ArticlePDF Available

Effect of surfactant protein A (SP-A) on the production of cytokines by human pulmonary macrophages

Authors:

Abstract

Surfactant protein A (SP-A) is thought to play a role in the modulation of lung inflammation during acute respiratory distress syndrome (ARDS). However, SP-A has been reported both to stimulate and to inhibit the proinflammatory activity of pulmonary macrophages (Mφ). Because of the interspecies differences and heterogeneity of Mφ subpopulations used may have influenced previous controversial results, in this study, we investigated the effect of human SP-A on the production of cytokines and other inflammatory mediators by two well-defined subpopulations of human pulmonary Mφ. Surfactant and both alveolar (aMφ) and interstitial (iMφ) macrophages were obtained from multiple organ donor lungs by bronchoalveolar lavage and enzymatic digestion. Donors with either recent history of tobacco smoking, more than 72 h on mechanical ventilation, or any radiological pulmonary infiltrate were discarded. SP-A was purified from isolated surfactant using sequential butanol and octyl glucoside extractions. After 24-h preculture, purified Mφ were cultured for 24 h in the presence or absence of LPS (10 μg/mL), SP-A (50 μg/mL), and combinations. Nitric oxide and carbon monoxide (CO) generation (pmol/μg protein), cell cGMP content (pmol/μg protein), and tumor necrosis factor alpha (TNFα), interleukin (IL)-1, and IL-6 release to the medium (pg/μg protein) were determined. SP-A inhibited the lipopolysaccharide (LPS)-induced TNFα response of both interstitial and alveolar human Mφ, as well as the IL-1 response in iMφ. The SP-A effect on TNFα production could be mediated by a suppression in the LPS-induced increase in intracellular cGMP. In iMφ but not in aMφ, SP-A also inhibited the LPS-induced IL-1 secretion and CO generation. These data lend further credit to a physiological function of SP-A in regulating alveolar host defense and inflammation by suggesting a fundamental role of this apoprotein in limiting excessive proinflammatory cytokine release in pulmonary Mφ during ARDS.
EFFECT OF SURFACTANT PROTEIN A (SP-A) ON THE PRODUCTION OF
CYTOKINES BY HUMAN PULMONARY MACROPHAGES
Javier Arias-Diaz,* Ignacio Garcia-Verdugo,
Cristina Casals,
Natalia Sanchez-Rico,
Elena Vara,
and Jose´ L. Balibrea*
*Department of Surgery, Hospital San Carlos, Madrid, Spain
Department of Biochemistry,
Faculty of Biology and
Medicine, Complutense University, Madrid, Spain
Received 28 Apr 2000; first review completed 5 May 2000; accepted in final form 16 Jun 2000
ABSTRACT—Surfactant protein A (SP-A) is thought to play a role in the modulation of lung inflammation
during acute respiratory distress syndrome (ARDS). However, SP-A has been reported both to stimulate and
to inhibit the proinflammatory activity of pulmonary macrophages (M
). Because of the interspecies differ-
ences and heterogeneity of M
subpopulations used may have influenced previous controversial results, in
this study, we investigated the effect of human SP-A on the production of cytokines and other inflammatory
mediators by two well-defined subpopulations of human pulmonary M
. Surfactant and both alveolar (aM
)
and interstitial (iM
) macrophages were obtained from multiple organ donor lungs by bronchoalveolar lavage
and enzymatic digestion. Donors with either recent history of tobacco smoking, more than 72 h on mechani-
cal ventilation, or any radiological pulmonary infiltrate were discarded. SP-A was purified from isolated
surfactant using sequential butanol and octyl glucoside extractions. After 24-h preculture, purified M
were
cultured for 24 h in the presence or absence of LPS (10 µg/mL), SP-A (50 µg/mL), and combinations. Nitric
oxide and carbon monoxide (CO) generation (pmol/µg protein), cell cGMP content (pmol/µg protein), and
tumor necrosis factor alpha (TNF
), interleukin (IL)-1, and IL-6 release to the medium (pg/µg protein) were
determined. SP-A inhibited the lipopolysaccharide (LPS)-induced TNF
response of both interstitial and
alveolar human M
, as well as the IL-1 response in iM
. The SP-A effect on TNF
production could be
mediated by a suppression in the LPS-induced increase in intracellular cGMP. In iM
but not in aM
, SP-A
also inhibited the LPS-induced IL-1 secretion and CO generation. These data lend further credit to a
physiological function of SP-A in regulating alveolar host defense and inflammation by suggesting a funda-
mental role of this apoprotein in limiting excessive proinflammatory cytokine release in pulmonary M
during
ARDS.
KEYWORDS—Nitric oxide, carbon monoxide, cyclic guanosin 35-monophosphate, pulmonary surfactants;
bronchoalveolar lavage fluid
INTRODUCTION
Cytokines seem to act predominantly in a paracrine way
when producing their deleterious effects during sepsis. There-
fore, the local release of cytokines by pulmonary M
would be
of importance in the pathogenesis of the adult respiratory
distress syndrome (ARDS). Recently, it has been found that
alveolar type II cells and some of their secretory products,
including surfactant-associated protein A (SP-A) and D, have
nonsurfactant-related functions and are actively associated
with various defense functions in the lung (1). SP-A is the most
abundant of the surfactant proteins. It is a calcium-dependent
lectin structurally similar to the complement component, C1q,
and the mannose-binding protein, an acute-phase protein with
opsonic function. Some of its nonsurfactant-related functions
include augmentation of alveolar macrophage migration,
enhancement of macrophage phagocytosis of opsonized or
nonopsonized bacteria or viruses (1), and regulation of reactive
oxygen species production (2).
SP-A seems also to influence the proinflammatory activity
of aM
, and it may participate this way in the modulation of
lung inflammation during the ARDS; however, it has been
reported both to stimulate (3) and to inhibit (4) the production
of cytokines by pulmonary M
.
Alveoli are lined with surfactant that consists of about 90%
lipids and 5–10% surfactant-specific proteins. The major func-
tion of this material is to reduce the surface tension in the
alveoli, thereby preventing alveolar collapse and edema.
Alveolar M
but not interstitial M
reside in this surfactant-
rich microenvironment and contain surfactant, being more
exposed to a potential influence of surfactant constituents.
However, during lung inflammation, alveolar epithelium is
disrupted and cells in the interstitium can also contact surfac-
tant. Furthermore, there have been several studies that have
detected circulating antibodies to the surfactant proteins or the
surfactant proteins themselves in the serum (5), suggesting that
these proteins may be introduced into the circulation in some
conditions and be able to interact with interstitial and even
circulating immune cells.
Besides different localizations, it is reasonable to assume
that the iM
and aM
populations exert distinct functions that
are altered upon exposure to inflammatory agents and that the
intensity and nature of these events have an impact on the net
Presented at the 20
th
annual meeting of the Surgical Infection Society, April
27–29, 2000, Providence RI.
