You are on page 1of 9

Cellular Microbiology (2009) 11(2), 208–216 doi:10.1111/j.1462-5822.2008.01271.

x
First published online 17 December 2008

Microreview

Cystic fibrosis and innate immunity: how chloride


channel mutations provoke lung disease

Gerd Döring1* and Erich Gulbins2** activation by microbial challenges. Given the essential
1
Institute of Medical Microbiology and Hygiene, role of the innate immune system in preventing infection,
Wilhelmstrasse 31, 72074 Tübingen, Germany. it is conceivable that dysfunction of even one of its many
2
Department of Molecular Biology and Center for Medical components can cause disease. This is demonstrated
Biology, Hufelandstrasse 55, 45122 Essen, Germany. by the fairly large number of primary immunodeficiency
diseases, in which genetic defects provoke a variety of
microbial infections (Notarangelo et al., 2006).
Summary
Although not regarded as a primary immunodeficiency
Innate immunity is essential for prevention of disease, a variety of innate immune functions have been
infection in vertebrates and plants and dysfunction reported to be dysregulated in cystic fibrosis (CF), a
of single components of innate immunity may hereditary disease caused by mutations in a membrane-
provoke severe disease. Here we describe how bound chloride channel, named CF transmembrane con-
mutations in the cystic fibrosis transmembrane ductance regulator (CFTR) (Kerem et al., 1989; Riordan
conductance regulator gene dysregulate a variety et al., 1989; Rommens et al., 1989). These concern
of components of the innate immune system in the MCC system, cationic antimicrobial (poly)peptides
individuals suffering from the hereditary disease (CAMPs), neutrophils, macrophages and CFTR itself. A
cystic fibrosis. In the airways of these individuals, new twist to the pathophysiology of CF was found only
functions of the mucociliary clearance system, cat- recently, when the effect of mutated CFTR on ceramide
ionic antimicrobial (poly)peptides and neutrophils accumulation was detected. Dysregulation of these innate
and macrophages are impaired and inflammatory immune functions causes chronic bacterial lung infections
signal transduction pathways exaggerated. Conse- which are mainly responsible for the reduced life expect-
quently, chronic airway colonization with opportu- ancy in CF patients (CF Foundation, 2005). CF provides
nistic bacterial pathogens develops and leads to not only a fascinating insight into biological mechanisms
life-threatening lung disease. of various innate immune functions, but also into the inter-
play of host defence functions and survival strategies of
opportunistic bacterial pathogens.
Introduction
In contrast to adaptive immunity, innate immunity provides Mucociliary clearance
rapid prevention of infection. This remarkable ability of
innate immunity is based on the rapid recruitment of The inability of mutated CFTR to effectively secrete chlo-
professional phagocytic cells, omnipresent germ-line- ride from respiratory epithelial cells into the airway surface
encoded pattern-recognition receptors, effective signal liquid causes excessive water absorption from the airway
transduction systems, immediate production of reactive surface liquid leading to impaired MCC in individuals with
oxygen species (ROS) and a large armatoire of preformed CF (Matsui et al., 1998; Boucher, 2007) (Fig. 1). This in
antimicrobial molecules, produced by various epithelial turn facilitates the colonization of the viscous mucus layer
cells. Another component of innate immunity, the muco- on the respiratory epithelium with bacterial pathogens
ciliary clearance (MCC) system, protecting the human (Ulrich et al., 1998; Worlitzsch et al., 2002). Although
respiratory tract, is permanently active and does not need studies using respiratory epithelial cell cultures from CF
individuals and controls convincingly demonstrate a
Received 26 September, 2008; revised 17 November, 2008; highly impaired MCC velocity, in vivo data from CF
accepted 20 November, 2008. For correspondence. *E-mail patients suggest MCC rates are heterogeneous (Boucher,
gerd.doering@med.uni-tuebingen.de; Tel. (+49) 7071 298 2069;
Fax (+49) 7071 29 3011; **E-mail: erich.gulbins@uni-due.de; Tel. 2007). The nature of compensatory mechanisms which
(+49) 201 723 3118; Fax (+49) 201 723 5974. allow at least partial function of the MCC system in CF
© 2008 Blackwell Publishing Ltd

cellular microbiology
Cystic fibrosis and innate immunity 209

Fig. 1. Bacterial killing mechanism of innate immunity in the respiratory tract of healthy individuals and cystic fibrosis (CF) 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 or
macrophages or within epithelial cells after uptake via functional CFTR. PMN, polymorphonuclear leukocytes.
B. In individuals with CF defective CFTR leads to a highly viscous mucus layer which impairs mucociliary clearance, the migration of CAMPs,
neutrophils and macrophages towards the bacterial targets, the uptake of bacteria by CFTR itself, induces a pH shift in epithelial lysosomes
which due to ceramide accumulation 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.

