airways. Indeed, single clones of P. aeruginosa have been COPD. Furthermore, unlike in CF, eradication of colonizing
identified in COPD patients that persisted over several years pathogenic bacteria, in the absence of exacerbation symp-
and developed an increasing number of mutations during toms, has not been shown to be beneficial in the clinical
the chronic course of the disease, resulting in reduced course of COPD. The questions as to why only some patients
motility, decreased protease production and increased anti- develop these chronic infections and why only selected
biotic resistance as also recognized in patients with CF bacterial pathogens establish these infections in COPD
(Martinez-Solano et al., 2008). Furthermore, different mor- remain unanswered at present. Multiple host factors includ-
photypes of P. aeruginosa, including SCVs and mucoid ing the severity of COPD, genetic polymorphisms as well as
phenotypes, were observed in sputum isolates from a single bacterial virulence factors in specific clones may play a role
COPD patient, suggesting that in this disease diversification in this context. However, a comparison of P. aeruginosa
also plays an important role in bacterial persistence. Addi- clones in the airways of patients with COPD with those in
tionally, strains that persisted over long periods of time the airways of chronically infected patients with CF revealed
tended to be hypermutable as in CF (Martinez-Solano et al., that despite the disparate geographic origin of the isolates,
2008). the dominant P. aeruginosa clones were found with similar
However, the similarity to CF reduces when larger patient frequencies in acutely and chronically infected respiratory
groups are investigated longitudinally. In a 10-year study tracts irrespective of the aetiology of the disease (Rakhimova
involving 126 COPD patients, only 13 strains persisted for et al., 2009) (Fig. 4).
4 6 months and only four of these were mucoid (Murphy
et al., 2008). These data suggest that most COPD patients
had sporadic and intermittent infection of airways with P.
Antimicrobial treatment for CF patients
aeruginosa, which were repeatedly cleared. Strains from one For chronic P. aeruginosa lung infection in CF patients,
clone were generally cleared by the patients’ immune system antibiotic therapy has been a cornerstone, allowing a largely
and a gap in time existed before the acquisition of a strain improved life expectancy of the affected patients due to
from a different clone (Rakhimova et al., 2009). Chronic stabilization of lung function; by contrast, lung function
carriage of a clone was observed in a minority of investigated rapidly decreases in untreated CF patients. Commonly
patients in this study. When a clone chronically persisted in applied antibiotics include the aminoglycoside tobramycin,
COPD-affected lungs for an extended period of time, the the b-lactam antibiotic ceftazidime, the fluoroquinolone
clone often altered the repertoire of its accessory genome. ciprofloxacine and the polymyxin E colistin (Döring et al.,
Hence, intraclonal microevolution and the frequent turn- 2000, 2004). However, this therapy regimen, even when
over or loss of clones are typical of infections with P. applied as recommended (Döring et al., 2000, 2004), i.e.,


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 137

with antibiotics immediately after the first detection of the


pathogen. In this case, reported eradication rates were above
95% (reviewed in Döring et al., 2000, 2004).
In addition to antibiotic therapy, other treatment strate-
gies may also be successfully used in CF patients to fight
bacterial lung infection. Based on the abnormal accumula-
tion of ceramide in CF airways, leading to DNA deposits on
the respiratory epithelium, the sphingomyelinase blocker
amitriptyline is currently under investigation in clinical
trials (Riethmüller et al., 2009). Furthermore, recombinant
Fig. 4. Distribution of Pseudomonas aeruginosa strains isolated from human DNAse may be helpful in this context, as evidenced
patients with CF and COPD. (a) Venn diagram of the number of P. in experimental mouse models of P. aeruginosa lung infec-
aeruginosa clonal complexes obtained from the airways of patients with
tion (Teichgräber et al., 2008) and in CF patients with regard
COPD and CF, respectively. Clonal complexes were calculated from the
to S. aureus (Frederiksen et al., 2006).
15-marker genotype of the core genome using the program EBURST
(Maiden, 2006). The intersections represent the clonal complexes that An important question is whether antibiotic therapy
had been identified in both diseases. (b) Clonal complex structure of P. directed against P. aeruginosa is also effective against the
aeruginosa strains collected from various different habitats and geo- large number of strict anaerobes present in the mucus plugs
graphic origins. The EBURST analysis was carried out with the informative within the airways of CF patients. Two studies suggest that
SNPs of a microassay of the core genome. Coloured clonal complexes this is not the case (Tunney et al., 2008; Worlitzsch et al.,
contain airway isolates from patients with COPD (yellow), from patients
2009). If strict anaerobes actually did contribute to lung
with CF (blue) and from both patient groups (green). The size of the
disease in CF patients, additional antibiotics, specifically
symbol indicates the number of typed strains with an identical genotype.
Clones C and PA14 are the most abundant clones (Wiehlmann et al., active against these pathogens, should be administered to CF
2007). Genotypes only observed in other habitats are depicted in grey. patients.
(Figure adapted from Rakhimova et al., 2009.) In the case of Bcc infections, there is no doubt that these
microorganisms cause lung disease in CF patients. Because
of the intrinsic resistance of Bcc strains to many antibiotics,
aggressively every 3 months with high doses of inhaled combination therapy with two to three different antibiotics
antibiotics, does not eradicate the chronically persisting P. according to minimal inhibition concentration assessment is
aeruginosa from the airways of CF patients. The highly recommended (Döring et al., 2004).
viscous mucus plugs and the biofilm-like macrocolonies of
P. aeruginosa within these plugs, which impede the penetra-
Antimicrobial treatment for COPD
tion of antibiotics, are assumed to be responsible for this
negative outcome (Xu et al., 1998; Wimpenny et al., 2000;
patients
Worlitzsch et al., 2002; Döring et al., 2004). As mentioned Antibiotic therapy regimens in COPD have focused on the
above, the stressful environment in the CF lung also favours treatment of exacerbations. However, only approximately
the emergence of hypermutable (or mutator) strains of P. 50% of acute exacerbations in COPD patients are caused by
aeruginosa, which are deficient in the DNA mismatch repair bacterial pathogens, a situation that complicates the inter-
system. Mutator strains produce antibiotic-resistant mu- pretation of results in such studies. Nevertheless, the data
tants at elevated rates (Oliver et al., 2000; Hogardt et al., available strongly suggest that antibiotic therapy for acute
2007). Combination therapy using antibiotics having differ- exacerbations in COPD patients reduces complications and
ent modes of action against P. aeruginosa could be superior hastens recovery. In a placebo-controlled study, including
to monotherapy in this situation. Recently, the combination 173 COPD patients who suffered from 362 acute exacerba-
of the fluoroquinolone ciprofloxacin and the amphipathic tions during the study period, the success rate was 68.1% in
oligopeptide polymyxin E (colistin) has yielded promising the antibiotic group vs. 55% in the placebo group (Antho-
results in killing P. aeruginosa biofilms in vitro, based on the nisen et al., 1987). Most of the success was seen in patients
concept of combining antibiotics that have different targets with more severe exacerbations, indicating that patients
in the biofilm cells. While biofilm cells exhibiting low with more severe exacerbations benefit more from antibiotic
metabolic activity were killed by colistin, ciprofloxacin was therapy. A systematic review analysed 11 placebo-controlled
found to specifically kill the subpopulation of metabolically trials (917 patients) of antibiotics in exacerbations of COPD.
active biofilm cells (Pamp et al., 2008). Whether this strategy Combining the data from three trials that used objective
is successful in CF patients still needs to be tested. definitions of COPD to qualify for inclusion showed a
To avoid the development of resistance and in an attempt reduction in mortality with antibiotic use and reduction in
to eradicate P. aeruginosa early, CF patients were treated the rates of treatment failure (Ram et al., 2006). Also, other

