ArticlePDF Available

Proteome analysis reveals adaptation of Pseudomonas aeruginosa to the cystic fibrosis lung environment

Authors:
  • Shres Consultancy (India) & Ataturk University - Medical School (Turkey)

Abstract and Figures

Pseudomonas aeruginosa is known for the chronic lung colonization of cystic fibrosis (CF) patients in addition to eye, ear and urinary tract infections. With the underlying disease CF patients are predisposed to P. aeruginosa chronic lung infection, which leads to morbidity and mortality. In this study, we compared the protein expression profile of a CF lung-adapted P. aeruginosa strain C with that of the burn-wound isolate PAO. Differentially expressed proteins from the whole-cell, membrane, periplasmic as well as extracellular fraction were identified. The whole-cell proteome of strain C showed down-regulation of several proteins involved in amino acid metabolism, fatty acid metabolism, energy metabolism and adaptation leading to a highly distinct proteome pattern for strain C in comparison to PAO. Analysis of secreted proteins by strain C compared to PAO revealed differential expression of virulence factors under non-inducing conditions. The membrane proteome of strain C showed modulation of the expression of porins involved in nutrient and antibiotic influx. The proteome of the periplasmic space of strain C showed retention of elastase despite that the equal amounts were secreted by strain C and PAO. Altogether, our results elucidate adaptive strategies of P. aeruginosa towards the nutrient-rich CF lung habitat during the course of chronic colonization.
Content may be subject to copyright.
REGULAR ARTICLE
Proteome analysis reveals adaptation of Pseudomonas
aeruginosa to the cystic fibrosis lung environment
Dinesh Diraviam Sriramulu1, 2, Manfred Nimtz3and Ute Romling1
1Microbiology and Tumor Biology Center (MTC), Karolinska Institutet, Stockholm, Sweden
2Department of Cell Biology and Immunology, Gesellschaft für Biotechnologische Forschung,
Braunschweig, Germany
3Department of Structural Biology, Gesellschaft für Biotechnologische Forschung,
Braunschweig, Germany
Pseudomonas aeruginosa is known for the chronic lung colonization of cystic fibrosis (CF) patients
in addition to eye, ear and urinary tract infections. With the underlying disease CF patients are
predisposed to P. aeruginosa chronic lung infection, which leads to morbidity and mortality. In
this study, we compared the protein expression profile of a CF lung-adapted P. aeruginosa strain
C with that of the burn-wound isolate PAO. Differentially expressed proteins from the whole-cell,
membrane, periplasmic as well as extracellular fraction were identified. The whole-cell proteome
of strain C showed down-regulation of several proteins involved in amino acid metabolism, fatty
acid metabolism, energy metabolism and adaptation leading to a highly distinct proteome pat-
tern for strain C in comparison to PAO. Analysis of secreted proteins by strain C compared to
PAO revealed differential expression of virulence factors under non-inducing conditions. The
membrane proteome of strain C showed modulation of the expression of porins involved in
nutrient and antibiotic influx. The proteome of the periplasmic space of strain C showed reten-
tion of elastase despite that the equal amounts were secreted by strain C and PAO. Altogether,
our results elucidate adaptive strategies of P. aeruginosa towards the nutrient-rich CF lung habitat
during the course of chronic colonization.
Received: June 1, 2004
Revised: November 20, 2004
Accepted: December 27, 2004
Keywords:
Periplasmic proteome / Adaptation / Proteome minimalism / Cystic fibrosis / Pseudo-
monas aeruginosa
3712 Proteomics 2005, 5, 3712–3721
1 Introduction
Pseudomonas aeruginosa is a common environmental bacte-
rium possessing a rich gene pool, which enables it to colo-
nize multiple niches and to utilize a variety of compounds as
energy source. A large number of virulence factors from this
opportunistic pathogen contribute to the infection of burn
wounds, lungs, eyes and also the urinary tract. In addition to
acute lung infections manifested in patients in intensive care
units, P. aeruginosa chronic lung infection in cystic fibrosis
(CF) accounts for morbidity and mortality. Once taken resi-
dence in the lung, P. aeruginosa cannot be eradicated with
most aggressive antibiotic therapies [1]. CF isolates of P. aer-
uginosa converge towards a common phenotype such as lack
of flagella, amino acid auxotrophy, changes in the lipopoly-
saccharide molecule like the loss of O-antigen and modifica-
tion of lipid A, reduced secretion of virulence factors, the
mutator phenotype and mucoidy regardless of their genetic
background [2–7]. The genetic basis of such differences can
be point mutations in structural or regulatory genes like
Correspondence: Dr. Ute Romling, Box 280, Microbiology and
Tumor Biology Center, Karolinska Institutet, SE-17177 Stock-
holm, Sweden
E-mail: Ute.Romling@mtc.ki.se
Fax: 146-8-330-744
Abbreviations: CF, cystic fibrosis; TSB, trypticase soy broth; TSA,
trypticase soy agar; SOD, superoxide dismutase; ROS, reactive
oxygen species
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
DOI 10.1002/pmic.200401227
Proteomics 2005, 5, 3712–3721 Microbiology 3713
mutS,mucA and rpoN or gene knockouts accompanied by
large chromosomal inversions [7–10]. Aforementioned fea-
tures of CF lung isolates of P. aeruginosa distinct from their
environmental counterpart reflect the selective pressure
offered by the unusual niche of the CF lung to modulate
gene expression.
A decade-long epidemiological study revealed that a pre-
dominant lineage of P. aeruginosa, clone C was found in the
lungs of CF patients, in other diseases and in clinical and
nonclinical environments such as rivers, swimming pool
and drinking water throughout Europe [5, 11, 12]. In this
study we analyzed the proteome of a member of P. aerugi-
nosa clone C, designated as strain C, a CF lung isolate which
has a genotype [5] frequently occurring in CF patients of the
Medical School of Hannover. P. aeruginosa strain C was iso-
lated during the onset of the colonization process in the
CF lung. P. aeruginosa strain C has been reported to harbor
600 kb additional genetic material compared to the reference
strain P. aeruginosa PAO due to insertions which ranged
from 23 to 155 kb, resulting in a mosaic-like structure of the
chromosome [13]. Here we present a detailed comparison of
the strain C and PAO proteome, which gave an overview of
the overall protein expression pattern. A detailed protein
expression profile was established by fractionation proce-
dures, which revealed localization of membrane and secreted
proteins and proteins in the periplasmic space in a P. aeru-
ginosa strain that inhabits the unusual niche, the CF lung.
Interpreting the data obtained, we hypothesize that P. aeru-
ginosa strain C exerts ‘proteome minimalism’ by turning off
metabolic pathways not required in the nutrient-rich CF
lung habitat.
2 Materials and methods
2.1 Bacterial strains and growth conditions
Pseudomonas aeruginosa strains PAO1 (ATCC 15692) and C
were pre-cultured in trypticase soy broth (TSB) (1% Tryp-
tone, 0.5% sodium chloride) overnight at 377C with shaking.
The pre-culture was used to prepare a lawn culture on tryp-
ticase soy agar (TSA) (TSB containing 1.5% agar) and incu-
bated for 16 h at 377C.
2.2 Preparation and purification of whole-cell
proteins
All preparative steps were carried out at 47C unless otherwise
mentioned. Cultures were scraped from the plate, washed
twice with 0.01 MPBS, pH 7.5 and centrifuged (6000 6gfor
10 min. at 47C) to obtain the bacterial pellet. Whole-cell pro-
teins were extracted as described [14]. A 1 mL volume of cell
pellet was thoroughly dispersed in 2 mL of solubilization
buffer (7 Murea, 2 Mthiourea, 4% w/v CHAPS, 30 mMDTT,
2mMleupeptin, 1 mMPefaBloc SC, 20 mL1MTris (pH 9.5),
0.5% w/v Pharmalyte (pH 3–10)) and lysed step-wise using a
Branson probe sonicator (Danbury, CT, USA) on ice (40%
duty cycle; Microtip limit 5) for 30 s. Further sonications
were carried out for 3 620 s with an interval of 30 s which
ensured efficient lysis. The sample mixture was then cen-
trifuged at 50 000 6gfor 30 min at 107C. The supernatant
was separated carefully from the pellet and used immedi-
ately for phenol-acetone purification of proteins.
2.3 Phenol-acetone purification
The crude protein extract was purified by a procedure
described by Hancock and Nikaido [15]. One volume of buf-
fer-saturated phenol was added to the protein extract, vor-
texed vigorously, incubated at 707C for 10 min and snap-
cooled on ice. To this mixture, an equal volume of water was
added, vortexed vigorously and warmed and cooled as before.
After centrifugation at 4000 6gfor 10 min at 47C, the upper
aqueous phase was discarded and proteins in the lower
organic phase were precipitated using 5 volumes of ice-cold
acetone. The pure protein pellet was recovered after low-
speed centrifugation at 47C. The final protein pellet was
resuspended in an appropriate volume of solubilization buf-
fer and centrifuged at 10 000 6gfor 3 min to remove insol-
uble materials. Finally, the supernatant corresponding to
600 mg of protein was used immediately for IEF across the
linear pH range 4–7 after quantification using the Bio-Rad
protein assay kit (Bio-Rad).
2.4 Isolation of secretory proteins
Secreted proteins were isolated as described [16]. Bacteria
grown in 100 mL TSB until the stationary phase was reached
were centrifuged at 6000 6g for 15 min and the super-
natant was passed through a 0.22 mm pore size membrane
filter to remove bacterial cells. Deoxycholic acid (0.2 mg/mL)
was added and the mixture was incubated for 30 min. on ice.