Address reprint requests to Javier Arias-Diaz, MD, PhD, Departamento de
Cirugia Hospital Clinico San Carlos, Ciudad Universitaria, 28040-Madrid,
Spain.
SHOCK, Vol. 14, No. 3, pp. 300–306, 2000
300
outcome of the inflammatory response. Accordingly, the ques-
tion arises of whether SP-A affects differently both M
popu-
lations in humans. To explore this possibility we studied the
lipopolysaccharide (LPS)-induced production of tumor necro-
sis factor alpha (TNF
), interleukin (IL)-1
, and IL-6 by
human interstitial and alveolar M
. To gain further insight into
the mechanisms underlying the SP-A effect, we also investi-
gated the cyclic guanosine 35-monophosphate (cGMP) path-
way of LPS-induced cytokine secretion (6).
MATERIALS AND METHODS
Human lung tissue procurement
This study was approved by the review board and the ethic committee of the
Hospital San Carlos. Human lung tissue was obtained from male multiple
organ donors. A written informed consent was obtained from the next-of-kin
of each donor. Ages ranged from 22 to 46 years, and cranial trauma or spon-
taneous intracranial hemorrhage was the cause of death in all donors. Donors
with either recent history of tobacco smoking, more than 72 h receiving
mechanical ventilation or any radiological pulmonary infiltrate were excluded
from this study. Immediately after obtaining the organs that were going to be
used for transplantation, the left lung was excised. A bronchoalveolar lavage
(BAL) was performed through the main bronchus using a total of 500 mL of
0.9% normal saline at 4°C, and the lung was then placed in cold saline solution.
Cold ischemia period was less than3hinallcases.
Isolation of SP-A
SP-A was purified from the fluid obtained at lung lavage as described
previously in detail (7). Briefly, the whole surfactant pellet from lung lavage
fluids was extracted with butanol; butanol-insoluble proteins were resolubi-
lized with octylglucopyranoside (OGP); and subsequently, SP-A was solubi-
lized in 5 mM Tris-buffered water, pH 7.4. The purity of SP-A was checked
by one-dimensional SDS/PAGE in 12% acrylamide under reducing conditions
(50 mM dithiothreitol). Quantification of SP-A was carried out by amino acid
analysis in a Beckman System 6300 High Performance analyzer (7). The
protein hydrolysis was performed with 0.2 mL of 6 M HCl, containing 0.1%
(w/v) phenol in evacuated and sealed tubes at 108°C for 24 h. Norleucine was
added to each sample as internal standard.
Cell isolation and culture
Bronchoalveolar cells were separated from lavage fluid by centrifugation.
The sedimented cells were washed twice with Hank’s balanced salt solution
(HBSS) and then centrifuged (250 g, 10 min). To isolate iM
(6), pulmonary
tissue was washed with a calcium-free solution and then minced into small
fragments (1 to 2 mm). After copious washing with the same solution, to
remove most blood cells and remaining airway M
, tissue fragments under-
went an enzymatic digestion with elastase (27 orcein-elastin U/mL elastase) in
a 37°C shaking bath. Digestion was stopped by addition of 4°C fetal calf
serum, the tissue was filtered through nylon mesh (200 and 20
m), and the
cell suspension was washed twice with HBSS and centrifuged (250 g, 10 min).
The cell pellets were resuspended in RPMI-1640 medium (10% heat-
inactivated fetal calf serum, 100 IU/mL penicillin G, 50
g/mL gentamycin),
and poured into 75 cm
2
culture flasks. After 90 min of incubation (37°C, under
O
2
/CO
2
atmosphere), the supernatants were removed and the cells were
washed four times with phosphate-buffered saline (PBS) to remove contami-
nating non-adherent cells. Adherent cells were found to be 99.5 ± 0.3% viable
(Trypan blue exclusion test), and to be composed of 92.5 ± 3.1% M
,as
judged by Wright–Giemsa stained cytocentrifuge preparations, being most of
the remaining cells neutrophils or fibroblasts. The cells were gently scraped,
plated onto collagen-coated 96-well plastic dishes (5 × 10
5
cells per well) and
precultured for 24 h. Under these conditions, approximately 95% of the cell
were attached; cell viability was higher than 97% and macrophage purity was
always greater than 98%. Cells were cultured for another 24 h in the presence
or absence of LPS (Escherichia Coli 055:B5, 10
g/mL), 8-Br-cGMP (1
mmol/L), methylene blue (MB, 10
−5
mol/L), hemin (1
mol/L), SP-A (50
g/mL), and combinations. Macrophage cultures from each individual donor
were plated in duplicate wells and each series of experiments was repeated
with a minimum of at least six donors.
Biochemical determinations
Supernatants and cells were recovered, and the cGMP content of the cells
and TNF
, IL-1, IL-6, carbon monoxide (CO), and nitric oxide (NO) release
to the medium were determined. Cell cGMP content was measured with
specific RIA (
125
I-RIA Kit, Radiochemical Centre, Amersham, Bucks, UK).
The cytokines measurements were performed by specific ELISA kits. An
aliquot of the cell suspension was used for protein quantitation, which was
performed spectrophotometrically by the Coomassie brilliant blue dye method.
Release of NO was measured by the Griess reaction as nitrite (NO
2
)
concentration after nitrate (NO
3
) reduction to NO
2
. Briefly, samples were
deproteinized by the addition of sulfosalicylic acid. They were then incubated
for 30 min at 4°C, and subsequently centrifuged for 20 min at 12000 g. After
incubation of the supernatants with E. coli NO
3
reductase (37°C, 30 min), 1
mL of Griess reagent (0.5% naphthylethylenediamine dihydrochloride, 5%
sulfonilamide, 25% H
3
PO
4
) was added. The reaction was performed at 22°C
for 20 min, and the absorbance at 546 nm was measured, using NaNO
2
solution
as standard.
To quantify the amount of CO formed, hemoglobin (Hb) was added to bind
CO as carboxyhemoglobin (CO-Hb) and the proportion of CO-Hb was esti-
mated (6). For that, Hb (4
mol/L) was mixed gently into the sample and 1 min
was allowed to ensure maximum CO binding. Then, samples were diluted with
buffer phosphate (0.01 mol/L KH
2
PO
4
/K
2
HPO
4
, pH 6.85) containing 2 mg/mL
sodium hydrosulfite, gently mixed in stoppered cuvettes and allowed to stand
at room temperature for 10 min. Absorbance was read at 420 and 432 nm
against a matched cuvette containing only buffer.
Statistical analyses
N represents the number of separate macrophage preparations employed
(each from a different donor lung). The different assays were performed in at
least duplicates, and the means were calculated. The results are presented as
the means standard error of the mean (SEM)], obtained by combining the
results from each cell preparation. Mean comparison was done by the Fried-
man’s analysis of variance of ranks, followed by a two-tailed Wilcoxon’s rank
sum test for paired data to identify the source of the found differences; a
confidence level 95% or greater (P< 0.05) was considered significant.