patients needs to be studied. Ideally, this should be Consequently, preformed bacteriolytic enzymes and
studied in uninfected airways of CF individuals which are CAMPs such as b-defensins and cathelicidins (Lehrer and
rather difficult to find. Studies in CF mice, on the other Ganz, 2002; Selsted and Quellette, 2003), produced by
hand, may be less informative, since the MCC system submucosal glands, may not be secreted onto the epithe-
differs anatomically from that in humans. lium (Verkman et al., 2003; Joo et al., 2004) (Fig. 1),
resulting in facilitated colonization and infection of the
respiratory epithelium with bacterial pathogens. Attempts
Preformed antimicrobial molecules
to determine the concentration of CAMPs or their traffick-
Viscous secretions may obstruct submucosal gland ducts ing in airways of CF individuals are still missing. In con-
in CF due to mutated CFTR (Engelhardt et al., 1992). trast to the notion that defective CFTR would result in

© 2008 Blackwell Publishing Ltd, Cellular Microbiology, 11, 208–216


210 G. Döring and E. Gulbins

hypotonic salt concentrations which become isotonic after How mutated CFTR mediates ceramide accumulation is
volume depletion, others have hypothesized that hyper- still discussed. Acidification of intracellular vesicles medi-
tonic salt concentrations are present on the respiratory ated by Cftr seems to be critical for the concerted regula-
epithelium (Smith et al., 1996). Such concentrations tion of ceramide by the activities of Asm and acid
would inactivate salt-sensitive CAMPs, thereby leading to ceramidase (Fig. 2) (Teichgräber et al., 2008). According
bacterial multiplication and subsequent infection. to current models, sphingomyelin is constitutively meta-
However, it has been difficult to prove that the airway bolized to ceramide by Asm and further degraded to
surface liquid in CF is indeed hypertonic (Boucher, 2007). sphingosine by acid ceramidase (Kolesnick et al., 2000).
The alkalinization of Cftr-deficient vesicles in respiratory
cells of CF mice from pH 4.5 to a pH of 5.9 causes an
Ceramide accumulation
imbalance of the Asm and acid ceramidase activities,
Biological membranes contain predominantly cholesterol, resulting in a net accumulation of ceramide. Two previous
phospholipids and sphingomyelin. The latter is hydrolysed reports support the notion of defective acidification as a
by the activities of acid, neutral and alkaline sphingomyeli- result of diminished Cl- conductance in other CF cell types.
nases to ceramide. The acid sphingomyelinase (Asm), These studies demonstrated defective acidification in the
residing within pre-lysosomes, lysosomes and secretory Golgi/trans-Golgi network, pre-lysosomes and endosomes
lysosomes and on the outer leaflet of the cell membrane of CF cell lines, leading to abnormal glycosylation of mem-
(Grassmé et al., 2001), is involved in cell death and is brane proteins (Barasch et al., 1991), and showed the
upregulated upon infection (Grassmé et al., 1997; 2003). same phenomenon in phagolysosomes of alveolar macro-
Recently, an abnormal age-dependent accumulation of phages from cftr-null mice, leading to impaired bactericidal
ceramide in the lungs of cftr-deficient mice and in epithelial activity (Di et al., 2006). However, other studies that inves-
cells from CF patients has been detected (Teichgräber tigated the pH in phagosomes in wild type and cftr-deficient
et al., 2008). As a consequence, the rate of cell death cells employing zymosan conjugates containing fluores-
increased in respiratory epithelial cells of uninfected CF cein and tetramethylrhodamine did not report a failure of
mice, resulting in the formation of DNA deposits on the res- these vesicles in CF cells to acidify, suggesting that a
piratory epithelium, which facilitated bacterial adherence. distinct expression pattern of ion channels including Cftr
The link between accumulated ceramide, cell death and determines the pH in different vesicle populations (see
infection with Pseudomonas aeruginosa had been demon- below).
strated by treating CF mice with amitriptyline which blocked
ceramide accumulation, reduced cell death and normal- Epithelial cells
ized the susceptibility of CF mice to P. aeruginosa infection
(Teichgräber et al., 2008). Further evidence for this notion Epithelial cells use CFTR as a receptor for internalization of
was provided by experiments in Cftr-/-/Asm+/- mice. In P. aeruginosa via endocytosis and subsequent removal
these mice ceramide accumulation, cell death rates and of bacteria from the airway. Thus, CFTR itself can be
susceptibility to P. aeruginosa infection were normalized. regarded as a substantial part of the innate immune system
Ceramide accumulation also provoked a pro- and mutant or missing CFTR has been linked to the
inflammatory status in the respiratory tissue of mice with initiation of P. aeruginosa infection (Pier, 2000), since the
CF, which preceded bacterial infection: an increased syn- CFTR–P. aeruginosa interaction does not occur, allowing
thesis and release of cytokines and a subsequent, age- for increased bacterial loads in the lungs. Binding occurs
dependent, recruitment of macrophages and neutrophils between the outer core of the bacterial lipopolysaccharide
had been observed in lungs of uninfected CF mice (Fig. 1) and amino acids 108–117 in the first predicted extracellular
(Teichgräber et al., 2008). That these phenomena are trig- domain of CFTR. In experimentally infected mice, inhibiting
gered by accumulated ceramide was demonstrated by CFTR-mediated endocytosis of P. aeruginosa by inclusion
experiments revealing that partial Asm inhibition by ami- in the bacterial inoculum of either free bacterial lipopoly-
triptyline or Asm heterozygosity abrogated effector cells saccharide or CFTR peptide 108–117 resulted in increased
accumulation in CF mouse lung tissues. How the accu- bacterial counts in the lungs suggesting that internalization
mulation of macrophages and neutrophils in lung tissues contributes – via still unknown mechanisms – to the clear-
of uninfected CF mice contributes to the hypersuscepti- ance of the bacteria in the lung (Fig. 1) (Pier, 2000).
bility of CF mice to P. aeruginosa lung infections remains
to be defined. The results unify several previously pub- Professional phagocytes
lished observations in CF mouse strains with regard to
Cell migration
inflammation (Zahm et al., 1997), apoptosis (Cannon
et al., 2003) and hypersusceptibility to P. aeruginosa The high viscosity of CF secretions negatively affects
infection (Pier, 2000; Coleman et al., 2003). migration of neutrophils (Matsui et al., 2005) (Fig. 1). The

