ArticlePDF Available

Expression of Surfactant Protein D in the Human Gastric Mucosa and during Helicobacter pylori Infection

American Society for Microbiology
Infection and Immunity
Authors:
  • Revolo Biotherapeutics Ltd. Gaithersburg USA

Abstract and Figures

Helicobacter pylori establishes persistent infection of gastric mucosa with diverse clinical outcomes. The innate immune molecule surfactant protein D (SP-D) binds selectively to microorganisms, inducing aggregation and phagocytosis. In this study, we demonstrated the expression of SP-D in gastric mucosa by reverse transcription-PCR and immuohistochemical analysis. SP-D is present at the luminal surface and within the gastric pits, with maximal expression at the surface. Levels of expression are significantly increased in H. pylori-associated gastritis compared to those in the normal mucosa. Immunofluorescence microscopy was used to demonstrate binding and agglutination of H. pylori by SP-D in a lectin-specific manner. These activities resulted in a 50% reduction in the motility of H. pylori, as judged on the basis of curvilinear velocity measured by using a Hobson BacTracker. Lipopolysaccharides extracted from three H. pylori strains were shown to bind SP-D in a concentration-dependent manner, and there was marked variation in the avidity of binding among the strains. SP-D may therefore play a significant role in the innate immune response to H. pylori infection.
This content is subject to copyright. Terms and conditions apply.
INFECTION AND IMMUNITY, Mar. 2002, p. 1481–1487 Vol. 70, No. 3
0019-9567/02/$04.000 DOI: 10.1128/IAI.70.3.1481–1487.2002
Copyright © 2002, American Society for Microbiology. All Rights Reserved.
Expression of Surfactant Protein D in the Human Gastric Mucosa and
during Helicobacter pylori Infection
Emma Murray,
1
Wafa Khamri,
2
Marjorie M. Walker,
2
Paul Eggleton,
1
Anthony P. Moran,
3
John A. Ferris,
3
Susanne Knapp,
3
Q. Najma Karim,
3
Mulegata Worku,
3
Peter Strong,
1
Kenneth B. M. Reid,
1
and Mark R. Thursz
3
*
MRC Immunochemistry Unit, Oxford,
1
and Faculty of Medicine, Imperial College of Science, Technology and
Medicine, St. Mary’s Hospital Campus, London,
2
United Kingdom, and Department of Microbiology,
National University of Ireland, Galway, Ireland
3
Received 18 January 2001/Returned for modification 11 April 2001/Accepted 15 October 2001
Helicobacter pylori establishes persistent infection of gastric mucosa with diverse clinical outcomes. The
innate immune molecule surfactant protein D (SP-D) binds selectively to microorganisms, inducing aggrega-
tion and phagocytosis. In this study, we demonstrated the expression of SP-D in gastric mucosa by reverse
transcription-PCR and immuohistochemical analysis. SP-D is present at the luminal surface and within the
gastric pits, with maximal expression at the surface. Levels of expression are significantly increased in H.
pylori-associated gastritis compared to those in the normal mucosa. Immunofluorescence microscopy was used
to demonstrate binding and agglutination of H. pylori by SP-D in a lectin-specific manner. These activities
resulted in a 50% reduction in the motility of H. pylori, as judged on the basis of curvilinear velocity measured
by using a Hobson BacTracker. Lipopolysaccharides extracted from three H. pylori strains were shown to bind
SP-D in a concentration-dependent manner, and there was marked variation in the avidity of binding among
the strains. SP-D may therefore play a significant role in the innate immune response to H. pylori infection.
Helicobacter pylori is a gram-negative bacterium which col-
onizes gastric mucosa. It is one of the most common patho-
gens, with a prevalence of up to 90% in developing countries
(17). Once infection is established, gastric mucosal inflamma-
tion develops, and although infection persists for life, only 30%
of those infected become symptomatic. The outcome of infec-
tion is diverse and includes duodenal ulcer, gastric ulcer, and
gastric malignancy—both carcinoma and lymphoma. Such het-
erogeneous consequences are dependent on the time span and
topography of mucosal inflammation (28). The route of infec-
tion is not proven, but the most likely event is direct ingestion
of gastric contents. Gastric mucosa is markedly adverse to
bacterial colonization, as the physical and chemical barriers
encountered (mucus, enzymes, and acid) inhibit colonization
by common bacteria.
H.pylori-related gastritis is characterized by both lympho-
cytic and neutrophil infiltrates. In addition, there is a strong
humoral immune response with specific antibodies of both
immunoglobulin G (IgG) and IgA classes. Despite evidence of
an immune response, H.pylori infection is frequently persis-
tent, suggesting that the organism may evade conventional
innate and adaptive immune responses. A number of factors
appear to contribute to the virulence of H.pylori, including the
possession of flagella, which confer motility (6).
Surfactant protein D (SP-D) is a collagenous glycoprotein
which contains trimeric arrays of C-type (calcium-dependent)
lectin domains and which belongs to a family of proteins im-
plicated in innate immunity, termed the collectins (5, 25, 27).
The protein and molecular structures for SP-D are well char-
acterized, and the gene has been localized to human chromo-
some 10q22.2-23.1 (8, 13, 18). SP-D has been shown to recog-
nize and bind selectively to the surfaces of viruses, bacteria,
protozoa, and fungi and is currently being considered for use as
a therapeutic agent for the treatment of both bacterial infec-
tions and allergies in humans (4, 19, 23, 24, 26). SP-D is
believed to bind directly to the lipopolysaccharide (LPS) on
the surface of gram-negative bacteria via the carbohydrate
recognition domain (16). This process may result in the aggre-
gation of microorganisms followed by enhanced phagocytosis
by neutrophils and macrophages (11). Although phagocytosis
of microorganisms appears to be enhanced by interaction with
SP-D or SP-A, it is unclear whether such interactions promote
further immunological or inflammatory responses (2, 9, 11, 30).
In humans, SP-D has been shown to be synthesized in the
lungs, specifically alveolar type II and Clara cells, and to be
present in the fluid phase of the airways. SP-D has also been
detected at other sites, including tears, amniotic fluid, and fetal
membranes. In the rat, SP-D has been found in mucus-secret-
ing cells of the gastric mucosa but not in the duodenum or
remaining intestine, and a speculative role in mucus barrier
assembly and possibly host defense has been proposed (7).
In order to establish whether SP-D may play a role in de-
fense against H.pylori infection or in the pathogenesis of es-
tablished H.pylori infection, the aim of this study was to de-
termine whether SP-D is present in the human gastric mucosa
and whether SP-D interacts with H.pylori. Further studies were
performed to determine if the motility of H.pylori is impaired
by SP-D. In addition, SP-D binding studies were performed to
establish whether SP-D binds to LPS from bacterial cell walls.
* Corresponding author. Mailing address: Faculty of Medicine, Im-
perial College of Science, Technology and Medicine, St. Mary’s Hos-
pital Campus, Norfolk Place, London W2 1NY, United Kingdom.
Phone: 44 207 594 3851. Fax: 44 207 706 9161. E-mail: m.thursz
@ic.ac.uk.
1481
MATERIALS AND METHODS
Preparation of SP-D and antisera to SP-D. SP-D was puried from lung lavage
uid from patients with alveolar proteinosis by the method of Strong et al. (29).
Briey, lavage uid was centrifuged at 10,000 gfor 40 min. The supernatant
was applied to a maltosyl-agarose column, and bound SP-D was specically
eluted by using MnCl
2
. SP-D was further puried by gel ltration on Superose-6.
