Download Protein Tuf Is a Factor H and Plasminogen Binding

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Complement component 4 wikipedia , lookup

Complement system wikipedia , lookup

Transcript
This information is current as
of June 11, 2017.
Immune Evasion of the Human Pathogen
Pseudomonas aeruginosa: Elongation Factor
Tuf Is a Factor H and Plasminogen Binding
Protein
Anja Kunert, Josephine Losse, Christin Gruszin, Michael
Hühn, Kerstin Kaendler, Stefan Mikkat, Daniela Volke, Ralf
Hoffmann, T. Sakari Jokiranta, Harald Seeberger, Ute
Moellmann, Jens Hellwage and Peter F. Zipfel
References
Subscription
Permissions
Email Alerts
This article cites 63 articles, 34 of which you can access for free at:
http://www.jimmunol.org/content/179/5/2979.full#ref-list-1
Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscription
Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts
The Journal of Immunology is published twice each month by
The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2007 by The American Association of
Immunologists All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
J Immunol 2007; 179:2979-2988; ;
doi: 10.4049/jimmunol.179.5.2979
http://www.jimmunol.org/content/179/5/2979
The Journal of Immunology
Immune Evasion of the Human Pathogen
Pseudomonas aeruginosa: Elongation Factor Tuf
Is a Factor H and Plasminogen Binding Protein
Anja Kunert,* Josephine Losse,* Christin Gruszin,* Michael Hühn,* Kerstin Kaendler,*
Stefan Mikkat,† Daniela Volke,‡ Ralf Hoffmann,‡ T. Sakari Jokiranta,§ Harald Seeberger,*
Ute Moellmann,* Jens Hellwage,* and Peter F. Zipfel1*¶
T
he opportunistic pathogen Pseudomonas aeruginosa
causes life-threatening infections in humans, including
pneumonia and bloodstream infections. This Gram-negative bacterium is a major cause of hospital-acquired infections especially in immunocompromised patients (1). In a recent report of
the Infectious Diseases Society of America, P. aeruginosa is listed
as one of the most dangerous human pathogens (2). Due to antibiotic resistance, P. aeruginosa infections are difficult to treat and
suitable drugs controlling P. aeruginosa infections are currently
limited (3). Lung colonization of cystic fibrosis patients by P.
aeruginosa is an important cause of chronic infections and results
in high morbidity and mortality. Several virulence factors such as
exotoxin A, hemolysin, and phospholipase C have been identified,
which enable the bacterium to evade the host immune attack (4).
However, it is unclear how this bacterium controls innate immunity and evades complement attack.
P. aeruginosa isolates and strains differ in resistance to complement-mediated lysis (5). The human complement system plays
an important role in clearance of early pulmonary P. aeruginosa
infection (6). Deposition of the complement component C3b at the
*Department of Infection Biology, Leibniz Institute for Natural Product Research and
Infection Biology, Jena, Germany; †University of Rostock, Medical Faculty, Rostock,
Germany; ‡University of Leipzig, Center for Biotechnology and Biomedicine,
Leipzig, Germany; §Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland; and ¶Faculty of Biology, Friedrich-Schiller-University, Jena, Germany
Received for publication May 3, 2007. Accepted for publication May 25, 2007.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
Address correspondence and reprint requests to Dr. Peter F. Zipfel, Leibniz Institute
for Natural Product Research and Infection Biology (Hans-Knoell-Institute), Beutenbergstrasse 11a, 07745 Jena, Germany. E-mail address: [email protected]
www.jimmunol.org
bacterial surface is important for induction of host responses and
elimination of the pathogen. Complement-deficient mice when
challenged with P. aeruginosa showed an amplified inflammatory
response and did not clear the pathogen (7). P. aeruginosa exploit
multiple strategies for immune evasion (4). The pathogen secretes
catalytic enzymes like alkaline protease and elastase, which degrade the complement activation product C3b deposited at the bacterial surface (8, 9). In addition, P. aeruginosa expresses LPS variants that interfere with C3b deposition at the surface (5, 10).
Complement evasion is particularly important during the initial
phase of infection, when individual bacteria are in contact with the
body fluids of the host. In this phase, the alternative pathway of
complement activation is central for immune recognition (11).
Similarly, the pathogen forms an alginate layer, which limits accessibility and the action of host plasma factors like complement
(12). During late infection, P. aeruginosa can form biofilms, which
protect the bacteria from complement attack and prevent complement-mediated lysis, phagocytosis, and access of antibiotics (13).
Factor H is the major host fluid-phase complement regulator that
controls alternative pathway activation and the amplification reaction at the level of C3 (14). Factor H promotes cleavage of surfacebound C3b by acting as a cofactor for the plasma serine protease
Factor I. In addition, Factor H accelerates the decay of the alternative pathway C3 convertase, C3bBb, and competes with Factor
B for binding to C3b (15). Factor H is a member of a protein
family that includes the Factor H-like protein (FHL)-1,2 which is
an alternatively spliced product of the Factor H gene, and five
2
Abbreviations used in this paper: FHL, Factor H-like protein; FHR, Factor H-related
protein; SCR, short consensus repeat; HiNHS, heat-inactivated normal human serum;
uPA, urokinase plasminogen activator.
Copyright © 2007 by The American Association of Immunologists, Inc. 0022-1767/07/$2.00
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
Pseudomonas aeruginosa is an opportunistic human pathogen that can cause a wide range of clinical symptoms and infections that
are frequent in immunocompromised patients. In this study, we show that P. aeruginosa evades human complement attack by
binding the human plasma regulators Factor H and Factor H-related protein-1 (FHR-1) to its surface. Factor H binds to intact
bacteria via two sites that are located within short consensus repeat (SCR) domains 6 –7 and 19 –20, and FHR-1 binds within SCR
domain 3–5. A P. aeruginosa Factor H binding protein was isolated using a Factor H affinity matrix, and was identified by mass
spectrometry as the elongation factor Tuf. Factor H uses the same domains for binding to recombinant Tuf and to intact bacteria.
Factor H bound to recombinant Tuf displayed cofactor activity for degradation of C3b. Similarly Factor H bound to intact P.
aeruginosa showed complement regulatory activity and mediated C3b degradation. This acquired complement control was rather
effective and acted in concert with endogenous proteases. Immunolocalization identified Tuf as a surface protein of P. aeruginosa.
Tuf also bound plasminogen, and Tuf-bound plasminogen was converted by urokinase plasminogen activator to active plasmin.
Thus, at the bacterial surface Tuf acts as a virulence factor and binds the human complement regulator Factor H and plasminogen.
Acquisition of host effector proteins to the surface of the pathogen allows complement control and may facilitate tissue
invasion. The Journal of Immunology, 2007, 179: 2979 –2988.
2980
Materials and Methods
P. aeruginosa strains and growth conditions
P. aeruginosa strains 27853 (American Type Culture Collection (ATCC)),
10662, SG137, PAO1 (National Collection of Type Cultures (NCTC)), the
PAO1 derivative AH377 (31), and various clinical isolates were cultivated
overnight at 37°C in enriched Nutrient Broth (Serva). Eight clinical isolates
(Is1-Is8) were derived from patients with different diseases including pyelonephritis, leukopenia, T cell lymphoma, urosepsis, or myeloma. All cultures were grown to an OD550 of 1. Cells were collected by centrifugation
and washed three times with PBS supplemented with 20 mM EDTA.
Expression of recombinant proteins and generation of antiserum
Factor H deletion constructs were expressed in Sf9 insect cells following
infection with recombinant baculovirus coding for Factor H SCRs 1–5,
1– 6, 1–7, 15–18, 15–19, and 15–20 (15, 32, 33). The Factor H construct
SCR19 –20, the cDNA of FHR-1, and its C-terminal deletion construct
FHR-1/SCR3–5 were cloned in pPICZ␣B (Invitrogen Life Technologies)
and expressed in the yeast Pichia pastoris strain X-33 according to standard protocols.
The tufB gene from P. aeruginosa was amplified from genomic DNA of
strain PAO1 using primers tuf (forward) 5⬘-GCGGATCCATGGCTAAA
GAAAAATTTGAACGG-3⬘ and (reverse) 5⬘-TTCTGCAGTTATTCGAT
GATCTTGGCAACCAC-3⬘. The amplicon was cloned into expression
vector pQE-9 (Qiagen) and transformed into Escherichia coli strain
SG13009 (pREP4; Qiagen), and protein expression was induced by isopropyl b-D-thiogalactoside. His-tagged Tuf was purified by chromatography using a zinc-loaded HiTrap chelating column (GE Healthcare) and
dialyzed against PBS using Slide-A-Lyzer (Pierce). All other recombinant
proteins were His-tagged and were purified by HisTrap columns (GE
Healthcare).