Then TCA acid was added (6% w/v), incubated overnight at
47C to precipitate proteins and centrifuged at 18 000 6gfor
30 min. The pellet was resuspended in a small volume of
water and precipitated with 8 volumes of ice-cold acetone.
After incubation at 2207C for 2 h the mixture was cen-
trifuged at 3500 6gfor 20 min and the pellet was dried at
room temperature for 5 min. Dried pellet was dissolved
thoroughly in an appropriate volume of solubilization buffer
and centrifuged at 50 000 6gfor 40 min at 107C to remove
insoluble materials. The supernatant was treated with phe-
nol-acetone as described above. Four hundred micrograms of
protein was separated by 2-DE across the linear pH range 3–
10.
2.5 Isolation and purification of membrane proteins
A method described by Hancock and Nikaido [15] was fol-
lowed with modifications. Harvested bacterial pellet was
washed twice with 10 mMTris-Cl (pH 7.5) and resuspended
in 0.5 Msucrose buffer containing protease inhibitors
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
3714 D. D. Sriramulu et al. Proteomics 2005, 5, 3712–3721
(1 tablet Complete Mini cocktail (Roche) in 30 mL solution).
The suspension was sonicated (Microtip limit 5; 40% duty
cycle) three times for 30 s on ice with an interval of 30 s each.
A discontinuous sucrose density gradient (0, 0.5, 1.0, 1.5 and
2.0 Msucrose) was prepared, the suspension was carefully
layered on the top of the gradient and centrifuged at
50 000 6gfor 1 h. Fractions at the interfaces of 0.5:1.0 M
(inner membrane) and 1.0:1.5 M(outer membrane) were
collected by successively sucking the liquid from the top and
pooled together. The pooled fractions were diluted with at
least 2 volumes of deionized water in order to achieve a
sucrose concentration below 20% and centrifuged at
50 000 6gfor 1 h to pellet the membranes.
The pellet was resuspended thoroughly in 1 MTr i s -C l
(pH 7.8) containing appropriate amount of Complete mini
protease inhibitor cocktail. To remove contaminants that inter-
fere with IEF, phenol-acetone purification was carried out as
described above. The resulting pellet was treated with
2 volumes of ether and the precipitated protein was resus-
pended in a modified solubilization buffer (7 Murea,
2Mthiourea, 2 mMtributyl phosphine, 1% amidosulfobe-
taine-14, 1tablet Complete Mini protease inhibitor cocktail
(Roche) per 5mL,20mL1MTris (pH 9.5), 0.5% w/v Pharma-
lyte (pH 3–10)). The protein preparation was centrifuged to
achieve a clear supernatant and used for IEF; 400 mgofprotein
was separated by 2-DE across the linear pH range 3–10.
2.6 Extraction of proteins from the periplasmic space
Proteins from the bacterial periplasm were isolated by the
chloroform shock procedure as described by Ames [17].
Briefly, harvested bacteria grown in 100 mL TSB until the
stationary phase was reached were washed, dispersed
homogenously in 0.5 mL chloroform and allowed to stay for
15 min at room temperature. 10 mL of 10 mMTris-Cl
(pH 8.0) was added and mixed thoroughly. The cells were
then removed by centrifugation at 10 000 6gfor 20 min to
obtain a clear supernatant. Proteins were extracted from the
resulting ‘chloroform shock fluid’ by following the procedure
described for the isolation of secretory proteins. Finally
400 mg of purified protein was separated by 2-DE across the
non-linear pH range 3–10.
2.7 2-DE
Isoelectric focussing was carried out using IPGphor
(Amersham Pharmacia) at 207C. IPG strips were rehydrated
for 4 h and the following scheme was used for focussing:
30 V constant for 10 h, 150 V gradient for 2 h, 300 V gradient
for 2 h, 600 V gradient for 2 h, 1500 V gradient for 4 h,
3500 V gradient for 8 h, 8000 V gradient for 3 h, and a final
8000 V constant for a total of 250 kVh. IPG strips were
reduced (6 Murea, 30% glycerol, 1% w/v DTT, 2% w/v SDS
in 0.05 MTris-Cl (pH 8.8)) for 12 min and free SH-groups
were blocked with iodoacetamide (6 Murea, 30% glycerol,
260 mMiodoacetamide, 2% w/vw/v SDS in 0.05 MTris-Cl
(pH 8.8) for 12 min. Then the strips were embedded on top
(using 0.5% agarose in running buffer colored with bromo-
phenol blue) of a 12–15% pore-gradient SDS-PAGE and run
in 24 mMTris, 0.2 Mglycine, 0.1% SDS at 6 V/cm at 107C
until the bromophenol blue dye front reached the end of the
gel. The gels were fixed in 40% ethanol and 10% acetic acid
mixture for at least 4 h to overnight, successively stained
overnight with CBB G-250 (Biomol, Hamburg, Germany) as
described by Neuhoff [18] and destained overnight in dis-
tilled water. After scanning (Umax Powerlook III) the gels,
protein spots, which showed at least three-fold different
intensities as evident from the comparison by using Phor-
etix 2D software (Nonlinear Dynamics, USA) and by visual
examination between comparative gels were excised and
processed for mass spectrometry or archived for future use.
All experiments were carried out at least twice, such as, pro-
tein extracts for the whole-cell proteome and sub-proteome
analyses were prepared from at least two independent bacte-
rial cultures and at least two gels were analyzed.
2.8 Protein identification and characterization
Preparation of protein spots for mass spectrometry was car-
ried out as described [19]. Briefly, a protein spot was excised
from the gel and chopped into tiny pieces. After washing
with deionized water the pieces were shrunk with 50 mL
ACN. The supernatant was discarded and the gel pieces were
reswollen in 50 mLof10m
MDTT in 0.1 MNH4HCO3to
reduce the disulfide bonds of the proteins. After incubation
at 567C for 30 min, 50 mL of ACN was added to shrink gel
pieces. The supernatant was removed and the gel pieces were
reswollen in 50 mLof55m
Miodoacetamide in
0.1 MNH4HCO3. After incubation for 20 min in the dark at
room temperature, the supernatant was removed completely.
Gel pieces were shrunk using ACN as before, dried briefly,
reswollen in trypsin solution (2 ng/mL trypsin in
0.05 MNH4HCO3) and incubated overnight at 377C for
digestion. After incubation 10–15 mL25m
MNH4HCO3was
added to the gel pieces and incubated at 377C for 15 min with
shaking. Then 50 mL ACN was added to the previous mixture
and incubated at 377C for 15 min with shaking. Supernatant
was collected and stored. To the gel pieces 40–50 mL 5% for-
mic acid was added and incubated at 377C for 15 min with
shaking. Then, 50 mL of ACN was added to the previous
mixture and incubated at 377C for 15 min with shaking. The
gel particles were spun down to collect the supernatant,
pooled with previously collected supernatant and dried com-
pletely in a SpeedVac (Eppendorf). Dried peptides were
recovered in 10 mL of 0.5% HCOOH in 5.0% methanol and
desalted using C18 ZipTip (Millipore). Elution of an aliquot of
peptides on MALDI target was done with an equal volume of
saturated solution of alpha-cyano-4-hydroxy cinnamic acid in
0.5% formic acid/65% methanol mixture. PMF maps of
trypsin-digested peptides generated by MALDI-Tof Reflex II
(Bruker-Franzen-Analytik) mass spectrometer were com-
pared to a database of all translated ORF from P. aeruginosa
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
Proteomics 2005, 5, 3712–3721 Microbiology 3715
PAO1 (www.pseudomonas.com) using MS-Fit of Protein
Prospector package (http://falcon.ludwig.ucl.ac.uk/
ucsfhtml3.2/msfit.htm). A minimum of 25% sequence cov-
erage was considered sufficient to identify a protein with
confidence. If the sequence coverage was less than 25%, at
least three peptide-sequence tags were sequenced by ESI
MS/MS using Q-Tof-2 (Micromass) to identify the protein. In
some cases peptide sequencing of tryptic digests was done
without prior MALDI-Tof analysis. If unambiguous assign-
ment was not possible, N-terminal sequencing (Procise Pro-
tein Sequencer) was carried out with protein spots excised
from a PVDF-blotted 2-D gel and the sequences were
screened against protein databases using Fasta3 (www.ebi.
ac.uk/fasta33).
3 Results and discussion
3.1 Whole-cell proteome comparison of P. aeruginosa
PAO and C
2-D separation of whole-cell proteins revealed characteristic
protein expression profiles for the burn wound isolate
P. aeruginosa PAO (Fig. 1a) and the CF-lung isolate C
Figure 1. Whole-cell proteome of P. aeruginosa (a) PAO and
(b) strain C. Numbered arrows indicate the identity of protein
spots listed in Table 1.
(Fig. 1b). The proteome of strain C as visible in the pH range
4–7 showed a large number of protein spots with lower
intensity as compared to PAO. Such a unique proteome pat-
tern was not only shown by strain C (Fig. 1b) isolated during
the onset of the colonization process but also by a few other
CF isolates belonging to clone C, from the mid- and late-
phase of chronic lung infection (data not shown). Due to the
huge number of differences between 2-D whole-cell protein
gels from P. aeruginosa PAO and strain C some spots had to
be selected. Therefore protein spots, which could not be vis-
ualized in one of the compared gels and showed at least
three-fold different intensities that could be visually dis-
tinguished between strain C and PAO were chosen for fur-
ther analysis by PMF and/or by MS/MS and/or by sequenc-
ing the N-terminal region. A total of 192 proteins (see Table 1
and Supplementary table) belonging to different functional
categories were identified with the help of the available
whole genome sequence database (www.pseudomonas.
com).