FEMS Microbiol Rev 35 (2011) 124–146 


c2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
138 G. Döring et al.

meta-analyses of placebo-controlled trials revealed a sub- standing the pathogenesis of P. aeruginosa infection in
stantial benefit for the antibiotic treatment of acute exacer- COPD and in the clinical management of these patients.
bations in COPD patients, emphasizing the pathogenic role
of bacteria in the course of this disease (Quon et al., 2008;
Roede et al., 2009).
Concluding remarks
Another goal of antimicrobial therapy is to slow or to The pathophysiology of CF and COPD lung disease is
prevent the worsening of pulmonary function. The use of complex. While chronic pulmonary inflammation triggered
antibiotics in stable COPD patients to prevent progressive by opportunistic pathogens is the cause of the more or less
worsening of lung function in the absence of exacerbations rapidly decreasing lung function in CF, the role of bacterial
has yielded unclear results due to the lack of large studies infection in the pathogenesis of COPD is less well under-
with stratified COPD patients. Nevertheless, a recent meta- stood. Chronic P. aeruginosa infection plays a critical role in
analysis of data from trials conducted in 1950s and 1960s the course of CF, whereas only a small minority of adults
showed that ‘prophylactic antibiotics’ produced a relative with COPD experience chronic P. aeruginosa infection. In
risk reduction of 9% for the occurrence of exacerbations COPD, the predominant pathogens are H. influenzae and
(Black et al., 2003). However, even this modest gain should M. catarrhalis. While in CF airways single P. aeruginosa
be interpreted with caution because the trials varied mark- clones, which adapt genotypically and phenotypically to the
edly in methodology and duration and many of the anti- CF niche, persist in the majority of patients due to a heavily
biotics used in these trials would not be expected to be impaired innate immune system, a frequent turnover and
effective with the current susceptibility patterns of bacteria. loss of clones are typical of infections with P. aeruginosa in
In a randomized trial, prophylactic erythromycin reduced COPD. However, once the extensive tissue remodelling and
the frequency of exacerbations in a group of COPD patients tissue destruction has reached a certain threshold (which has
and increased the mean interval between exacerbations not yet been determined), opportunistic pathogens such as
(Seemungal et al., 2008). Whether the benefits are due to P. aeruginosa may persist and develop epidemiological
antimicrobial, anti-inflammatory or both effects of erythro- signatures similar to those in CF. To prevent this point of
mycin is not clear. In a recent pilot study involving 13 COPD no return is a challenge for microbiologists and clinicians.
patients, colonized with multi-drug-resistant P. aeruginosa,
inhalation of tobramycin (300 mg twice daily for 2 weeks)
reduced the sputum density of P. aeruginosa and the exacer- Acknowledgements
bation rate (Dal Negro et al., 2008). This small uncontrolled
The authors want to thank Peter Michael Weber, Children’s
study is promising, but needs to be replicated in larger
Clinic, University of Tübingen, Germany, for excellent
controlled trials that examine the impact of such a treatment
graphic art.
on lung function and clinical outcomes.
The success of early treatment with antibiotics in redu-
cing the incidence of airway infection in CF raises the
question of whether a similar approach might be effective
References
in adults with COPD, particularly with regard to P. aerugi- Andersen DH (1938) Cystic fibrosis of the pancrease and its
nosa infection. The majority of adults with COPD who relation to coelic disease. Am J Dis Child 56: 344–399.
acquire P. aeruginosa clear the organism without antibiotic Anthonisen NR (2002) Bacteria and exacerbations of chronic
therapy (Murphy et al., 2008). However, patients with more obstructive pulmonary disease. New Engl J Med 347:
advanced COPD, persistent infection with P. aeruginosa and 526–527.
severe exacerbations are more likely to benefit from anti- Anthonisen NR, Manfreda J, Warren CP, Hershfield ES, Harding
biotic therapy. Therefore, COPD patients hospitalized with GK & Nelson NA (1987) Antibiotic therapy in exacerbations of
chronic obstructive pulmonary disease. Ann Intern Med 106:
exacerbations, particularly with severe exacerbations reveal-
196–204.
ing purulent sputum masses, and patients with severe
Anthony M, Rose B, Pegler MB et al. (2002) Genetic analysis of
underlying COPD having exacerbations should receive anti-
Pseudomonas aeruginosa isolates from the sputa of
biotics, according to a decision-making algorithm (Sethi & Australian adult cystic fibrosis patients. J Clin Microbiol 40:
Murphy, 2008). 2772–2778.
It is not known whether the subset of COPD patients who Baird RM, Brown H, Smith AW & Watson ML (1999)
have persistently positive sputum cultures for P. aeruginosa Burkholderia cepacia is resistant to the antimicrobial activity of
experience adverse consequences of chronic infection, ana- airway epithelial cells. Immunopharmacol 44: 267–272.
logous to patients with CF. This is an important question Barasch J, Kiss B, Prince A, Saiman L, Gruenert D & al-Awqati Q
that needs to be addressed in carefully designed clinical (1991) Defective acidification of intracellular organelles in CF.
trials. The results will have important implications in under- Nature 352: 70–73.