Serum and protein adsorption experiments with live
P. aeruginosa
P. aeruginosa (2 ⫻ 1010 cells) was resuspended in 150 ␮l of PBS supplemented with 75 mM NaCl, 100 mM MgSO4, and 1% BSA and incubated
with 750 ␮l of 50% heat-inactivated normal human serum (HiNHS) or PBS
for 1 h at room temperature. For mapping of the Factor H or FHR-1 binding
domains, bacteria were incubated in culture supernatant containing Factor
H deletion (i.e., SCR 1–5, 1– 6, 1–7, 15–18, 15–19, and 15–20) or purified
proteins (i.e., SCR 19 –20, FHR-1, FHR-1/SCR 3–5). Following incubation, bacteria were washed four times with PBS, and bound proteins were
eluted using 3 M potassium thiocyanate. The last wash and the elution
fractions were separated by SDS-PAGE and transferred onto a nitrocellulose membrane. Proteins were identified by Western blotting using a polyclonal goat Factor H antiserum (18).
For flow cytometry bacteria were incubated in 50% HiNHS or PBS.
Cells were resuspended in 100 ␮l of PBS and incubated for 1 h with the
monoclonal Factor H Ab C18 (1/200) (33). After washing three times in
PBS, bacteria were incubated with 1/100 Alexa Fluor 488-labeled rabbit
anti-mouse antiserum (Molecular Probes) for 1 h at 4°C in the dark. The
cells were washed again in PBS, diluted to a concentration of 1 ⫻ 108
cells/ml, and analyzed using a FACSCalibur (BD Biosciences). Some
10,000 events were routinely counted.
Biotinylation and isolation of P. aeruginosa surface proteins
Surface biotinylation of intact P. aeruginosa was performed as described
(34). In short, bacteria (2 ⫻ 1011 cells) were washed in buffer A (PBS, 1
mM CaCl2, 0.5 mM MgCl2) and resuspended in buffer B (buffer A supplemented with 1.6 mM D-biotin). Bacteria were pelleted and surface proteins were labeled with biotin by incubation with 500 ␮l of 400 ␮M EZLink Sulfo-NHS-LC-Biotin (Pierce) for 30 min on ice. Cells were washed
extensively in buffer C (50 mM Tris (pH 7.4), 100 mM NaCl, 27 mM KCl,
1 mM CaCl2, 0.5 mM MgCl2) and resuspended in buffer C supplemented
with Protease Inhibitor Cocktail Complete (Roche). Bacteria were lysed by
sonication and biotinylated envelope proteins were purified by affinity
chromatography using ImmunoPure Immobilized Monomeric Avidin
(Pierce). Proteins were eluted with D-Biotin (2 mM in PBS) according to
the manufacturer’s recommendations. The elute fractions were separated
by SDS-PAGE and transferred to a membrane, and biotinylated proteins
were identified by HRP-conjugated avidin (Roche). Positive fractions were
pooled.
Isolation of Factor H ligands with magnetic beads
Factor H SCR 8 –20 (40 ␮g) was covalently coupled to magnetic beads
(100 ␮l of suspension) according to the manufacturer’s instructions (Invitrogen Life Technologies). These beads were incubated with purified
biotinylated P. aeruginosa surface proteins for 2 h at 37°C. After washing
three times (50 mM HEPES (pH 7.5), 1% Nonidet P-40, 1 mM dithioerythritol, 1 mM MgCl2, 1 mM CaCl2), proteins were eluted with 40 ␮l of
1 M NaCl for 10 min at 37°C, separated by SDS-PAGE, and visualized by
silver staining. Individual bands were excised from the gel and submitted
to mass spectrometry.
Protein identification by peptide mass fingerprinting
Silver-stained bands were destained using the ProteoSilver Plus silver stain
kit (Sigma-Aldrich) according to the manufacturer’s protocol. Subsequently, the proteins were reduced and alkylated using 10 mM DTT and
100 mM iodoacetamide in 25 mM NH4HCO3, washed with 25 mM
NH4HCO3, and dehydrated with acetonitrile. Digestion with trypsin,
MALDI-TOF-MS, and protein identification by peptide mass fingerprinting were performed as described (35).
Protein interaction assays
For binding studies human Factor H (Merck), recombinant Factor H deletion constructs, recombinant FHR-1 and plasminogen (Chromogenix) were
used. Bound proteins were identified with goat antiserum specific for Factor H (Merck), goat antiserum recognizing plasminogen (Acris Antibodies), Tuf antiserum generated in rabbit against recombinant Tuf, or mouse
anti-Tuf mAb (36).
For ligand affinity blotting recombinant Tuf was either separated by
SDS-PAGE and transferred to a membrane or directly blotted onto a membrane under native conditions. Membranes were blocked with Rotiblock
(Roth) supplemented with 2.5% BSA overnight and incubated with the
corresponding binding proteins. Bound proteins were identified with specific antisera and visualized by ECL.
For ELISA recombinant Tuf (0.25 ␮g in carbonate buffer) was coated to
the wells of Costar half-area plates (Corning) overnight at 4°C. Wells were
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
additional Factor H-related proteins (FHR) (specifically, FHR-1
to FHR-5), which are encoded by distinct genes (14). All members of the Factor H protein family represent structurally and
immunologically related proteins that are exclusively composed
of repetitive structural domains, termed short consensus repeat
(SCR) domains (16).
Pathogens use a common strategy for immune evasion and acquire the fluid-phase complement regulators Factor H, FHL-1, and
C4BP from host serum. When bound to the surface of the pathogen, each host regulator maintains complement regulatory activity,
which aids in survival of the pathogen (17). This type of complement evasion was shown for Gram-negative bacteria, e.g., Borrelia
burgdorferi (18, 19) or Neisseria gonorrhoeae (20), for Grampositive bacteria, e.g., Streptococcus pneumoniae (21, 22) or S.
pyogenes (23), for the eukaryotic yeast Candida albicans (24), for
the multicellular parasites Onchocerca volvulus (25) or Echinococcus granulosus (26), and for viruses (27, 28). In several cases
the corresponding surface proteins were identified and cloned (14).
Pathogens like P. aeruginosa require proteolytic activity for tissue invasion, which is provided by endogenous or host acquired
proteases. P. aeruginosa bind host plasminogen, and at the bacterial surface, the inactive precursor is converted to proteolytically
active plasmin, which causes fibrin degradation and aids in, for
example, tissue invasion. So far plasminogen receptors of P.
aeruginosa were postulated but not identified (29, 30).
In this study, we identify the elongation factor Tuf as a surface
protein of P. aeruginosa that binds the host plasma molecules Factor H and plasminogen. Factor H and plasminogen bound to Tuf,
both maintain their functional activity. P. aeruginosa inhibits host
complement activity by endogenous and acquired mechanisms: the
bacterium uses endogenous proteases for complement control and
also binds host regulators to its surface. For the analyzed P.
aeruginosa strains, the acquired C3b degrading activity mediated
by Factor H was stronger and more efficient than the endogenous
activity.
COMPLEMENT AND TISSUE INVASION OF P. aeruginosa
The Journal of Immunology
2981
blocked with PBS containing 1% BSA for 2 h at room temperature. After
washing with PBS, the indicated ligands (10 ␮g/ml in 75 ␮l) were added
and the mixture was incubated for 1 h at room temperature. Bound proteins
were identified with appropriate antisera. After addition of orthophenylenediamine (DakoCytomation) the absorption at 490 nm was measured.
Competition of Factor H and plasminogen to immobilized Tuf was analyzed by ELISA. A total of 0.2 ␮g of plasminogen was mixed with different quantities of Factor H resulting in various molar ratios and added to
each well containing immobilized Tuf. Both serum proteins were detected
individually using specific antisera.
For whole cell ELISA, intact bacteria (108/well) were resuspended in
carbonate buffer and immobilized to Maxisorp plates (Nunc). ELISA was
performed as described. HiNHS was used at a 20-fold dilution. The amino
acid analogs ␧-aminocaproic acid and imidazol were added to a final concentration of 1 mM. Incubations were performed in a volume of 150 ␮l.
For surface plasmon resonance assays a Biacore 3000 instrument was
used. Briefly, Factor H SCR 8 –20, or recombinant Tuf, was immobilized
via standard amine coupling to the flow cells of a CM5 sensor chip as
described (37). The surface of the flow cells was activated and the analyte
diluted in coupling buffer (10 mM acetate buffer (pH 5.0)) was injected
until an appropriate level of coupling was reached (⬃3400 resonance
units). Recombinant Tuf or plasminogen was used as analytes at the indicated concentrations at a flow rate of 5 ␮l/min.