3.2 Down-regulation of proteins involved in general
metabolic pathways in strain C
Enzymes involved in biosynthesis as well as degradation of
several amino acids were down-regulated in the strain C
proteome. Most prominently down-regulated were the
enzymes involved in the biosynthesis and metabolism of
branched-chain amino acids (leucine, isoleucine and valine
(LIV)), serine, threonine, phenylalanine, lysine, methionine,
glutamine and tryptophan (Table 1). Acetyl-Coenzyme A (A-
CoA) is a central metabolite and is used in several metabolic
processes like amino acid biosynthesis and fatty acid biosyn-
thesis. Three proteins of a multi-enzyme complex (pyruvate
dehydrogenase (AceE), lipoamide dehydrogenase-Val (LpdV)
and branched-chain alpha-keto acid dehydrogenase (BkdB))
that catalyzes the formation of A-CoA from pyruvate
(www.kegg.org) were found down-regulated in the strain C
proteome. AceE in complex with acetolactate synthase is also
required for the conversion of pyruvate to 2-hydroxyethyl
thiamine diphosphate, which enters into valine and iso-
leucine biosynthetic pathways. Ketol-acid reductoiosmerase
(IlvC), the second enzyme involved in two catalytic reactions
in the biosynthetic pathway leading to the formation of
valine and isoleucine was also down-regulated in the strain C
proteome. 2-Oxoisovalerate dehydrogenase, a complex en-
zyme with two subunits (BkdA1 and BkdA2) and beta-keto-
adipyl CoA thiolase (PcaF) involved in the initial step and
final steps, respectively, of the LIV degradation pathway were
down-regulated in the strain C proteome. Other differentially
expressed proteins involved in amino acid metabolism are
shown in Table 1. Amino acids were found at higher con-
centrations in the CF sputum compared to sputum of heal-
thy individuals and proposed to play a significant role in the
emergence and maintenance of P. aeruginosa mutants auxo-
trophic for amino acids during chronic colonization of the
lung [6, 20, 21]. Differential expression of proteins
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
3716 D. D. Sriramulu et al. Proteomics 2005, 5, 3712–3721
Table 1. Identity of protein spots shown in Figures 1 through 4
Spot
no.
PA
no.
Function Gene Mass
(kDa)
pIExpression
level
Amino acid biosynthesis and metabolism
1 PA2250 Lipoamide dehydrogenase-Val lpdV 48.6 6.16 ;
2 PA5413 Low specificity L-threonine aldolase ltaA 38.2 5.18 ;
3 PA2248 2-Oxoisovalerate dehydrogenase-Beta subunit bkdA2 38.3 5.37 ;
4 PA0865 4-Hydroxyphenylpyruvate dioxygenase hpd 39.9 5.10 ;
5 PA2249 Branched-chain alpha-keto acid dehydrogenase bkdB 45.8 5.84 ;
6 PA5171 Arginine deiminase arcA 46.4 5.52 ;
7 PA5015 Pyruvate dehydrogenase aceE 99.6 5.56 ;
8 PA2247 2-Oxoisovalerate dehydrogenase-Alpha subunit bkdA1 45.3 5.67 ;
9 PA4938 Adenylosuccinate synthetase purA 46.8 5.70 ;
10 PA0432 S-adenosyl-L-homocysteine hydrolase sahH 51.4 5.71 ;
11 PA4759 Dihydrodipicolinate reductase dapB 28.3 5.74 ;
12 PA5172 Ornithine carbamoyl transferase, catabolic arcB 38.1 6.13 ;
13 PA0609 Anthranilate sythetase - component I trpE 54.9 5.00 ;
14 PA5119 Glutamine synthetase (glutamate-ammonia ligase) glnA 52.1 5.10 ;
15 PA0871 Pterin-4-alpha-carbinolamine dehydratase phhB 13.3 6.89 ;
16 PA0316 D-3-phosphoglycerate dehydrogenase serA 44.2 6.52 ;
17 PA0870 Aromatic amino acid aminotransferase phhC 43.2 6.24 ;
18 PA0447 Glutaryl-CoA dehydrogenase gcdH 43.3 6.12 ;
19 PA4694 Ketol-acid reductoisomerase ilvC 36.4 5.65 ;
Fatty acid biosynthesis and metabolism
20 PA4848 Biotin carboxylase accC 48.9 5.92 ;
21 PA1609 Beta-ketoacyl-ACP synthase I fabB 42.8 5.40 ;
22 PA3639 Acetyl coenzyme A carboxylase carboxyl transferase-alpha accA 35.0 5.15 ;
23 PA0228 Beta-ketoadipyl CoA thiolase pcaF 42.1 5.52 ;
Membrane proteins
24 PA1092 Flagellin type B fliC 49.2 5.40 *
\;
25 PA0958 Porin D precursor oprD 48.4 4.75 ;
26 PA2760 Probable outer membrane protein oprQ 46.8 5.51 ;
27 PA1777 Outer membrane protein OprF precursor oprF 37.6 4.98 :
28 PA4370 Insulin-cleavage metalloproteinase icmP 47.2 4.68 :
29 PA0766 Serine protease MucD precursor mucD 50.4 7.77 :
30 PA3262 Probable peptidyl-prolyl cis-trans isomerase FkbP-type 26.9 6.80 :
31 PA1041 Probable outer membrane protein 21.8 5.06 :
32 PA2853 Major OM lipoprotein precursor (murein-lipoprotein) oprI 8.80 8.51 :
33 PA1178 PhoP/Q & low Mg21inducible OMP H1 precursor oprH 21.6 9.43 :
Secreted proteins
34 PA5112 Lipase/esterase precursor estA 71.7 4.66 *
\;
35 PA1080 Flagellar hook protein flgE 48.3 4.42 *
\;
36 PA1092 Flagellin type B fliC 49.2 5.40 *
\;
37 PA2939 Probable aminopeptidase 59.3 4.97 ;
38 PA3724 Propeptide fragment of elastase lasB 18.0 5.90 :
39 PA4175 Protease IV prpL 26.0 8.70 :
40 PA3724 Elastase lasB 53.7 6.28 nc
Periplasmic proteins
41 PA1092 Flagellin type B fliC 49.2 5.40 *
\;
42 PA1074 Branched chain amino acid binding protein braC 39.8 5.80 ;
43 PA0301 Polyamine transport protein PotF3 potF3 40.1 5.51 ;
44 PA3250 Putative spermidine/putrescine substrate-binding protein 38.3 5.92 ;
45 PA4913 Probable binding component of ABC transporter 39.8 6.20 :
46 PA0852 Chitin binding protein precursor cbpD 42.0 6.60 ;
47 PA2442 Glycine cleavage system protein T2 gcvT2 39.9 5.95 *
\;
48 PA0300 Polyamine transport protein PotF2 potF2 40.6 6.97 ;
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
Proteomics 2005, 5, 3712–3721 Microbiology 3717
Table 1. Continued
Spot
no.
PA
no.
Function Gene Mass
(kDa)
pIExpression
level
49 PA1493 Sulfate binding protein of ABC transporter cysP 36.5 7.76 ;
50 PA1946 Binding protein component precursor of ABC ribose transporter rbsB 33.9 7.78 :
51 PA3724 Elastase lasB 53.7 6.28 :
52 PA5489 Disulphide oxidoreductase/thiol:disulphide interchange protein dsbA 23.4 5.98 :
53 PA5153 Probable periplasmic binding protein 27.7 5.13 ;
54 PA2513 Anthranilate dioxygenase small subunit antB 19.4 6.65 *
\;
55 PA2614 Lipoprotein-specific chaperone lolA 23.1 5.75 :
56 PA2300 Chitinase chiC 53.0 5.23 *
57 PA4356 Xenobiotic reductase xenB 37.8 5.11 :
58 PA5018 Peptide methionine sulfoxide reductase msrA 23.7 5.20 :
59 PA0852 Chitin binding protein (cleavage product) cbpD 30.0 6.40 :
60 PA0888 Arginine/ornithine binding protein aotJ 28.0 6.43 ;
61 PA0139 Alkyl hydroperoxide reductase subunit C ahpC 20.6 5.89 ;
62 PA4468 Superoxide dismutase (manganese cofactored) sodM 22.5 5.81 ;
63 PA4366 Superoxide dismutase (iron cofactored) sodB 21.4 5.27 ;
64 PA4922 Azurin precursor azu 16.0 6.39 ;
Symbols represent differential expression of proteins in strain C compared to PAO
;down regulated
:up regulated
*
\Not found in the gel of strain C
*Spot hidden by an overlapping protein
nc, no change
involved in amino acid metabolism has also been reported
for P. aeruginosa during the course of biofilm formation [22].
In Bacillus subtilis, it has been shown that addition of
0.2% casamino acids in the medium had a pronounced effect
on the expression of proteins involved in amino acid metab-
olism [23]. Based on the global changes in amino acid me-
tabolism in strain C, we speculate that strain C, though a
prototroph, has developed partial auxotrophy during the
course of chronic lung infection. It has been known for a
long time that P. aeruginosa develops auxotrophy for various
amino acids in the CF lung [6].
Beta-ketoadipyl CoA thiolase (PcaF), required for the LIV
degradation, is also required for the first step in the fatty acid
biosynthesis and metabolism. In addition to PcaF, other
enzymes such as FabB and AccAB involved in the fatty acid
biosynthetic pathway were down-regulated by strain C. Fatty
acids are essential constituents of bacterial membrane phos-
pholipids and lipid A and broad-spectrum antimicrobials tar-
get fatty acid biosynthetic pathway enzymes [24]. Down-reg-
ulation of proteins involved in fatty acid biosynthesis might be
one of the adaptation strategies of strain C to the CF lung en-
vironment, where there is a constant antimicrobial pressure.