© 2008 Blackwell Publishing Ltd, Cellular Microbiology, 11, 208–216


Cystic fibrosis and innate immunity 211

failure of neutrophils to rapidly encounter the pathogens in


the viscous mucus may allow bacterial multiplication and
profound changes on the bacterial phenotypes which
facilitate chronic infection (see below). The unaccom-
plished mission to kill mucus-encased bacteria will result
in increased endobronchial DNA deposition, further
increasing mucus viscosity, bacterial adhesion and biofilm
formation (Whitchurch et al., 2002; Worlitzsch et al., 2002;
Matsui et al., 2006) in addition to ceramide-driven DNA
deposition, as well as the liberation of granule-stored pro-
teases and antimicrobial peptides which finally leads a
lung tissue remodelling (Nathan, 2006). Increasing mucus
viscoelasticity then also impairs antimicrobial compounds
such as lactoferrin to encounter their bacterial targets
(Matsui et al., 2006). Impaired neutrophil migration may
also be partly responsible for the reduced efficacy of
vaccines against P. aeruginosa in individuals with CF,
despite the development of high and long-lasting
P. aeruginosa-specific antibody titres after vaccination
(Döring et al., 2007).

Defective acidification
A recent study demonstrated that lysosomal CFTR par-
ticipates in phagosomal pH control (Fig. 2) (Di et al.,
2006). Due to its counterion effect of Cl- in promoting
lumenal H+ accumulation by the lysosomal V-type
H+-ATPase, CFTR ensures acidification of late phago-
somes to pH < 5 and provides optimal bactericidal activi-
ties for degradative enzymes when lysosomes fuse with
phagosomes during the maturation process of these
organelles. Alveolar macrophages from CFtr -/- mice
retained the ability to phagocytose and generate an oxi-
dative burst, but exhibited defective killing of internalized
bacteria (Di et al., 2006). The contribution of CFTR to the
regulation of pH in intracellular organelles, first described
in trans-Golgi vesicles (Barasch et al., 1991), has been
Fig. 2. Consequences of defective acidification in lysosomes of controversially discussed (Seksek et al., 1996; Haggie
epithelial cells and the phagolysosome of macrophages for
bacterial infection. and Verkman, 2007), possibly due to experimental diffi-
A. In lysosomes and phagosomes of normal respiratory epithelial culties to ensure targeting of identical vesicle populations
cells and macrophages, respectively, sphingomyelin is constitutively by pH-sensitive fluorescent dyes.
metabolized to ceramide by acid sphingomyelinase (Asm) and
further degraded to sphingosine by acid ceramidase (Ac). Due to
its counterion effect of Cl- in promoting lumenal H+ accumulation by
the lysosomal V-type H+-ATPase, CFTR ensures acidification of Oxidative killing
lysosomes and late phagosomes to pH < 5. In macrophages this
pH provides optimal bactericidal activities for degradative enzymes In the bronchi, the highly viscous mucus generates a
when lysosomes fuse with phagosomes during the maturation microaerobic milieu which may become anaerobic by
process of these organelles. a rapid oxygen consumption by pathogens such as
B. In individuals with CF defective CFTR leads to an imbalance of
Asm and Ac activities which leads in turn to ceramide P. aeruginosa (Worlitzsch et al., 2002). Consequently, the
accumulation. As a consequence of the alkalinization, increased generation of ROS by neutrophils and other cells is abol-
cytokine release, cell death and decreased bacterial killing are ished and bacterial killing impaired (Fig. 3). The observa-
observed leading to fibrosis and bacterial infection.
tion that anaerobic conditions impair apoptosis (Hannah
et al., 1995; Zhang et al., 2003) and NETosis (Brinkmann
and Zychlinsky, 2007) may explain why neutrophil necro-
sis is so dominant in infected airways of individuals with