Bacteria of P. aeruginosa strain AH377 (2 ⫻ 107) were washed three times
in PBSB (PBS supplemented with 0.2% BSA and 100 ␮l) and incubated
for 1 h at room temperature with 1/100 diluted primary Ab (polyclonal goat
antiserum raised against Factor H). Following washing in PBSB, bacteria
were resuspended in 20 ␮l of buffer, applied to microscopic slides and
dried overnight. After fixation with 100 ␮l of acetone for 15 min, bacteria
were incubated with an Alexa Fluor 647-labeled anti-mouse or anti-goat
immune serum (1/500 in PBSB; Molecular Probes) for 1 h at room temperature. After extensive washing in PBSB, bacteria were embedded in
ProTaqs Mount Fluor (Biocyc) and fluorescence was analyzed using an
Olympus BX51 microscope. For detection of plasminogen binding, intact
bacteria were incubated with 0.5 ␮g of plasminogen in 100 ␮l of PBS for
1.5 h at room temperature and then treated with goat plasminogen antiserum (Acris Antibodies).
FIGURE 1. Factor H and FHR-1 bind to intact P. aeruginosa. A, P.
aeruginosa strains PAO1, ATCC 27853, NCTC 10662, and SG137 were
incubated in HiNHS. Bacteria were washed, and bound proteins were
eluted, separated by SDS-PAGE, and analyzed by Western Blotting. Factor
H was detected in the eluate as a 150-kDa protein, and FHR-1 was identified as the doublet of 37 and 43 kDa. The mobility of the marker proteins
is indicated on the right side. e, elution fraction; w, final wash fraction. B,
Binding of Factor H as assayed by flow cytometry. P. aeruginosa strains
PAO1 and ATCC 27853 were treated with HiNHS, and after extensive
washing bacteria were incubated with Factor H-specific antiserum. Bacteria incubated in PBS served as controls. Representative experiments of five
are shown.
Fibrinogen degradation of P. aeruginosa
P. aeruginosa strain PAO1 (108 cells) were thoroughly washed with PBS
and incubated in reaction buffer (64 mM Tris, 350 mM NaCl, 0.15% (w/v)
Triton X-100 (pH 7.5)) supplemented with plasminogen (2 ␮g) and urokinase plasminogen activator (uPA, 20 ng) for 1 h at room temperature. After
extensive washing fibrinogen (0.5 ␮g plasminogen-depleted; Calbiochem)
was added and cells were incubated at 37°C. At the indicated times (4 and
16 h), aliquots were removed and separated by SDS-PAGE. Fibrinogen
degradation was analyzed after transfer to a membrane by Western blotting
using a polyclonal fibrinogen Ab (Calbiochem) and an HRP-conjugated
anti-rabbit antiserum.
C3b cleavage assay
The C3b cleavage capacity of P. aeruginosa was assayed after incubation
of the bacteria in 50% HiNHS or PBS for 1 h. After washing with PBS cells
were resuspended in 40 ␮l PBS, supplemented with 10 ␮g/ml C3b and 20
␮g/ml Factor I (Merck), and incubated at room temperature either for 2 h
or overnight. Bacteria were pelleted, lysed, and separated by SDS-PAGE.
After transfer onto a nitrocellulose membrane, C3b degradation products
were identified by Western blotting using an anti-C3c antiserum raised in
rabbits (Merck).
To assay cofactor activity of Tuf-bound Factor H, recombinant Tuf was
coated to microtiter plates and Factor H (0.2 ␮g/well) was added. After 1 h
incubation, C3b and Factor I were applied and the mixture was incubated
for 15 min at 37°C. The fluid phase was removed and C3b degradation
products were analyzed as described.
Plasminogen activation and plasmin activity
Recombinant Tuf was immobilized onto Costar half-area plates (Corning).
After blocking with PBS containing 2% BSA, plasminogen (0.6 ␮g/well)
was added for 2 h at room temperature. Unbound plasminogen was removed by washing with PBS. The activator uPA (4 ng per well; Millipore)
and the chromogenic substrate S-2251 (D-valyl-leucyl-lysine-p-nitroanilide
dihydrochloride, 150 ␮g; Sigma-Aldrich) dissolved in reaction buffer (64
mM Tris, 350 mM NaCl, 0.15% Triton X-100 (pH 7.5)) were added. Plasmin activity was measured as described (38). The activity of the generated
plasmin was measured at 37°C over an incubation period of 15 h by recording the absorbance at 405 nm.
Serum sensitivity assay
P. aeruginosa strain SG137 (107 cells) were incubated at room temperature
in 50% complement active normal human plasma or 50% Factor H-depleted human plasma (39) in reaction buffer (20 mM HEPES, 144 mM
NaCl, 10 mM EGTA, 7 mM MgCl2, (pH 7.4), final volume 160 ␮l). After
1 h bacteria were harvested and thoroughly washed, and serial dilutions
were plated on Nutrient Broth agar. After 24 h incubation at 37°C, colonies
were counted and CFUs were calculated. In additional experiments, Factor
H-depleted human plasma was reconstited with purified Factor H
(Calbiochem).
Statistical analysis
Where appropriate, data points were subjected to the unpaired Student’s
t test.
Results
Binding of Factor H and FHR-1 to P. aeruginosa
Binding of Factor H to intact P. aeruginosa was assayed by serum
absorption experiments. Bacteria were incubated in HiNHS and
after extensive washing bound proteins were eluted, separated by
SDS-PAGE and analyzed by Western blotting. A band of 150 kDa
representing Factor H and a doublet of 37 and 43 kDa representing
the two forms of FHR-1 (40) were identified in the elution fractions of all tested strains: i.e., serum-resistant PAO1, ATCC
27853, NCTC 10662, and serum-sensitive SG137 (Fig. 1A). For
strains PAO1 and ATCC 27853, Factor H binding was further
assayed by flow cytometry. Following incubation in human serum
Factor H binding was visualized by an increase in fluorescence
(Fig. 1B).
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
Immunofluorescence studies
2982
COMPLEMENT AND TISSUE INVASION OF P. aeruginosa
FIGURE 2. Factor H and FHR-1 bind to clinical isolates of P. aeruginosa. Clinical isolates of P. aeruginosa
derived from patients with pyelonephritis, leukopenia, T
cell lymphoma, urosepsis, or myeloma (Is1-Is8) were incubated with human serum. Bound proteins were eluted,
separated by SDS-PAGE, and analyzed by Western Blotting. Factor H was detected in the elution fraction as a
150-kDa protein, and FHR-1 was identified as the doublet
of 37 and 43 kDa. The mobility of the marker proteins is
indicated on the right side. A representative experiment of
five is shown. e, elution fraction; w, final wash fraction.
beads and incubated with purified biotinylated surface proteins of
strain PAO1. Bacterial proteins that bind to the Factor H/SCRs
8 –20 matrix were eluted, separated by SDS-PAGE and visualized
by silver staining (data not shown). Three bands were excised from
the gel and analyzed by peptide mass fingerprinting, and the band
of 43 kDa was positively identified. Of 19 ion signals labeled in the
assigned mass spectrum, 10 signals matched to the amino acid
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
In addition, binding of host immune regulators to eight clinical
P. aeruginosa isolates was tested, which differ in respect to pigment production, morphology, and mucosity. All isolates (Is1 to
Is8) bound Factor H and FHR-1 (Fig. 2). Additional bands were
detected with weaker intensity for Is1, Is3, Is5, and Is6. These
nonspecific bands were not detectable when bacteria were incubated in PBS instead of HiNHS. Thus, binding of Factor H and
FHR-1 seems to be a general feature of P. aeruginosa.
Mapping of Factor H and FHR-1 binding domains
For the two human immune regulators Factor H and FHR-1, the
binding regions for interaction with live P. aeruginosa were localized using recombinant deletion mutants. N-terminal (SCR domains 1– 6 and 1–7) and C-terminal deletion constructs of Factor
H (SCR domains 15–19, 15–20, and 19 –20) bound to intact bacteria (strain ATCC 27853), whereas constructs SCRs 1–5 and
15–18 did not bind (Fig. 3, A and B). In addition, complete FHR-1
and a deletion construct representing the three C-terminal SCR
domains (FHR-1/SCRs 3–5) did bind to P. aeruginosa (Fig. 3C).
The same binding profile was identified for Factor H and FHR-1
binding to P. aeruginosa strain PAO1 (data not shown). The additional bands observed for several recombinant Factor H fragments most likely represent dimeric or even multimeric forms and
are also detectable with purified recombinant fragments. Thus,
Factor H binds with two domains to the surface of P. aeruginosa,
i.e., SCRs 6 –7 and 19 –20 and FHR-1 binds with one domain,
which is contained in SCRs 3–5.