3.3 Membrane subproteome of P. aeruginosa strain C
Identification of 15 differentially expressed protein spots
from the membrane fraction revealed that there was no con-
tamination with cytoplasmic proteins (Fig. 2a and b and
Supplementary table). As a prominent spot in the 2-D gel,
the major nonspecific outer membrane porin (OprF) [25] was
up-regulated in strain C. OprF is a major multifunctional
outer membrane protein, which was shown to have a large
exclusion limit allowing molecules with molecular weight up
to 3000 which includes amino acids, di- and tri-saccharides to
pass into periplasmic space [25–27]. Recently, OprF was
shown to be significantly up-regulated by P. aeruginosa
growing in the CF lung [28]. OprH involved in the resistance
to polycationic antibiotic polymyxin B [29, 30] and the major
outer membrane lipoprotein OprI (spot 32 in Fig. 1a and b)
were also up-regulated, as was a peptidyl-prolyl-cis-trans
isomerase (PA3262) associated with outer membrane
involved in the folding of proteins. MucD, a serine protease
and a repressor of mucoid phenotype [31], was also up-regu-
lated by strain C. This observation is consistent with the non-
mucoid phenotype exhibited by strain C on a solid culture
medium (data not shown). About five protein spots (Table 1
and Supplementary table) identified as proteins of unknown
function were up-regulated in the strain C membrane sub-
proteome.
Conversely, OprD, a porin specific for antibiotic influx
[32] was expressed at a lower level by strain C. This observa-
tion seems considerable as the resistance mechanism to car-
bapenem antibiotics requires down-regulation of this porin
which serves as the primary route of entry [32, 33]. Moreover,
down-regulation or loss of OprD has been shown by P. aeru-
ginosa isolates derived from the CF lung environment [34].
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
3718 D. D. Sriramulu et al. Proteomics 2005, 5, 3712–3721
Figure 2. Membrane sub-proteome of P. aeruginosa (a) PAO and
(b) strain C. Numbered arrows indicate the identity of protein
spots listed in Table 1.
3.4 Secretory proteome
Extracellular proteins were extracted from the stationary
phase rich medium culture of P. aeruginosa PAO and C
(Fig. 3a and b) and 13 protein spots (see Supplementary
table) were identified. Protease IV, an important corneal
virulence factor [35] was expressed in higher quantities by
strain C. This protease was shown to be a virulence factor
in corneal infection and it can digest biologically important
proteins such as immunoglobulin, complement compo-
nents, fibrinogen and plasminogen [36, 37]. Over-
expression and secretion of proteases by strain C might be
necessary to alleviate host immune factors, for example,
transferrin and lactoferrin [38]. Elastase is one of the major
virulence factors produced by P. aeruginosa [39, 40]. Both
strain C and PAO produced equal amount of the secreted
form of elastase.
Figure 3. Secretory proteome of P. aeruginosa (a) PAO and
(b) strain C. Numbered arrows indicate the identity of protein
spots listed in Table 1.
The cell surface appendage protein flagellin (FliC) and its
associated hook protein (FlgE) were found in the extra-
cellular fraction of PAO1 as the flagellum can easily be
detached from the cell especially in shaking cultures and is
not excluded by the filtration procedure [39]. However, FliC
was not detected in the secretory proteome of strain C, con-
sistent with strain C being non-motile (data not shown).
3.5 Proteome of the periplasmic space
Periplasm not only serves as a buffer zone that controls the
influx and efflux of nutrients, antibiotics and other compo-
nents but also serves as a milieu where a number of viru-
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
Proteomics 2005, 5, 3712–3721 Microbiology 3719
lence factors are processed for subsequent secretion [40–42].
Proteins in the periplasmic space were extracted from sta-
tionary phase cultures by the ‘chloroform shock’ procedure.
A large number of differentially expressed protein spots
could be observed from the periplasmic sub-proteome of
P. aeruginosa PAO and C (Fig. 4a and b). About 39 protein
spots (Table 1 and Supplementary table) were chosen for
further characterization. The predicted and reported peri-
plasmic location of the identified proteins established the
absence of contaminating proteins from other compart-
ments. Prominently visible protein spots from the P. aerugi-
nosa PAO periplasmic subproteome and down-regulated in
the strain C periplasmic subproteome were identified as fla-
gellin type B (FliC), branched-chain amino acid binding
protein (BraC), arginine/ornithine binding protein (AotJ),
superoxide dismutase (SodB) and azurin precursor (Azu).
ATP-binding cassette transport systems in Gram-nega-
tive bacteria are basically composed of a substrate-binding
protein located in the periplasm and a membrane-bound
complex which consists of a permease located in the inner
Figure 4. Sub-proteome of the periplasmic space of P. aerugi-
nosa (a) PAO and (b) strain C. Numbered arrows indicate the
identity of protein spots listed in Table 1.
membrane and a ATP-binding protein associated with inner
membrane. Seven proteins (spots 42, 43, 44, 48, 49, 53 and
60 in Table 1) down-regulated in strain C were identified as
the substrate-binding component of ATP-binding cassette
transport systems of amino acids, amines, salts and sugar
(www.pseudomonas.com). Only two periplasmic substrate-
binding proteins were found up-regulated, one of them
being the binding protein component of ribose transporter
(RbsB).
In the P. aeruginosa PAO genome, there exist the well-
characterized high-affinity branched-chain amino acid (LIV)
ABC transporter, BraCDEFG [43]. The periplasmic binding
component BraC (PA1074), is down-regulated in strain C.
Surprisingly, a paralogous protein encoded by PA4913 which
shows 42.7% identity to BraC (PA1074) is up-regulated in the
strain C.
In the CF-lung, P. aeruginosa is exposed to reactive oxygen
species (ROS) produced by neutrophils. Superoxide dis-
mutase (SOD) plays an important role in scavenging oxygen
radicals. Two isoforms of SOD discriminated by the metal ion
at the active site have been reported in P. aeruginosa PAO.
Strain C expressed both the manganese- (SodM) and the iron-
cofactored (SodB) form of SOD whereas PAO expressed SodB
only, although at higher levels than in strain C. The C-subunit
of alkyl hydroperoxide reductase (AhpCF), another protein
involved in the oxidative stress response against exogenous
ROS [44], was up-regulated by strain C.
A highly up-regulated protein spot identified as the
mature form of elastase was found in the periplasmic sub-
proteome of strain C. Retention of elastase in the peri-
plasmic space has been shown to have a biological role,
namely, to participate in the cleavage of the 16 kDa form of
nucleoside diphosphate kinase (Ndk) to the 12 kDa form [40,
45] which predominantly synthesizes GTP necessary for
protein synthesis and virulence factor production. Accumu-
lation of enzymatically active elastase in the periplasmic
space was shown to indirectly influence the energy metabo-
lism of P. aeruginosa [40]. Although the 12 kDa form of Ndk
was not identified, the lower quantities of the 16 kDa form of
Ndk from the whole-cell proteome of strain C compared to
PAO could serve as an indirect evidence for an enhanced
conversion of Ndk from the 16 kDa to the 12 kDa counter-
parts. As an alternative function, however not mutually
exclusive, elastase is retained in the periplasm to be secreted
via membrane vesicles [46].
Chitin-binding protein (CbpD) precursor is one of the
major secreted proteins by P. aeruginosa PAO [47]. CbpD was
identified as a 43 kDa protein spot in addition to a 30 kDa
fragment, which has been shown to be cleaved by elastase
[47]. Though there was only a minor difference in the level of
expression of the full-length CbpD by P. aeruginosa C and
PAO, the 30 kDa fragment was found in huge amounts in
the strain C periplasmic sub-proteome. Chitin-binding pro-
tein (CbpD), which is expressed by many clinical isolates of
P. aeruginosa, plays a significant role as an adhesin, mediat-
ing colonization of eukaryotic cells [47].
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
3720 D. D. Sriramulu et al. Proteomics 2005, 5, 3712–3721
A variety of proteins involved in proper folding of dis-
ulfide-bond-containing proteins have been found up-regu-
lated in strain C. DsbA, a periplasmic protein that functions
as a thiol:disulfide oxidoreductase, is required for catalyzing
the oxidative folding and assembly of many secreted factors
in P. aeruginosa [42, 48]. Up-regulation of DsbA by strain C
might be important for proper folding of virulence factors
such as elastase and protease IV, which have disulfide bonds.
DsbA was also shown to affect the twitching motility pheno-
type mediated by type IV pili, which contain an intrastrand
disulfide loop [49]. A probable peptidyl-prolyl cis-trans isom-
erase FkbP type (PA3262) was also up-regulated in
strain C, which might participate in the maturation of peri-
plasmic and secreted factors. LolA, a lipoprotein-specific
chaperone [50] known to participate in the translocation of
lipoproteins from inner membrane to the outer membrane
was produced in significant amounts by strain C. Up-reg-
ulation of trigger factor, a ribosome-associated chaperone
involved in the folding of nascent secretory and non-secre-
tory polypeptide chains [51] might be necessary under
stressful growth conditions. From the above observation, it
can be speculated that up-regulation of chaperones involved
in protein-folding plays a significant role in the proper pro-
duction and localization of several virulence factors and
membrane proteins under stressful conditions such as in the
CF-lung.
Peptide methionine-sulfoxide reductase (MsrA) reduces
methionine sulfoxide produced as a consequence of oxidation
by ROS to methionine with subsequent restoration of the bio-
logical activity of proteins [52]. MsrA was also reported to be
important for the production and maintenance of cell surface
structures especially adhesins [53]. Up-regulation of MsrA by
strain C might have a protective role on proteins, which inter-
act closely with the CF-lung environment, where bacterial cells
face enhanced concentration of exogenous ROS.