© 2008 Blackwell Publishing Ltd, Cellular Microbiology, 11, 208–216


212 G. Döring and E. Gulbins

Fig. 3. Bacterial sensitivity and resistance to neutrophil-mediated killing in aerobic and anaerobic environments.
A. On the respiratory epithelium of healthy individuals, neutrophils effectively kill the CF-related bacterial pathogens P. aeruginosa, S. aureus
and B. cepacia by reactive oxygen species (ROS) and defensins.
B. Neutrophils in the anaerobic milieu in the mucus layer overlaying the respiratory epithelium in individuals with CF are devoid of ROS,
leading to intra-neutrophil survival of S. aureus and B. cepacia which are resistant to non-oxidative killing. The anaerobic milieu also triggers
biofilm formation of S. aureus and P. aeruginosa which prevents phagocytosis.

© 2008 Blackwell Publishing Ltd, Cellular Microbiology, 11, 208–216


Cystic fibrosis and innate immunity 213

CF. Whether lack of ROS also impairs the non-oxidative Limitation of innate and adaptive immune function
innate defence mechanisms (Segal, 2005) has been during chronic infection
controversially discussed (Nathan, 2006). Nevertheless,
The exaggerated inflammatory response to persistent
microaerobic/anaerobic environmental conditions on the
infection not only facilitates tissue destruction but also
respiratory epithelium allow microbial pathogens, intrins-
induces tissue remodelling, in which a number of pro-
ingly resistant to non-oxidative killing or capable to
teases, growth factors and other compounds from various
change their phenotype to become rapidly resistant to
cells are involved. During chronic inflammation, several
non-oxidative killing, to persist in airways of individuals
immune cell functions are impaired. For instance, human
with CF.
neutrophil elastase cleaves immunoglobulins, comple-
This notion is in line with the observed microbial species
ment components (Brozna et al., 1977; Döring et al.,
which cause disease in individuals with CF: the
1986; Berger et al., 1989), and a variety of cell receptors
microaerobic/anaerobic environmental conditions on the
(Sommerhoff et al., 1990; Döring et al., 1995; Ferry et al.,
respiratory epithelium in CF provoke a switch from single
1997; Voynow et al., 1999; Döring et al., 2000; Walsh
cells of P. aeruginosa which are sensitive to both oxidative
et al., 2001; Henriksen et al., 2004; Caldwell et al., 2005;
and non-oxidative killing by neutrophils (Sousa et al.,
Suzuki et al., 2005; Hartl et al., 2007) and surfactant pro-
2007) to a mucoid phenotype (Worlitzsch et al., 2002;
teins (Griese et al., 2004; Hirche et al., 2004; Rubio et al.,
Matsui et al., 2006) now resistant to non-oxidative killing
2004). Persistent levels of active elastase also contribute
by phagocytes. Additionally, structural changes of the
to ineffective removal of apoptotic cells and continuous
outer membrane occur during anaerobic growth of
low-grade inflammation by cleavage of the phosphati-
P. aeruginosa (Sabra et al., 2003). This change provides
dylserine receptor (Vandivier et al., 2002). Further, human
enhanced resistance to cationic antimicrobials, such as
neutrophil elastase downregulated the expression of
aminoglycoside antibiotics, and potentially to naturally
CD40, CD80 and CD86 on DCs and inhibits DC matura-
produced cationic peptides due to inhibition of charge-
tion (Roghanian et al., 2006). Finally, neutrophil elastase
mediated uptake. Similarly, Staphylococcus aureus
cleaves its own endogenous inhibitor, a-1-antitrypsin
changes its phenotype under microaerobic/anaerobic
when protease concentrations are higher than inhibitor
environmental conditions forming polysaccharide-
concentrations (Goldstein and Döring, 1986).
encased cells which confer resistance to non-oxidative
killing (McKenney et al., 1999; Cramton et al., 2001;
Ulrich et al., 2007). Finally, strains of the Burkholderia
Conclusion
cepacia complex, intrinsingly resistant to non-oxidative
killing, cause disease in individuals with CF (Sousa Cystic fibrosis is unique in its remarkable number of dys-
et al., 2007). Since the anaerobic environment is locally regulated innate immune functions which are conse-
restricted to the lung epithelium, bacterial sepsis is virtu- quences of mutations in CFTR. Synergistically, they may
ally not observed in CF and chronic microbial infections provoke lung disease in virtually all subjects caused by a
remain localized to the endobronchial site. Interestingly, limited number of microbial pathogens adapted to this
S. aureus and B. cepacia complex strains but not specific environmental niche. Combined, defective innate
P. aeruginosa strains are pathogens in chronic granulo- immunity leads to chronic disease in which a vicious cycle
matous disease, caused by mutations in the NADPH still worsens the function of innate defence mechanisms.
oxidase (Heyworth et al., 2003). For instance, accumulation of ceramide on the respiratory
Besides P. aeruginosa, S. aureus and B. cepacia epithelium may still increase after the onset of chronic
complex strains, also other microbial species colonize the P. aeruginosa infection since the pathogen itself triggers
airways of CF patients. The microaerobic/anaerobic envi- Asm activation (Grassmé et al., 2003), and, hence, may
ronmental conditions in the sputum let strict anaerobic further facilitate bacterial adhesion by increasing the depo-
species grow to cell numbers comparable to those of sition of DNA, impair MCC by increasing the viscoelasticity
P. aeruginosa (Tunney et al., 2008). A large number of of the airway surface liquid, and augment tissue destruc-
different species have been detected and future studies tion and remodelling by increasing inflammation. Neu-
will provide information whether these bacteria contribute trophil necrosis will further enhance extracellular DNA
to the pathophysiology of CF lung disease. Taken concentrations. The anaerobic niche, in which biofilm
together, only bacteria that are able to protect themselves bacteria surrounded by viscous mucus persist, will impair
from non-oxidative killing by the production of toxins or apoptosis and induce necrosis of neutrophils, thus
biofilm formation and that can sufficiently grow rapidly to enhancing the volume and the viscosity of mucus plugs.
high cell numbers in this specific niche become dominant Increased sputum production then will attract strict anaero-
pathogens in CF lungs. Apparently, enteric bacteria lack bic bacteria which further complicate the pathophysiology
some of these characteristics. of lung disease in CF. Unravelling dysregulation of innate