RESULTS
LPS significantly increased the release of TNF
(Fig. 1A),
IL-1 (Fig. 2A), and IL-6 (Fig. 3A) by both iM
and aM
. The
increase in TNF
production was more pronounced in iM
than in aM
.
The baseline release of TNF
, IL-1, and IL-6 by unstimu-
lated M
was not affected by 50
g/mL SP-A, which,
however, significantly decreased the LPS-induced TNF
release by both iM
and aM
(Fig. 1A). SP-A also decreased
the LPS-induced IL-1 release by iM
. However, it did not
modify the IL-1 release by aM
(Fig. 2A). SP-A did not
modify the LPS-induced IL-6 release by aM
and iM
(Fig.
3A).
LPS also increased the content of cGMP on aM
as well as
on iM
(Fig. 4). SP-A significantly reduced the increase on
cGMP content induced by LPS (Fig. 4A).
MB, a guanylate cyclase (GC) inhibitor, was effective in
suppressing the LPS-induced increase in cGMP content (Fig.
4A). This inhibitory effect was accompanied by an inhibition
in the TNF
secretion (Fig. 1A) without modifying IL-6
release (Fig. 3A). Neither LPS nor SP-A showed any detect-
able effect on the NO release to the medium (Fig. 5). In
contrast, LPS significantly increased CO release to the medium
by both iM
an aM
.IniM
, this increase was significantly
SHOCK SEPTEMBER 2000 EFFECT OF SP-A ON PULMONARY MACROPHAGES 301
reduced in the presence of SP-A, which, however, did not
modify the LPS-induced CO production on aM
(Fig. 6A).
The addition of 8-Br-cGMP to the medium increased TNF
(Fig. 1B) and IL-1 (Fig. 2B) release by both aM
and iM
but
did not modify IL-6 release (Fig. 3b). The addition of hemin,
the substrate for heme oxygenase, induced similar changes.
The hemin effects on TNF
and IL-1 release were accompa-
nied by an increase in cGMP content (Fig. 4B) and CO release
to the medium (Fig. 6B). SP-A did not modify hemin nor
cGMP effects.
In additional experiments, we studied the dose-dependent
activity of SP-A by using a lower concentration (10
g/mL)
and another one higher (100
g/mL) than 50
g/mL. Even 100
g/mL SP-A did not affect either the baseline concentrations
of cytokines, CO and cGMP, or the LPS-stimulated release of
IL-6 by M
.IniM
, SP-A dose-dependently inhibited the the
LPS-stimulated increases in TNF
(to 6.54 ± 0.05 at 10
g/mL
SP-A, and 2.94 ± 0.01 at 100
g/mL SP-A; pg/
g protein; N
3), IL-1 (to 4.25 ± 0.08 at 10
g/mL SP-A, and 2.87 ± 0.04
at 100
g/mL SP-A; pg/
g protein; N 3), CO (to 5.15 ±
0.03 at 10
g/mL SP-A and 2.68 ± 0.09 at 100
g/mL SP-A;
pmol/
g protein; N 3), and cGMP (to 0.26 ± 0.02 at 10
g/mL SP-A, and 0.11 ± 0.01 at 100
g/mL SP-A; pmol/
g
protein; N 3). In aM
, SP-A did not affect the LPS-
stimulated production of either IL-1 or CO even at the maximal
concentration used. The effect on LPS-stimulated TNF
and
cGMP increases was related to the SP-A concentration also in
aM
(to 4.34 ± 0.28 at 10
g/mL SP-A, and 3.02 ± 0.11 at 100
g/mL SP-A; pg/
g protein; for TNF
; and to 0.17 ± 0.01 at
10
/mL SP-A, and 0.13 ± 0.01 at 100
g/mL SP-A; pmol/
g
protein; for cGMP; N 3) . There were no significant differ-
ences between the effects of 50
g/mL and 100
g/mL SP-A
in any case.
DISCUSSION
The main findings of the present study indicate that SP-A
can modulate lung inflammation in humans by decreasing the
LPS-induced production of proinflammatory cytokines by both
iM
and aM
. They also show some differences in the
response to SP-A between both M
populations.
Pulmonary M
are considered to be present in at least two
anatomically different compartments (8, 9). Alveolar M
are
found in the bronchoalveolar lumen, where they act as a
primary defensive line phagocytosing inhaled material. Inter-
stitial M
, similar in number, reside within the interstitial
space and are thought to be precursors of the former. To our
knowledge, all previous studies involving interactions between
SP-A and pulmonary M
were carried out using exclusively
the alveolar variety, easily accessible by BAL. The method
used by us permits also the recovery of iM
, which, in fact,
seem to be more active from the immunological viewpoint
(10). As a source of lung tissue, we used multiple organ donors
because pulmonary M
from patients with lung cancer, whose
surgical resection specimen would have been a more accessible
source of pulmonary tissue, have shown a higher TNF
and
FIG.1. Secretion of TNF
by human pulmonary macrophages in
the presence or absence of LPS (10 µg/mL), SP-A (50 µg/mL), meth-
ylene blue (MB, 10
−5
mol/L), hemin (1 µmol/L), 8-Br-cGMP (1 mmol/
L), and combinations. Each column represents the mean ± SE of
duplicate samples from 6 different experiments. *
P
< 0.01 vs. all other
groups, **
P
< 0.01 vs. the rest (upper panel); *
P
<0.01vs
.
control and
SP-A, **
P
<0.05vs
.
Br-cGMP groups (lower panel).
FIG.2. Secretion of IL-1 by human pulmonary macrophages in
the presence or absence of LPS (10 µg/mL), SP-A (50 µg/mL), meth-
ylene blue (MB, 10
−5
mol/L), hemin (1 µmol/L), 8-Br-cGMP (1 mmol/
L), and combinations. Each column represents the mean ± SE of
duplicate samples from 6 different experiments. *
P
< 0.01 vs. all other
groups, **
P
< 0.01 vs. the rest (upper panel); *
P
< 0.01 vs. control and
SP-A (lower panel).
302 SHOCK VOL. 14, NO.3 ARIAS-DIAZ ET AL.
IL-1 secretion in vitro than healthy controls (11). Similarly, we
discarded the donors with recent history of tobacco smoking in
view that functional changes have been reported in aM
from
smokers (12). In bronchoalveolar fluid, the concentration of
SP-A is normally 1–2
g/mL (13). However, dilution due to
the lavage procedure probably causes a 10- to 100-fold reduc-
tion in the concentrations normally present in the alveolar
hypophase. Therefore, we believe that the SP-A concentration
we have used is in the physiological range. On the other hand,
the concentration of LPS used as a stimulus was selected
according to previous dose-response studies in human pulmo-
nary M
(14).