4 Concluding remarks
In this study, we compared the protein expression pattern of
P. aeruginosa strains derived from different niches. CF iso-
lates of P. aeruginosa are known to exhibit multiple pheno-
typic changes depending on the extent of chronic coloniza-
tion in the CF lung. Analysis of proteins from different cel-
lular compartments and the milieu (secreted proteins)
under predefined culture conditions gave us an overview
about the general adaptation strategy of bacteria living in
the unusual niche of the CF lung (Fig. 5). This adaptation
strategy, which is maintained even under in vitro conditions,
suggests that P. aeruginosa strain C performs ‘proteome
minimalism’, by reducing the expression of all unnecessary
metabolic pathways with the probable involvement of global
regulatory gene(s). Differential regulation of proteins
involved in general metabolism, virulence and substrate
acquisition by the CF lung isolate strain C indicate that
bacteria can tailor themselves according to the nutrient sta-
tus of the microenvironment of the host. Moreover, by fol-
lowing a compartmental proteomic approach it is possible
to characterize and to localize proteins involved in novel and
unknown functions and to find effective antibacterial tar-
gets. Particularly, elaboration of proteins from the peri-
plasmic space provided better understanding of the proces-
sing of virulence factors by a well-adapted P. aeruginosa
CF lung isolate.
We thank Dirk Wehmhoener and Maja Baumgaertner for
their technical guidance. S.D.D was a member of the European
Graduate College ‘Pseudomonas: Pathogenicity and Biotechnol-
ogy’. This work was further supported by the Mukoviszidose e. V.
the Karolinska Institutet (Elitforskartjänst to U.R) and the
Anna-Greta and Holger Crafoords fund.
Figure 5. Adaptation strategy
shown by P. aeruginosa strain C
during the course of chronic
colonization in the CF lung.
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
Proteomics 2005, 5, 3712–3721 Microbiology 3721
5 References
[1] Hoiby, N., Annu. Rev. Med. 1993, 44, 1–10.
[2] Hancock, R. E., Mutharia, L. M., Chan, L., Darveau, R. P. et al.,
Infect. Immun. 1983, 42, 170–177.
[3] Luzar, M. A., Thomassen, M. J., Montie, T. C., Infect. Immun.
1985, 50, 577–582.
[4] Doring, G., Horz, M., Ortelt, J., Grupp, H., Wolz, C., Epide-
miol. Infect. 1993, 110, 427–436.
[5] Romling, U., Wingender, J., Muller, H., Tummler, B., Appl.
Environ. Microbiol. 1994, 60, 1734–1738.
[6] Barth, A. L., Pitt, T. L., J. Clin. Microbiol. 1995, 33, 37–40.
[7] Oliver, A., Canton, R., Campo, P., Baquero, F., Blazquez, J.,
Science 2000, 288, 1251–1254.
[8] Mahenthiralinga m, E., Campbell, M. E., Speert, D. P., Infect.
Immun. 1994, 62, 596–605.
[9] Mathee, K., Ciofu, O., Sternberg, C., Lindum, P. W. et al.,
Microbiology 1999, 145 ( Pt 6), 1349–1357.
[10] Kresse, A., , U., Dinesh, S. D., Larbig, K., Romling, U., Mol.
Microbiol. 2003, 47, 145–158.
[11] Romling, U., Fiedler, B., Bosshammer, J., Grothues, D. et al.,
J. Infect. Dis. 1994, 170, 1616–1621.
[12] Dinesh, S. D., Grundmann, H., Pitt, T. L., Romling, U., Clin.
Microbiol. Infect. 2003, 9, 1228–1233.
[13] Schmidt, K. D., Tummler, B., Romling, U., J. Bacteriol. 1996,
178, 85–93.
[14] Go
00rg, A., Obermaier, C., Boguth, G., Harder, A. et al.,Elec-
trophoresis 2000, 21, 1037–1053.
[15] Hancock, R. E., Nikaido, H., J. Bacteriol. 1978, 136, 381–390.
[16] Wehmhoner, D., Haussler, S., Tummler, B., Jansch, L. et al.,
J. Bacteriol. 2003, 185, 5807–5814.
[17] Ames, G. F., Prody, C., Kustu, S., J. Bacteriol. 1984, 160,
1181–1183.
[18] Neuhoff, V., Arold, N., Taube, D., Ehrhardt, W., Electropho-
resis 1988, 9, 255–262.
[19] Shevchenko, A., Wilm, M., Vorm, O., Mann, M., Anal. Chem.
1996, 68, 850–858.
[20] Barth, A. L., Pitt, T. L., J. Med. Microbiol. 1996, 45, 110–119.
[21] Thomas, S. R., Ray, A., Hodson, M. E., Pitt, T. L., Thorax 2000,
55, 795–797.
[22] Sauer, K., Camper, A. K., Ehrlich, G. D., Costerton, J. W.,
Davies, D. G., J. Bacteriol. 2002, 184, 1140–1154.
[23] Mader, U., Homuth, G., Scharf, C., Buttner, K. et al.,J. Bac-
teriol. 2002, 184, 4288–4295.
[24] Freiberg, C., Brunner, N. A., Schiffer, G., Lampe, T. et al.,J.
Biol. Chem. 2004 279, 26066–26013.
[25] Hancock, R. E., Brinkman, F. S., Annu. Rev. Microbiol. 2002,
56, 17–38.
[26] Yoshihara, E., Nakae, T., J. Biol. Chem. 1989, 264, 6297–6301.
[27] Bellido, F., Martin, N. L., Siehnel, R. J., Hancock, R. E., J.
Bacteriol. 1992, 174, 5196–5203.
[28] Yoon, S. S., Hennigan, R. F., Hilliard, G. M., Ochsner, U. A. et
al.,Dev. Cell 2002, 3, 593–603.
[29] Young, M. L., Bains, M., Bell, A., Hancock, R. E., Antimicrob.
Agents Chemother. 1992, 36, 2566–2568.
[30] Bell, A., Bains, M., Hancock, R. E., J. Bacteriol. 1991, 173,
6657–6664.
[31] Boucher, J. C., Martinez-Salazar, J., Schurr, M. J., Mudd, M.
H. et al.,J. Bacteriol. 1996, 178, 511–523.
[32] Lister, P. D., Am. J. Pharmacogenomics 2002, 2, 235–243.
[33] Trias, J., Nikaido, H., Antimicrob. Agents Chemother. 1990,
34, 52–57.
[34] Pai, H., Kim, J., Lee, J. H., Choe, K. W., Gotoh, N., Anti-
microb. Agents Chemother. 2001, 45, 480–484.
[35] O’Callaghan, R. J., Engel, L. S., Hobden, J. A., Callegan, M.
C. et al.,Invest. Ophthalmol. Vis. Sci. 1996, 37, 534–543.
[36] Engel, L. S., Hill, J. M., Caballero, A. R., Green, L. C., O’Call-
aghan, R. J., J. Biol. Chem. 1998, 273, 16792–16797.
[37] Engel, L. S., Hill, J. M., Moreau, J. M., Green, L. C. et al.,
Invest. Ophthalmol. Vis. Sci. 1998, 39, 662–665.
[38] Britigan, B. E., Hayek, M. B., Doebbeling, B. N., Fick, R. B., Jr.,
Infect. Immun. 1993, 61, 5049–5055.
[39] Nouwens, A. S., Willcox, M. D., Walsh, B. J., Cordwell, S. J.,
Proteomics 2002, 2, 1325–1346.
[40] Kamath, S., Kapatral, V., Chakrabarty, A. M., Mol. Microbiol.
1998, 30, 933–941.
[41] Raivio, T. L., Silhavy, T. J., Annu. Rev. Microbiol. 2001, 55,
591–624.
[42] Urban, A., Leipelt, M., Eggert, T., Jaeger, K. E., J. Bacteriol.
2001, 183, 587–596.
[43] Hoshino, T., Kose, K., J. Bacteriol. 1990, 172, 5540–5543.
[44] Ochsner, U. A., Vasil, M. L., Alsabbagh, E., Parvatiyar, K.,
Hassett, D. J., J. Bacteriol. 2000, 182, 4533–4544.
[45] Chakrabarty, A. M., Mol. Microbiol. 1998, 28, 875–882.
[46] Kadurugamuwa, J. L., Beveridge, T. J., J. Bacteriol. 1995,
177, 3998–4008.
[47] Folders, J., Tommassen, J., van Loon, L. C., Bitter, W., J.
Bacteriol. 2000, 182, 1257–1263.
[48] Ha, U. H., Wang, Y., Jin, S., Infect. Immun. 2003, 71, 1590–
1595.
[49] Malhotra, S., Silo-Suh , L. A., Mathee, K., Ohman, D. E., J.
Bacteriol. 2000, 182, 6999–7006.
[50] Tajima, T., Yokota, N., Matsuyama, S., Tokuda, H., FEBS Lett.
1998, 439, 51–54.
[51] Houry, W. A., Curr. Protein Pept. Sci. 2001, 2, 227–244.
[52] Moskovitz, J., Rahman, M. A., Strassman, J., Yancey, S. O. et
al.,J. Bacteriol. 1995, 177, 502–507.
[53] Wizemann, T. M., Moskovitz, J., Pearce, B. J., Cundell, D. et
al.,Proc. Natl. Acad. Sci. USA 1996, 93, 7985–7990.