© 2008 Blackwell Publishing Ltd, Cellular Microbiology, 11, 208–216


214 G. Döring and E. Gulbins

immunity in CF may allow developing drugs with the aim to Döring, G., Knight, R., and Bellon, G. (2000) Immunology of
normalize innate immune functions. CF. In Cystic Fibrosis. Hodson, M.E., and Geddes, D.
(eds). London: Arnold, pp. 109–140.
Döring, G., Meisner, C., Stern, M., for the Flagella Vaccine
Acknowledgements Trial Study Group (2007) A double-blind randomized
placebo-controlled phase III study of a Pseudomonas
The authors want to thank Peter Michael Weber, Children’s
aeruginosa flagella vaccine in CF patients. Proc Natl Acad
Clinic, University of Tübingen, Germany, for excellent graphic art.
Sci USA 104: 11020–11025.
Engelhardt, J.F., Yankaskas, J.R., Ernst, S.A., Yang, Y.,
References Marino, C.R., Boucher, R.C., et al. (1992) Submucosal
glands are the predominant site of CFTR expression in the
Barasch, J., Kiss, B., Prince, A., Saiman, L., Gruenert, D., human bronchus. Nat Genet 2: 240–248.
and al-Awqati, Q. (1991) Defective acidification of intracel- Ferry, G., Lonchampt, M., Pennel, L., de Nanteuil, G., Canet,
lular organelles in CF. Nature 352: 70–73. E., and Tucker, G.C. (1997) Activation of MMP-9 by neu-
Berger, M., Sorensen, R.U., Tosi, M.F., Dearborn. D.G., and trophil elastase in an in vivo model of acute lung injury.
Döring, G. (1989) Complement receptor expression on FEBS Lett 402: 111–115.
neutrophils at an inflammatory site, the Pseudomonas- Goldstein, W., and Döring, G. (1986) Lysosomal enzymes
infected lung in CF. J Clin Invest 84: 1302–1313. from polymorphonuclear leukocytes and proteinase inhibi-
Boucher, R.C. (2007) Airway surface dehydration in CF: tors in patients with CF. Am Rev Respir Dis 134: 49–56.
pathogenesis and therapy. Annu Rev Med 58: 157–170. Grassmé, H., Gulbins, E., Brenner, B., Ferlinz, K., Sandhoff,
Brinkmann, V., and Zychlinsky, A. (2007) Beneficial suicide: K., Harzer, K., et al. (1997) Acidic sphingomyelinase medi-
why neutrophils die to make nets. Nat Rev Microbiol 5: ates entry of N. gonorrhoeae into nonphagocytic cells. Cell
577–582. 91: 605–615.
Brozna, J.P., Senior, R.M., Kreutzer, D.L., and Ward, P.A. Grassmé, H., Jekle, A., Riehle, A., Schwarz, H., Berger, J.,
(1977) Chemotactic factor inactivators of human granulo- Sandhoff, K., et al. (2001) CD95 signaling via ceramide-
cytes. J Clin Invest 60: 1280–1288. rich membrane rafts. J Biol Chem 276: 20589–20596.
Caldwell, R.A., Boucher, R.C., and Stutts, M.J. (2005) Neu- Grassmé, H., Jendrossek, V., Riehle, A., von Kurthy, G.,
trophil elastase activates near-silent epithelial Na+ chan- Berger, J., Schwarz, H., et al. (2003) Host defense against
nels and increases airway epithelial Na+ transport. Am J Pseudomonas aeruginosa requires ceramide-rich mem-
Physiol 288: L813–L819. brane rafts. Nat Med 9: 322–330.
Cannon, C.L., Kowalski, M.P., Stopak, K.S., and Pier, G.B. Griese, M., Essl, R., Schmidt, R., Rietschel, E., Ratjen, F.,