A recombinant preparation of human SP-D head and neck regions expressed
in Escherichia coli was used to raise polyclonal antisera in rabbits. The IgG
fraction was puried from rabbit serum by sodium sulfate precipitation followed
by protein A chromatography as previously described (20). The specicity of the
antisera for native human SP-D was conrmed by an enzyme-linked immunosor-
bent assay (ELISA) and Western blotting.
Detection of SP-D mRNA. mRNA for SP-D was detected by reverse transcrip-
tion (RT)-PCR. Twenty-eight patients with nonulcer dyspepsia were selected at
the time of endoscopy and gave informed consent. Thirteen patients had normal
histological ndings, and 7 had H.pylori-associated antral gastritis. RNA was
extracted from three gastric antral biopsy specimens taken from each patient.
The biopsy specimens were snap-frozen in liquid nitrogen and stored at 70°C
until processed. When required, biopsy specimens were homogenized in saline
and RNA was isolated by using a total RNA isolation kit according to the
manufacturers protocol (Ambion, AMS Biotechnology, Europe Ltd.). cDNA
synthesis and PCR amplication were carried out as a single step by using a
one-step RT-PCR kit according the manufacturers protocol (ABgene, Surrey,
United Kingdom). Primers from exons 3 and 4 of the SP-D gene were designed
to give product sizes of 475 bp from genomic DNA and 155 bp from cDNA. The
primer sequences were as follows: exon 3, 5-GAACATAGGACCTCAGGGC
A-3, and exon 4, 5-TGTGTTTCCAGGGACTCCAC-3. Parallel RT-PCRs
were performed with primers for the human -actin gene to serve as a reference.
First-strand synthesis was performed with a single cycle at 47°C for 30 min. The
reverse transcriptase was then inactivated by denaturation at 94°C for 2 min.
PCR amplication was performed with 43 cycles of annealing at 63°C for 30 s,
extension at 72°C for 1 min, and denaturation at 94°C for 30 s. There was a nal
extension step at 72°C for 5 min. Negative and positive control amplications
were performed in every experiment. RT-PCR products for SP-D and -actin
were run on a 2% (wt/vol) agarose gel, stained with ethidium bromide, and
visualized under UV light. Digital images were obtained, and densitometry was
performed by using a Syngene gel documentation system (Synoptics Ltd., Cam-
bridge, United Kingdom) and Genetools v3.0 software (Synoptics). Semiquanti-
tative measurement of SP-D mRNA was achieved by comparing the area under
the peak of the SP-D band with the area under the peak of the -actin band and
expressing the result as a simple ratio.
Immunohistochemical analysis. Immunohistochemical analysis was per-
formed with the anti-SP-D polyclonal antisera described above and with mono-
clonal antibody 245.2 (a gift from U. Holmskov, Odense, Denmark). The spec-
icity of this antibody has been conrmed by Western blotting and ELISA
analyses (21).
Formalin-xed, parafn-embedded gastric biopsy specimens were randomly
selected from the pathology archives for immunocytochemical staining, and 24
specimens were classied according to histological appearances. Of the 24 cases
selected, 11 were H.pylori-associated antral gastritis, 4 were H.pylori-negative
chemical antral gastritis, and 9 were H.pylori-negative normal antral mucosa.
Two 5-mm sections were cut from each biopsy specimen; one of these was
stained with the rabbit anti-SP-D polyclonal antisera, and the other was stained
with the SP-D-specic monoclonal antibody. Staining was demonstrated by using
a routine avidin-biotin-horseradish peroxidase system. Negative controls were
stained with the monoclonal antibody which had been preincubated with puried
SP-D, with an irrelevant antibody, and with preimmune sera from the same
rabbits.
SP-D expression was evaluated by a quantitative method with a standard grid
(10). Positive immunostaining was assessed by site in the gastric mucosa (lumen,
foveola, and pit) by counting cells expressing SP-D in a given area delineated by
the grid.
Binding and agglutination. H.pylori strain J178 (cagA positive) was cultured in
brain heart infusion broth at 37°C under microaerophilic conditions to a con-
centration of 10
8
to 10
9
bacteria ml
1
. The organisms were centrifuged at 3,000
gand resuspended in Tris-buffered saline (pH 7.4) (TBS).
Binding of SP-D to H.pylori was demonstrated by immunouorescence. H.
pylori was incubated with SP-D (10 g/ml) in TBS containing 10 mM CaCl
2
or 10
mM EDTA to demonstrate calcium-dependent binding. Controls were incubated
in calcium buffer alone. To assess lectin specicity, 100 mM maltose was added
to bacteria treated with SP-D and 10 mM CaCl
2
. Finally, SP-D binding to
bacteria was detected by probing with biotinylated anti-human SP-D antisera
(1:200 dilution of a 1-mg/ml IgG stock solution) followed by streptavidin-uo-
rescein isothiocyanate (1:200; Sigma Chemical Co., Dorset, United Kingdom).
Agglutination was demonstrated by direct observation of the bacteria with phase-
contrast microscopy. H.pylori organisms resuspended in TBS containing 10 mM
CaCl
2
at 10
8
organisms/ml were incubated for 30 min with buffer alone, with 10
g of SP-D/ml, with 10 g of SP-D/ml and 10 mM EDTA, or with 10 gof
SP-D/ml and 100 mM maltose. Phase-contrast microscopy and uorescence
microscopy were performed by using a Zeiss Axioskop transmitted-light micro-
scope. H.pylori was photographed with a Zeiss MC 100 camera (nal magni-
cation, 400) by using TX 400-ASA black and white lm for phase contrast and
Provia 1600 color slide lm for uorescence.
Quantitative estimates of SP-D-mediated agglutination were obtained for clin-
ical isolates of H.pylori. Bacteria were placed in optical cuvettes suspended in
TBS plus 10 mM CaCl
2
. Two aliquots of each isolate were adjusted to an optical
density at 700 nm (OD
700
) of 0.7, and SP-D at a nal concentration of 2.5 g/ml
was added to one aliquot. OD
700
values were measured at 15-min intervals up to
60 min and again at 90 min. Agglutination was expressed as the difference in
OD
700
units between control and SP-D-containing aliquots at 60 min.
Inhibition of motility. Assessment of H.pylori motility was made by using a
Hobson BacTracker as previously described (15). The technique gives a quanti-
tative measure of motility by using an image-processing computer combined with
a phase-contrast microscope and video equipment. Motility was assessed in the
presence and absence of 5 g of SP-D/ml in calcium buffer with and without the
addition of 100 mM maltose. Motility is expressed as curvilinear velocity, which
is the length of a track (total path length) divided by the time taken to travel it
in micrometers per second.
LPS binding studies. Binding of SP-D to LPS was demonstrated in a compet-
itive inhibition ELISA. LPS was extracted from three strains of H.pylori: clinical
strain J178, cagA-positive reference strain 007, and mouse-adapted strain SS1.
After pretreatment of bacterial biomass with pronase (Calbiochem, Los Angeles,
Calif.), LPS was extracted by the hot phenol-water technique. The resulting
crude LPS preparations, recovered from the water phase of extracts, were puri-
ed by treatment with RNase, DNase II, and proteinase K (Sigma) and by
ultracentrifugation as described previously (22).
Mannan (10 g/ml) in carbonate buffer (pH 9.6) was used to coat a Maxisorb
Immunoplate (Life Technologies, Paisley, United Kingdom) overnight at 4°C.