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 139

Barnes PJ (2000) Chronic obstructive pulmonary disease. New Burns JL (2005) Hypermutability in CF pathogens. Pediatr Pulm
Engl J Med 343: 269–280. Suppl 28: 115–116.
Barnes PJ (2004) Alveolar macrophages as orchestrators of Celli BR & MacNee W (2004) Standards for the diagnosis and
COPD. COPD 1: 59–70. treatment of patients with COPD: a summary of the ATS/ERS
Barnes PJ, Shapiro SD & Pauwels RA (2003) Chronic obstructive position paper. Eur Respir J 23: 932–946.
pulmonary disease: molecular and cellular mechanisms. Eur Chin CL, Manzel LJ, Lehman EE et al. (2005) Haemophilus
Respir J 22: 672–688. influenzae from patients with chronic obstructive pulmonary
Berenson CS, Garlipp MA, Grove LJ, Maloney J & Sethi S (2006a) disease exacerbation induce more inflammation than
Impaired phagocytosis of nontypeable Haemophilus influenzae colonizers. Am J Resp Crit Care 172: 85–91.
by human alveolar macrophages in chronic obstructive Chu KK, Davidson DJ, Halsey TK, Chung JW & Speert DP (2002)
pulmonary disease. J Infect Dis 194: 1375–1384. Differential persistence among genomovars of the
Berenson CS, Wrona CT, Grove LJ et al. (2006b) Impaired Burkholderia cepacia complex in a murine model of
alveolar macrophage response to Haemophilus antigens in pulmonary infection. Infect Immun 70: 2715–2720.
chronic obstructive lung disease. Am J Resp Crit Care 174: Ciofu O, Riis B, Pressler T, Poulsen HE & Høiby N (2005)
31–40. Occurrence of hypermutable Pseudomonas aeruginosa in cystic
Berger M, Sorensen RU, Tosi MF, Dearborn DG & Döring G fibrosis patients is associated with the oxidative stress caused
(1989) Complement receptor expression on neutrophils at an by chronic lung inflammation. Antimicrob Agents Ch 49:
inflammatory site, the Pseudomonas-infected lung in cystic 2276–2282.
fibrosis. J Clin Invest 84: 1302–1313. Ciofu O, Lee B, Johannesson M, Hermansen NO, Meyer P &
Biswas L, Biswas R, Schlag M, Bertram R & Gotz F (2009) Small- Hoiby N for the Scandinavian Cystic Fibrosis Study
colony variant selection as a survival strategy for Consortium (2008) Investigation of the algT operon sequence
Staphylococcus aureus in the presence of Pseudomonas in mucoid and non-mucoid Pseudomonas aeruginosa isolates
aeruginosa. Appl Environ Microb 75: 6910–6912. from 115 Scandinavian patients with cystic fibrosis and in 88
Bittar F, Riichet H, Dubus J-C et al. (2008) Molecular detection of in vitro non-mucoid revertants. Microbiology 154: 103–113.
multiple emerging pathogens in sputa from cystic fibrosis Ciofu O, Mandsberg LF, Bjarnsholt T, Wassermann T & Høiby N
patients. Plos ONE 3: e2908. (2010) Genetic adaptation of Pseudomonas aeruginosa during
Black P, Staykova T, Chacko E, Ram FS & Poole P (2003) chronic lung infection of patients with cystic fibrosis: strong
Prophylactic antibiotic therapy for chronic bronchitis. and weak mutators with heterogeneous genetic backgrounds
Cochrane Db Syst Rev 1: CD004105. emerge in mucA and/or lasR mutants. Microbiology 154:
Boles BR & Horswill AR (2008) Agr-mediated dispersal of 1003–1013.
Staphylococcus aureus biofilms. PLoS Pathog 4: e1000052. Coenye T, Vandamme P, Govan JR & LiPuma JJ (2001) Taxonomy
Boles BR, Thoendel M & Singh PK (2004) Self-generated and identification of the Burkholderia cepacia complex. J Clin
diversity produces ‘insurance effects’ in biofilm communities. Microbiol 39: 3427–3436.
P Natl Acad Sci USA 101: 16630–16635. Conway BA, Venu V & Speert DP (2002) Biofilm formation and
Boucher JC, Yu H, Mudd MH & Deretic V (1997) Mucoid acyl homoserine lactone production in the Burkholderia
Pseudomonas aeruginosa in cystic fibrosis: characterization of cepacia complex. J Bacteriol 184: 5678–5685.
muc mutations in clinical isolates and analysis of clearance in a Corey M & Farewell V (1996) Determinants of mortality from
mouse model of respiratory infection. Infect Immun 65: cystic fibrosis in Canada. Am J Epidemiol 43: 1007–1017.
3838–3846. Cornelis P, Matthijs S & Van Oeffelen L (2009) Iron uptake
Boucher RC (2007) Airway surface dehydration in regulation in Pseudomonas aeruginosa. BioMetals 22: 15–22.
CF: pathogenesis and therapy. Annu Rev Med 58: Cosio MG, Saetta M & Agusti A (2009) Immunologic aspects of
157–170. chronic obstructive pulmonary disease. New Engl J Med 360:
Bragonzi A, Wiehlmann L, Klockgether J, Cramer N, 2445–2454.
Worlitzsch D, Döring G & Tümmler B (2006) Sequence Cox AD & Wilkinson SG (1991) Ionizing groups in
diversity of the mucABD locus in Pseudomonas aeruginosa lipopolysaccharides of Pseudomonas cepacia in relation to
isolates from patients with cystic fibrosis. Microbiology 152: antibiotic resistance. Mol Microbiol 5: 641–646.
3261–3269. Cramton SE, Gerke C, Schnell NF et al. (1999) The intercellular
Bragonzi A, Paroni M, Nonis A, Cramer N, Montanari A, Rejman adhesion (ica) locus is present in Staphylococcus aureus
J, Di Serio C, Döring G & Tümmler B (2009) Pseudomonas and is required for biofilm formation. Infect Immun 67:
aeruginosa microevolution during cystic fibrosis lung infection 5427–5433.
establishes clones with adapted virulence. Am J Resp Crit Care Cramton SE, Ulrich M, Götz F & Döring G (2001) Anaerobic
Dis 180: 138–145. conditions induce expression of the polysaccharide
Brook I & Fink R (1983) Transtracheal aspiration in pulmonary intercellular adhesin in Staphylococcus aureus and
infection in children with cystic fibrosis. Eur J Respir Dis 64: Staphylococcus epidermidis. Infect Immun 69:
51–57. 4079–4085.

FEMS Microbiol Rev 35 (2011) 124–146 


c2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
140 G. Döring et al.