Leukocytes obtained from the alveolar compartment by
lavage techniques repeatedly have been shown to be hypore-
sponsive to inflammatory stimuli as compared with leukocytes
isolated from peripheral blood. This relative “dampening” of
leukocyte activity within the alveolar space is thought to
protect the host from persistent immune cell activation via
inhaled antigens. Surfactant has since long been implicated in
this suppression, since surfactant lipid mixtures and individual
lipid components were noted to inhibit lymphocyte prolifera-
tion and Ig secretion, as well as phagocyte oxygen radical
production. Recently, immunosuppressive activity has also
been demonstrated for SP-A, since this major surfactant
component inhibited lymphocyte proliferation and IL-2
production (15), and reduced TNF
generation in M
(4, 16).
However, others have reported that SP-A per se stimulated
proinflammatory cytokine production in mononuclear cells,
secretion of Igs by splenocytes, and proliferation of lympho-
cytes (3, 17).
Our observation that SP-A decreased TNF
secretion from
LPS-stimulated M
appears to contradict the data from Krem-
lev and Phelps (3), who showed that SP-A could stimulate the
production of TNF
by rat aM
and TNF
, IL-1, and IL-6 by
human monocytes. Although our study focused on examining
the effects of SP-A on LPS-activated M
, we did test the
ability of SP-A to induce production of TNF
, IL-1, or IL-6 by
unstimulated cells and we could not detect any significant
increase in the baseline liberation of these cytokines. We
currently have no unequivocal explanation for these contrast-
ing results, although there were some differences in experi-
FIG.3. Secretion of IL-6 by human pulmonary macrophages in
the presence or absence of LPS (10 µg/mL), SP-A (50 µg/mL), meth-
ylene blue (MB, 10
−5
mol/L), hemin (1 µmol/L), 8-Br-cGMP (1 mmol/
L), and combinations. Each column represents the mean±SE of dupli-
cate samples from 6 different experiments. *
P
< 0.01 vs. non-LPS
groups.
FIG.4. Levels of cGMP in human pulmonary macrophages in the
presence or absence of LPS (10 µg/mL), SP-A (50 µg/mL), methy-
lene blue (MB, 10
−5
mol/L), hemin (1 µmol/L), and combinations.
Each column represents the mean ± SE of duplicate samples from 6
different experiments. *
P
< 0.01 vs. all other groups (upper panel); *
P
<
0.01 vs. control and SP-A (lower panel).
FIG.5. Production of nitric oxide by human pulmonary macro-
phages in the presence or absence of LPS (10 µg/mL) ± SP-A (50
µg/mL). Each column represents the mean ± SE of duplicate samples
from 6 different experiments.
SHOCK SEPTEMBER 2000 EFFECT OF SP-A ON PULMONARY MACROPHAGES 303
mental design. For example, the cells were isolated and plated
under slightly different conditions. It is possible that these
subtle differences in design induced differences in the respon-
siveness of the aM
. Major differences in the two studies are
the different origin of pulmonary M
, the different source of
human SP-A and the different methods of SP-A isolation. Vari-
ous cell types might respond differentially to SP-A, as well as
the cells from different animals, and it is well known that
interspecies variation in the mediator-induced behavior poten-
tially is a problem when extrapolating from animal studies to
humans. In an attempt to address this issue, we have employed
M
from human origin and we tested two different subpopu-
lations of pulmonary M
. The origin and chemical features of
the SP-A batches employed in these studies must also be taken
into consideration. We isolated SP-A from previously healthy
organ donors. The SP-A used in the Kremlev and Phelps study
(3) was from patients with alveolar proteinosis, whose SP-A
are known to differ from normal subjects (18), and was isolated
by a different technique. Also, SP-A appears to avidly bind
LPS, and it even may enhance presentation of LPS to aM
(19); thus, even a trace contamination of endotoxin with SP-A
might alter the M
response. In agreement with our results,
Rosseau et al. (4) have found that SP-A strongly suppress the
proinflammatory cytokine response of human aM
and mono-
cytes to Candida albicans, effecting down-regulation of proin-
flammatory cytokine synthesis at the transcriptional level. In
this study, internalization of SP-A by M
seemed necessary
for the interference with the cytokine response. In vivo,aM
are continuously in contact with surfactant and SP-A and yet
are not permanently activated. However, it is also recognized
that SP-A likely functions differently in vitro than in the
complex lipid-rich milieu of the alveolar spaces where any
potential proinflammatory effect could be overcome by an
antagonistic effect of another surfactant component. In fact, the
previous in vivo surfactant exposure could also influence the in
vitro behavior of aM
after isolation. This potential influence
of previous surfactant exposure should be negligible in the
iM
subpopulation.
Our findings, that exogenously added cGMP increases the
release of TNF
and IL-1 and that LPS-stimulated TNF
and
IL-1 release can be abolished by the GC inhibitor MB, are in
agreement with the hypothesis of cGMP playing a role in the
regulation of both TNF
and IL-1 production by macrophages
(20). In contrast, a different regulatory mechanism seems to be
present for IL-6. Cyclic GMP is generated by the enzyme GC,
and the best known activator of the soluble isoform of this
enzyme is NO (21). Nevertheless, a high output, inducible
NO-generating system similar to that reported in rats and mice
has not been found in human M
under a variety of conditions
(22, 23). In fact, we did not find any increase in nitrite concen-
tration in the supernatants of LPS-stimulated macrophages
despite their showing an increased production of cGMP and
cytokines, confirming previous findings of our group (6) as
well as others (22, 23). Since GC can be activated also by CO
(21) and we have found this gas can participate in the LPS-
induced cytokine production in human M
(6), we chose to
examine the production of CO in our system. We found an
LPS-induced increase in CO generation by both iM
and aM
.
Hemin, a precursor of CO elicited the same effect serving as a
control. SP-A was able to suppress the LPS-induced produc-
tion of CO on iM
but not on aM
.
At least two sources of CO are available, one of which is the
metabolism of heme (24), catalysed by heme oxygenase. A
second source is lipid peroxidation. Induction of heme oxygen-
ase has been observed in the presence of several oxidant
stresses, especially agents that deplete glutathione (25), and
LPS has been found to induce heme oxygenase activity in rat
macrophages and Kupffer cells (26). Our finding that hemin
increases CO, TNF
, and IL-1 production and cGMP content
indicates that endogenous heme oxygenase-dependent CO
production is effective in triggering the production of both
TNF
and IL-1 by macrophages, and SP-A seems not to inter-
fere with this pathway.