(2003) Pseudomonas aeruginosa-induced apoptosis is Ballmann, M., et al. (2004) Pulmonary surfactant, lung
defective in respiratory epithelial cells expressing mutant function, and endobronchial inflammation in CF. Am J
CF transmembrane conductance regulator. Am J Respir Respir Crit Care Med 170: 1000–1005.
Cell Mol Biol 29: 188–197. Haggie, P.M., and Verkman, A.S. (2007) CF transmembrane
CF Foundation (2005) Patient Registry Annual Report conductance regulator-independent phagosomal acidifica-
2004. Bethesda, MD [WWW document]. URL http://www. tion in macrophages. J Biol Chem 282: 31422–31428.
cff.org/LivingWithCF/CareCenterNetwork/PatientRegistry/. Hannah, S., Mecklenburgh, K., Rahman, I., Bellingan, G.J.,
Coleman, F.T., Mueschenborn, S., Meluleni, G., Ray, C., Greening, A., Haslett, C., and Chilvers, E.R. (1995)
Carey, V.J., Vargas, S.O., et al. (2003) Hypersusceptibility Hypoxia prolongs neutrophil survival in vitro. FEBS Lett
of CF mice to chronic Pseudomonas aeruginosa oropha- 372: 233–237.
ryngeal colonization and lung infection. Proc Natl Acad Sci Hartl, D., Latzin, P., Hordijk, P., Marcos, V., Rudolph, C.,
USA 100: 1949–1954. Woischnik, M., et al. (2007) Cleavage of CXCR1 on neu-
Cramton, S.E., Ulrich, M., Gotz, F., and Doring, G. (2001) trophils disables bacterial killing in CF lung disease. Nat
Anaerobic conditions induce expression of polysaccharide Med 13: 1423–1430.
intercellular adhesin in Staphylococcus aureus and Sta- Henriksen, P.A., Hitt, M., Xing, Z., Wang, J., Haslett, C.,
phylococcus epidermidis. Infect Immun 69: 4079–4085. Riemersma, R.A., et al. (2004) Adenoviral gene delivery of
Di, A., Brown, M.E., Deriy, L.V., Li, C., Szeto, F.L., Chen, Y., elafin and secretory leukocyte protease inhibitor attenuates
et al. (2006) CFTR regulates phagosome acidification in NF-kappa B-dependent inflammatory responses of human
macrophages and alters bactericidal activity. Nat Cell Biol endothelial cells and macrophages to atherogenic stimuli.
8: 933–944. J Immunol 172: 4535–4544.
Döring, G., Goldstein, W., Botzenhart, K., Kharazmi, A., Heyworth, P.G., Cross, A.R., and Cornutte, J.T. (2003)
Schiøtz, P.O., Høiby, N., and Dasgupta, M. (1986) Chronic granulomatous disease. Curr Opin Immunol 15:
Elastase from polymorphonuclear leukocytes – a regula- 578–584.
tory enzyme in immune complex disease. Clin Exp Hirche, T.O., Crouch, E.C., Espinola, M., Brokelman, T.J.,
Immunol 64: 597–605. Mecham, R.P., DeSilva, N., et al. (2004) Neutrophil serine
Döring, G., Frank, F., Boudier, C., Herbert, S., Fleischer, B., proteinases inactivate surfactant protein D by cleaving
and Bellon, G. (1995) Cleavage of lymphocyte surface within a conserved subregion of the carbohydrate recogni-
antigens CD2, CD4, and CD8 by polymorphonuclear leu- tion domain. J Biol Chem 279: 27688–27698.
kocyte elastase and cathepsin G in patients with CF. Joo, N.S., Lee, D.J., Winges, K.M., Rustagi, A., and Wine,
J Immunol 154: 4842–4850. J.J. (2004) Regulation of antiprotease and antimicrobial