The plate was washed three times in TBS and then blocked with TBS plus 3%
(wt/vol) bovine serum albumin for1hat37°C. Between each step, the plate was
washed three times in TBS plus 0.05% Tween 20 (TBST). LPS (200 to 1 g/ml)
from H.pylori (J178, 007, or SS1) was incubated with a xed concentration of
SP-D (1 g/ml) for1hatroom temperature before addition to the mannan-
coated, blocked plate. The LPSSP-D mixtures and SP-D alone (1,000 to 0
ng/ml) were incubated on the plate for3hat37°C. An aliquot (100 l) of
biotinylated rabbit anti-SP-D polyclonal antisera (1:1,000 dilution of a 1-mg/ml
stock) in TBST was added to each well and incubated for2hat37°C. After
washing, 100 l of ExtrAvidin-peroxidase conjugate (Sigma) diluted 1:10,000 in
TBST was added and incubated for 30 min at 37°C. The plate was developed with
tetramethylbenzidine substrate (Bio-Rad, Hemel Hempstead, United Kingdom),
the reaction was stopped with 100 lof1NH
2
SO
4
, and readings were carried
out at 450 nm (Titertek Multiscan PLUS MKII). The resulting graphs (see Fig.
5) were generated by using Prism software (GraphPad Software Inc., San Diego,
Calif.), and the data were used to calculate the concentration of LPS required for
50% inhibition of SP-D binding.
RESULTS
Detection of SP-D mRNA. SP-D expression at the mRNA
level was assessed in gastric mucosa by use of RT-PCR. mRNA
for SP-D could be detected in 12 (100%) of 12 gastric tissue
samples where H.pylori infection was present and in 10
(62.5%) of 16 samples from individuals who did not have the
infection (Fig. 1A). The levels of gastric SP-D mRNA expres-
sion in individuals with H.pylori infection were signicantly
higher than those in individuals without the infection. The
median SP-D/-actin ratios were 0.34 (range, 0.01 to 0.79) in
individuals with H.pylori infection and 0.08 (range, 0 to 0.45)
in individuals without the infection (Fig. 1B) (P0.004)
(Mann-Whitney U test).
Immunohistochemical analysis. In order to assess, at the
cellular level, the site of SP-D expression, immunocytochemi-
1482 MURRAY ET AL. INFECT.IMMUN.
cal analysis was performed on endoscopic gastric mucosal bi-
opsy specimens taken routinely from 24 patients with dyspep-
sia. Immunohistochemical analysis was performed with rabbit
polyclonal antisera and the SP-D-specic monoclonal anti-
body. Negative controls, performed with the monoclonal anti-
body preabsorbed with SP-D, an irrelevant antibody, and pre-
immune sera from the same rabbits, revealed no
immunohistochemical staining. Staining with the anti-SP-D
monoclonal antibody gave results similar to those seen with the
polyclonal antisera.
The staining demonstrated SP-D protein expression in epi-
thelial cells, and SP-D coating of H.pylori was observed in situ
FIG. 1. SP-D mRNA is expressed in gastric mucosa. (A) RT-PCR results for RNA extracted from gastric mucosal tissue samples taken from
patients with dyspepsia. Bands specic for actin and SP-D are seen at 325 and 155 bp, respectively. Helicobacterindicates the presence ()or
absence ()ofH.pylori infection, as determined by histological examination; s, standards. (B) Level of SP-D mRNA expression, assessed by
densitometry of the RT-PCR bands and presented as plots (mean and standard error of the mean) of the SP-D/-actin ratio. H.pylori-positive and
H.pylori-negative gastric samples were compared.
VOL. 70, 2002 SP-D AND H.PYLORI 1483
(Fig. 2). The level of SP-D expression was observed to vary at
different epithelial levels in the mucosa, with maximal intensity
in the luminal mucosa compared to the foveolar region and the
gastric pits. No staining was observed in sections stained with
preimmune sera.
Semiquantitative analysis of cellular expression at different
levels in the mucosa was performed by comparing the intensi-
ties of expression in patients with H.pylori-associated gastritis,
reactive (chemical) gastritis, and normal histological ndings.
SP-D was expressed at equal intensities in the basal region of
epithelial cells at each of the anatomical sites examined in all
three patient groups. However, in H.pylori-associated gastritis,
there was a signicant increase in cell surface expression at all
sites (P0.05) (Mann-Whitney U test) (Fig. 3). Increased cell
surface expression was not seen in reactive gastritis.
Binding and agglutination. Binding of SP-D to heat-xed H.
pylori was clearly seen with immunouorescence staining. The
calcium dependence and lectin-specic nature of this binding
FIG. 2. SP-D expression demonstrated by immunohistochemical analysis. Immunohistochemical analysis was performed with an anti-SP-D
monoclonal antibody and antral gastric mucosa by using a standard avidin-biotin technique as described in Materials and Methods. (A) Negative
control after the antibody was preabsorbed with native SP-D. (B) Low-level expression of SP-D (as judged by the brown staining) in the basal
regions of epithelial cells on the luminal surface of the mucosa. (C) Luminal epithelial cells of H.pylori-positive antral gastric mucosa, with strong
SP-D expression on the surface and in the basal regions of the cells. (D) Coating of H.pylori with SP-D (arrowheads). Original magnications,
1,000 (A, B, and C) and 3,000 (D).
1484 MURRAY ET AL. INFECT.IMMUN.
were conrmed by the lack of binding in the absence of cal-
cium and competitive inhibition by maltose (Fig. 4A to D).
SP-D caused agglutination of live H.pylori into large clumps
that was inhibited by both EDTA and maltose (Fig. 4E to H).
Agglutination of clinical isolates of H.pylori was measured
as the difference in the OD
700
after 60 min of incubation with
2.5 g of SP-D/ml (Fig. 5). There was a wide range of values,
from 0.100 to 0.475; the mean was 0.218. After 60 min of
incubation, the difference in the OD
700
reached a plateau in
some isolates.
Motility. The motility studies demonstrated that SP-D re-
duced the curvilinear velocity of H.pylori by approximately
50%. H.pylori in TBS plus 10 mM CaCl
2
had a curvilinear
velocity of 17.1 m/s; this value fell to 9.65 m/s in the pres-
ence of 5 g of SP-D/ml. H.pylori motility was not reduced in
the presence of maltose or in the absence of calcium. Direct
observation of binding showed that the bacteria formed encir-
cling clusters. Free movement was impeded by agglutination
but was not halted.
LPS binding. SP-D bound to mannan in a concentration-
dependent fashion with a linear relationship to the absorbance
at 450 nm at between 50 and 1,000 ng/ml. LPS inhibited SP-D
binding in a dose-dependent manner. There was clear variation
in the ability of each LPS preparation to inhibit SP-D binding,
indicating variability in the avidity of SP-D for LPS binding.
The 50% inhibitory concentration for H.pylori J178 LPS was
5.3 g/ml, that for SS1 LPS was 13.4 g/ml, and that for 007
LPS was 91.5 g/ml. The 50% inhibitory concentration ob-
tained with E.coli LPS was 73.7 g/ml (Fig. 6).
DISCUSSION
This study has demonstrated the expression of SP-D in hu-
man gastric antral mucosa at both the mRNA and the protein
levels. Expression at this site suggests that SP-D may play a
role either in the structure of the gastric mucus layer or in host
defense. Low-level SP-D expression in the stomach was re-
cently reported by Madsen et al. (21). However, this study did
not take into account the H.pylori status or the anatomical site
of tissue collection. In contrast, our data suggest that the gas-
tric epithelium is capable of high levels of SP-D expression in
the context of H.pylori infection.
Recent studies with SP-D gene knockout mice have indi-
cated that SP-D is not an essential component of the mucus
layer in respiratory epithelium, as these mice develop without
signicant respiratory pathology (3). However, these experi-
ments do not completely rule out a structural role for SP-D in
the gastric mucus layer. SP-D is related to the collectin family
of proteins, which are components of the innate immune sys-
tem. Evidence suggests that the major role of SP-D is the
recognition of foreign carbohydrate structures expressed on
the cell walls of microorganisms (26). SP-D may therefore be
one of the rst lines of defense in the gastric mucosa.