2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de
... Due to the high sequence identity (39.4%) with E. coli FkpA (EcFkpA), in this study, we denote the Mip-like protein as PaFkbA. Proteomic analysis of PaFkbA revealed that it is involved in adapting the P. aeruginosa cells to the cystic fibrosis (CF) lung microenvironment [30]. However, literature related to the PaFkbA function is minimal, and its underlying molecular mechanism remains elusive. ...
... Previous studies have addressed their overlapping roles and physiological significance in bacterial adhesion, motility, biofilm formation, and virulence and implicated their crosstalk and connection during bacterial infections [17,18,52]. EcFkpA and DegP orthologs (PaFkbA and MucD) in P. aeruginosa were found to be associated with bacterial adaptation in host niches [30]. In this study, for the first time, we proved the chaperone activity of PaFkbA for MucD folding; besides, we biochemically demonstrated the direct correlation between these two chaperones. ...
Article
Full-text available
Bacterial Mip-like FK506-binding proteins (FKBPs) mostly exhibit peptidyl–prolyl-cis/trans-isomerase (PPIase) and chaperone activities. These activities are associated with various intracellular functions with diverse molecular mechanisms. Herein, we report the PA3262 gene-encoded crystal structure of the Pseudomonas aeruginosa PAO1′s Mip-like protein PaFkbA. Biochemical characterization of PaFkbA demonstrated PaFkbA’s chaperone activity for periplasmic protein MucD, a negative regulator of alginate biosynthesis. Furthermore, structural analysis of PaFkbA was used to describe the key features of PaFkbA chaperone activity. The outcomes of this analysis showed that the hinge region in the connecting helix of PaFbkA leads to the crucial conformational state transition for PaFkbA activity. Besides, the N-terminal domains participated in dimerization, and revealed its potential connection with FKBP domain and substrate binding. Mutagenesis and chaperone activity assay supported the theory that inter-domain motions are essential for PaFkbA function. These results provide biochemical and structural insights into the mechanism for FKBP’s chaperone activity and establish a plausible correlation between PaFkbA and P. aeruginosa MucD.
... Mutations in these genes have been linked to antibiotic tolerance [65,66], which may help explain persistence despite adequate antimicrobial treatment. Similarly, S. aureus isolates from persistent cystic fibrosis infections have the downregulation of many proteins involved in amino acid metabolism, fatty acid metabolism, and energy metabolism [113,131]. These studies also found a reduction in the expression of proteins in the phosphoenolpyruvate carbohydrate phosphotransferase systems, which are responsible for the transport of glucose into the bacterial cell, a necessary precursor to glycolysis. ...
Article
Full-text available
Staphylococcus aureus bacteremia continues to be associated with significant morbidity and mortality, despite improvements in diagnostics and management. Persistent infections pose a major challenge to clinicians and have been consistently shown to increase the risk of mortality and other infectious complications. S. aureus, while typically not considered an intracellular pathogen, has been proven to utilize an intracellular niche, through several phenotypes including small colony variants, as a means for survival that has been linked to chronic, persistent, and recurrent infections. This intracellular persistence allows for protection from the host immune system and leads to reduced antibiotic efficacy through a variety of mechanisms. These include antimicrobial resistance, tolerance, and/or persistence in S. aureus that contribute to persistent bacteremia. This review will discuss the challenges associated with treating these complicated infections and the various methods that S. aureus uses to persist within the intracellular space.
... Mutations in these genes have been linked to antibiotic tolerance [56,57], which may help explain persistence despite adequate antimicrobial treatment. Similarly, S. aureus isolates from persistent cystic fibrosis infections have downregulation of many proteins involved in amino acid metabolism, fatty acid metabolism, and energy metabolism [103,121]. These studies also found a reduction in expression of proteins in the phosphoenolpyruvate carbohydrate phosphotransferase systems, which are responsible for the transport of glucose into the bacterial cell, a necessary precursor to glycolysis. ...
Preprint
Full-text available
Staphylococcus aureus bacteremia continues to be associated with significant morbidity and mortality, despite improvements in diagnostics and management. Persistent infections pose a major challenge to clinicians and have been consistently shown to increase the risk of mortality and other infectious complications. S. aureus, while typically not considered an intracellular pathogen, has been proven to utilize an intracellular niche, through several phenotypes including small colony variants, as a means for survival that has been linked to chronic, persistent, and recurrent infections. This intracellular persistence allows for protection from the host immune system and leads to reduced antibiotic efficacy through a variety of mechanisms. These include antimicrobial resistance, tolerance, and/or persistence in S. aureus that contribute to persistent bacteremia. This review will discuss the challenges associated with treating these complicated infections and the various methods that S. aureus uses to persist within the intracellular space.
... Other studies have also found OprH to be significantly transcriptionally upregulated in a mouse model of acute pneumonia by qRT-PCR analysis and in a cystic fibrosis-adapted strain by proteomic analysis. 69,70 Analysis of recent transcriptomic studies with P. aeruginosa derived from human infection samples also shows a slight, but significant up regulation (2-fold) of PhoQ, which in turn regulates OprH, and a down regulation (1.5-fold) of OprL, relative to in vitro grown cultures. 71 These expression changes in OprL and in PhoQ were even more apparent in sputum samples from cystic fibrosis patients compared to soft tissue infection samples. ...
Preprint
The surge of antimicrobial resistance threatens efficacy of current antibiotics, particularly against Pseudomonas aeruginosa , a highly resistant gram-negative pathogen. The asymmetric outer membrane (OM) of P. aeruginosa combined with its array of efflux pumps provide a barrier to xenobiotic accumulation, thus making antibiotic discovery challenging. We adapted PROSPECT ¹ , a target-based, whole-cell screening strategy, to discover small molecule probes that kill P. aeruginosa mutants depleted for essential proteins localized at the OM. We identified BRD1401, a small molecule that has specific activity against a P. aeruginosa mutant depleted for the essential lipoprotein, OprL. Genetic and chemical biological studies identified that BRD1401 acts by targeting the OM β-barrel protein OprH to disrupt its interaction with LPS and increase membrane fluidity. Studies with BRD1401 also revealed an interaction between OprL and OprH, directly linking the OM with peptidoglycan. Thus, a whole-cell, multiplexed screen can identify species-specific chemical probes to reveal novel pathogen biology.
... Similarly, evidence has existed for two decades that CbpD might play a role in P. aeruginosa virulence. The CbpD gene has extensively been found in pathogenic clinical isolates (Folders et al., 2000) and is upregulated in the lungs of cystic fibrosis patients (Sriramulu et al., 2005). ...
Article
Full-text available
Pseudomonas aeruginosa secretes diverse proteins via its type 2 secretion system, including a 39 kDa chitin-binding protein, CbpD. CbpD has recently been shown to be a lytic polysaccharide monooxygenase active on chitin and to contribute substantially to virulence. To date, no structure of this virulence factor has been reported. Its first two domains are homologous to those found in the crystal structure of Vibrio cholerae GbpA, while the third domain is homologous to the NMR structure of the CBM73 domain of Cellvibrio japonicusCjLPMO10A. Here, the 3.0 Å resolution crystal structure of CbpD solved by molecular replacement is reported, which required ab initio models of each CbpD domain generated by the artificial intelligence deep-learning structure-prediction algorithm RoseTTAFold. The structure of CbpD confirms some previously reported substrate-specificity motifs among LPMOAA10s, while challenging the predictive power of others. Additionally, the structure of CbpD shows that post-translational modifications occur on the chitin-binding surface. Moreover, the structure raises interesting possibilities about how type 2 secretion-system substrates may interact with the secretion machinery and demonstrates the utility of new artificial intelligence protein structure-prediction algorithms in making challenging structural targets tractable.
... Similar studies have shown that pilli expression does not contribute significantly to virulence in the absence of flagella. The expression of this gene is however downregulated in strains from cystic fibrosis isolates, allowing the pathogen to colonize the lungs [27] . With the significant diversion from the other biofilm formation genes, we speculate that the fliC gene undergoes necessary mutations to enable the pathogenesis of different strains of P. aeruginosa and survival within different ecological niches. ...
Article
Full-text available
Pseudomonas aeruginosa is an opportunistic pathogen associated with numerous nosoco-mial infections that are difficult to treat as a result of natural resistance to various antibiotics , particularly because of biofilm formation. The purpose of this study was to determine the evolutionary relationship of P. aeruginosa biofilm formation genes and their distribution within different strains. In total, 13 genes responsible for biofilm formation by P. aeruginosa were identified and used in the study. The BLAST Ring Image Generator (BRIG) analysis was used to visualize the distribution of biofilm formation genes between different strains of P. aeruginosa. Phylogenetic analyses of the biofilm formation genes indicated a strong co-evolution of a majority of these genes, n = 10. Exceptions were the genes fliC, algD, and algU which may have been exchanged by horizontal gene transfer or evolved faster than the other genes of this functional group as they are more important in terms of a proper response of the biofilm formation to specific environmental stimuli in different habitats. The BRIG analysis revealed that the pslE, pslG and pslJ genes were all located around the same locus (2450kbps) in a variable region of the genome of strains of P. aerug-inosa giving credence to the coevolution between these set of genes. Findings of this study could inform identification of potential target genes for therapeutic agents.
... Similar studies have shown that pilli expression does not contribute significantly to virulence in the absence of flagella. The expression of this gene is however downregulated in strains from cystic fibrosis isolates, allowing the pathogen to colonize the lungs [27] . With the significant diversion from the other biofilm formation genes, we speculate that the fliC gene undergoes necessary mutations to enable the pathogenesis of different strains of P. aeruginosa and survival within different ecological niches. ...