Identification of a Factor H binding protein in P. aeruginosa
To isolate Factor H binding proteins from P. aeruginosa, a matrix
representing Factor H/SCRs 8 –20 was immobilized to magnetic
FIGURE 3. Mapping of the binding domains within Factor H and
FHR-1. P. aeruginosa strain ATCC 27853 was incubated with recombinant
deletion constructs of Factor H and FHR-1. N-terminal (A) and C-terminal
fragments of Factor H (B), or recombinant FHR-1 and the C-terminal fragment SCRs 3–5 (C) were used. Bound proteins were eluted, separated by
SDS-PAGE, and analyzed by Western Blotting. Arrows indicate the position of the recombinant deletion constructs, which differ in mobility as they
include a variable number of SCR domains. The mobility of the marker
proteins is indicated on the right side. A representative experiment of five
is shown. e, elution fraction; w, final wash fraction.
FIGURE 4. Elongation Factor Tuf of P. aeruginosa is a surface protein.
A, Tuf of P. aeruginosa strain PAO1 was recombinantly expressed in E.
coli as a His-tagged protein and purified by affinity chromatography. The
E. coli lysate, flow through (FT), wash (w), and elution (e) fraction were
separated by SDS-PAGE and analyzed by silver staining (lanes 1– 4). The
elution fraction was analyzed by Western blotting using a mAb detecting
the N-terminal histidine tag (lane 5). The band of ⬃57 kDa is visualized by
silver staining but did not react with the histidine antiserum. B, Tuf is
detected at the surface of immobilized P. aeruginosa (strain PAO1) using
whole cell ELISA. Tuf was detected using a polyclonal antiserum, which
was raised against recombinant Tuf. Data show the mean of four experiments with SD indicated with error bars. ⴱ, p ⬍ 0.001. C, Surface expression of Tuf as identified by immunofluorescence microscopy. P. aeruginosa (strain PAO1) was incubated with a monoclonal anti-Tuf Ab and
stained with a Alexa Fluor 647-labeled antiserum (left). Treatment in the
absence of the mAb served as control (right). A representative experiment
of three is shown. D, Tuf is identified in the surface protein fraction of P.
aeruginosa. Purified biotinylated surface proteins of strains PAO1 (lanes 1
and 3) and SG137 (lanes 2 and 4) were separated by SDS-PAGE and
analyzed by Western blotting using a monoclonal anti-Tuf Ab or a polyclonal anti-GroEL antiserum. The mobility of the marker proteins is indicated on the right. A representative experiment of three is shown.
The Journal of Immunology
2983
sequence of the elongation factor Tuf (EF-Tu) from P. aeruginosa,
resulting in sequence coverage of 24%. Tuf is 397 aa long and has
a predicted molecular mass of 43.4 kDa (39).
Tuf is a surface protein of P. aeruginosa
The gene coding for Tuf was amplified using DNA of P. aeruginosa strain PAO1, cloned into an appropriate expression vector,
and expressed as a His-tagged protein in E. coli. Recombinant Tuf
was purified almost to homogeneity as indicated by silver staining
of SDS-PAGE-separated purified protein (Fig. 4A, lane 4). Recombinant Tuf reacted with a polyhistidine antiserum (Fig. 4A,
lane 5). In addition, a specific polyclonal antiserum was raised
against recombinant Tuf. With this antiserum, Tuf was detected on
the surface of P. aeruginosa, using a whole cell ELISA (Fig. 4B).
Surface-exposed Tuf was also identified by immunofluorescence
microscopy with P. aeruginosa (Fig. 4C). Surface expression of
Tuf was further confirmed analyzing surface protein fractions of P.
aeruginosa strains PAO1 and SG137. Surface proteins were labeled with biotin, isolated by avidin affinity chromatography,
and separated by SDS-PAGE. After Western blotting Tuf was
identified among the surface proteins. GroEL (heat shock protein
60), which was used as a marker for cytoplasmic proteins, was not
detected in these fractions (Fig. 4D, lanes 3 and 4).
Recombinant Tuf binds human complement regulators
Ligand affinity blotting with recombinant Tuf was used to show
binding to Factor H, recombinant FHL-1 (Fig. 5A), and FHR-1.
Binding was further assayed by ELISA. Tuf was immobilized,
and binding of Factor H, FHL-1, and FHR-1 was tested. Factor
H and FHL-1 binding was prominent. Binding of FHR-1 was
further assayed using ligand affinity blotting. When Tuf was subjected
to SDS-PAGE and blotted onto a membrane, binding of FHR-1 was
observed (Fig. 5B). FHR-1 binding was detected, but the signal
showed variations (Fig. 5C). Factor H interaction with Tuf was
further confirmed by surface plasmon resonance. A C-terminal
fragment of Factor H, i.e., SCRs 8 –20, was immobilized to the
chip surface and Tuf was applied as analyte. Tuf binding was
concluded based on the association curve and the pronounced dissociation profile following removal of the ligand. Binding was
dose-dependent as shown for two Tuf concentrations (Fig. 5D). To
localize the binding regions within Factor H, binding of recombinant Factor H deletion mutants to Tuf was analyzed by ELISA.
The N-terminal constructs SCRs 1– 6 and SCRs 1–7 bound to immobilized Tuf, and SCRs 1–5 showed weak binding. Similarly, the
C-terminal constructs SCRs 15–18 and SCRs 15–20 did bind, but
not the deletion constructs that represent the middle region, i.e.,
SCRs 8 –11 and SCRs 11–15 (Fig. 5E). These results show that the
human complement regulator Factor H binds to recombinant Tuf
of P. aeruginosa. Factor H has two binding regions for Tuf, one is
shared with FHL-1 and the second region is located in the C terminus, i.e., SCRs 18 –20.
Tuf is a plasminogen-binding protein of P. aeruginosa
Interaction of Tuf with other human serum proteins was tested, and
plasminogen was identified as an additional Tuf binding protein.
Plasminogen bound strongly to Tuf as demonstrated by ligand affinity blotting and by ELISA (Figs. 6, A and B). As Tuf interacts
with the two human plasma proteins Factor H and plasminogen,
we analyzed whether the two proteins bind simultaneously and
compete for binding. Immobilized Tuf was incubated with a mixture of Factor H and plasminogen in different molar ratios. Bound
human proteins were detected individually. Factor H affected the
interaction between plasminogen and Tuf, as increasing the concentration of Factor H resulted in decreased plasminogen binding.
When both human proteins were applied at a molar ratio that corresponds to the plasma situation, concurrent binding of both
plasma proteins was observed (Fig. 6C). Thus plasminogen and
Factor H bind simultaneously to bacterial Tuf and apparently use
similar or even overlapping binding sites. Binding of plasminogen
to Tuf was further characterized by surface plasmon resonance.
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
FIGURE 5. Factor H, FHL-1 and FHR-1 bind to recombinant Tuf. A, Recombinant Tuf or BSA was applied to a membrane by slot blotting and after
incubation with Factor H or FHL-1 bound proteins were detected with specific antisera. BSA was used as a negative control. B, Tuf or BSA were separated
by SDS-PAGE, blotted to a membrane, and incubated with FHR-1. Bound FHR-1 was detected with specific antisera. BSA was used as a negative control.
C, Binding of Factor H, FHL-1, and FHR-1 to immobilized Tuf was analyzed by ELISA. Bound proteins were detected using specific antisera. BSA was
used as a negative control. Data show the mean of three experiments, and SD is indicated by error bars. ⴱ, p ⬍ 0.03; ⴱⴱ, p ⬍ 0.001. D, Binding of Factor
H to Tuf was analyzed by surface plasmon resonance. The C-terminal Factor H construct SCR 8 –20 was immobilized on the chip surface and Tuf was
applied as an analyte within the fluid phase. Tuf binding was dose-dependent and resulted in elevated resonance units (RU). E, Localization of the Factor
H binding domains by ELISA. Recombinant Tuf was immobilized, and binding of the indicated Factor H deletion constructs was assayed using a polyclonal
anti-Factor H antiserum, which reacted similarly to all constructs. A representative experiment is shown. Data show the mean value and SD is indicated
by error bars shown.
2984
COMPLEMENT AND TISSUE INVASION OF P. aeruginosa
Tuf was immobilized to the surface of a sensor chip and binding of
plasminogen was followed. Plasminogen binding was dose-dependent over the range from 12.5 to 400 nM (Fig. 6D).