H. pylori is a common human pathogen which is capable of
establishing a chronic infection of gastric mucosa. The binding,
agglutination, and motility studies presented here indicate that
SP-D recognizes and interacts with H.pylori, causing aggluti-
nation of the organism and inhibition of bacterial motility.
Agglutination is thought to facilitate phagocytosis by polymor-
FIG. 3. Site-specic expression of SP-D in gastric mucosa. (Left) Schematic representation of the sites used for immunocytochemical assess-
ment. (Right) Semiquantitative comparison of SP-D expression in H.pylori-associated gastritis, reux-type gastritis, and samples with normal
histological ndings at the epithelial surface (top), the foveolar region (middle), and the gastric pit region (bottom).
VOL. 70, 2002 SP-D AND H.PYLORI 1485
phonuclear cells or macrophages (9). While some reports sug-
gest that there may be a specic SP-D receptor on neutrophils
and macrophages, this idea is controversial, and other studies
suggest that SP-D does not act as an opsonin (1, 12).
Motility is thought to be an important virulence determinant
in H.pylori, allowing the organism to migrate to suitable niches
within the gastric mucosa; inhibition of motility prevents col-
onization (6, 31). In these experiments, motility was signi-
cantly inhibited even in the absence of agglutination.
An attempt was made to measure the levels of SP-D in
gastric juice and to correlate these measurements with the
histological and immunocytochemical ndings. Although it was
possible to detect low concentrations of SP-D in gastric juice
(results not shown), SP-D appeared to be rapidly digested by
proteases. It has been suggested that SP-D secreted from sal-
ivary glands may also interact with H.pylori. However, our
observations suggest that SP-D from salivary secretions would
be rapidly digested in the gastric lumen, whereas SP-D ex-
pressed by gastric epithelial cells would be effectively protected
under the mucus-bicarbonate barrier.
SP-D is believed to bind to LPS, which is the major compo-
nent of the cell wall in gram-negative bacteria. LPS was there-
fore considered to be the most likely target for SP-D binding to
H.pylori, and the data from the competitive inhibition assays
appear to conrm this notion. Our data do not rule out the
possibility that other components of the H.pylori cell wall are
also involved in SP-D binding. Signicant interstrain variations
in the terminal O-chain structures of H.pylori LPS and in the
core oligosaccharide have been described (14). Wide variations
in agglutination measurements in clinical isolates and varia-
tions in binding afnities between LPSs extracted from differ-
ent H.pylori strains and SP-D were observed, potentially re-
ecting these structural differences. In the inhibition studies,
LPS of H.pylori J178 was the most inhibitory, with H.pylori SS1
LPS being next and H.pylori 007 LPS being the least inhibitory.
The structure of J178 LPS is presently under investigation, and
serological analysis indicates that LPS of H.pylori SS1 ex-
presses Le
y
(A. P. Moran et al., unpublished results); however,
the O chain of H.pylori 007 LPS consists of a polymeric Le
x
chain, lacking terminal Le
y
, thus indicating variations in the
FIG. 4. Binding of SP-D to H.pylori and agglutination. (A) Immu-
nouorescence detection of SP-D binding to H.pylori by biotin-labeled
anti-human SP-D antibody (1:200 dilution of a 1-mg/ml stock).
(B) Phase-contrast image of the same bacteria as in panel A. (C) Effect
of 100 M maltose on SP-D binding to H.pylori, as detected by
immunouorescence. (D) Phase-contrast image of the same bacteria
as in panel C. (E to H) Agglutination of H.pylori after incubation with
10 g of SP-D/ml and 10 mM CaCl
2
buffer (E), 10 mM EDTA (F), or
100 M maltose (G) or without treatment (H). Immunouorescence
microscopy and phase-contrast microscopy were performed with a
Zeiss Axioskop transmitted-light microscope with a Zeiss MC 100
camera. Final magnications, 400.
FIG. 5. Quantitative assessment of SP-D-mediated agglutination of
H.pylori clinical isolates. Agglutination was measured as the difference
in OD
700
between isolates incubated with or without SP-D at 2.5
g/ml.
FIG. 6. Inhibition of SP-D binding to mannan by H.pylori LPS.
Dose-dependent competitive inhibition of SP-Dmannan binding by
LPSs prepared from three H.pylori strains (J178, 007, and SS1) and
one E.coli strain (O128:B8) is shown. Curve tting was performed by
using GraphPad Prism software.
1486 MURRAY ET AL. INFECT.IMMUN.
O-chain structures of these strains. However, these later stud-
ies also have indicated variations in the core oligosaccharide of
these strains. Therefore, at present, no clear conclusions can
be made as to the structures within H.pylori LPS involved in
SP-D binding.
The expression of SP-D in human gastric mucosa and a
functional interaction between SP-D and H.pylori have been
demonstrated. It therefore remains to be explained how per-
sistent infection with this organism is established and how the
organism can evade this component of innate immunity.
REFERENCES
1. Benne, C. A., B. Benaissa Trouw, J. A. Van Strijp, C. A. Kraaijeveld, and
J. F. Van Iwaarden. 1997. Surfactant protein A, but not surfactant protein D,
is an opsonin for inuenza A virus phagocytosis by rat alveolar macrophages.
Eur. J. Immunol. 27:886890.
2. Borron, P. J., E. C. Crouch, J. F. Lewis, J. R. Wright, F. Possmayer, and L. J.
Fraher. 1998. Recombinant rat surfactant-associated protein D inhibits hu-
man T lymphocyte proliferation and IL-2 production. J. Immunol. 161:4599
4603.
3. Botas, C., F. Poulain, J. Akiyama, C. Brown, L. Allen, J. Goerke, J. Clements,
E. Carlson, A. M. Gillespie, C. Epstein, and S. Hawgood. 1998. Altered
surfactant homeostasis and alveolar type II cell morphology in mice lacking
surfactant protein D. Proc. Natl. Acad. Sci. USA 95:1186911874.
4. Brown Augsburger, P., K. Hartshorn, D. Chang, K. Rust, C. Fliszar, H. G.
Welgus, and E. C. Crouch. 1996. Site-directed mutagenesis of Cys-15 and
Cys-20 of pulmonary surfactant protein D. Expression of a trimeric protein
with altered anti-viral properties. J. Biol. Chem. 271:1372413730.
5. Crouch, E., A. Persson, D. Chang, and J. Heuser. 1994. Molecular structure
of pulmonary surfactant protein D (SP-D). J. Biol. Chem. 269:1731117319.
6. Eaton, K. A., D. R. Morgan, and S. Krakowka. 1992. Motility as a factor in
the colonisation of gnotobiotic piglets by Helicobacter pylori. J. Med. Mi-
crobiol. 37:123127.
7. Fisher, J. H., and R. Mason. 1995. Expression of pulmonary surfactant
protein D in rat gastric mucosa. Am. J. Respir. Cell Mol. Biol. 12:1318.
8. Hakansson, K., N. K. Lim, H. J. Hoppe, and K. B. Reid. 1999. Crystal
structure of the trimeric alpha-helical coiled-coil and the three lectin do-
mains of human lung surfactant protein D. Structure 7:255264.
9. Hartshorn, K. L., E. Crouch, M. R. White, M. L. Colamussi, A. Kakkanatt,
B. Tauber, V. Shepherd, and K. N. Sastry. 1998. Pulmonary surfactant
proteins A and D enhance neutrophil uptake of bacteria. Am. J. Physiol.
274:L958-L969.
10. Helander, H. F. 1985. Quantitative morphological methods in intestinal
research. Scand. J. Gastroenterol. Suppl. 112:15.