Article
Full-text available
Pseudomonas aeruginosa is an opportunistic pathogen associated with numerous nosocomial infections that are difficult to treat as a result of natural resistance to various antibiotics, particularly because of biofilm formation. The purpose of this study was to determine the evolutionary relationship of P. aeruginosa biofilm formation genes and their distribution within different strains. In total, 13 genes responsible for biofilm formation by P. aeruginosa were identified and used in the study. The BLAST Ring Image Generator (BRIG) analysis was used to visualize the distribution of biofilm formation genes between different strains of P. aeruginosa. Phylogenetic analyses of the biofilm formation genes indicated a strong co-evolution of a majority of these genes, n=10. Exceptions were the genes fliC, algD, and algU which may have been exchanged by horizontal gene transfer or evolved faster than the other genes of this functional group as they are more important in terms of a proper response of the biofilm formation to specific environmental stimuli in different habitats. The BRIG analysis revealed that the pslE, pslG and pslJ genes were all located around the same locus (2450kbps) in a variable region of the genome of strains of P. aeruginosa giving credence to the coevolution between these set of genes. Findings of this study could inform identification of potential target genes for therapeutic agents.
... A hallmark of the expression of gene products of the tLST island is their production during exponential growth, which is substantially increased upon entry in the stationary phase of growth at environmental or body temperature (Mercer et al., 2017a) or exclusively highly produced in the late growth phase (Lee et al., 2015). Even more, in E. coli and P. aeruginosa, tLSTencoded proteins can be among the most abundant proteins (Williams, 1995;Sriramulu et al., 2005;Lee et al., 2015Lee et al., , 2018Kamal et al., 2019;Li et al., 2020). This expression pattern is distinct from homologous core genome heat shock proteins, which are overexpressed upon exposure to sublethal heat stress. ...
Article
Full-text available
The transmissible locus of stress tolerance (tLST) is found mainly in beta- and gamma-Proteobacteria and confers tolerance to elevated temperature, pressure, and chlorine. This genomic island, previously referred to as transmissible locus of protein quality control or locus of heat resistance likely originates from an environmental bacterium thriving in extreme habitats, but has been widely transmitted by lateral gene transfer. Although highly conserved, the gene content on the island is subject to evolution and gene products such as small heat shock proteins are present in several functionally distinct sequence variants. A number of these genes are xenologs of core genome genes with the gene products to widen the substrate spectrum and to be highly (complementary) expressed thus their functionality to become dominant over core genome genes. In this review, we will present current knowledge of the function of core tLST genes and discuss current knowledge on selection and counter-selection processes that favor maintenance of the tLST island, with frequent acquisition of gene products involved in cyclic di-GMP signaling, in different habitats from the environment to animals and plants, processed animal and plant products, man-made environments, and subsequently humans.
... During infection, the secretion of CbpD mediates bacterial adhesion to N-acetylglucosamine (GlcNAc)-containing mucins, glycoproteins and glycolipids of host cells [28,32,33]. Many mucoid cystic fibrosis (CF) isolates express CbpD [34,35], and its high expression has been reported in free-living planktonic cells in the Australian epidemic CF strain-2 (AES-2). CbpD is downregulated at the early stage of biofilm development [36], however, during the late phase it is secreted in abundance and possibly involved in biofilm stability and pathogenesis [37]. ...
Article
Full-text available
Pseudomonas aeruginosa is a ubiquitous bacterium, which is able to change its physiological characteristics in response to different habitats. Environmental strains are presumably less pathogenic than clinical strains and whether or not the clinical strains originate from the environment or through inter-host transmission remains poorly understood. To minimize the risk of infection, a better understanding of proteomic profiling of P. aeruginosa is necessary for elucidating the correlation between environmental and clinical strains. Based on antimicrobial susceptibility and patterns of virulence, we selected 12 clinical and environmental strains: (i) environmental, (ii) multidrug resistant (MDR) clinical and (iii) susceptible clinical strains. Whole-cell protein was extracted from each strain and subjected to two-dimensional differential gel electrophoresis (2-D DIGE) and liquid chromatography tandem mass spectrometry quadrupole time-of-flight (LC-MS QTOF). All 12 strains were clustered into 3 distinct groups based on their variance in protein expression. A total of 526 matched spots were detected and four differentially expressed protein spots (p < 0.05) were identified and all differential spots were downregulated in MDR strain J3. Upregulation of chitin binding and BON domain proteins was present in the environmental and some MDR strains, whereas the clinical strains exhibited distinct proteomic profiles with increased expression of serine protein kinase and arginine/ornithine transport ATP-binding proteins. Significant difference in expression was observed between susceptible clinical and MDR strains, as well as susceptible clinical and environmental strains. Transition from an environmental saprophyte to a clinical strain could alter its physiological characteristics to further increase its adaptation.
... Oxidative stress can further enhance production of sHsp20 GI (Lee et al. 2015). Of note, sHsp20 GI is one of the most highly expressed proteins of P. aeruginosa clone C strains under standard growth conditions (Sriramulu, Nimtz and Römling 2005). ...
Article
Full-text available
One sentence summary: Molecular epidemiology of Pseudomonas aeruginosa unraveled the occurence of few predominant clones that can thrive in various habitats with clone C one of the most abundant group of closely related strains that harbor common and strain specific entities which provide unique features such as xenologues of core genome genes involved in protein homeostasis to clone C strains. ABSTRACT The environmental species Pseudomonas aeruginosa thrives in a variety of habitats. Within the epidemic population structure of P. aeruginosa, occassionally highly successful clones that are equally capable to succeed in the environment and the human host arise. Framed by a highly conserved core genome, individual members of successful clones are characterized by a high variability in their accessory genome. The abundance of successful clones might be funded in specific features of the core genome or, although not mutually exclusive, in the variability of the accessory genome. In clone C, one of the most predominant clones, the plasmid pKLC102 and the PACGI-1 genomic island are two ubiquitous accessory genetic elements. The conserved transmissible locus of protein quality control (TLPQC) at the border of PACGI-1 is a unique horizontally transferred compository element, which codes predominantly for stress-related cargo gene products such as involved in protein homeostasis. As a hallmark, most TLPQC xenologues possess a core genome equivalent. With elevated temperature tolerance as a characteristic of clone C strains, the unique P. aeruginosa and clone C specific disaggregase ClpG is a major contributor to tolerance. As other successful clones, such as PA14, do not encode the TLPQC locus, ubiquitous denominators of success, if existing, need to be identified.
Article
Full-text available
aeruginosa to undergo the typical adaptation to the intractable mu- coid form in the CF lung. These findings indicate that gene activation in bacteria by toxic oxygen radicals, similar to that found in plants and mammalian cells, may serve as a defence mechanism for the bacteria. This suggests that mucoid conversion is a response to oxygen radical exposure and that this response is a mechanism of defence by the bacteria. This is the first report to show that PMNs and their oxygen radicals can cause this phenotypic and genotypic change which is so typical of the intractable form of P. aeruginosa in the CF lung. These findings may provide a basis for the development of anti-oxidant and anti-inflammatory therapy for the early stages of infection in CF patients.
Article
Full-text available
A method for separating the outer and inner membranes of Pseudomonas aeruginosa PAO1 in the absence of added ethylenediaminetetraacetic acid was devised. The method yields two outer membrane fractions which show the same protein pattern on sodium dodecyl sulfate-polyacrylamide gel electrophoresis, but differ substantially in their relative contents of phospholipids. One of these outer membrane fractions and the inner membrane fraction are less than 4% cross-contaminated, as judged by the content of typical inner and outer membrane markers. The outer membrane contains four major protein bands with apparent molecular weights of 37,000, 35,000, 21,000 and 17,000. Vesicles reconstituted from lipopolysaccharide and phospholipids were impermeable to all saccharides included in the vesicles during vesicle formation. When the vesicles contained outer membrane proteins, they fully retained only those saccharides of greater than 9,000 molecular weight, suggesting that the exclusion limit of the outer membrane of P. aeruginosa for saccharides is substantially larger than the figure (500 to 600 daltons) obtained for certain enteric bacteria. The advantages and potential disadvantages of having an outer membrane with a higher exclusion limit for hydrophilic substances are discussed.
Article
Full-text available
Earlier studies that used model membrane reconstitution methods have come to different conclusions regarding the exclusion limit of the outer membrane of Pseudomonas aeruginosa and whether OprF is the major channel-forming protein in the outer membrane. In this study, a 6.2-kbp SalI fragment, encoding only two cytoplasmic enzymes, alpha-galactosidase and sucrose hydrolase, and the inner membrane raffinose permease, was cloned behind the m-toluate-inducible tol promoter of vector pNM185 to create plasmid pFB71. P. aeruginosa strains harboring pFB71, when grown with inducer, produced both enzymes encoded by the insert and had acquired the ability to grow on the disaccharide melibiose and the trisaccharide raffinose. The rate of growth was dependent on the concentration and size of the saccharide and was decreased three- to fivefold by the absence of OprF, as examined by measuring the growth on melibiose and raffinose of an isogenic OprF-deficient omega insertion derivative, H636(pFB71). At high concentrations, di-, tri-, and tetrasaccharides could pass across the outer membrane to plasmolyze P. aeruginosa, as measured by light scattering and confirmed by electron microscopy. The initial rate kinetics of light-scattering changes were dependent on the size of the saccharide being used. Furthermore, the rates of change in light scattering due to raffinose and stachyose uptake across the outer membrane for strain H636 were fivefold or more lower than for its OprF-sufficient parent H103. These data are consistent with model membrane studies showing that OprF is the most predominant porin for compounds larger than disaccharides in P. aeruginosa and suggest that the exclusion limit for this porin and the outer membrane is greater than the size of a tetrasaccharide. In addition, these data confirmed the existence of other porins with a predominant function in monosaccharide uptake and a more minor function in the uptake of larger saccharides.