© 2008 Blackwell Publishing Ltd, Cellular Microbiology, 11, 208–216


Cystic fibrosis and innate immunity 215
protein secretion by airway submucosal gland serous cells. Segal, A.J. (2005) How neutrophils kill microbes. Annu Rev
J Biol Chem 279: 38854–38860. Immunol 23: 197–223.
Kerem, B., Rommens, J.M., Buchanan, J.A., Markiewicz, D., Seksek, O., Biwersi, J., and Verkman, A.S. (1996) Evidence
Cox, T.K., Chakravarti, A., et al. (1989) Identification of the against defective trans-Golgi acidification in CF. J Biol
CF gene: genetic analysis. Science 245: 1073–1080. Chem 271: 15542–15548.
Kolesnick, R.N., Goni, F.M., and Alonso, A. (2000) Compart- Selsted, M.E., and Quellette, A.J. (2003) Mammalian
mentalization of ceramide signaling: physical foundations defensins in the antimicrobial immune response. Nat
and biological effects. J Cell Physiol 184: 285–300. Immunol 6: 551–557.
Lehrer, R.I., and Ganz, T. (2002) Defensins of vertebrate Smith, J.J., Travis, S.M., Greenberg, E.P., and Welsh, M.J.
animals. Curr Opin Immunol 14: 96–102. (1996) Cystic fibrosis airway epithelia fail to kill bacteria
McKenney, D., Tibbetts, K.L., Wang, Y., Murthy, V., Ulrich, because of abnormal airway surface fluid. Cell 85: 229–236.
M., Döring, G., et al. (1999) Broadly protective vaccine for Sommerhoff, C.P., Nadel, J.A., Basbaum, C.B., and
Staphylococcus aureus based on an in vivo-expressed Caughey, G.H. (1990) Neutrophil elastase and cathepsin G
antigen. Science 284: 1523–1527. stimulate secretion from cultured bovine airway gland
Matsui, H., Grubb, B.R., Tarran, R., Randell, S.H., Gatzy, serous cells. J Clin Invest 85: 682–689.
J.T., Davis, C.W., and Boucher, R.C. (1998) Evidence for Sousa, S., Ulrich, M., Bragonzi, A., Burke, M., Worlitzsch, D.,
periciliary liquid layer depletion, not abnormal ion compo- Leitão, J.H., et al. (2007) Virulence of Burkholderia cepacia
sition, in the pathogenesis of CF airways disease. Cell 95: complex strains in gp91phox-/- mice. Cell Microbiol 9: 2817–
1005–1015. 2825.
Matsui, H., Verghese, M.W., Kesimer, M., Schwab, U.E., Suzuki, T., Moraes, T.J., Vachon, E., Ginzberg, H.H., Huang,
Randell, S.H., Sheehan, J.K., et al. (2005) Reduced three- T.-T., Matthay, M.A., et al. (2005) Proteinase-activated
dimensional motility in dehydrated airway mucus prevents receptor-1 mediates elastase-mediated apoptosis of
neutrophil capture and killing bacteria on airway epithelial human lung epithelial cells. Am J Respir Cell Mol Biol 33:
surfaces. J Immunol 175: 1090–1099. 231–247.
Matsui, H., Wagner, V.E., Hill, D.B., Schwab, U.E., Teichgräber, V., Ulrich, M., Riethmüller, J., Grassme, H.,
Rogers, T.D., Button, B., et al. (2006) A physical linkage Wilker, B., De Oliveira-Munding, C.C., et al. (2008) Cera-
between CF airway surface dehydration and Pseudomo- mide accumulation mediates inflammation, cell death and
nas aeruginosa biofilms. Proc Natl Acad Sci USA 103: infection susceptibility in CF. Nat Med 14: 382–391.
18131–18136. Tunney, M.M., Field, T.R., Moriarty, T.F., Patrick, S., Döring,
Nathan, C. (2006) Neutrophils and immunity: challenges and G., Muhlebach, M.S., et al. (2008) Detection of anaerobic
opportunities. Nat Rev Immunol 6: 173–182. bacteria in high numbers in sputum from patients with
Notarangelo, L., Casanova, J.L., Conley, M.E., Chapel, H., cystic fibrosis. Am J Respir Crit Care Med 177: 995–1001.
Fischer, A., Puck, J., et al. (2006) Primary immunodefi- Ulrich, M., Herbert, S., Berger, J., Bellon, G., Louis, D.,
ciency diseases: an update from the International Union of Münker, G., and Döring, G. (1998) Localization of Staphy-
Immunological Societies Primary Immunodeficiency Dis- lococcus aureus in infected airways of patients with CF and