11. Hickling, T. P., R. B. Sim, and R. Malhotra. 1998. Induction of TNF-alpha
release from human buffy coat cells by Pseudomonas aeruginosa is reduced
by lung surfactant protein A. FEBS Lett. 437:6569.
12. Holmskov, U., J. Mollenhauer, J. Madsen, L. Vitved, J. Gronlund, I. Tornoe,
A. Kliem, K. B. Reid, A. Poustka, and K. Skjodt. 1999. Cloning of gp-340, a
putative opsonin receptor for lung surfactant protein D. Proc. Natl. Acad.
Sci. USA 96:1079410799.
13. Hoover, R. R., and J. Floros. 1998. Organization of the human SP-A and
SP-D loci at 10q22-q23. Physical and radiation hybrid mapping reveal gene
order and orientation physical and radiation hybrid mapping reveal gene
order and orientation. Am. J. Respir. Cell Mol. Biol. 18:353362.
14. Hynes, S. O., S. Hirmo, T. Wadstrom, and A. P. Moran. 1999. Differentiation
of Helicobacter pylori isolates based on lectin binding of cell extracts in an
agglutination assay. J. Clin. Microbiol. 37:19941998.
15. Karim, Q. N., R. P. Logan, J. Puels, A. Karnholz, and M. L. Worku. 1998.
Measurement of motility of Helicobacter pylori, Campylobacter jejuni, and
Escherichia coli by real time computer tracking by using the Hobson Bac-
Tracker. J. Clin. Pathol. 51:623628.
16. Kuan, S. F., K. Rust, and E. Crouch. 1992. Interactions of surfactant protein
D with bacterial lipopolysaccharides. Surfactant protein D is an Escherichia
coli-binding protein in bronchoalveolar lavage. J. Clin. Investig. 90:97106.
17. Logan, R. P. H., and A. M. Hirschl. 1996. Epidemiology of Helicobacter pylori
infection. Curr. Opin. Gastroenterol. 12:15.
18. Lu, J., A. C. Willis, and K. B. Reid. 1992. Purication, characterization and
cDNA cloning of human lung surfactant protein D. Biochem. J. 284:795802.
19. Madan, T., P. Eggleton, U. Kishore, P. Strong, S. S. Aggrawal, P. U. Sarma,
and K. B. Reid. 1997. Binding of pulmonary surfactant proteins A and D to
Aspergillus fumigatus conidia enhances phagocytosis and killing by human
neutrophils and alveolar macrophages. Infect. Immun. 65:31713179.
20. Madan, T., U. Kishore, A. Shah, P. Eggleton, P. Strong, J. Y. Wang, S. S.
Aggrawal, P. U. Sarma, and K. B. Reid. 1997. Lung surfactant proteins A and
D can inhibit specic IgE binding to the allergens of Aspergillus fumigatus
and block allergen-induced histamine release from human basophils. Clin.
Exp. Immunol. 110:241249.
21. Madsen, J., A. Kliem, I. Tornoe, K. Skjodt, C. Koch, and U. Holmskov. 2000.
Localization of lung surfactant protein D on mucosal surfaces in human
tissues. J. Immunol. 164:58665870.
22. Moran, A. P., I. M. Helander, and T. U. Kosunen. 1992. Compositional
analysis of Helicobacter pylori rough-form lipopolysaccharides. J. Bacteriol.
174:13701377.
23. Murray, E., and P. Eggleton. 2000. Protective roles of surfactant protein A
and D in infection and allergy. Mod. Asp. Immunobiol. 1:27.
24. ORiordan, D. M., J. E. Standing, K. Y. Kwon, D. Chang, E. C. Crouch, and
A. H. Limper. 1995. Surfactant protein D interacts with Pneumocystis carinii
and mediates organism adherence to alveolar macrophages. J. Clin. Investig.
95:26992710.
25. Persson, A., D. Chang, and E. Crouch. 1990. Surfactant protein D is a
divalent cation-dependent carbohydrate-binding protein. J. Biol. Chem. 265:
57555760.
26. Reid, K. B. 1998. Interactions of surfactant protein D with pathogens, aller-
gens and phagocytes. Biochim. Biophys. Acta 1408:290295.
27. Rust, K., L. Grosso, V. Zhang, D. Chang, A. Persson, W. Longmore, G. Z.
Cai, and E. Crouch. 1991. Human surfactant protein D: SP-D contains a
C-type lectin carbohydrate recognition domain. Arch. Biochem. Biophys.
290:116126.
28. Sipponen, P., and M. Stolte. 1997. Clinical impact of routine biopsies of the
gastric antrum and body. Endoscopy 29:671678.
29. Strong, P., U. Kishore, C. Morgan, A. Lopez Bernal, M. Singh, and K. B.
Reid. 1998. A novel method of purifying lung surfactant proteins A and D
from the lung lavage of alveolar proteinosis patients and from pooled am-
niotic uid. J. Immunol. Methods 220:139149.
30. Tino, M. J., and J. R. Wright. 1999. Surfactant proteins A and D specically
stimulate directed actin-based responses in alveolar macrophages. Am. J.
Physiol. 276:L164-L174.
31. Worku, M. L., R. L. Sidebotham, M. M. Walker, T. Keshavarz, and Q. N.
Karim. 1999. The relationship between Helicobacter pylori motility, mor-
phology and phase of growth: implications for gastric colonization and pa-
thology. Microbiology 145:28032811.
Editor: B. B. Finlay
VOL. 70, 2002 SP-D AND H.PYLORI 1487
... Its interaction with H. pylori Lewis variants in vitro can diminish the production of interleukin-6 and the development of Th1 cells (Bergman et al., 2004). Surfactant protein D (SP-D), another C-type lectin, is upregulated in the gastric mucosa during infection and can specifically bind to H. pylori, reducing its activity by 50% (Murray et al., 2002). Moreover, in vitro culture of the escape mutant J178V, which evades SP-D binding through focused glycosylation of the LPS O-chain, has been documented (Khamri et al., 2005). ...
Article
Full-text available
Helicobacter pylori ( H. pylori ) is the predominant pathogen causing chronic gastric mucosal infections globally. During the period from 2011 to 2022, the global prevalence of H. pylori infection was estimated at 43.1%, while in China, it was slightly higher at approximately 44.2%. Persistent colonization by H. pylori can lead to gastritis, peptic ulcers, and malignancies such as mucosa-associated lymphoid tissue (MALT) lymphomas and gastric adenocarcinomas. Despite eliciting robust immune responses from the host, H. pylori thrives in the gastric mucosa by modulating host immunity, particularly by altering the functions of innate and adaptive immune cells, and dampening inflammatory responses adverse to its survival, posing challenges to clinical management. The interaction between H. pylori and host immune defenses is intricate, involving evasion of host recognition by modifying surface molecules, manipulating macrophage functionality, and modulating T cell responses to evade immune surveillance. This review analyzes the immunopathogenic and immune evasion mechanisms of H. pylori , underscoring the importance of identifying new therapeutic targets and developing effective treatment strategies, and discusses how the development of vaccines against H. pylori offers new hope for eradicating such infections.
... 19,31 In addition, surfactant protein-D (SP-D) has been reported to play a crucial role in the elimination of Helicobacter from the luminal mucus layer. [32][33][34] In this study, the level of SP-D was increased during H felis infection and was much higher in H felis-infected Flt3 -/mice than in H felis-infected WT mice ( Figure 7F). Therefore, the increased gastric inflammation and SP-D accumulation may be associated with high clearance of H felis in Flt3 -/mice. ...