Dal Negro R, Micheletto C, Tognella S, Visconti M & Eller J, Ede A, Schaberg T, Niederman MS, Mauch H & Lode H
Turati C (2008) Tobramycin nebulizer solution in severe (1998) Infective exacerbations of chronic bronchitis. Chest
COPD patients colonized with Pseudomonas aeruginosa: 113: 1542–1548.
effects on bronchial inflammation. Adv Ther 25: Engelhardt JF, Yankaskas JR, Ernst SA et al. (1992) Submucosal
1019–1030. glands are the predominant site of CFTR expression in the
D’Argenio DA (2004) The pathogenic lifestyle of Pseudomonas human bronchus. Nat Genet 2: 240–248.
aeruginosa in model systems of virulence. Pseudomonas: Ernst RK, Yi EC, Guo L, Lim KB, Burns JL, Hackett M
Genomics, Life Style and Molecular Architecture, Vol. 1 (Ramos & Miller SI (1999) Specific lipopolysaccharide found in cystic
J-L, ed), pp. 477–504. Kluwer Academics/Plenum Publishers, fibrosis airway Pseudomonas aeruginosa. Science 286:
New York. 1561–1565.
Davies JC (2007) Genotype–phenotype correlations and modifier Erwin AL & Smith AL (2007) Nontypeable Haemophilus
genes. Cystic Fibrosis (Hodson M, Gededs D & Bush A, eds), influenzae: understanding virulence and commensal behavior.
pp. 81–86. Hodder Arnold, London. Trends Microbiol 15: 355–362.
Deretic V, Schurr MJ & Yu H (1995) Pseudomonas aeruginosa, Fagon JY, Chastre J, Trouillet JL, Domart Y, Dombret MC, Bornet
mucoidy and the chronic infection phenotype in cystic M & Gibert C (1990) Characterization of distal bronchial
fibrosis. Trends Microbiol 3: 351–356. microflora during acute exacerbation of chronic bronchitis.
Desai M, Buhler T, Weller PH & Brown MR (1998) Use of the protected specimen brush technique in 54
Increasing resistance of planktonic and biofilm cultures of mechanically ventilated patients. Am Rev Respir Dis 142:
Burkholderia cepacia to ciprofloxacin and ceftazidime 1004–1008.
during exponential growth. J Antimicrob Chemoth 42: Ferreira AS, Leitao JH, Silva IN, Pinheiro PF, Sousa SA, Ramos
153–160. CG & Moreira LM (2010) Distribution of cepacian
de Vries SP, Bootsma HJ, Hays JP & Hermans PW (2009) biosynthesis genes among environmental and clinical
Molecular aspects of Moraxella catarrhalis pathogenesis. Burkholderia strains and role of cepacian exopolysaccharide in
Microbiol Mol Biol R 73: 389–406. resistance to stress conditions. Appl Environ Microb 76:
Di A, Brown ME, Deriy LV et al. (2006) CFTR regulates 441–450.
phagosome acidification in macrophages and alters Ferrer M, Ioanas M, Arancibia F, Marco MA, de la Bellacasa JP &
bactericidal activity. Nat Cell Biol 8: 933–944. Torres A (2005) Microbial airway colonization is associated
di Sant’Agnese PA & Talamo RC (1967) Pathogenesis and with noninvasive ventilation failure in exacerbation of
physiopathology of cystic fibrosis of the pancreas. New Engl J chronic obstructive pulmonary disease. Crit Care Med 33:
Med 277: 1287–1294. 2003–2009.
Dodge JA, Lewis PA, Stanton M & Wilsher J (2007) Cystic fibrosis Frederiksen B, Pressler T, Hansen A, Koch C & Hoiby N (2006)
mortality and survival in the UK: 1947–2003. Eur Respir J 29: Effect of aerosolized rhDNase (pulmozyme) on pulmonary
522–526. colonization in patients with cystic fibrosis. Acta Paediatr 95:
Döring G (1999) Perspectives of serine proteinase inhibitor 1070–1074.
therapy for a1-antitrypsin inhibitor deficiency and cystic Gilles-Gonzalez MA & Gonzalez G (2005) Heme-based sensors:
fibrosis. Pediatr Pulm 28: 363–375. defining characteristics, recent developments, and regulatory
Döring G & Gulbins E (2009) Cystic fibrosis and innate hypotheses. J Inorg Biochem 99: 1–22.
immunity: how chloride channel mutations provoke lung Gilsdorf JR, Marrs CF & Foxman B (2004) Haemophilus
disease. Cell Microbiol 11: 208–216. influenzae: genetic variability and natural selection to identify
Döring G & Ratjen F (2005) Cystic fibrosis molecular cell biology. virulence factors. Infect Immun 72: 2457–2461.
Encyclopedia of Molecular Cell Biology and Molecular Medicine, Goerke C & Wolz C (2004) Regulatory and genomic plasticity of
Vol. 8, 2nd edn (Meyers RA, ed), pp. 623–640. Wiley-VCH, Staphylococcus aureus during persistent colonization and
Weinheim. infection. Int J Med Microbiol 294: 195–202.
Döring G & Ratjen F (2007) Immunology of cystic fibrosis. Cystic Goerke C, Campana S, Bayer MG et al. (2000) Direct quantitative
Fibrosis (Hodson M, Gededs D & Bush A, eds), pp. 69–80. transcript analysis of the agr regulon of Staphylococcus aureus
Hodder Arnold, London. during human infection in comparison to the expression
Döring G, Conway SP, Heijerman HGM et al. (2000) Antibiotic profile in vitro. Infect Immun 68: 1304–1311.
therapy against Pseudomonas aeruginosa in cystic fibrosis: a Goerke C, Matias y Papenberg S, Dasbach S et al. (2004)
European consensus. Eur Respir J 16: 749–767. Increased frequency of genomic alterations in Staphylococcus
Döring G & Høiby Nthe Consensus Study Group (2004) Early aureus during chronic infection is in part due to phage
intervention and prevention of lung disease in cystic fibrosis: a mobilization. J Infect Dis 189: 724–734.
European consensus. J Cyst Fibros 3: 67–91. Govan JRW, Brown PH, Maddison J, Doherty CJ, Nelson JW,
Duan K, Sibley CD, Davidson CJ & Surette MG (2009) Chemical Dodd M, Greening AP & Webb AK (1993) Evidence for
interactions between organisms in microbial communities. transmission of Pseudomonas cepacia by social contact in cystic
Contrib Microbiol 16: 1–17. fibrosis. Lancet 342: 15–19.


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 141

Green J & Paget MS (2004) Bacterial redox sensors. Nat Rev Hoffman LR, Deziel E, D’Argenio DA, Lepine F, Emerson J,
Microbiol 2: 954–966. McNamara S, Gibson RL, Ramsey BW & Miller SI (2006)
Greenberg SB, Allen M, Wilson J & Atmar RL (2000) Respiratory Selection for Staphylococcus aureus small-colony variants due
viral infections in adults with and without chronic obstructive to growth in the presence of Pseudomonas aeruginosa. P Natl
pulmonary disease. Am J Resp Crit Care 162: 167–173. Acad Sci USA 103: 19890–19895.
Haggie PM & Verkman AS (2007) Cystic fibrosis transmembrane Hoffmann N, Rasmussen TB, Jensen PO, Stub C, Hentzer M,
conductance regulator-independent phagosomal acidification Molin S, Ciofu O, Givskov M, Johansen HK & Hoiby N (2005)
in macrophages. J Biol Chem 282: 31422–31428. Novel mouse model of chronic Pseudomonas aeruginosa lung
Haggie PM & Verkman AS (2009) Defective organellar infection mimicking cystic fibrosis. Infect Immun 73:
acidification as a cause of cystic fibrosis lung disease – re- 2504–2514.
examination of a recurring hypothesis. Am J Physiol Lung-C Hogardt M, Roeder M, Schreff AM, Eberl L & Heesemann J
296: L859–L867. (2004) Expression of Pseudomonas aeruginosa exoS is
Hancock RE, Mutharia LM, Chan L, Darveau RP, Speert DP & controlled by quorum sensing and RpoS. Microbiology 150:
Pier GB (1983) Pseudomonas aeruginosa isolates from patients 843–851.
with cystic fibrosis: a class of serum-sensitive, nontypable Hogardt M, Hoboth C, Schmoldt S, Henke C, Bader L &
strains deficient in lipopolysaccharide O side chains. Infect Heesemann J (2007) Stage-specific adaptation of
Immun 42: 170–177. hypermutable Pseudomonas aeruginosa isolates during chronic
Harris JK, De Groote MA, Sagel SD, Zemanick ET, Kapsner R, pulmonary infection in patients with cystic fibrosis. J Infect Dis
Penvari C, Kaess H, Deterding RR, Accurso FJ & Pace NR 195: 70–80.
(2007) Molecular identification of bacteria in bronchoalveolar Hogg JC & Timens W (2009) The pathology of chronic
lavage fluid from children with cystic fibrosis. P Natl Acad Sci obstructive pulmonary disease. Annu Rev Pathol 4: 435–459.
USA 104: 20529–20533. Hogg JC, Chu F, Utokaparch S et al. (2004) The nature of small-
Hassett DJ, Sutton MD, Schurr MJ, Herr AB, Caldwell CC & airway obstruction in chronic obstructive pulmonary disease.
Matu JO (2009) Pseudomonas aeruginosa hypoxic or anaerobic New Engl J Med 350: 2645–2653.
biofilm infections within cystic fibrosis airways. Trends Høiby N & Frederiksen B (2000) Microbiology of cystic fibrosis.
Microbiol 17: 130–138. Cystic Fibrosis (Hodson ME & Geddes D, eds), pp. 83–108.
Häussler S, Tümmler B, Weissbrodt H, Rohde M & Steinmetz I Arnold, London.
(1999) Small-colony variants of Pseudomonas aeruginosa in Jain M, Ramirez D, Seshadri R, Cullina JF, Powers CA, Schulert
cystic fibrosis. Clin Infect Dis 29: 621–625. GS, Bar-Meir M, Sullivan CL, McColley SA & Hauser AR
Häussler S, Ziegler I, Lottel A, von Götz F, Rohde M, (2004) Type III secretion phenotypes of Pseudomonas
Wehmhöhner D, Saravanamuthu S, Tümmler B & Steinmetz I aeruginosa strains change during infection of individuals with
(2003) Highly adherent small-colony variants of Pseudomonas cystic fibrosis. J Clin Microbiol 42: 5229–5237.
aeruginosa in cystic fibrosis lung infection. J Med Microbiol 52: Jewes LA & Spencer RC (1990) The incidence of anaerobes in the
295–301. sputum of patients with cystic fibrosis. J Med Microbiol 31:
Herbert S, Worlitzsch D, Dassy B et al. (1997) Regulation of 271–274.
the Staphylococcus aureus capsular polysaccharide type 5: Jones AM, Dodd ME & Webb AK (2001) Burkholderia cepacia:
CO2 inhibition in vitro and in vivo. J Infect Dis 176: current clinical issues, environmental controversies and ethical
431–438. dilemmas. Eur Respir J 17: 295–301.
Hill AT, Bayley D & Stockley RA (1999a) The interrelationship of Joo NS, Lee DJ, Winges KM, Rustagi A & Wine JJ (2004)
sputum inflammatory markers in patients with chronic Regulation of antiprotease and antimicrobial protein secretion
bronchitis. Am J Resp Crit Care 160: 893–898. by airway submucosal gland serous cells. J Biol Chem 279:
Hill AT, Campbell EJ, Bayley DL, Hill SL & Stockley RA (1999b) 38854–38860.
Evidence for excessive bronchial inflammation during an acute Keatings VM, Collins PD, Scott DM & Barnes PJ (1996)
exacerbation of chronic obstructive pulmonary disease in Differences in interleukin-8 and tumor necrosis factor-alpha
patients with alpha(1)-antitrypsin deficiency (PiZ). Am J Resp in induced sputum from patients with chronic obstructive
Crit Care 160: 1968–1975. pulmonary disease or asthma. Am J Resp Crit Care 153:
Hill AT, Campbell EJ, Hill SL, Bayley DL & Stockley RA (2000) 530–534.
Association between airway bacterial load and markers of Keller L & Surette MG (2006) Communication in bacteria: an
airway inflammation in patients with stable chronic ecological and evolutionary perspective. Nat Rev Microbiol 4:
bronchitis. Am J Med 109: 288–295. 249–258.
Hoboth C, Hoffmann R, Eichner A, Henke C, Schmoldt S, Imhof Kerem BT, Rommens JM, Buchanan JA et al. (1989)
A, Heesemann J & Hogardt M (2009) Dynamics of adaptive Identification of the cystic fibrosis gene: genetic analysis.
microevolution of hypermutable Pseudomonas aeruginosa Science 245: 1073–1080.
during chronic pulmonary infection in patients with cystic Klepac-Ceraj V, Lemon KP, Marti TR et al. (2010) Relationship
fibrosis. J Infect Dis 200: 118–130. between cystic fibrosis respiratory tract bacterial communities