eases Classification Committee meeting in Budapest 2005. in a cell culture model of S. aureus adherence. Am J Respir
J Allergy Clin Immunol 117: 883–896. Crit Care Med 19: 83–91.
Pier, G.B. (2000) Role of the CF transmembrane conduc- Ulrich, M., Bastian, M., Cramton, S.E., Ziegler, K., Pragman,
tance regulator in innate immunity to Pseudomonas aerugi- A.A., Bragonzi, A., et al. (2007) The staphylococcal respi-
nosa infections. Proc Natl Acad Sci USA 97: 8822–8828. ratory response regulator SrrAB induces ica gene tran-
Riordan, J.R., Rommens, J.M., Kerem, B., Alon, N., Rozma- scription and PIA expression, protecting Staphylococcus
hel, R., Grzelczak, Z., et al. (1989) Identification of the CF aureus from neutrophil killing under anaerobic growth con-
gene: cloning and characterization of complementary DNA. ditions. Mol Microbiol 65: 1276–1287.
Science 245: 1066–1073. Vandivier, R.W., Fadok, V.A., Hoffmann, P.R., Bratton, D.L.,
Roghanian, A., Williams, S.E., Sheldrake, T.A., Brown, T.I., Penvari, C., Brown, K.K., et al. (2002) Elastase-mediated
Oberheim, K., Xing, Z., et al. (2006) The antimicrobial/ phosphatidylserine receptor cleavage impairs apoptotic
elastase inhibitor elafin regulates lung dendritic cells and cell clearance in CF and bronchiectasis. J Clin Invest 109:
adaptive immunity. Am J Respir Cell Mol Biol 34: 634–642. 661–670.
Rommens, J.M., Iannuzzi, M.C., Kerem, B., Drumm, M.L., Verkman, A.S., Song, Y., and Thiagarajah, J.R. (2003) Role
Melmer, G., Dean, M., et al. (1989) Identification of the CF of airway surface liquid and submucosal glands in CF lung
gene: chromosome walking and jumping. Science 245: disease. Am J Physiol Cell Physiol 284: C2–C15.
1059–1065. Voynow, J.A., Young, L.R., Wang, Y., Horger, T., Rose, M.C.,
Rubio, F., Cooley, J., Accurso, F.J., and Remold-O’Donnell, and Fischer, B.M. (1999) Neutrophil elastase increases
E. (2004) Linkage of neutrophil serine proteases and MUC5AC mRNA and protein expression in respiratory epi-
decreased surfactant protein-A (SP-A) levels in inflamma- thelial cells. Am J Physiol 276: L835–L843.
tory lung disease. Thorax 59: 318–323. Walsh, D.E., Greene, C.M., Carroll, T.P., Taggart, C.C., Gal-
Sabra, W., Lunsdorf, H., and Zeng, A.P. (2003) Alterations lagher, P.M., O’Neill, S.J., and McElvaney, N.G. (2001)
in the formation of lipopolysaccharide and membrane Interleukin-8 up-regulation by neutrophil elastase is medi-
vesicles on the surface of Pseudomonas aeruginosa PAO1 ated by MyD88/IRAK/TRAF-6 in human bronchial epithe-
under oxygen stress conditions. Microbiology 149: 2789– lium. J Biol Chem 276: 354–394.
2795. Whitchurch, C.B., Tolker-Nielsen, T., Ragas, P.C., and

© 2008 Blackwell Publishing Ltd, Cellular Microbiology, 11, 208–216


216 G. Döring and E. Gulbins
Mattick, J.S. (2002) Extracellular DNA required for bacterial D., Dorin, J.R., et al. (1997) Early alterations in airway
biofilm formation. Science 295: 1487. mucociliary clearance and inflammation of the lamina
Worlitzsch, D., Tarran, R., Ulrich, M., Schwab, U., Cekici, A., propria in CF mice. Am J Physiol 272: C853–C859.
Meyer, K.C., et al. (2002) Reduced oxygen concentrations Zhang, B., Hirahashi, J., Cullere, X., and Mayadas, T.N.
in airway mucus contribute to the early and late pathogen- (2003) Elucidation of molecular events leading to neutro-
esis of Pseudomonas aeruginosa CF airways infection. phil apoptosis following phagocytosis: cross-talk between
J Clin Invest 109: 317–325. caspase 8, reactive oxygen species, and MAPK/ERK acti-
Zahm, J.M., Gaillard, D., Dupuit, F., Hinnrasky, J., Porteous, vation. J Biol Chem 278: 28443–28454.

© 2008 Blackwell Publishing Ltd, Cellular Microbiology, 11, 208–216

You might also like