Article
Full-text available
Background & aims: Helicobacter pylori has been reported to modulate local immune responses in order to colonize persistently in gastric mucosa. Although the induced expression of programmed cell death ligand 1 (PD-L1) has been suggested as an immune modulatory mechanism for persistent infection of Helicobacter pylori, the main immune cells expressing PD-L1 and their functions in Helicobacter-induced gastritis still remain to be elucidated. Methods: The blockades of PD-L1 with antibody or PD-L1 deficient bone marrow transplantation were performed in Helicobacter-infected mice. The main immune cells expressing PD-L1 in Helicobacter-infected stomach were determined by flow cytometry and immunofluorescence staining. Helicobacter felis or pylori-infected dendritic cell (DC)-deficient mouse models including Flt3-/-, Zbtb46-DTR, and BDCA2-DTR mice were analyzed for pathological changes and colonization levels. Finally, the location of PD-L1-expressing DCs and the correlation with Helicobacter pylori infection were analyzed in human gastric tissues using multiplexed immunohistochemistry. Results: Genetic or antibody-mediated blockade of PD-L1 aggravated Helicobacter-induced gastritis with mucosal metaplasia. Gastric classical DCs (cDCs) expressed considerably higher levels of PD-L1 than other immune cells and co-localized with T cells in gastritis lesions from Helicobacter-infected mice and humans. Helicobacter felis or pylori-infected Flt3-/- or cDC-depleted mice showed aggravated gastritis with severe T cell and neutrophil accumulation with low bacterial loads compared with that in control mice. Finally, PD-L1 expressing DCs are co-localized with T cells and showed positive correlation with Helicobacter pylori infection in human subjects. Conclusions: The PD-1/PD-L1 pathway may be responsible for the immune modulatory function of gastric DCs that protects the gastric mucosa from Helicobacter-induced inflammation, but allows persistent Helicobacter colonization.
... Helicobacter pylori infection, an increased level of SP-D has been detected, suggesting the possible role of SP-D in the mucosal defense outside the lungs (Murray et al. 2002), eg. gastrointestinal tract. ...
Chapter
Full-text available
CLEC5A is a spleen tyrosine kinase (Syk)-coupled C-type lectin that is highly expressed by monocytes, macrophages, neutrophils, and dendritic cells and interacts with virions directly, via terminal fucose and mannosemoieties of viral glycans. CLEC5A also binds to N-acetylglucosamine (GlcNAc) and N-acetylmuramic acid (MurNAc) disaccharides of bacterial cellwalls. Compared to other C-type lectins (DC-SIGN and DC-SIGNR) and TLRs, CLEC5A binds its ligands with relatively low affinities. However, CLEC5A forms a multivalent hetero-complex with DCSIGN and other C-type lectins upon engagement with ligands, and thereby mediates microbe-induced inflammatory responses via activation of Syk. For example, in vivo studies in mouse models have demonstrated that CLEC5A is responsible for flaviviruses-induced hemorrhagic shock and neuroinflammation, and a CLEC5A polymorphism in humans is associated with disease severity following infection with dengue virus. In addition, CLEC5A is co-activated with TLR2 by Listeria and Staphylococcus. Furthermore, CLEC5A-postive myeloid cells are responsible for Concanavilin A-induced aseptic inflammatory reactions. Thus, CLEC5A is a promiscuous pattern recognition receptor in myeloid cells and is a potential therapeutic target for attenuation of both septic and aseptic inflammatory reactions.
... Expression of MBL, SP-A and SP-D at the mucosal surfaces suggest the innate immune roles of these collectins against invading pathogens. During Helicobacter pylori infection, an increased level of SP-D has been detected, suggesting the possible role of SP-D in the mucosal defense outside the lungs (Murray et al. 2002), eg. gastrointestinal tract. ...
Chapter
Full-text available
Collectins are collagen-containing C-type (calcium-dependent) lectins which are important pathogen pattern recognising innate immune molecules. Their primary structure is characterised by an N-terminal, triple-helical collagenous region made up of Gly-X-Y repeats, an alpha-helical coiled-coil trimerising neck region, and a C-terminal C-type lectin or carbohydrate recognition domain (CRD). Further oligomerisation of this primary structure can give rise to more complex and multimeric structures that can be seen under electron microscope. Collectins can be found in serum as well as in a range of tissues at the mucosal surfaces. Mannan-binding lectin can activate the complement system while other members of the collectin family are extremely versatile in recognising a diverse range of pathogens via their CRDs and bring about effector functions designed at the clearance of invading pathogens. These mechanisms include opsonisation, enhancement of phagocytosis, triggering super oxidative burst and nitric oxide production. Collectins can also potentiate the adaptive immune response via antigen presenting cells such as macrophages and dendritic cells through modulation of cytokines and chemokines, thus they can act as a link between innate and adaptive immunity. This chapter describes the structure-function relationships of collectins, their diverse functions, and their interaction with viruses, bacteria, fungi and parasites.
... CRTAC1 is upregulated (>10-fold) in type 2 epithelial cells as they differentiate in culture [38]. Previous studies have shown that binding of SP-D to phosphatidylinositol [39], toll like receptor (TLR)2 and TLR4 [40] and carbohydrate structures on the surface of bacteria, viruses and fungi is calcium dependent [41][42][43][44][45][46]. Calcium may also cause SP-D oligomers to self-aggregate, as has been shown previously with surfactant protein A [47]. ...
Article
Surfactant protein D (SP-D) is produced primarily in the lung and is involved in regulating pulmonary surfactants, lipid homeostasis and innate immunity. Circulating SP-D levels in blood are associated with chronic obstructive pulmonary disease (COPD), although causality remains elusive. In 4061 subjects with COPD, we identified genetic variants associated with serum SP-D levels. We then determined whether these variants affected lung tissue gene expression in 1037 individuals. A Mendelian randomisation framework was then applied, whereby serum SP-D-associated variants were tested for association with COPD risk in 11 157 cases and 36 699 controls and with 11 years decline of lung function in the 4061 individuals. Three regions on chromosomes 6 (human leukocyte antigen region), 10 ( SFTPD gene) and 16 ( ATP2C2 gene) were associated with serum SP-D levels at genome-wide significance. In Mendelian randomisation analyses, variants associated with increased serum SP-D levels decreased the risk of COPD (estimate −0.19, p=6.46×10 ⁻⁰³ ) and slowed the lung function decline (estimate=0.0038, p=7.68×10 ⁻³ ). Leveraging genetic variation effect on protein, lung gene expression and disease phenotypes provided novel insights into SP-D biology and established a causal link between increased SP-D levels and protection against COPD risk and progression. SP-D represents a very promising biomarker and therapeutic target for COPD.
Article
The intestine is an important immune organ consisting of a complex cellular network, secreted peptides and proteins and other host defenses. Innate immunity plays a central role in intestinal immune defense against invading pathogens. It also serves as a bridge to the activation of the adaptive immune system. Pattern recognition molecules of microorganisms are an essential component for identifying invading pathogens. Toll‐like receptors (TLRs), CARD15/NOD2 and scavenger receptors all serve as the pattern recognition receptors in the innate immune defense system. Secreted bactericidal peptides or defensins produced by the intestinal epithelia represent another crucial element of innate mucosal immune defense. Mutations in pattern recognition receptors and dysfunction of secretory bactericidal peptides may impair host immune defenses leading to an invasion of pathogens resulting in chronic inflammation of the gut. This review updates our current understanding of innate immunity of the gastrointestinal tract.