FEMS Microbiol Rev 35 (2011) 124–146 


c2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
142 G. Döring et al.

and age, genotype, antibiotics and Pseudomonas aeruginosa. Martinez-Solano L, Macia MD, Fajardo A, Oliver A & Martinez
Environ Microbiol 12: 1293–1303. JL (2008) Chronic Pseudomonas aeruginosa infection in
Klockgether J, Würdemann D, Reva O, Wiehlmann L & Tümmler chronic obstructive pulmonary disease. Clin Infect Dis 47:
B (2007) Diversity of the abundant pKLC102/PAGI-2 family of 1526–1533.
genomic islands in Pseudomonas aeruginosa. J Bacteriol 189: Matsui H, Grubb BR, Tarran R, Randell SH, Gatzy JT, Davis CW
2443–2459. & Boucher RC (1998) Evidence for periciliary liquid
Kolpen M, Hansen CR, Bjarnsholt T et al. (2010) The respiratory layer depletion, not abnormal ion composition, in the
burst of polymorphonuclear leukocytes accelerates oxygen pathogenesis of cystic fibrosis airways disease. Cell 95:
depletion in sputum from CF patients with chronic 1005–1010.
Pseudomonas aeruginosa lung infection. Thorax 65: Matsui H, Verghese MW, Kesimer M et al. (2005) Reduced three-
57–62. dimensional motility in dehydrated airway mucus prevents
Kong KF, Vuong C & Otto M (2006) Staphylococcus quorum neutrophil capture and killing bacteria on airway epithelial
sensing in biofilm formation and infection. Int J Med Microbiol surfaces. J Immunol 175: 1090–1099.
296: 133–139. McKenney D, Pouliot KL, Wang Y et al. (1999) Broadly protective
Konstan MW, Hilliard KA, Norvell TM & Berger M (1994) vaccine for Staphylococcus aureus based on an in vivo-
Bronchoalveolar lavage findings in cystic fibrosis patients with expressed antigen. Science 284: 1523–1527.
stable, clinically mild lung disease suggest ongoing infection Miravitlles M, Espinosa C, Fernandez-Laso E, Martos JA,
and inflammation. Am J Resp Crit Care 150: 448–454. Maldonado JA & Gallego MStudy Group of Bacterial Infection
Kummerli R, Griffin AS, West SA, Buckling A & Harrison F in COPD (1999) Relationship between bacterial flora in
(2009) Viscous medium promotes cooperation in the sputum and functional impairment in patients with acute
pathogenic bacterium Pseudomonas aeruginosa. P Roy Soc
exacerbations of COPD. Chest 116: 40–46.
Lond B 276: 3531–3538. Mitchell G, Brouillette E, Lalonde Séguin D, Asselin A-E, Lebeau
Lee DG, Urbach JM, Wu G et al. (2006) Genomic analysis reveals
Jacob C & Malouin F (2010) A role for sigma factor B in the
that Pseudomonas aeruginosa virulence is combinatorial.
emergence of Staphylococcus aureus small-colony variants and
Genome Biol 7: R90.
elevated biofilm production resulting from an exposure to
Lee VT, Smith RS, Tummler B & Lory S (2005) Activities of
aminoglycosides. Microbial Pathogenesis 48: 18–27.
Pseudomonas aeruginosa effectors secreted by the Type III
Monso E, Ruiz J, Rosell A, Manterola J, Fiz J, Morera J & Ausina V
secretion system in vitro and during infection. Infect Immun
(1995) Bacterial infection in chronic obstructive pulmonary
73: 1695–1705.
disease. A study of stable and exacerbated outpatients using
Lehrer RI & Ganz T (2002) Defensins of vertebrate animals. Curr
the protected specimen brush. Am J Resp Crit Care 152:
Opin Immunol 14: 96–102.
1316–1320.
Lopez AD, Shibuya K, Rao C et al. (2006) Chronic obstructive
Montanari S, Oliver A, Salerno P, Mena A, Cariani L, Bretoni G,
pulmonary disease: current burden and future projections. Eur
Conese M, Tümmler B, Döring G & Bragonzi A (2007)
Respir J 27: 397–412.
Lory S, Merighi M & Hyodo M (2009) Multiple activities of c-di- Biological cost of hypermutation in Pseudomonas aeruginosa
GMP in Pseudomonas aeruginosa. Nucleic Acids Symp Ser 53: strains from patients with cystic fibrosis. Microbiology 153:
51–52. 1445–1454.
Luzar MA, Thomassen MJ & Montie TC (1985) Flagella and Murphy TF & Parameswaran G (2009) Moraxella catarrhalis,
motility alterations in Pseudomonas aeruginosa strains from a human respiratory tract pathogen. Clin Infect Dis 49:
patients with cystic fibrosis: relationship to patient clinical 124–131.
condition. Infect Immun 50: 577–582. Murphy TF, Brauer AL, Schiffmacher AT & Sethi S (2004)
Mahenthiralingam E, Campbell ME & Speert DP (1994) Persistent colonization by Haemophilus influenzae in chronic
Nonmotility and phagocytic resistance of Pseudomonas obstructive pulmonary disease. Am J Resp Crit Care 170:
aeruginosa isolates from chronically colonized patients with 266–272.
cystic fibrosis. Infect Immun 62: 596–605. Murphy TF, Brauer AL, Grant BJ & Sethi S (2005) Moraxella
Mahenthiralingam E, Urban TA & Goldberg JB (2005) The catarrhalis in chronic obstructive pulmonary disease: burden
multifarious,multireplicon Burkholderia cepacia complex. Nat of disease and immune response. Am J Resp Crit Care 172:
Rev Microbiol 3: 144–156. 195–199.
Maiden MC (2006) Multilocus sequence typing of bacteria. Annu Murphy TF, Brauer AL, Eschberger K, Lobbins P, Grove L, Cai X
Rev Microbiol 60: 561–588. & Sethi S (2008) Pseudomonas aeruginosa in chronic
Mandsberg LF, Ciofu O, Kirkby N, Christiansen LE, Poulsen HE obstructive pulmonary disease. Am J Resp Crit Care 177:
& Høiby N (2009) Antibiotic resistance in Pseudomonas 853–860.
aeruginosa strains with increased mutation frequency due to Nguyen D & Singh PK (2006) Evolving stealth: genetic adaptation
inactivation of the DNA oxidative repair system. Antimicrob of Pseudomonas aeruginosa during cystic fibrosis infections.
Agents Ch 53: 2483–2491. P Natl Acad Sci USA 103: 8305–8306.