Article
Full-text available
The cloned oprH gene of Pseudomonas aeruginosa was mutated by inserting a 1.4-kbp fragment that encodes tetracycline resistance. This mutated gene was then incorporated into the P. aeruginosa chromosome by homologous recombination. Growth of the resultant oprH::tet mutant in Mg(2+)-deficient medium had little effect on susceptibility to polymyxin B, gentamicin, or EDTA unless the mutant was complemented by the cloned oprH gene in plasmid pGB25. In contrast, growth of the parent strain on Mg(2+)-deficient medium resulted in resistance to all three agents. These data support the hypothesis that overexpression of OprH under Mg(2+)-deficient growth conditions is conditionally required for resistance to these three agents.
Article
Full-text available
Overexpression of major outer membrane protein OprH of Pseudomonas aeruginosa as a result of mutation (in strain H181) or adaptation to low Mg2+ concentrations (in parent strain H103) is accompanied by increased resistance to polymyxin B, gentamicin, and EDTA. A 2.8-kb EcoRI fragment containing the oprH gene was subcloned into several different expression plasmids in Escherichia coli. These experiments showed that significant levels of OprH could be produced from a promoter on the EcoRI fragment; that the cloned oprH gene was not regulated by Mg2+ deficiency; that there were no differences in the expression of OprH in any construction, regardless of whether the gene from strain H103 or its OprH-overexpressing, polymyxin B-resistant derivative, strain H181, was used; and that overexpression of OprH in E. coli to the level observed in P. aeruginosa H181 did not result in a resistance phenotype. These results favored the conclusion that the mutation in strain H181 was a regulatory rather than a promoter mutation. The oprH gene was cloned behind the benzoate-inducible pm promoter in plasmid pGB25 and transferred to P. aeruginosa H103. Overexpression of OprH from the cloned gene in H103/pGB25 resulted in EDTA resistance but not polymyxin B resistance. This result suggested that another factor, possibly lipopolysaccharide, was affected by the mutation in strain H181. Consistent with this suggestion was the demonstration that mutants of strain H181 with alterations in lipopolysaccharide had reverted to wild-type polymyxin B susceptibility but had unaltered gentamicin and EDTA resistance. These data were consistent with the hypothesis that OprH replaces outer membrane-stabilizing divalent cations.
Article
Strains of Pseudomonas aeruginosa can be phenotypically classified by their mode of pathogenicity as either invasive, where the bacterium is internalised by host cells, or cytotoxic, where the host cell is killed without internalisation through the expression of cytotoxicity factors. These phenotypes are thought to depend primarily on the interactions of pseudomonal membrane and secreted proteins with host cells. We report here comparisons of outer membrane and extracellular protein‐enriched fractions from invasive (PAO1) and cytotoxic (6206) strains of P. aeruginosa separated by two‐dimensional (2‐D) gel electrophoresis. Gel image comparisons revealed the two strains express essentially identical membrane protein profiles under the conditions investigated. Membrane protein strain differences were typically the result of minor amino acid sequence variations resulting in small mass and isoelectric point shifts visible on 2‐D gels. Analysis of extracellular proteins from stationary phase growth, however, revealed significantly different protein profiles. Extracellular fractions from the invasive PAO1 strain were dominated by extracellular proteases including elastase (LasB), LasA protease and chitin‐binding protein, as well as several previously designated ‘hypothetical’ proteins. LasB appeared to be highly processed with 28 discrete mass and isoelectric point forms detected in this study. The significant number of active extracellular proteases (including LasB itself) may account for this processing. Conversely, extracellular fractions from strain 6206 consisted mainly of cellular and membrane exposed proteins including GroEL, DnaK and flagellar subunits. These are thought to result from cellular turnover during growth and the reliance on the secretory mechanisms of this strain to produce high levels of cytotoxicity factors, such as ExoU, which may be produced only upon specific interactions with host cells. These studies will aid in elucidating the differences between invasive and cytotoxic P. aeruginosa at the proteome level.
Article
The original protocol of two-dimensional electrophoresis with immobilized pH gradient (IPG-Dalt; Görg et al., Electrophoresis 1988, 9, 531—546) is updated. Merits and limits of different methods for sample solubilization, sample application (by cup-loading or in-gel rehydration) with respect to the pH interval used for IPG-isoelectric focusing are critically discussed. Guidelines for running conditions of analytical and micropreparative IPG-Dalt, using wide IPGs up to pH 12 for overview patterns, or narrow IPGs for zoom-in gels for optimum resolution and detection of minor components, are stated. Results with extended separation distances as well as automated procedures are demonstrated, and a comparison between protein detection by silver staining and fluorescent dyes is given. A brief trouble shooting guide is also included.
Article
Earlier studies that used model membrane reconstitution methods have come to different conclusions regarding the exclusion limit of the outer membrane of Pseudomonas aeruginosa and whether OprF is the major channel-forming protein in the outer membrane. In this study, a 6.2-kbp SalI fragment, encoding only two cytoplasmic enzymes, alpha-galactosidase and sucrose hydrolase, and the inner membrane raffinose permease, was cloned behind the m-toluate-inducible tol promoter of vector pNM185 to create plasmid pFB71. P. aeruginosa strains harboring pFB71, when grown with inducer, produced both enzymes encoded by the insert and had acquired the ability to grow on the disaccharide melibiose and the trisaccharide raffinose. The rate of growth was dependent on the concentration and size of the saccharide and was decreased three- to fivefold by the absence of OprF, as examined by measuring the growth on melibiose and raffinose of an isogenic OprF-deficient omega insertion derivative, H636(pFB71). At high concentrations, di-, tri-, and tetrasaccharides could pass across the outer membrane to plasmolyze P. aeruginosa, as measured by light scattering and confirmed by electron microscopy. The initial rate kinetics of light-scattering changes were dependent on the size of the saccharide being used. Furthermore, the rates of change in light scattering due to raffinose and stachyose uptake across the outer membrane for strain H636 were fivefold or more lower than for its OprF-sufficient parent H103. These data are consistent with model membrane studies showing that OprF is the most predominant porin for compounds larger than disaccharides in P. aeruginosa and suggest that the exclusion limit for this porin and the outer membrane is greater than the size of a tetrasaccharide. In addition, these data confirmed the existence of other porins with a predominant function in monosaccharide uptake and a more minor function in the uptake of larger saccharides.
Article
BACKGROUND Pseudomonas aeruginosa may undergo a phenotypic change from the wild (prototrophic) type to an auxotrophic phenotype in the course of respiratory infection in patients with cystic fibrosis. The clinical significance of this is unclear. A study was undertaken to investigate whether the presence of auxotrophs of P aeruginosa in the sputum of patients with cystic fibrosis correlated with severity of respiratory disease, and whether increased sputum concentrations of amino acids were associated with the emergence of these forms. METHODS Sixty adult patients with cystic fibrosis, colonised by P aeruginosa, were recruited and baseline clinical data including lung function were recorded. Serial sputum samples were obtained before, during, and after infective exacerbations where possible. These samples were used for routine microbiological culture, assessment of auxotrophy of P aeruginosa, measurement of amino acid content, and neutrophil elastase assay. RESULTS Auxotrophy was common in patients with cystic fibrosis and 20 (33%) had a mean percentage auxotroph count of more than 50% total cfu/ml. The mean percentage auxotroph count was inversely correlated with forced expiratory volume in one second (FEV1; τ = –0.194, p = 0.031). The median sputum amino acid concentration of the group was 12.5 mmol/l (range 0.13–40.6). The mean amino acid concentration in 33 subjects during infective exacerbations was 18.2 mmol/l (95% CI 15.1 to 21.3) compared with 12.3 mmol/l (95% CI 9.8 to 14.8) when well (p = 0.001). The amino acid content of sputum was inversely correlated with FEV1 (τ = –0.253, p = 0.005). CONCLUSIONS P aeruginosa frequently exhibits auxotrophy in patients with cystic fibrosis, particularly in those with severe underlying pulmonary disease. The sputum amino acid content of patients with cystic fibrosis is high during infective exacerbations and correlates with pulmonary disease severity.
Article
A Pseudomonas mutant deficient in protease IV has significantly reduced virulence in experimental keratitis. In the present study, the corneal toxicity of purified protease IV and its ability to augment the virulence of protease-IV-deficient bacteria were analyzed. The toxicity of purified protease IV was determined by intrastromally injecting the exoenzyme (20-200 ng) into the cornea. The effects of protease IV on the corneal virulence of the protease-IV-deficient strain, PA103-29::Tn9, were determined by injecting eyes with 1000 CFU of log phase bacteria plus either 200 ng active purified protease IV or 200 ng heat-inactivated protease IV. Changes in ocular disease, determined by slit-lamp examination, were measured at 3, 16, 22, and 27 hours after infection. Colony-forming units per cornea were quantified at 27 hours after infection. Purified protease IV at doses from 50 to 200 ng induced epithelial defects within 3 hours of injection. Injection of 20 ng active protease IV or heat-inactivated protease IV (200 ng) had no effect on ocular tissue. Corneal virulence of the protease-IV-deficient strain was augmented by intrastromal injection with purified protease IV but not with heat-inactivated protease IV (P < or = 0.0001). Neither active nor heat-inactivated protease IV altered the growth of bacteria in the cornea (6 log units; P = 0.81). The important role of protease IV in corneal virulence was demonstrated by direct toxicity and by its ability to significantly augment the virulence of protease-IV-deficient Pseudomonas.