Binding of human plasminogen to intact P. aeruginosa
Binding of plasminogen to live P. aeruginosa of strain PAO1 was
assayed. Plasminogen binding was first demonstrated using immu-
nofluorescence microscopy. Bacteria were incubated with plasminogen and binding was visualized with the specific antiserum (Fig.
7A). Binding of purified plasminogen and serum-derived plasminogen to P. aeruginosa was also observed using a whole-cell
ELISA. Apparently, lysine residues are relevant for the interaction
as the lysine analog ␧-aminocaproic acid inhibited plasminogen
binding, but the histidine analog imidazol had no effect (Fig. 7B).
FIGURE 7. Plasminogen binds to live P. aeruginosa. A, Binding of plasminogen to intact P. aeruginosa (strain AH377) was analyzed by immunofluorescence microscopy. Bound plasminogen was detected using specific antisera (left). Treatment in the absence of plasminogen served as control (right).
A representative experiment of three is shown. B, Whole cell ELISA demonstrating plasminogen binding to live bacteria (strain PAO1) that were
immobilized to a microtiter plate. Binding of purified plasminogen and of plasminogen derived from HiNHS was detected with specific antiserum. Amino
acid analogs ␧-aminocaproic acid (␧ACA) and imidazol (imi) were added to a final concentration of 1 mM. Binding of the antiserum to the bacteria in the
absence of a ligand was used as control. Data show the mean with SD indicated with error bars. ⴱ, p ⬍ 0.01 significance relative to the control. Experiments
were repeated at least three times. C, Plasminogen attached to P. aeruginosa cleaves fibrinogen. P. aeruginosa strain PAO1 was incubated with plasminogen
in the presence of the activator uPA. Following incubation and extensive washing, fibrinogen was added. After 4 and 16 h of incubation of the bacteria,
samples were withdrawn, separated by SDS-PAGE, and transferred to a membrane. Fibrinogen was identified using a polyclonal fibrinogen antiserum in
combination with HRP-conjugated rabbit antiserum.
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
FIGURE 6. Plasminogen binds to recombinant Tuf.
A, Recombinant Tuf was blotted onto a membrane and
incubated with purified plasminogen. Bound protein
was detected with specific antisera. BSA was used as a
negative control. A representative experiment of three is
shown. B, Binding of plasminogen to immobilized Tuf
was analyzed by ELISA. Bound plasminogen was detected using a specific antiserum. BSA was used as a
negative control. Data show the mean and error bars
indicate SD. ⴱ, p ⬍ 0.01. C, Plasminogen and Factor H
share binding sites for Tuf. Binding of plasminogen to
immobilized Tuf was assayed in the presence of increasing molar concentrations of Factor H. Both serum
proteins were detected by specific antisera. In human
plasma, plasminogen has a concentration of ⬃100 nM
and Factor H of 140 nM. Thus the overall molar ratio is
1:1.4 (Plasma data), which reflects the in vivo situation.
Data show the mean of four independent experiments,
and the SD is indicated by error bars. D, Surface plasmon resonance analyses of plasminogen binding to immobilized Tuf. Recombinant Tuf was immobilized to
the surface of a sensor chip, and plasminogen was applied at the indicated concentrations. Binding was dosedependent over a range from 0 to 400 nM.
The Journal of Immunology
2985
To characterize the function of bound plasminogen, plasminogen
was attached to the surface of intact bacteria and activated with
uPA. Following extensive washing, the cells with the attached protease were incubated with fibrinogen. In the presence of plasminogen, fibrinogen was completely degraded after 16 h (Fig. 7C).
This response shows that surface bound plasminogen is proteolytically active and aids in fibrinolysis and degradation of the extracellular matrix components.
Factor H attached to the surface of P. aeruginosa displays
cofactor activity
To determine the regulatory role of surface-attached Factor H, degradation of C3b was analyzed directly at the surface of P. aeruginosa after incubation in human serum. C3b processing was visualized following separation by SDS-PAGE and Western blotting.
Incubation of P. aeruginosa strain PAO1 in human serum resulted
in acquisition of complement regulatory activity as demonstrated
by the disappearance of the 110 kDa ␣⬘ chain of C3b and the
appearance of the cleavage products ␣⬘ 68, ␣⬘ 46, and ␣⬘ 43. As P.
aeruginosa uses endogenous proteases to inactivate complement
(8), the C3b degradation was compared following incubation of the
bacteria in PBS in the absence of human serum. Endogenous C3b
cleavage was revealed for strain PAO1 by the appearance of the ␣⬘
43, ␣⬘ 41, and ␣]prime] 35 fragments of C3b, which were detected
after 18 h of incubation (Fig. 8A, lane 3). A comparison of the
intensities of the degradation products showed that the acquired
C3b cleavage activity from human serum is stronger than the endogenous activity (Fig. 8A, compare lane 3 with lane 4). Similar
results were observed for the P. aeruginosa strains ATCC 27853
and NCTC 10662 (Fig. 8, B and C). The three analyzed strains
showed endogenous C3b cleaving activity as well as strain-specific
C3b degradation products. For all analyzed strains the acquired
activity was more pronounced.
Functional activity of Tuf-bound human serum proteins
To assay whether Tuf-bound host effector proteins are functionally
active, plasminogen was bound to immobilized Tuf and treated
with the activator uPA, and plasmin activity was assayed. Tufbound plasminogen was converted to proteolytically active plasmin as evidenced by cleavage of the chromogenic substrate
S-2251. The proteolytic activity was dose-dependent and correlated with the amount of immobilized Tuf (Fig. 8D). Thus, plasminogen attached to Tuf is accessible for the activator uPA, and
the activated Tuf-bound plasmin has proteolytic activity.
To assay whether Factor H attached to Tuf maintains complementregulating activity, Factor I-mediated cleavage of C3b was analyzed.
Following binding of Factor H to immobilized Tuf, C3b and Factor I
were added. After incubation the mixture was separated by SDSPAGE and C3b cleavage products were identified by Western blotting. Tuf-bound Factor H displayed cofactor activity as demonstrated
by the appearance of the ␣⬘ 46 and ␣⬘ 43 bands of C3b (Fig. 8B, lanes
1–3). The cofactor activity of Tuf-bound Factor H was comparable to
that of Factor H immobilized (Fig. 8E, compare lane 3 with lane 5).
Attachment of Factor H affects survival in human plasma
To assay the protective role of surface attached Factor H, survival of bacterial was compared following incubation in Factor
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
FIGURE 8. Tuf-bound Factor H and plasminogen are functionally active. A–C, C3b cleavage due to acquired and endogenous activity. P. aeruginosa
strains PAO1 (A), ATCC 27853 (B), or NCTC 10662 (C) were incubated in PBS or HiNHS. Following addition of C3b and Factor I, the reaction was
incubated at 37°C for 2 or 18 h. Aliquots were separated by SDS-PAGE and the appearance of C3b degradation products was analyzed by Western blotting.
The endogenous C3b cleavage activity was assayed in the absence of serum (PBS). The position of the ␣⬘ chain and the ␤-chain of C3b is indicated.
Degradation of C3b is evidenced by appearance of cleavage products and by disappearance of the ␣⬘ chain. Degradation products from Factor H-mediated
cofactor activity, i.e., ␣⬘ 46 and ␣⬘ 43, are represented by filled arrows. C3b fragments generated by endogenous strain-specific proteases are indicated by
arrowheads and their apparent product size is underlined and shown with boldface. A representative experiment of three is shown. Experiments were
repeated at least four times. D, Tuf was immobilized to a microtiter plate and plasminogen was added. After extensive washing, the tissue-type
plasminogen activator and the chromogenic substrate S-2251 were added and the absorbance of the converted substrate was measured. Data show
the mean and error bars represent SD. E, Tuf-bound Factor H displays cofactor activity. Factor H bound to immobilized Tuf was incubated with C3b
and Factor I. The supernatant was removed and separated by SDS-PAGE, and the appearance of C3b cleavage products was analyzed by Western
blotting. C3b is represented by the ␣⬘ chain and the ␤-chain, as indicated. C3b degradation is visualized by the appearance of the 46- and 43-kDa
bands. In the absence of Factor H, no degradation products were detected (lane 4). As a positive control, immobilized Factor H was directly incubated
with C3b and Factor I (lane 5).
2986
H-deficient and -sufficient complement-active human plasma. Incubation in Factor H-depleted plasma results in poor survival of P.
aeruginosa strain SG137. However in Factor H-containing plasma
bacterial survival was observed (Fig. 9A). The damaging effect of
plasma was dependent on Factor H, as addition of this regulator to
depleted plasma resulted in a dose-dependent increase in survival
(Fig. 9B). Thus demonstrating that acquisition of Factor H to the
bacterial surface has a protective effect.