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 143

Nocker A, Burr M & Camper AK (2007) Genotypic microbial Rakhimova E, Wiehlmann L, Brauer AL, Sethi S, Murphy TF &
community profiling: a critical technical review. Microb Ecol Tummler B (2009) Pseudomonas aeruginosa population
54: 276–289. biology in chronic obstructive pulmonary disease. J Infect Dis
Novick RP (2003) Autoinduction and signal transduction in the 200: 1928–1935.
regulation of staphylococcal virulence. Mol Microbiol 48: Ram FS, Rodriguez-Roisin R, Granados-Navarrete A, Garcia-
1429–1449. Aymerich J & Barnes NC (2006) Antibiotics for exacerbations
Novick RP, Ross HF, Projan SJ, Kornblum J, Kreiswirth B & of chronic obstructive pulmonary disease. Cochrane Db Syst
Moghazeh S (1993) Synthesis of staphylococcal virulence Rev 2: CD004403.
factors is controlled by a regulatory RNA molecule. EMBO J Ramsey DM & Wozniak DJ (2005) Understanding the control of
12: 3967–3975. Pseudomonas aeruginosa alginate synthesis and the prospects
Novick RP, Projan SJ, Kornblum J et al. (1995) The agr P2 operon: for management of chronic infections in cystic fibrosis. Mol
an autocatalytic sensory transduction system in Staphylococcus Microbiol 56: 309–322.
aureus. Mol Gen Genet 248: 446–458. Ratjen F, Hartog CM, Paul K, Wermelt J & Braun J (2002) Matrix
Nseir S, Di Pompeo C, Cavestri B et al. (2006) Multiple-drug- metalloproteases in BAL fluid of cystic fibrosis patients and
resistant bacteria in patients with severe acute exacerbation their modulation by treatment with dornase alpha. Thorax 57:
of chronic obstructive pulmonary disease: prevalence, 930–934.
risk factors, and outcome. Crit Care Med 34: Rau MH, Hansen SK, Johansen HK, Thomsen LE, Workman CT,
2959–2966. Nielsen KF, Jelsbak L, Høiby N, Yang L & Molin S (2010) Early
Oliver A, Cantón R, Campo P, Baquero F & Blázquez J (2000) adaptive developments of Pseudomonas aeruginosa after the
High frequency of hypermutable Pseudomonas aeruginosa in transition from life in the environment to persistent
cystic fibrosis lung infection. Science 288: 1251–1254. colonization in the airways of human cystic fibrosis hosts.
Oliver A, Baquero F & Blazquez J (2002) The mismatch repair Environ Microbiol April 7. Epub ahead of print.
system (mutS, mutL and uvrD genes) in Pseudomonas Riedel K, Hentzer M, Geisenberger O, Huber B, Steidle A, Wu H,
aeruginosa: molecular characterization of naturally occurring Høiby N, Givskov M, Molin S & Eberl L (2001) N-
mutants. Mol Microbiol 43: 1641–1650. acylhomoserine-lactone-mediated communication between
Pamp SJ, Gjermansen M, Johansen HK & Tolker-Nielsen T (2008) Pseudomonas aeruginosa and Burkholderia cepacia in mixed
Tolerance to the antimicrobial peptide colistin in Pseudomonas biofilms. Microbiology 147: 3249–3262.
aeruginosa biofilms is linked to metabolically active cells, and Riethmüller J, Anthonysamy J, Serra E, Schwab M, Döring G &
depends on the pmr and mexAB-oprM genes. Mol Microbiol Gulbins E (2009) Therapeutic efficacy and safety of amitriptyline
68: 223–240. in patients with cystic fibrosis. Cell Physiol Biochem 24: 65–72.
Parameswaran G, Wrona C, Murphy T & Sethi S (2009) Riordan JR, Rommens JM, Kerem BS et al. (1989) Identification
Moraxella catarrhalis acquisition, airway inflammation and of the Cystic Fibrosis Gene: Cloning and characterization of
protease-antiprotease balance in chronic obstructive complementary DNA. Science 245: 1066–1073.
pulmonary disease. BMC Infect Dis 9: 178. Roede BM, Bresser P, Prins JM, Schellevis F, Verheij TJ & Bindels
Perez Vidakovics ML & Riesbeck K (2009) Virulence mechanisms PJ (2009) Reduced risk of next exacerbation and mortality
of Moraxella in the pathogenesis of infection. Curr Opin Infect associated with antibiotic use in COPD. Eur Respir J 33:
Dis 22: 279–285. 282–288.
Pier GB (2000) Role of the CF transmembrane conductance Rogers GB, Hart CA, Mason JR et al. (2003) Bacterial diversity in
regulator in innate immunity to Pseudomonas aeruginosa cases of lung infection in cystic fibrosis patients: 16S ribosomal
infections. P Natl Acad Sci USA 97: 8822–8828. DNA (rDNA) length heterogeneity PCR and 16S rDNA
Pollock JD, Williams DA, Gifford MAC et al. (1995) Mouse terminal reobligation fragment length polymorphism
model of X-linked chronic granulomatous disease, an profiling. J Clin Microbiol 41: 3548–3558.
inherited defect in phagocyte superoxide production. Nat Rogers GB, Carroll MP, Serisier DJ et al. (2004) Characterization
Genet 9: 202–209. of bacterial community diversity in cystic fibrosis lung
Prunier AL, Malbruny B, Laurans M, Brouard J, Duhamel JF & infections by use of 16S ribosomal DNA terminal reobligation
Leclercq R (2003) High rate of macrolide resistance in fragment length polymorphism profiling. J Clin Microbiol 42:
Staphylococcus aureus strains from patients with cystic fibrosis 5176–5183.
reveals high proportions of hypermutable strains. J Infect Dis Rogers GB, Carroll MP, Serisier DJ et al. (2006) Use of 16S rRNA
187: 1709–1716. gene profiling by terminal restriction fragment length
Quon BS, Gan WQ & Sin DD (2008) Contemporary polymorphism analysis to compare bacterial communities in
management of acute exacerbations of COPD: a systematic sputum and mouthwash samples from patients with cystic
review and metaanalysis. Chest 133: 756–766. fibrosis. J Clin Microbiol 44: 2601–2604.
Rahman I & MacNee W (1996) Oxidant/antioxidant imbalance Rogers GB, Carroll MP & Bruce KD (2009) Studying bacterial
in smokers and chronic obstructive pulmonary disease. Thorax infections through culture-independent approaches. J Med
51: 348–350. Microbiol 58: 1401–1418.