On the other hand, an increase in the generation of oxygen
free radicals is a well-known component in the pathophysiol-
ogy of sepsis. Several specific intracellular signaling mecha-
nism have been found to be influenced by redox changes,
including calcium shifts, tyrosine kinase activation, and the
nuclear transcription factor NF-
B. It has been shown that the
LPS stimulatory pathway in the alveolar macrophage possesses
a signal transduction mechanism that is sensitive to redox
changes (27). Given that SP-A did not interfere with the effect
of exogenously added cGMP or hemin, the inhibitory effect of
SP-A on LPS-induced CO production in iM
might be
explained by an SP-A-mediated attenuation of the LPS-
induced oxygen radical production in iM
. Although SP-A
does not have a scavenger effect for superoxide anion (28),
FIG.6.Production of carbon monoxide by human pulmonary
macrophages in the presence or absence of LPS (10 µg/mL), SP-A
(50 µg/mL), methylene blue (MB, 10
−5
mol/L), hemin (1 µmol/L),
8-Br-cGMP (1 mmol/L), and combinations. Each column represents
the mean ± SE of duplicate samples from 6 different experiments. *
P
<
0.01 vs. all other groups.
304 SHOCK VOL. 14, NO.3 ARIAS-DIAZ ET AL.
several studies suggest that SP-A alters oxygen radical produc-
tion (28). Alveolar M
incubated with SP-A have a decrease in
superoxide production, indicating a dampening of the respira-
tory burst (15, 28) and suggesting that SP-A has a protective
role against the oxidant injury caused by aM
in the lung. In
contrast with these results, van Iwaarden et al. (2) found an
increase in the oxygen radical production by rat aM
induced
by human SP-A.
Collectively, our findings suggest functional differences
between iM
and aM
. The LPS-induced increase in IL-1
release and CO generation was inhibited by SP-A in iM
, but
not in aM
. These findings are also in agreement with earlier
reports of the functional differences between aM
and M
from digested lung tissue (9). Nevertheless, we cannot exclude
a possible influence of the previous in vivo surfactant exposi-
tion of the aM
subpopulation. In addition, there exists the
possibility that the enzyme treatment per se could have influ-
ence the phenotype of the cells, although Johansson et al. (8)
showed that enzyme treatment did not modify the receptor
expression of rat iM
.
The LPS-elicited synthesis of IL-1 in aM
, and IL-6 in both
types of M
was not suppressed by SP-A. This selective effect
together with the finding that SP-A did not affect the baseline
levels of cytokine generation in our M
suggest a distinct
immunomodulatory rather than a general inhibitory effect of
SP-A on M
inflammatory responsiveness.
In conclusion, the major surfactant protein SP-A was found
to inhibit the LPS-induced TNF
response of both interstitial
and alveolar human M
, as well as the IL-1 response in iM
.
The SP-A effect on TNF
production could be mediated by a
suppression in the LPS-induced increase in intracellular
cGMP. In iM
, but not in aM
, SP-A also inhibited the LPS-
induced IL-1 secretion and CO generation. These data lend
further credit to a physiological function of SP-A in regulating
alveolar host defense and inflammation and suggest a funda-
mental role of this apoprotein in limiting excessive proinflam-
matory cytokine release in pulmonary M
during the ARDS.
ACKNOWLEDGMENTS
This report was partially financed by grants from Fondo de Investigacio´nde
la Seguridad Social (FISS 98/1397), and Direccio´n General De Investigacio´n
Cientı´fica Y Te´cnica (DGICYT PB98-0769-C02). We thank the transplant
coordinators and transplant team staff nurses of the Hospital Clinico “San
Carlos” for their kind cooperation in obtaining the lung tissue.
REFERENCES
1. McCormack F: The structure and function of surfactant protein-A. Chest
111:114S–119S, 1997.
2. van Iwaarden F, Welmers B, Verhoef J, Haagsman HP, van Golde LM:
Pulmonary surfactant protein A enhances the host-defense mechanism of
rat alveolar macrophages. Am J Respir Cell Mol Biol 2:91–98, 1990.
3. Kremlev SG, Phelps DS: Surfactant protein A stimulation of inflammatory
cytokine and immunoglobulin production. Am J Physiol 267:L712–L719,
1994.
4. Rosseau S, Hammerl P, Maus U, Gu¨ nther A, Seeger W, Grimminger F,
Lohmeyer J: Surfactant protein A down-regulates proinflammatory cyto-
kine production evoked by Candida albicans in human alveolar macro-
phages and monocytes. J Immunol 163:4495–4502, 1999.
5. Chida S, Phelps DS, Soll RF, Taeusch HW: Surfactant proteins and anti-
surfactant antibodies in sera from infants with respiratory distress
syndrome with and without surfactant treatment. Pediatrics 88:84–89,
1991.
6. Arias-Diaz J, Vara E, Garcia C, Villa N, Balibrea JL: Evidence for a cyclic
guanosine monophosphate-dependent, carbon monoxide-mediated, signal-
ing system in the regulation of TNF
production by human pulmonary
macrophages. Arch Surg 130:1287–1293, 1995.
7. Casals C, Miguel E, Perez-Gil J: Tryptophan fluorescence study on the
interaction of pulmonary surfactant protein A with phospholipid vesicles.
Biochem J 296(Pt 3):585–593, 1993
8. Johansson A, Lundborg M, Sko¨ld CM, Lundahl J, Tornling G, Eklund A,
Camner P: Functional, morphological, and phenotypical differences
between rat alveolar and interstitial macrophages. Am J Respir Cell Mol
Biol 16:582–588, 1997.
9. Zetterberg G, Johansson A, Lundahl J, Lundborg M, Sko¨ld CM, Tornling
G, Camner P, Eklund A: Differences between rat alveolar and interstitial
macrophages 5 wk after quartz exposure. Am J Physiol 274:L226–L234,
1998.
10. Weissler JC, Lyons CR, Lipscomb MF, Toews GB: Human pulmonary
macrophages. Functional comparison of cells obtained from whole lung
and by bronchoalveolar lavage. Am Rev Respir Dis 133:473–477, 1986.
11. Siziopikou KP, Harris JE, Casey L, Nawas Y, Braun DP: Impaired tumori-
cidal function of alveolar macrophages from patients with non-small cell
lung cancer. Cancer 68:1035–1044, 1991.
12. Sherman MP, Campbell LA, Gong HJr, Roth MD, Tashkin DP: Antimi-
crobial and respiratory burst characteristics of pulmonary alveolar macro-
phages recovered from smokers of marijuana alone, smokers of tobacco
alone, smokers of marijuana and tobacco, and nonsmokers. Am Rev Respir
Dis 144:1351–1356, 1991.
13. Phelps DS, Rose RM: Increased recovery of surfactant protein A (SP-A)
in AIDS related pneumonia. Am Rev Respir Dis 143:1072–1075, 1991.
14. Balibrea JL, Arias-Diaz J, Garcia C, Vara E: Effect of pentoxifylline and
somatostatin on tumour necrosis factor production by human pulmonary
macrophages. Circ Shock 43:51–56, 1994.