Discussion
The Gram-negative bacterium P. aeruginosa exploits host plasma
proteins like Factor H and plasminogen for immune evasion and
tissue destruction. Binding of the host proteins Factor H and
FHR-1 was observed for four laboratory strains (i.e., P. aeruginosa
PAO1, ATCC 27853, NCTC 10662, and SG137) and eight clinical
isolates. Factor H bound to P. aeruginosa shows cofactor activity
for the cleavage of C3b and thus mediates complement evasion of
the pathogen. P. aeruginosa is, to our knowledge, the first pathogen for which a combination of acquired and endogenous complement regulatory activity has been shown. For all strains analyzed
in this study, the acquired regulatory activity is stronger than the
endogenous activity. In addition, the acquired and the endogenous
proteases generate different C3b cleavage products. Factor H uses
two domains for attachment to the bacterial surface that are located
in SCR domains 6 –7 and 19 –20, and similarly FHR-1 binds via
the C terminus, i.e., SCR 3–5. The translational elongation factor
Tuf of P. aeruginosa was identified as a bacterial Factor H binding
protein. A newly generated antiserum identified Tuf at the surface
of P. aeruginosa. Factor H binds to recombinant Tuf, which in its
bound form maintains cofactor activity for C3b degradation. In
addition, Tuf binds plasminogen and Tuf-bound plasminogen is
converted by uPA to functionally active plasmin. Thus, acquisition
of plasminogen may enhance extracellular matrix interaction and
degradation and may support tissue invasion of the pathogen. In
summary, the pathogen P. aeruginosa acquires human plasma proteins Factor H and plasminogen via the surface protein Tuf and
uses the attached host proteins for immune evasion and tissue
interaction.
All analyzed P. aeruginosa strains as well as different clinical
isolates bind the soluble complement regulator Factor H and the
related FHR-1 to their surfaces, suggesting that binding of complement regulators is a general feature of pathogenic P. aeruginosa. The three C-terminal SCRs of Factor H and FHR-1, which
show near sequence identity, include heparin and glycosaminoglycan binding sites, thus explaining the comparable binding (41, 42).
The absence of FHL-1 binding to intact bacteria can be explained
either by the different serum concentrations of Factor H and FHL-1
or by the higher binding affinity of the C-terminal SCR 19 –20 vs
SCR 6 –7.
The same two domains of Factor H, which bind to P. aeruginosa, do also interact with virulence factors of other pathogens,
like the M protein of S. pyogenes (43), BbCRASP-1 and
BbCRASP-2 of B. burgdorferi (44), and CaCRASP-1 of C. albicans (P. Poltermann, A. Kunnert, M. vonder Heide, R. Eck, A.
Hartmann, and P. F. Zipfel, manuscript in preparation).
Factor H bound to P. aeruginosa is functionally active and
together with the serine protease Factor I degrades C3b. P. aeruginosa uses a combination of endogenous and acquired complementdegrading activity. The endogenous C3b-degrading activity generates unique strain-specific cleavage products that differ from
those generated by the host protease Factor I (Figs. 8, A and B).
The endogenous regulatory activity, which is mediated by elastase
and alkaline protease, was initially described for several clinical P.
aeruginosa isolates (8, 9, 45). The endogenous C3b cleavage activity varied within the analyzed strains. However, in all strains the
acquired C3b-degrading activity was more potent than the endogenous activity.
Complement, in particular the alternative pathway, is essential
for clearance of P. aeruginosa in vertebrate hosts. Using a P.
aeruginosa pulmonary infection model, C3 knockout mice showed
high mortality and increased bacterial loads in lungs and blood. A
central role of the alternative pathway is concluded by the high
mortality of mice deficient for the alternative complement component Factor B, but survival of mice deficient in the classical pathway protein C4 (11). Thus alternative pathway-induced C3 activation and C3b generation is crucial for immune sensing and
host-mediated elimination of P. aeruginosa. As complement is
part of innate immunity and is particularly active during the initial
phase of the infection, binding of the host complement regulator
Factor H represents a potent strategy of P. aeruginosa for innate
immune evasion.
The elongation factor Tuf was identified as a Factor H binding
surface protein of P. aeruginosa using a combination of surface
protein extraction, affinity chromatography, and mass spectrometry. A second independent approach, which was based on crosslinking of P. aeruginosa surface proteins to Factor H, immunoprecipitation and mass spectrometry also identified Tuf as a Factor
H binding protein (data not shown). The translational elongation
factor Tuf (EF-Tu) was initially identified as a GTP binding protein, which is associated with the 50 S ribosomal subunit (39). Tuf,
which lacks classical signal and transport sequences, has a cytoplasmic location, but was identified at the surface of P. aeruginosa
(Fig. 4, B–D). Surface localization of Tuf is also reported for Mycobacterium leprae and Mycoplasma pneumoniae (46, 47). Recombinant Tuf of P. aeruginosa binds the host plasma proteins
Factor H, FHL-1, FHR-1, and plasminogen (Figs. 5 and 6). In
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
FIGURE 9. Survival of P. aeruginosa in Factor H-depleted human
plasma. A, P. aeruginosa strain SG137 was incubated for 1 h either in
Factor H-depleted complement active human plasma (DHP) or in complement active normal human plasma (NHP). Afterward, bacteria were plated
onto agar plates at various dilutions, and CFUs were determined. This
experiment was repeated three times, and the mean data and error bars for
SD are indicated. ⴱ, p ⬍ 0.005. B, P. aeruginosa strain SG137 was incubated for 1 h in Factor H-depleted human plasma, which was supplemented
with the indicated concentrations of purified Factor H. The plasma concentration of Factor H is about 500 ␮g/ml. CFU obtained under physiological Factor H concentrations was set at 100%. This experiment was
repeated four times and data indicate mean values and SD as error bars.
COMPLEMENT AND TISSUE INVASION OF P. aeruginosa
The Journal of Immunology
Acknowledgments
We thank Tim Tolker-Nielsen (The Technical University of Denmark) for
providing the P. aeruginosa strain AH377, Jorge Koehl (University of
Cincinnati, OH) for providing the monoclonal Tuf Ab, and Stefan Heinen
(Jena, Germany) for Factor H-depleted human plasma. We also thank Steffi
Hälbich for surface plasmon resonance analysis.
Disclosures
The authors have no financial conflict of interest.
References
1. Lyczak, J. B., C. L. Cannon, and G. B. Pier. 2000. Establishment of Pseudomonas
aeruginosa infection: lessons from a versatile opportunist. Microbes Infect. 2:
1051–1060.
2. Talbot, G. H., J. Bradley, J. E. Edwards, Jr., D. Gilbert, M. Scheld, and
J. G. Bartlett. 2006. Bad bugs need drugs: an update on the development pipeline
from the Antimicrobial Availability Task Force of the Infectious Diseases Society
of America. Clin. Infect. Dis. 42: 657– 668.
3. Endimiani, A., F. Luzzaro, B. Pini, G. Amicosante, G. M. Rossolini, and
A. Q. Toniolo. 2006. Pseudomonas aeruginosa bloodstream infections: risk factors and treatment outcome related to expression of the PER-1 extended-spectrum
beta-lactamase. BMC Infect. Dis. 6: 52.
4. Buret, A., and A. W. Cripps. 1993. The immunoevasive activities of Pseudomonas aeruginosa: relevance for cystic fibrosis. Am. Rev. Respir. Dis. 148:
793– 805.
5. Schiller, N. L., and K. A. Joiner. 1986. Interaction of complement with serumsensitive and serum-resistant strains of Pseudomonas aeruginosa. Infect. Immun.
54: 689 – 694.
6. Gross, G. N., S. R. Rehm, and A. K. Pierce. 1978. The effect of complement
depletion on lung clearance of bacteria. J. Clin. Invest. 62: 373–378.
7. Younger, J. G., S. Shankar-Sinha, M. Mickiewicz, A. S. Brinkman, G. A.
Valencia, J. V. Sarma, E. M. Younkin, T. J. Standiford, F. S. Zetoune, and
P. A. Ward. 2003. Murine complement interactions with Pseudomonas aeruginosa and their consequences during pneumonia. Am. J. Respir. Cell Mol. Biol. 29:
432– 438.
8. Hong, Y. Q., and B. Ghebrehiwet. 1992. Effect of Pseudomonas aeruginosa
elastase and alkaline protease on serum complement and isolated components
C1q and C3. Clin. Immunol. Immunopathol. 62: 133–138.
9. Schmidtchen, A., E. Holst, H. Tapper, and L. Björck. 2003. Elastase-producing
Pseudomonas aeruginosa degrade plasma proteins and extracellular products of
human skin and fibroblasts, and inhibit fibroblast growth. Microb. Pathog. 34:
47–55.