Article
Background: Surfactant protein D (SP-D) is an innate host defense protein that clears infectious pathogens from the lung and regulates pulmonary host defense cells. SP-D is also detected in lower concentrations in plasma and many other non-pulmonary tissues. Plasma levels of SP-D increase during infection and other proinflammatory states; however, the source and functions of SP-D in the systemic circulation are largely unknown. We hypothesized that systemic SP-D may clear infectious pathogens and regulate host defense cells in extrapulmonary systems. Methods: To determine if SP-D inhibited inflammation induced by systemic lipopolysaccharide (LPS), E.coli LPS was administered to mice via tail vein injection with and without SP-D and the inflammatory response was measured. Results: Systemic SP-D has a circulating half-life of 6hours. Systemic IL-6 levels in mice lacking the SP-D gene were similar to wild type mice at baseline but were significantly higher than wild type mice following LPS treatment (38,000 vs 29,900ng/ml for 20mg/kg LPS and 100,700 vs 73,700ng/ml for 40mg/kg LPS). In addition, treating wild type mice with purified intravenous SP-D inhibited LPS induced secretion of IL-6 and TNFα in a concentration dependent manner. Inhibition of LPS induced inflammation by SP-D correlated with SP-D LPS binding suggesting SP-D mediated inhibition of systemic LPS requires direct SP-D LPS interactions. Conclusions: Taken together, the above results suggest that circulating SP-D decreases systemic inflammation and raise the possibility that a physiological purpose of increasing systemic SP-D levels during infection is to scavenge systemic infectious pathogens and limit inflammation-induced tissue injury.
Chapter
The human stomach is a harsh and fluctuating environment for bacteria with hazards such as gastric acid and flow through of gastric contents into the intestine. H. pylori gains admission to a stable niche with nutrient access from exudates when attached to the epithelial cells under the mucus layer, whereof adherence to glycolipids and other factors provides stable and intimate attachment. To reach this niche, H. pylori must overcome mucosal defence mechanisms including the continuously secreted mucus layer, which provides several layers of defence: (1) mucins in the mucus layer can bind H. pylori and transport it away from the gastric niche with the gastric emptying, (2) mucins can inhibit H. pylori growth, both via glycans that can have antibiotic like function and via an aggregation-dependent mechanism, (3) antimicrobial peptides (AMPs) have antimicrobial activity and are retained in a strategic position in the mucus layer and (4) underneath the mucus layer, the membrane-bound mucins provide a second barrier, and can function as releasable decoys. Many of these functions are dependent on H. pylori interactions with host glycan structures, and both the host glycosylation and concentration of antimicrobial peptides change with infection and inflammation, making these interactions dynamic. Here, we review our current understanding of mucin glycan and antimicrobial peptide-dependent host defence mechanisms against H. pylori infection.
Article
Aim: This study aimed to study the effects of intratracheal instillation of budesonide on lung maturity of premature fetal rabbits. The developmental pattern of pulmonary alveoli in rabbits is similar to that in humans. Method: Fetal rabbits were taken out from female rabbits on the 28th day of pregnancy (full term = 31 days) by cesarean section (c-section). The fetal rabbits were divided into four groups: control (normal saline, NS), budesonide (budesonide, BUD), calf pulmonary surfactant for injection (pulmonary surfactant, PS), and calf pulmonary surfactant + budesonide for injection (pulmonary surfactant + budesonide, PS + BUD). All premature rabbits were kept warm after c-section. After 15-min autonomous respiration, a tracheal cannula was implemented for instilling NS, BUD, PS, and PS + BUD. The morphology of lung tissues of premature fetal rabbits was analyzed using optical and electron microscopes. Surfactant protein B (SP-B) mRNA and protein levels in lung tissues were determined using polymerase chain reaction and Western blotting, respectively. Result: Intratracheal instillation of BUD could increase the alveolar area of the fetal rabbits (P < 0.01), decrease the alveolar wall thickness (P < 0.01), and increase the mean density of lamellar bodies (P < 0.05) and SP-B protein levels in type II epithelial cells of pulmonary alveoli (P < 0.05). Conclusion: Intratracheal instillation of BUD during early postnatal life is effective in promoting alveolarization and increasing SP-B expression, the pro-pulmonary maturity of BUD combined with PS is superior to that of BUD or PS alone. However, the long-term effect of BUD on lung development needs further exploration.
Article
Full-text available
The human surfactant protein (SP) A locus has been assigned to chromosome 10q22-q23 and consists of two very similar genes, SP-A1 and SP-A2, as well as a truncated pseudogene. SP-A belongs to the family of collagenous C-type lectins along with mannose binding protein (MBP) and SP-D, both of which have also been mapped to the long arm of chromosome 10. In this article we report the relative location and orientation of each of the SP-A and SP-D genomic sequences. Characterization of two overlapping genomic clones revealed that the SP-A pseudogene lies in a reverse orientation 15 kb away from the 5′ side of SP-A1. This finding was verified by the amplification of the entire SP-A pseudogene/SP-A1 intergenic region using long-range polymerase chain reaction. The relative location of SP-A2 and SP-D was then ascertained by testing a number of sequence tagged sites against the Stanford TNG3 and G3 radiation hybrid panels. The radiation hybrid mapping data showed that both SP-A2 and SP-D are on the 5′ side of SP-A...
Article
Full-text available
Surfactant protein D (SP-D) molecules are preferentially assembled as dodecamers consisting of trimeric subunits associated at their amino termini. The NH2-terminal sequence of each monomer contains two conserved cysteine residues, which participate in interchain disulfide bonds. In order to study the roles of these residues in SP-D assembly and function, we employed site-directed mutagenesis to substitute serine for cysteine 15 and 20 in recombinant rat SP-D (RrSP-D), and have expressed the mutant (RrSP-Dser15/20) in Chinese hamster ovary (CHO-K1) cells. The mutant, which was efficiently secreted, bound to maltosyl-agarose, but unlike RrSP-D, was assembled exclusively as trimers. The constituent monomers showed a decreased mobility on SDS-polyacrylamide gel electrophoresis resulting from an increase in the size and sialylation of the N-linked oligosaccharide at Asn-70. Although RrSP-Dser15/20 contained a pepsin-resistant triple helical domain, it showed a decreased Tm, and acquired susceptibility to proteolytic degradation. Like RrSP-D, RrSP-Dser15/20 bound to the hemagglutinin of influenza A. However, it showed no viral aggregation and did not enhance the binding of influenza A to neutrophils (PMN), augment PMN respiratory burst, or protect PMNs from deactivation. These studies indicate that amino-terminal disulfides are required to stabilize dodecamers, and support our hypothesis that the oligomerization of trimeric subunits contributes to the anti-microbial properties of SP-D.
Article
Full-text available
Surfactant protein D (SP-D) is one of two collectins found in the pulmonary alveolus. On the basis of homology with other collectins, potential functions for SP-D include roles in innate immunity and surfactant metabolism. The SP-D gene was disrupted in embryonic stem cells by homologous recombination to generate mice deficient in SP-D. Mice heterozygous for the mutant SP-D allele had SP-D concentrations that were approximately 50% wild type but no other obvious phenotypic abnormality. Mice totally deficient in SP-D were healthy to 7 months but had a progressive accumulation of surfactant lipids, SP-A, and SP-B in the alveolar space. By 8 weeks the alveolar phospholipid pool was 8-fold higher than wild-type littermates. There was also a 10-fold accumulation of alveolar macrophages in the null mice, and many macrophages were both multinucleated and foamy in appearance. Type II cells in the null mice were hyperplastic and contained giant lamellar bodies. These alterations in surfactant homeostasis were not associated with detectable changes in surfactant surface activity, postnatal respiratory function, or survival. The findings in the SP-D-deficient mice suggest a role for SP-D in surfactant homeostasis.