15. Borron P, Veldhuizen RA, Lewis JF, Possmayer F, Caveney A, Inchley K,
McFadden RG, Fraher LJ: Surfactant associated protein-A inhibits human
lymphocyte proliferation and IL-2 production. Am J Respir Cell Mol Biol
15:115–121, 1996.
16. McIntosh JC, Mervin-Blake S, Conner E, Wright JR: Surfactant protein A
protects growing cells and reduces TNF
activity from LPS-stimulated
macrophages. Am J Physiol 271:L310–L319, 1996.
17. Kremlev SG, Umstead TM, Phelps DS: Effects of surfactant protein A and
surfactant lipids on lymphocyte proliferation in vitro.Am J Physiol
267:L357–L364, 1994.
18. Voss T, Melchers K, Scheirle G, Schaefer KP: Structural comparison of
recombinant pulmonary surfactant protein SP-A derived from two human
coding sequences: implications for the chain composition of natural
human SP-A. Am J Respir Cell Mol Biol 4:88–92, 1991.
19. Stamme C, Wright JR: Surfactant protein A enhances the binding and
deacylation of E. coli LPS by alveolar macrophages. Am J Physiol
276:L540–L547, 1999.
20. Renz H, Gong JH, Schmidt A, Nain M, Gemsa D: Release of tumor
necrosis factor-
from macrophages. Enhancement and suppression are
dose-dependently regulated by prostaglandin E
2
and cyclic nucleotides. J
Immunol 141:2388–2393, 1988
21. Furchgott RF, Jothianandan D: Endothelium-dependent and -independent
vasodilation involving cyclic GMP: relaxation induced by nitric oxide,
carbon monoxide and light. Blood Vessels 28:52–61, 1991.
22. Sakai N, Milstien S: Availability of tetrahydrobiopterin is not a factor in
the inability to detect nitric oxide production by human macrophages.
Biochem Biophys Res Commun 193:378–383, 1993.
23. Schneemann M, Schoedon G, Hofer S, Blau N, Guerrero L, Schaffner A:
Nitric oxide synthase is not a constituent of the antimicrobial armature of
human mononuclear phagocytes. J Infect Dis 167:1358–1363, 1993.
24. Coburn RF, Williams WJ, White P, Kahn SB: The production of carbon
monoxide from hemoglobin in vivo.J Clin Invest 46:346–356, 1967.
25. Lautier D, Luscher P, Tyrrell RM: Endogenous glutathione levels modu-
late both constitutive and UVA radiation/hydrogen peroxide inducible
expression of the human heme oxygenase gene. Carcinogenesis 13:227–
232, 1992.
SHOCK SEPTEMBER 2000 EFFECT OF SP-A ON PULMONARY MACROPHAGES 305
26. Gemsa D, Woo CH, Fudenberg HH, Schmid R: Stimulation of heme
oxygenase in macrophages and liver by endotoxin. J Clin Invest 53:647–
651, 1974.
27. Mendez C, Garcia I, Maier RV: Oxidants augment endotoxin-induced
activation of alveolar macrophages. Shock 6:157–163, 1996.
28. Katsura H, Kawada H, Konno K: Rat surfactant apoprotein A (SP-A)
exhibits antioxidant effects on alveolar macrophages. Am J Respir Cell
Mol Biol 9:520–525, 1993.
DISCUSSION
DR. MAIER: I appreciate the opportunity to discuss this well-performed,
extensive study. As the authors have shown, there is an impact of human SPA
on pulmonary macrophage function in the lung with a selective effect on the
intra-alveolar versus the interstitial populations. A significant advantage to
their studies is that they used human tissues as a source for cells and awesome
products. They used normal tissue and, as such, avoided many of the potential
artifacts and complicating factors that have been reported in the literature.
They show that SPA inhibits the production of both macrophage population’s
production of TNF and a selective inhibition of IL-1 in the interstitial macro-
phage, while there is no effect on IL-6 or nitric oxide production.
As published previously, the authors have demonstrated a carbon monox-
ide-dependent induction in cGMP, which appears to be the mechanism for
upregulating TNF in the alveolar macrophage. In the current study SPA blocks
cGMP and TNF in the alveolar macrophage population, yet does not block
carbon monoxide production. Therefore inhibition of CO production in the
alveolar macrophage is not the cause of the decreased GMP. Do the authors
have any suggestions or data to help us understand at what point SPA is
working in the alveolar macrophage that inhibits TNF production but not
carbon monoxide.
I would also ask the authors to extend their comments on the LPS interac-
tions in their experiments. The amount of LPS used was extremely high, at 15
mcg/ml. We and others have shown maximal production after 10 to 100 ng of
LPS in human cells rather than these enormous doses. There are recent data
showing that at extremely high doses, LPS may signal the macrophage through
both the CD-14-dependent pathway and also a Toll protein-independent path-
way. Do the authors have data at lower doses of LPS which would avoid this
complicating dual pathway signaling to confirm SPA has an effect on the
CD-14-dependent pathway? In addition, in their experimental methods I could
not tell if they provided serum and therefore a source for lipopolysaccharide-
binding protein (LPB). Was there serum present in their experimental condi-
tions? Without LBP, it is highly likely that LPS was not signaling through
CD-14.
Lastly, while SPA appears to have an effect, SPA, as they pointed out,
constitutes only 5 to 10% of total surfactant in vivo. It is well known that the
other phospholipids in surfactant also have significant LPS-binding and
immunomodulatory effects. Have the authors added back increasing doses of
SPA to depleted surfactant to establish the relative contribution of SPA to
intact surfactant? This would establish a clinical relevance to SPA separate
from the effect of surfactant overall?
I would like to thank the Society for the opportunity to discuss this paper.
DR. ARIAS-DIAZ: Thank you, Dr. Maier, for your interesting comments.
In the first place, previous studies of our group about carbon monoxide regu-
lating TNF production were performed exclusively using the interstitial variety
of macrophages. So the different behavior of carbon monoxide in alveolar
macrophages was an unexpected finding. SP-A was able to suppress the LPS-
induced production of carbon monoxide on interstitial macrophages, but not on
alveolar macrophages. However, SP-A antagonized the LPS-induced increase
in cGMP and TNF in both types of cells. In our opinion, other guanylate-
cyclase-stimulating molecule, different from nitric oxide and carbon monox-
ide, could be the responsible for the cGMP increase in alveolar macrophages.
We currently haven’t any other explanation for this finding.
With respect to the LPS concentration used by us, I agree it is a high LPS
concentration, but we think it may be closer to the one present in pathological
conditions. In the purulent exudate of a gram-negative pneumonia, LPS
concentration is known to be very high. At 10 micrograms per mL, LPS might
bind to membranes and different receptors than CD-14. Nevertheless, it contin-
ues to be a valid conclusion that SP-A is able to overcome some of the LPS
effects either CD-14-specific or unspecific.