10. Engels, W., J. Endert, and C. P. Van Boven. 1985. A quantitative method for
assessing the third complement factor (C3) attached to the surface of opsonized
Pseudomonas aeruginosa: interrelationship between C3 fixation, phagocytosis
and complement consumption. J. Immunol. Methods 81: 43–55.
11. Mueller-Ortiz, S. L., S. M. Drouin, and R. A. Wetsel. 2004. The alternative
activation pathway and complement component C3 are critical for a protective
immune response against Pseudomonas aeruginosa in a murine model of pneumonia. Infect. Immun. 72: 2899 –2906.
12. Marrie, T. J., G. K. Harding, A. R. Ronald, J. Dikkema, J. Lam, S. Hoban, and
J. W. Costerton. 1979. Influence of mucoidy on antibody coating of Pseudomonas
aeruginosa. J. Infect. Dis. 139: 357–361.
13. Kharazmi, A. 1991. Mechanisms involved in the evasion of the host defence by
Pseudomonas aeruginosa. Immunol. Lett. 30: 201–205.
14. Zipfel, P. F., C. Skerka, J. Hellwage, S. T. Jokiranta, S. Meri, V. Brade,
P. Kraiczy, M. Noris, and G. Remuzzi. 2002. Factor H family proteins: on complement, microbes and human diseases. Biochem. Soc. Trans. 30: 971–978.
15. Kühn, S., C. Skerka, and P. F. Zipfel. 1995. Mapping of the complement regulatory domains in the human factor H-like protein 1 and in factor H1. J. Immunol.
155: 5663–5670.
16. Krushkal, J., O. Bat, and I. Gigli. 2000. Evolutionary relationships among proteins encoded by the regulator of complement activation gene cluster. Mol. Biol.
Evol. 17: 1718 –1730.
17. Würzner, R. 1999. Evasion of pathogens by avoiding recognition or eradication
by complement, in part via molecular mimicry. Mol. Immunol. 36: 249 –260.
18. Kraiczy, P., C. Skerka, M. Kirschfink, V. Brade, and P. F. Zipfel. 2001. Immune
evasion of Borrelia burgdorferi by acquisition of human complement regulators
FHL-1/reconectin and Factor H. Eur. J. Immunol. 31: 1674 –1684.
19. Kurtenbach, K., S. De Michelis, S. Etti, S. M. Schäfer, H. S. Sewell, V. Brade,
and P. Kraiczy. 2002. Host association of Borrelia burgdorferi sensu lato: the key
role of host complement. Trends Microbiol. 10: 74 –79.
20. Ram, S., D. P. McQuillen, S. Gulati, C. Elkins, M. K. Pangburn, and P. A. Rice.
1998. Binding of complement factor H to loop 5 of porin protein 1A: a molecular
mechanism of serum resistance of nonsialylated Neisseria gonorrhoeae. J. Exp.
Med. 188: 671– 680.
21. Dave, S., A. Brooks-Walter, M. K. Pangburn, and L. S. McDaniel. 2001. PspC,
a pneumococcal surface protein, binds human factor H. Infect. Immun. 69:
3435–3447.
22. Janulczyk, R., F. Iannelli, A. G. Sjöholm, G. Pozzi, and L. Björck. 2000. Hic, a
novel surface protein of Streptococcus pneumoniae that interferes with complement function. J. Biol. Chem. 275: 37257–37263.
23. Johnsson, E., K. Berggård, H. Kotarsky, J. Hellwage, P. F. Zipfel, U. Sjöbring,
and G. Lindahl. 1998. Role of the hypervariable region in streptococcal M proteins: binding of a human complement inhibitor. J. Immunol. 161: 4894 – 4901.
24. Meri, T., A. Hartmann, D. Lenk, R. Eck, R. Würzner, J. Hellwage, S. Meri, and
P. F. Zipfel. 2002. The yeast Candida albicans binds complement regulators
Factor H and FHL-1. Infect. Immun. 70: 5185–5192.
25. Meri, T., T. S. Jokiranta, J. Hellwage, A. Bialonski, P. F. Zipfel, and S. Meri.
2002. Onchocerca volvulus microfilariae avoid complement attack by direct binding of factor H. J. Infect. Dis. 185: 1786 –1793.
26. Diaz, A., A. Ferreira, and R. B. Sim. 1997. Complement evasion by Echinococcus granulosus: sequestration of host factor H in the hydatid cyst wall. J. Immunol. 158: 3779 –3786.
27. Stoiber, H., C. Ebenbichler, R. Schneider, J. Janatova, and M. P. Dierich. 1995.
Interaction of several complement proteins with gp120 and gp41, the two envelope glycoproteins of HIV-1. AIDS 9: 19 –26.
28. Sahu, A., S. N. Isaacs, A. M. Soulika, and J. D. Lambris. 1998. Interaction of
vaccinia virus complement control protein with human complement proteins:
factor I-mediated degradation of C3b to iC3b1 inactivates the alternative complement pathway. J. Immunol. 160: 5596 –5604.
29. da Silva, C. M., L. de Abreu Vidipó, R. Nishi, and M. Cristina Plotkowski. 2004.
Binding of plasminogen to Pseudomonas aeruginosa results in formation of surface-associated plasmin and enhanced bacterial invasiveness. Microb. Pathog.
36: 59 – 66.
30. Ullberg, M., G. Kronvall, I. Karlsson, and B. Wiman. 1990. Receptors for human
plasminogen on Gram-negative bacteria. Infect. Immun. 58: 21–25.
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
addition, Tuf of M. pneumoniae binds fibronectin and Tuf of Lactobacillus johnsonii mediates attachment of the bacteria to mucins
and to human intestinal cells (47, 48). An additional role of Tuf for
immune response was reported for the plant Arabidopsis thaliana,
which recognizes Tuf at the surface of the pathogen Pseudomonas
syringae. Treatment of plant leaves with Tuf-derived peptides initiates defense reactions and induces resistance of the plant (49). In
summary, Tuf is a multifunctional protein that is located in the
cytoplasm and at the bacterial surface. Therefore Tuf is a member
of a still expanding family of “moonlighting proteins” (50), which
represent virulence factors that are localized in the cytoplasm as
well at the cell surface. The exact export pathway and the surface
anchoring mechanisms of these proteins are currently unknown
(51). However, for bacteria there is evidence for an additional
protein transport mechanism mediated by membrane vesicles (52).
In this study, we identify Tuf as a bacterial plasminogen binding
protein. Plasminogen binding to P. aeruginosa was recently reported (29), and to our knowledge Tuf is the first plasminogen
receptor identified for P. aeruginosa. The translational elongation
factor Tef1p of the pathogenic yeast C. albicans, which shares
29% sequence identity on the amino acid level, was also identified
as a plasminogen binding surface protein (53). P. aeruginosa Tuf
lacks the plasminogen binding motif that was identified for enolase
of S. pneumoniae (54) and shows no homology to other bacterial
plasminogen binding proteins such as GAPDH of S. pneumoniae
(55), OspA of B. burgdorferi (56), or M protein of S. pyogenes
(57). Plasminogen bound to recombinant Tuf is accessible for the
activator uPA and is converted to proteolytically active plasmin
(Fig. 8A).
Acquisition of plasminogen has been reported for several pathogens and seems crucial for extracellular matrix interaction, degradation, and systemic dissemination of a pathogen from superficial
skin lesions (38, 58 – 60). The proenzyme plasminogen is converted by the human plasminogen activators uPa and tissue-type
plasminogen activator to plasmin, which forms a key protease for
intravascular fibrinolysis. However, plasmin also displays extravascular functions like degradation of extracellular matrix components such as laminin, vitronectin, and fibronectin (61). Binding
of plasminogen via Tuf to the surface of P. aeruginosa may
therefore facilitate invasion of host tissue. Thus, Tuf as a bacterial surface protein binds the central host effector proteins
plasminogen and the complement inhibitor Factor H, which are
relevant for tissue disintegration and complement inactivation
by P. aeruginosa.
2987
2988
46. Kraiczy, P., C. Skerka, V. Brade, and P. F. Zipfel. 2001. Further characterization
of complement regulator-acquiring surface proteins of Borrelia burgdorferi. Infect. Immun. 69: 7800 –7809.
47. Tosi, M. F., H. Zakem, and M. Berger. 1990. Neutrophil elastase cleaves C3bi on
opsonized Pseudomonas as well as CR1 on neutrophils to create a functionally
important opsonin receptor mismatch. J. Clin. Invest. 86: 300 –308.
48. Marques, M. A., S. Chitale, P. J. Brennan, and M. C. Pessolani. 1998. Mapping
and identification of the major cell wall-associated components of Mycobacterium leprae. Infect. Immun. 66: 2625–2631.