Article
Full-text available
Non-motile variants of Helicobacter pylori (strain 26695) occurred with a frequency of 1.6 (SD 0.4) x 10(-4) variants/cell/division cycle, and reversion to the motile form occurred with a frequency of less than 10(-7) variants/cell/division cycle. The two forms remained greater than 90% pure for up to 50 cell divisions and differed only in the presence or absence of motility and flagella. Bacteria were recovered from nine of 10 gnotobiotic piglets inoculated orally with motile H. pylori, but from only two of eight inoculated with the non-motile variant. The motile form survived for 21 days in infected piglets, but the non-motile variant survived for only 6 days. Bacteria recovered from piglets inoculated with the non-motile variant were non-motile. These data support the hypothesis that motility is a colonisation factor for H. pylori.
Article
Lung surfactant proteins A and D (SP-A and SP-D), together with serum derived mannan-binding lectin (MBL), are human proteins that belong to the collectin subgroup of the C-type lectin family. The collectins play an important role in innate immunity by binding to carbohydrate moieties on the surface of microorganisms, promoting their uptake and killing by phagocytes in vitro. Recent studies have also demonstrated that SP-A and SP-D may have the potential to modify allergy-induced inflammation by inhibiting the production and release of inflammatory mediators by allergic effector cells.
Article
The seroprevalence of Helicobacter pylori infection increases with age. Incidence rates in children are higher than in adults and are consistent with a cohort effect reflecting primary infection during childhood. Preliminary data would suggest that horizontal transmission of infection may occur both in adults and in children. Occasionally, this may result in transient multiple infection, because the observation of mosaicism has to imply horizontal transfer of genetic information between infecting strains of H. pylori. The very close association between H. pylori infection and socio-economic conditions in childhood means that the possibility of confounding factors should be considered when studying routes of transmission or assessing the extra-gastric consequences of infection. Future studies will clarify these important issues. (C) Lippincott-Raven Publishers.
Article
A simple procedure has been developed for the purification of the surfactant proteins SP-A and SP-D from lung lavage of patients with alveolar proteinosis. The SP-D is purified by fractionation of the supernatant obtained after spinning the lavage at 10 000×g for 40 min, while the bulk of the SP-A is purified by fractionation of the pellet. The supernatant is applied to a maltosyl–agarose column and the bound SP-D is specifically eluted using MnCl2. The pellet is solubilised in 6 M urea and, following renaturation, the solubilised proteins are applied to maltosyl–agarose and SP-A eluted using a gradient of EDTA. Both SP-A and SP-D are further purified by gel-filtration on Superose-6. This procedure has also been used to prepare successfully human SP-A and SP-D from amniotic fluid and may be generally applicable to the isolation of these surfactant proteins from lung washings obtained from other species.
Article
Human pulmonary surfactant protein D (SP-D) was identified in lung lavage by its similarity to rat SP-D in both its molecular mass and its Ca(2+)-dependent-binding affinity for maltose [Persson, Chang & Crouch (1990) J. Biol. Chem. 265, 5755-5760]. For structural studies, human SP-D was isolated from amniotic fluid by affinity chromatography on maltose-Sepharose followed by f.p.l.c. on Superose 6, which showed it to have a molecular mass of approx. 620 kDa in non-dissociating conditions. On SDS/PAGE the human SP-D behaved as a single band of 150 kDa or 43 kDa in non-reducing or reducing conditions respectively. The presence of a high concentration of glycine (22%), hydroxyproline and hydroxylysine in the amino acid composition of human SP-D indicated that it contained collagen-like structure. Collagenase digestion yielded a 20 kDa collagenase-resistant globular fragment which retained affinity for maltose. Use of maltosyl-BSA as a neoglycoprotein ligand in a solid-phase binding assay showed that human SP-D has a similar carbohydrate-binding specificity to rat SP-D, but a clearly distinct specificity from that of other lectins, such as conglutinin, for a range of simple saccharides. Amino acid sequence analysis established the presence of collagen-like Gly-Xaa-Yaa triplets in human SP-D and also provided sequence data from the globular region of the molecule which was used in the synthesis of oligonucleotide probes. Screening of a human lung cDNA library with the oligonucleotide probes, and also with rabbit anti-(human SP-D), allowed the isolation of two cDNA clones which overlap to give the full coding sequence of human SP-D. The derived amino acid sequence indicates that the mature human SP-D polypeptide chain is 355 residues long, having a short non-collagen-like N-terminal section of 25 residues, followed by a collagen-like region of 177 residues and a C-terminal C-type lectin domain of 153 residues. Comparison of the human SP-D and bovine serum conglutinin amino acid sequences indicated that they showed 66% identity despite their marked differences in carbohydrate specificity.
Article
Surfactant protein D (SP-D) is a collagenous glycoprotein that is secreted into the pulmonary airspaces by alveolar type II and nonciliated bronchiolar cells. SP-D exhibits Ca(++)-dependent carbohydrate binding in vitro and is structurally related to the collagenous C-type lectins, including serum conglutinin, serum mannose-binding proteins, and surfactant protein A. Preliminary studies showed calcium- and saccharide-dependent binding of fluorescein-conjugated or radioiodinated SP-D to a variety of microorganisms, including Gram-negative bacteria and fungi. A laboratory strain of Escherichia coli (Y1088) was chosen to further examine the mechanism(s) of binding. Binding of SP-D to Y1088 was time dependent, saturable, and inhibited by cold SP-D or competing saccharides; Scatchard analysis gave a Kd of 2 x 10(-11) M. At higher concentrations, SP-D also caused Ca(++)-dependent agglutination of Y1088 that was inhibited by alpha-glucosyl-containing saccharides, antisera to the carbohydrate-binding domain of SP-D, or Y1088 LPS. Lectin blots showed specific binding of 125I-SP-D to Y1088 LPS, as well as LPS from other several strains of enteric Gram-negative bacteria. Immunogold studies demonstrated strong and uniform surface labeling of the bacteria. Rat and human bronchoalveolar lavage (BAL) caused Ca(++)-dependent agglutination of E. coli that was dose dependent and inhibited by competing saccharides or anti-SP-D. SP-D was selectively and efficiently adsorbed from rat BAL by incubation with E. coli, and incubation of E. coli with radiolabeled rat type II cell medium revealed that SP-D is the major E. coli-binding protein secreted by freshly isolated cells in culture. We suggest that SP-D plays important roles in the lung's defense against Gram-negative bacteria.
Article
Sodium dodecyl sulfate-polyacrylamide gel electrophoresis was used to analyze the macromolecular heterogeneity of lipopolysaccharides (LPS) from seven fresh clinical isolates and three culture collection strains of the human pathogen Helicobacter pylori. All the clinical isolates produced smooth-form LPS with O side chains of relatively homogeneous chain length, whereas the culture collection strains yielded rough-form LPS. A better yield of the latter LPS was obtained when combined protease pretreatment and hot phenol-water extraction were used than when the conventional phenol-water technique alone was used for extraction. The LPS of the three culture collection strains (S-24, C-5437, and NCTC 11637) were chemically characterized. Constituents common to all the LPS were fucose, D-mannose, D-glucose, D-galactose, D-glycero-D-manno-heptose, L-glycero-D-manno-heptose, and 3-deoxy-D-manno-2-octulosonic acid. The molar ratios of the hexoses differed between different strains, thereby reflecting structural differences. Phosphate, phosphorylethanolamine, and pyrophosphorylethanolamine were present also. Free lipid A contained D-glucosamine and fatty acids, with phosphate and a minor amount of ethanolamine. The major fatty acids were ester- and amide-bound 3-hydroxyoctadecanoic acid and ester-bound octadecanioc and 3-hydroxyhexadecanoic acids, with minor amounts of ester-bound tetradecanoic and hexadecanoic acids. In addition to the uncommonly long 3-hydroxy fatty acids, an unusual phosphorylation pattern was deduced to be present in the lipid A.