With regard to the presence of serum in the cultures, we add 10% fetal calf
serum to the RPMI-1640 media.
Finally, regarding whether we have studied the effect of SP-A in combi-
nation with the lipid component of surfactant, yes we did. We have repeated
some of these experiments with SP-A with and without either phosphatidyl-
choline or dipalmitoylphosphatidylcholine. We found that neither phosphati-
dylcholine nor dipalmitoylphosphatidylcholine had any effect on TNF produc-
tion in the absence of SP-A. Interestingly, they did not interfere with the effect
of SP-A on the production of cytokines by LPS-stimulated pulmonary macro-
phages, in spite that we had previously found that SP-A binds and aggregates
phospholipid vesicles in the RPMI-1640 medium.
306 SHOCK VOL. 14, NO.3 ARIAS-DIAZ ET AL.
... cGMP has different effects on inflammation under stimulated versus unstimulated conditions [28]. In unstimulated conditions, treatment with GC activators (e.g., NO donor, ANP) enhances expression of inflammatory mediators including NFκB activity and proinflammatory cytokines [28][29][30][31][32]. Whereas these cGMP modulating treatments attenuate inflammatory responses in LPS-or TNF-α-stimulated conditions [28,[31][32][33][34][35][36][37][38][39][40][41][42]. ...
... In unstimulated conditions, treatment with GC activators (e.g., NO donor, ANP) enhances expression of inflammatory mediators including NFκB activity and proinflammatory cytokines [28][29][30][31][32]. Whereas these cGMP modulating treatments attenuate inflammatory responses in LPS-or TNF-α-stimulated conditions [28,[31][32][33][34][35][36][37][38][39][40][41][42]. Decreasing GC activity and thus cGMP formation by using GC inhibitors suppresses cGMP effects on inflammation in both unstimulated and LPS-stimulated conditions [29,30,39]. However, there were controversial results; for instance in LPS stimulated conditions cGMP treatment increases TNF-α protein levels [43] and downregulating cellular cGMP with GC inhibitors reduced LPS-induced TNF-α production [29,30]. ...
... Decreasing GC activity and thus cGMP formation by using GC inhibitors suppresses cGMP effects on inflammation in both unstimulated and LPS-stimulated conditions [29,30,39]. However, there were controversial results; for instance in LPS stimulated conditions cGMP treatment increases TNF-α protein levels [43] and downregulating cellular cGMP with GC inhibitors reduced LPS-induced TNF-α production [29,30]. Notably, the impact of NO on inflammation appears to be more complicated. ...
Article
Full-text available
Interleukin-1 receptor-associated kinase-3 (IRAK3) is a critical checkpoint molecule of inflammatory responses in the innate immune system. The pseudokinase domain of IRAK3 contains a guanylate cyclase (GC) centre that generates small amounts of cyclic guanosine monophosphate (cGMP) associated with IRAK3 functions in inflammation. However, the mechanisms of IRAK3 actions are poorly understood. The effects of low cGMP levels on inflammation are unknown, therefore a dose–response effect of cGMP on inflammatory markers was assessed in THP-1 monocytes challenged with lipopolysaccharide (LPS). Sub-nanomolar concentrations of membrane permeable 8-Br-cGMP reduced LPS-induced NFκB activity, IL-6 and TNF-α cytokine levels. Pharmacologically upregulating cellular cGMP levels using a nitric oxide donor reduced cytokine secretion. Downregulating cellular cGMP using a soluble GC inhibitor increased cytokine levels. Knocking down IRAK3 in THP-1 cells revealed that unlike the wild type cells, 8-Br-cGMP did not suppress inflammatory responses. Complementation of IRAK3 knockdown cells with wild type IRAK3 suppressed cytokine production while complementation with an IRAK3 mutant at GC centre only partially restored this function. Together these findings indicate low levels of cGMP form a critical component in suppressing cytokine production and in mediating IRAK3 action, and this may be via a cGMP enriched nanodomain formed by IRAK3 itself.
... SP-A-enhanced degradation of LPS-induced TLR4 is associated with a Rab7dependent decrease in TNF-a release. Previous data demonstrated that SP-A reduces LPS-induced TNF-a release in vitro and in vivo (14,(44)(45)(46)(47)(48). To ascertain whether Rab7 is required for SP-A-mediated inhibition of LPS-induced TNF-a release, AMs from wildtype and SP-A 2/2 mice were treated with LPS, or SP-A prior to LPS in the absence or presence of Rab7 blocking peptides and TNF-a release was determined by ELISA. ...
Article
Full-text available
Respiratory infections by Gram-negative bacteria are a major cause of global morbidity and mortality. Alveolar macrophages (AMs) play a central role in maintaining lung immune homeostasis and host defense by sensing pathogens via pattern recognition receptors (PRR). The PRR Toll-like receptor (TLR) 4 is a key sensor of lipopolysaccharide (LPS) from Gram-negative bacteria. Pulmonary surfactant is the natural microenvironment of AMs. Surfactant protein A (SP-A), a multifunctional host defense collectin, controls LPS-induced pro-inflammatory immune responses at the organismal and cellular level via distinct mechanisms. We found that SP-A post-transcriptionally restricts LPS-induced TLR4 protein expression in primary AMs from healthy humans, rats, wild-type and SP-A -/- mice by further decreasing cycloheximide-reduced TLR4 protein translation and enhances the co-localization of TLR4 with the late endosome/lysosome. Both effects as well as the SP-A-mediated inhibition of LPS-induced TNFα release are counteracted by pharmacological inhibition of the small GTPase Rab7. SP-A-enhanced Rab7 expression requires β-arrestin2 and, in β-arrestin2 -/- AMs and after intratracheal LPS challenge of β-arrestin2 -/- mice, SP-A fails to enhance TLR4/lysosome co-localization and degradation of LPS-induced TLR4. In SP-A -/- mice, TLR4 levels are increased after pulmonary LPS challenge. SP-A-induced activation of mechanistic target of rapamycin complex 1 (mTORC1) kinase requires β-arrestin2 and is critically involved in degradation of LPS-induced TLR4. The data suggest that SP-A post-translationally limits LPS-induced TLR4 expression in primary AMs by lysosomal degradation comprising Rab7, β-arrestin2, and mTORC1. This study may indicate a potential role of SP-A-based therapeutic interventions in unrestricted TLR4-driven immune responses to lower respiratory tract infections caused by Gram-negative bacteria.
... Macrophage cultures were plated in triplicate wells, and each series of experiments was repeated at least three times. Measurement of TNF-α production in supernatants of rat aMϕs was performed using specific ELISA kit following the supplier's instructions (BD Biosciences, San Diego, CA) [34,[38][39][40]. Statistics: Data are presented as means ± SEM. ...