49. Dallo, S. F., T. R. Kannan, M. W. Blaylock, and J. B. Baseman. 2002. Elongation
factor Tu and E1 ␤ subunit of pyruvate dehydrogenase complex act as fibronectin
binding proteins in Mycoplasma pneumoniae. Mol. Microbiol. 46: 1041–1051.
50. Granato, D., G. E. Bergonzelli, R. D. Pridmore, L. Marvin, M. Rouvet, and
I. E. Corthésy-Theulaz. 2004. Cell surface-associated elongation factor Tu mediates the attachment of Lactobacillus johnsonii NCC533 (La1) to human intestinal cells and mucins. Infect. Immun. 72: 2160 –2169.
51. Kunze, G., C. Zipfel, S. Robatzek, K. Niehaus, T. Boller, and G. Felix. 2004. The
N terminus of bacterial elongation factor Tu elicits innate immunity in Arabidopsis plants. Plant Cell. 16: 3496 –3507.
52. Jeffery, C. J. 1999. Moonlighting proteins. Trends Biochem. Sci. 24: 8.
53. Chhatwal, G. S. 2002. Anchorless adhesins and invasins of Gram-positive bacteria: a new class of virulence factors. Trends Microbiol. 10: 205–208.
54. Kadurugamuwa, J. L., and T. J. Beveridge. 1995. Virulence factors are released
from Pseudomonas aeruginosa in association with membrane vesicles during
normal growth and exposure to gentamicin: a novel mechanism of enzyme secretion. J. Bacteriol. 177: 3998 – 4008.
55. Crowe, J. D., I. K. Sievwright, G. C. Auld, N. R. Moore, N. A. Gow, and
N. A. Booth. 2003. Candida albicans binds human plasminogen: identification of
eight plasminogen-binding proteins. Mol. Microbiol. 47: 1637–1651.
56. Bergmann, S., D. Wild, O. Diekmann, R. Frank, D. Bracht, G. S. Chhatwal, and
S. Hammerschmidt. 2003. Identification of a novel plasmin(ogen)-binding motif
in surface displayed ␣-enolase of Streptococcus pneumoniae. Mol. Microbiol. 49:
411– 423.
57. Bergmann, S., M. Rohde, and S. Hammerschmidt. 2004. Glyceraldehyde-3-phosphate dehydrogenase of Streptococcus pneumoniae is a surface-displayed plasminogen-binding protein. Infect. Immun. 72: 2416 –2419.
58. Fuchs, H., R. Wallich, M. M. Simon, and M. D. Kramer. 1994. The outer surface
protein A of the spirochete Borrelia burgdorferi is a plasmin(ogen) receptor.
Proc. Natl. Acad. Sci. USA 91: 12594 –12594.
59. Ringdahl, U., and U. Sjöbring. 2000. Analysis of plasminogen-binding M proteins of Streptococcus pyogenes. Methods 21: 143–150.
60. Coleman, J. L., E. J. Roemer, and J. L. Benach. 1999. Plasmin-coated Borrelia
burgdorferi degrades soluble and insoluble components of the mammalian extracellular matrix. Infect. Immun. 67: 3929 –3936.
61. Jong, A. Y., S. H. Chen, M. F. Stins, K. S. Kim, T. L. Tuan, and S. H. Huang.
2003. Binding of Candida albicans enolase to plasmin(ogen) results in enhanced
invasion of human brain microvascular endothelial cells. J. Med. Microbiol. 52:
615– 622.
62. Sun, H., U. Ringdahl, J. W. Homeister, W. P. Fay, N. C. Engleberg, A. Y. Yang,
L. S. Rozek, X. Wang, U. Sjöbring, and D. Ginsburg. 2004. Plasminogen is a
critical host pathogenicity factor for group A streptococcal infection. Science
305: 1283–1286.
63. Lähteenmäki, K., P. Kuusela, and T. K. Korhonen. 2001. Bacterial plasminogen
activators and receptors. FEMS Microbiol. Rev. 25: 531–552.
Downloaded from http://www.jimmunol.org/ by guest on June 11, 2017
31. Klausen, M., A. Heydorn, P. Ragas, L. Lambertsen, A. Aaes-Jørgensen, S. Molin,
and T. Tolker-Nielsen. 2003. Biofilm formation by Pseudomonas aeruginosa
wild type, flagella and type IV pili mutants. Mol. Microbiol. 48: 1511–1524.
32. Kühn, S., and P. F. Zipfel. 1995. The baculovirus expression vector pBSV-8His
directs secretion of histidine-tagged proteins. Gene 162: 225–229.
33. Jokiranta, T. S., J. Hellwage, V. Koistinen, P. F. Zipfel, and S. Meri. 2000. Each
of the three binding sites on complement factor H interacts with a distinct site on
C3b. J. Biol. Chem. 275: 27657–27662.
34. Oppermann, M., T. Manuelian, M. Józsi, E. Brandt, T. S. Jokiranta, S. Heinen,
S. Meri, C. Skerka, O. Götze, and P. F. Zipfel. 2006. The C-terminus of complement regulator Factor H mediates target recognition: evidence for a compact
conformation of the native protein. Clin. Exp. Immunol. 144: 342–352.
35. Sabarth, N., S. Lamer, U. Zimny-Arndt, P. R. Jungblut, T. F. Meyer, and
D. Bumann. 2002. Identification of surface proteins of Helicobacter pylori by
selective biotinylation, affinity purification, and two-dimensional gel electrophoresis. J. Biol. Chem. 277: 27896 –27902.
36. Fulda, S., S. Mikkat, F. Huang, J. Huckauf, K. Marin, B. Norling, and
M. Hagemann. 2006. Proteome analysis of salt stress response in the cyanobacterium Synechocystis sp. strain PCC 6803. Proteomics 6: 2733–2745.
37. Weber, S., F. Lottspeich, and J. Köhl. 1995. An epitope of elongation factor Tu
is widely distributed within the bacterial and archaeal domains. J. Bacteriol. 177:
11–19.
38. Wieland, G. D., N. Nehmann, D. Müller, H. Eibel, U. Siebenlist, J. Sühnel,
P. F. Zipfel, and C. Skerka. 2005. Early growth response proteins EGR-4 and
EGR-3 interact with immune inflammatory mediators NF-␬B p50 and p65.
J. Cell. Sci. 118: 3203–3292.
39. Heinen, S., M. Jozsi, A. Hartmann, M. Noris, G. Remuzzi, C. Skerka, and
P. F. Zipfel. 2007. Hemolytic uremic syndrome: a Factor H mutation
(E1172Stop) causes defective complement control at the surface of endothelial
cells. J. Am. Soc. Nephrol. 18: 506 –514.
40. Bergmann, S., M. Rohde, K. T. Preissner, and S. Hammerschmidt. 2005. The
nine residue plasminogen-binding motif of the pneumococcal enolase is the major
cofactor of plasmin-mediated degradation of extracellular matrix, dissolution of
fibrin and transmigration. Thromb. Haemost. 94: 304 –311.
41. Timmann, C., M. Leippe, and R. D. Horstmann. 1991. Two major serum components antigenically related to complement factor H are different glycosylation
forms of a single protein with no factor H-like complement regulatory functions.
J. Immunol. 146: 1265–1270.
42. Mukhopadhyay, S., S. Shankar, W. Walden, and A. M. Chakrabarty. 1997. Complex formation of the elongation factor Tu from Pseudomonas aeruginosa with
nucleoside diphosphate kinase modulates ribosomal GTP synthesis and peptide
chain elongation. J. Biol. Chem. 272: 17815–17820.
43. Estaller, C., V. Koistinen, W. Schwaeble, M. P. Dierich, and E. H. Weiss. 1991.
Cloning of the 1.4-kb mRNA species of human complement factor H reveals a
novel member of the short consensus repeat family related to the carboxy terminal of the classical 150-kDa molecule. J. Immunol. 146: 3190 –3196.
44. Blackmore, T. K., T. A. Sadlon, H. M. Ward, D. M. Lublin, and D. L. Gordon.
1996. Identification of a heparin binding domain in the seventh short consensus
repeat of complement Factor H. J. Immunol. 157: 5422–5427.
45. Kotarsky, H., J. Hellwage, E. Johnsson, C. Skerka, H. G. Svensson, G. Lindahl,
U. Sjöbring, and P. F. Zipfel. 1998. Identification of a domain in human Factor
H and Factor H-like protein-1 required for the interaction with streptococcal M
proteins. J. Immunol. 160: 3349 –3354.
COMPLEMENT AND TISSUE INVASION OF P. aeruginosa