FEMS Microbiol Rev 35 (2011) 124–146 


c2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
144 G. Döring et al.

Roman F, Canton R, Perez-Vazquez M, Baquero F & Campos J Selsted ME & Quellette AJ (2003) Mammalian defensins in the
(2004) Dynamics of long-term colonization of respiratory antimicrobial immune response. Nat Immunol 6: 551–557.
tract by Haemophilus influenzae in cystic fibrosis patients Sethi S & Murphy TF (2001) Bacterial infection in chronic
shows a marked increase in hypermutable strains. J Clin obstructive pulmonary disease in 2000: a state-of-the-art
Microbiol 42: 1450–1459. review. Clin Microbiol Rev 14: 336–363.
Rommens JM, Iannuzzi MC, Kerem BS et al. (1989) Sethi S & Murphy TF (2008) Infection in the pathogenesis and
Identification of the Cystic Fibrosis Gene: Chromosome course of chronic obstructive pulmonary disease. New Engl J
walking and jumping. Science 245: 1059–1065. Med 359: 2355–2365.
Rosell A, Monso E, Soler N et al. (2005) Microbiologic Sethi S, Evans N, Grant BJ & Murphy TF (2002) New strains of
determinants of exacerbation in chronic obstructive bacteria and exacerbations of chronic obstructive pulmonary
pulmonary disease. Arch Intern Med 165: 891–897. disease. New Engl J Med 347: 465–471.
Sabra W, Lunsdorf H & Zeng AP (2003) Alterations in the Sethi S, Wrona C, Eschberger K, Lobbins P, Cai X & Murphy TF
formation of lipopolysaccharide and membrane vesicles on the (2008) Inflammatory profile of new bacterial strain
surface of Pseudomonas aeruginosa PAO1 under oxygen stress exacerbations of chronic obstructive pulmonary disease. Am J
conditions. Microbiology 149: 2789–2795. Resp Crit Care 177: 491–497.
Sadowska B, Bonar A, von Eiff C et al. (2002) Characteristics of Sibley CD, Parkins MD, Rabin HR, Duan K, Norgaard JC &
Staphylococcus aureus, isolated from airways of cystic fibrosis Surette MG (2008) A polymicrobial perspective of pulmonary
patients, and their small colony variants. FEMS Immunol Med infections exposes an enigmatic pathogen in cystic fibrosis
Mic 32: 191–197. patients. P Natl Acad Sci USA 105: 15070–15075.
Sahly H, Schubert S, Harder J, Rautenberg P, Ullmann U, Sibley CD, Parkins MD, Rabin HR & Surette MG (2009) The
Schroder H & Podschun R (2003) Burkholderia is highly relevance of the polymicrobial nature of airway infection in the
resistant to human beta-defensin 3. Antimicrob Agents Ch 47: acute and chronic management of patients with cystic fibrosis.
1739–1741. Curr Opin Investig D 10: 787–794.
Salunkhe P, Smart CH, Morgan JA, Panagea S, Walshaw MJ, Hart Silo-Suh L, Suh SJ, Phibbs PV & Ohman DE (2005) Adaptations
CA, Geffers R, Tümmler B & Winstanley C (2005a) A cystic of Pseudomonas aeruginosa to the cystic fibrosis lung
fibrosis epidemic strain of Pseudomonas aeruginosa displays environment can include deregulation of zwf, encoding
enhanced virulence and antimicrobial resistance. J Bacteriol glucose-6-phosphate dehydrogenase. J Bacteriol 187:
187: 4908–4920. 7561–7568.
Salunkhe P, Töpfer T, Buer J & Tümmler B (2005b) Genome-wide Silverman EK, Spira A & Pare PD (2009) Genetics and genomics
transcriptional profiling of the steady-state response of of chronic obstructive pulmonary disease. Proc Am Thorac Soc
Pseudomonas aeruginosa to hydrogen peroxide. J Bacteriol 187: 6: 539–542.
2565–2572. Slevogt H, Zabel S, Opitz B et al. (2008) CEACAM1 inhibits toll-
Schneider M, Muhlemann K, Droz S, Couzinet S, Casaulta C & like receptor 2-triggered antibacterial responses of human
Zimmerli S (2008) Clinical characteristics associated with pulmonary epithelial cells. Nat Immunol 9: 1270–1278.
isolation of small-colony variants of Staphylococcus aureus Smith EE, Buckley DG, Wu Z et al. (2006) Genetic adaptation by
and Pseudomonas aeruginosa from respiratory secretions Pseudomonas aeruginosa to the airways of cystic fibrosis
of patients with cystic fibrosis. J Clin Microbiol 46: patients. P Natl Acad Sci USA 103: 8487–8492.
1832–1834. Smolonska J, Wijmenga C, Postma DS & Boezen HM (2009)
Schweda EK, Richards JC, Hood DW & Moxon ER (2007) Meta-analyses on suspected chronic obstructive pulmonary
Expression and structural diversity of the lipopolysaccharide disease genes: a summary of 20 years’ research. Am J Resp Crit
of Haemophilus influenzae: implication in virulence. Int J Med Care 180: 618–631.
Microbiol 297: 297–306. Soler N, Torres A, Ewig S et al. (1998) Bronchial microbial
Seemungal T, Harper-Owen R, Bhowmik A et al. (2001) patterns in severe exacerbations of chronic obstructive
Respiratory viruses, symptoms, and inflammatory markers in pulmonary disease (COPD) requiring mechanical ventilation.
acute exacerbations and stable chronic obstructive pulmonary Am J Resp Crit Care 157: 1498–1505.
disease. Am J Resp Crit Care 164: 1618–1623. Soler N, Ewig S, Torres A, Filella X, Gonzalez J & Zaubet A (1999)
Seemungal TAR, Wilkinson TMA, Hurst JR, Perera WR, Airway inflammation and bronchial microbial patterns in
Sapsford RJ & Wedzicha JA (2008) Long-term erythromycin patients with stable chronic obstructive pulmonary disease.
therapy is associated with decreased chronic obstructive Eur Respir J 14: 1015–1022.
pulmonary disease exacerbations. Am J Resp Crit Care 178: Soler N, Agusti C, Angrill J, Puig De la Bellacasa J & Torres A
1139–1147. (2007) Bronchoscopic validation of the significance of sputum
Segal BH, Ding L & Holland SM (2003) Phagocyte NADPH purulence in severe exacerbations of chronic obstructive
oxidase, but not inducible nitric oxide synthase, is essential pulmonary disease. Thorax 62: 29–35.
for early control of Burkholderia cepacia and Chromobacterium Sousa S, Ulrich M, Bragonzi A, Burke M, Worlitzsch D, Leitão JH,
violaceum infection in mice. Infect Immun 71: 205–210. Meisner C, Eberl L, Sá-Correia I & Döring G (2007) Virulence


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Bacterial infections in CF and COPD 145

of Burkholderia cepacia complex strains in gp91phox/ mice. Ulrich M, Herbert S, Berger J, Bellon G, Louis D, Münker G &
Cell Microbiol 9: 2817–2825. Döring G (1998) Localization of Staphylococcus aureus in
Speert DP, Bond M, Woodman RC & Curnutte JT (1994) infected airways of patients with CF and in a cell
Infection with Pseudomonas cepacia in chronic granulomatous culture model of S. aureus adherence. Am J Resp Crit Care 19:
disease: role of nonoxidative killing by neutrophils in host 83–91.
defense. J Infect Dis 170: 1524–1531. Ulrich M, Bastian M, Cramton SE et al. (2007) The
Srikhanta YN, Fox KL & Jennings MP (2010) The phasevarion: staphylococcal respiratory response regulator SrrAB induces
phase variation of type III DNA methyltransferases controls ica gene transcription and PIA expression, protecting
coordinated switching in multiple genes. Nat Rev Microbiol 8: Staphylococcus aureus from neutrophil killing under anaerobic
196–206. growth conditions. Mol Microbiol 65: 1276–1287.
Starkey M, Hickman JA, Ma L et al. (2009) Pseudomonas Ulrich M, Worlitzsch D, Viglio S et al. (2010)
aeruginosa Rugose Small-Colony Variants Have Adaptations Alveolar inflammation in cystic fibrosis. J Cyst Fibros 9:
That Likely Promote Persistence in the Cystic Fibrosis Lung. 217–227.
J Bacteriol 191: 3492–3503. van Delden C (2004) Virulence factors of Pseudomonas
Starner TD, Zhang N, Kim GH, Apicella MA & McCray PB Jr aeruginosa. Pseudomonas: Virulence and Gene Regulation, Vol. 2
(2006) Haemophilus influenzae forms biofilms on airway (Ramos J-L, ed), pp. 3–46. Kluwer Academics/Plenum
epithelia: implications in cystic fibrosis. Am J Resp Crit Care Publishers, New York.
174: 213–220. Verkman AS, Song Y & Thiagarajah JR (2003) Role of airway
Stockley RA (1999) Neutrophils and protease/antiprotease surface liquid and submucosal glands in CF lung disease. Am J
imbalance. Am J Resp Crit Care 160: S49–S52. Physiol-Cell Ph 284: C2–C15.
Stover CK, Pham XQ, Erwin AL et al. (2000) Complete genome Vinion-Dubiel AD & Goldberg JB (2003) Lipopolysaccharide
sequence of Pseudomonas aeruginosa PAO1, an opportunistic of Burkholderia cepacia complex. J Endotoxin Res 9:
pathogen. Nature 406: 959–964. 201–213.
Suh SJ, Silo-Suh L, Woods DE, Hassett DJ, West SE & Ohman DE von Eiff C (2008) Staphylococcus aureus small colony variants: a
(1999) Effect of rpoS mutation on the stress response and challenge to microbiologists and clinicians. Int J Antimicrob
expression of virulence factors in Pseudomonas aeruginosa. J Agents 31: 507–510.
Bacteriol 181: 3890–3897. Watson ME Jr, Burns JL & Smith AL (2004) Hypermutable
Taddei F, Radman M, Maynard-Smith J, Toupance B, Gouyon PH Haemophilus influenzae with mutations in mutS are found in
& Godelle B (1997) Role of mutator alleles in adaptive cystic fibrosis sputum. Microbiology 150: 2947–2958.
evolution. Nature 387: 700–702. Wehmhöner D, Häussler S, Tümmler B, Jänsch L, Bredenbruch F,
Taylor RF, Hodson ME & Pitt TL (1993) Adult cystic fibrosis: Wehland J & Steinmetz I (2003) Inter- and intraclonal
association of acute pulmonary exacerbations and increasing diversity of the Pseudomonas aeruginosa proteome manifests
severity of lung disease with auxotrophic mutants of within the secretome. J Bacteriol 185: 5807–5814.
Pseudomonas aeruginosa. Thorax 48: 1002–1005. Whitchurch CB, Tolker-Nielsen T, Ragas PC & Mattick JS (2002)
Teichgräber V, Ulrich M, Riethmüller J et al. (2008) Ceramide Extracellular DNA required for bacterial biofilm formation.
accumulation mediates inflammation, cell death and infection Science 295: 1487.
susceptibility in CF. Nat Med 14: 382–391. White AJ, Gompertz S, Bayley DL, Hill SL, O’Brien C,
Thomas SR, Ray A, Hodson ME & Pitt TL (2000) Increased Unsal I & Stockley RA (2003) Resolution of bronchial
sputum amino acid concentrations and auxotrophy of inflammation is related to bacterial eradication following
Pseudomonas aeruginosa in severe cystic fibrosis lung disease. treatment of exacerbations of chronic bronchitis. Thorax 58:
Thorax 55: 795–797. 680–685.
Thomassen MJ, Demko CA, Boxerbaum B, Stern RC & Wiehlmann L, Wagner G, Cramer N et al. (2007) Population
Kuchenbrod PJ (1979) Multiple of isolates of Pseudomonas structure of Pseudomonas aeruginosa. P Natl Acad Sci USA 104:
aeruginosa with differing antimicrobial susceptibility patterns 8101–8106.
from patients with cystic fibrosis. J Infect Dis 140: 873–880. Williams P & Camara M (2009) Quorum sensing and
Tosi MF, Zakem H & Berger M (1990) Neutrophil elastase cleaves environmental adaptation in Pseudomonas aeruginosa: a tale of
C3bi on opsonized Pseudomonas as well as CR1 on neutrophils regulatory networks and multifunctional signal molecules.
to create a functionally important opsonin receptor mismatch. Curr Opin Microbiol 12: 182–191.
J Clin Invest 86: 300–308. Wimpenny J, Manz W & Szewzyk U (2000) Heterogeneity in
Tümmler B (2006) Clonal variations in Pseudomonas aeruginosa. biofilms. FEMS Microbiol Rev 24: 661–671.
Pseudomonas: Molecular Biology of Emerging Issues (Ramos JL Winstanley C & Fothergill JL (2009) The role of quorum sensing
& Levesque R, eds), pp. 35–68. Springer, Heidelberg. in chronic cystic fibrosis Pseudomonas aeruginosa infections.
Tunney MM, Field TR, Moriarty TF et al. (2008) Detection of FEMS Microbiol Lett 290: 1–9.
anaerobic bacteria in high numbers in sputum from patients Wood AM & Stockley RA (2006) The genetics of chronic
with cystic fibrosis. Am J Resp Crit Care 177: 995–1001. obstructive pulmonary disease. Respir Res 7: 130.

FEMS Microbiol Rev 35 (2011) 124–146 


c2010 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
146 G. Döring et al.

Woods DE, Schaffer MS, Rabin HR, Campbell GD & Sokol PA Worlitzsch D, Rintelen C, Böhm K et al. (2009) Antibiotic-
(1986) Phenotypic comparison of Pseudomonas aeruginosa resistant obligate anaerobes during exacerbations of cystic
strains isolated from a variety of clinical sites. J Clin Microbiol fibrosis patients. Clin Microbiol Infec 15: 454–460.
24: 260–264. Xu KD, Stewart PS, Xia F, Huang CT & McFeters GA (1998)
Worlitzsch D, Tarran R, Ulrich M et al. (2002) Effects of reduced Spatial physiological heterogeneity in Pseudomonas aeruginosa
mucus oxygen concentration in airway Pseudomonas infections biofilm is determined by oxygen availability. Appl Environ
of cystic fibrosis patients. J Clin Invest 109: 317–325. Microb 64: 4035–4039.


c 2010 Federation of European Microbiological Societies FEMS Microbiol Rev 35 (2011) 124–146
Published by Blackwell Publishing Ltd. All rights reserved
Copyright of FEMS Microbiology Reviews is the property of Wiley-Blackwell and its content may not be
copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written
permission. However, users may print, download, or email articles for individual use.

S-ar putea să vă placă și