Article
Full-text available
The coating composition of nanomedicines is one of the main features in determining the medicines' fate, clearance, and immunoresponse in the body. To highlight the coatings' impact in pulmonary administration, two micellar superparamagnetic iron oxide nanoparticles (SPION) were compared. These nanoparticles are similar in size and charge but have different coatings: either phosphatidylcholine (PC-SPION) or bovine serum albumin (BSA-SPION). The aim of the study was to increase the understanding of the nano-bio interaction with the cellular and non-cellular components of the lung and underline valuable coatings either for local lung-targeted drug delivery in theranostic application or patient-friendly route systemic administration. PC-SPION and BSA-SPION were deposited in the alveoli by in vivo instillation and, despite the complexity of imaging the lung, SPION were macroscopically visualized by MRI. Impressively, PC-SPION were retained within the lungs for at least a week, while BSA-SPION were cleared more rapidly. The different lung residence times were confirmed by histological analysis and supported by a flow cytometry analysis of the SPION interactions with different myeloid cell populations. To further comprehend the way in which these nanoformulations interact with lung components at the molecular level, we used fluorescence spectroscopy, turbidity measurements, and dynamic light scattering to evaluate the interactions of the two SPION with surfactant protein A (SP-A), a key protein in setting up the NP behavior in the alveolar fluid. We found that SP-A induced aggregation of PC-SPION, but not BSA-SPION, which likely caused PC-SPION retention in the lung without inducing inflammation. In conclusion, the two SPION show different outcomes from interaction with SP-A leading to distinctive fate in the lung. PC-SPION hold great promise as imaging and theranostic agents when prolonged pulmonary drug delivery is required.
... 22 The remaining surfactant fraction is composed of two classes of surfactant proteins (SPs), the hydrophilic SP-A and SP-D and the hydrophobic SP-B and SP-C. 23 While the larger hydrophilic SPs mainly have a role in the innate immune system and therefore are removed from the clinical preparations, [24][25][26] their smaller hydrophobic counterparts play an essential role in surface tension reduction. [27][28][29][30] PS dynamics at the alveolar interface have been investigated from both a biophysical and clinical point of view. ...
Article
RNA therapeutics are poised to revolutionize medicine. To unlock the full potential of RNA drugs, safe and efficient (nano)formulations to deliver them inside target cells are required. Endosomal sequestration of nanocarriers represents a major bottleneck in nucleic acid delivery. Gaining more detailed information on the intracellular behavior of RNA nanocarriers is crucial to rationally develop delivery systems with improved therapeutic efficiency. Surfactant protein B (SP-B) is a key component of pulmonary surfactant (PS), essential for mammalian breathing. In contrast to the general belief that PS should be regarded as a barrier for inhaled nanomedicines, we recently discovered the ability of SP-B to promote gene silencing by siRNA-loaded and lipid-coated nanogels. However, the mechanisms governing this process are poorly understood. The major objective of this work was to obtain mechanistic insights into the SP-B-mediated cellular delivery of siRNA. To this end, we combined siRNA knockdown experiments, confocal microscopy, and focused ion beam scanning electron microscopy imaging in an in vitro non-small-cell lung carcinoma model with lipid mixing assays on vesicles that mimic the composition of (intra)cellular membranes. Our work highlights a strong correlation between SP-B-mediated fusion with anionic endosomal membranes and cytosolic siRNA delivery, a mode of action resembling that of certain viruses and virus-derived cell-penetrating peptides. Building on these gained insights, we optimized the SP-B proteolipid composition, which dramatically improved delivery efficiency. Altogether, our work provides a mechanistic understanding of SP-B-induced perturbation of intracellular membranes, offering opportunities to fuel the rational design of SP-B-inspired RNA nanoformulations for inhalation therapy.
... Macrophage cultures were plated in triplicate wells, and each series of experiments was repeated at least three times. Measurement of TNF-α production in supernatants of rat aMϕs was performed using specific ELISA kit following the supplier's instructions (BD Biosciences, San Diego, CA) [34,[38][39][40]. Statistics: Data are presented as means ± SEM. ...
... The results revealed that inhibited SP-A and SP-B caused decreased cell viability, increased apoptosis, and induced inflammatory reaction in cells. A number of studies have concluded that SP-A and SP-B can inhibit inflammatory reaction (23)(24)(25). In addition, SP-A also plays an important role in cell apoptosis (11). ...
Article
Full-text available
This study explored how interleukin-8 (IL-8) causes acute lung injury (ALI) through pulmonary surfactant protein A (SP-A) and surfactant protein B (SP-B). Serum was collected from 53 ALI patients and further 56 healthy subjects who underwent physical examination. The IL-8, SP-A, and SP-B levels were determined using enzyme-linked immunosorbent assay (ELISA). An ALI model was constructed using lipopolysaccharide (LSP)-induced normal A549 cells. siRNA was employed to interfere with the expression of IL-8, SP-A and SP-B. Western blot analysis was carried out to determine the protein levels, and MTT assay to determine the cell activity. In addition, co-immunoprecipitation (Co-IP) assay was used to verify the interaction between IL-8, SP-A and SP-B. ALI patients showed high expression of serum IL-8, and low expression of SP-A and SP-B, and IL-8 was negatively correlated with SP-A and SP-B, respectively. LSP-induced normal A549 cells showed increased expression of IL-8 and decreased expression of SP-A and SP-B. Silencing IL-8 led to increased expression levels of SP-A, SP-B and Bcl2, decreased expression levels of caspase-9, caspase-3, Bax, TNF-α, IL-17 and IL-1β, reduced cell apoptosis rate, and enhanced cell viability. Silencing SP-A and SP-B resulted in increased expression of IL-8, caspase-9, caspase-3, Bax, TNF-α, IL-17 and IL-1β, and decreased expression of Bcl2. Co-IP assay revealed that IL-8 could interact with SP-A and SP-B, respectively. IL-8 induces apoptosis by inhibiting SP-A and SP-B, and intensifies cellular inflammatory reaction, leading eventually to ALI.
... This is in accordance with a previous study that concluded that SP-A can modulate lung inflammation in humans by modulating LPSinduced production of pro-inflammatory cytokines by alveolar macrophage and increase the antibacterial and antiviral functions of macrophages. 38 We have also shown that the presence of SP-A significantly increased the phagocytic capacity of alveolar macrophages. SP-A can enhance phagocytosis through opsonization and also directly stimulate phagocytosis by the upregulation of cell surface phagocytic receptors in macrophages. ...
Article
Full-text available
Surfactant protein A (SP-A) plays an important role in innate immune response and host defense against various microorganisms through opsonization and complement activation. To investigate the role of SP-A in non-typeable Haemophilus influenzae (NTHi)-induced acute otitis media, this study used wild type C57BL/6 (WT) and SP-A knockout (KO) mice. We divided mice into an infection group in which the middle ear (ME) was injected with NTHi and a control group that received the same treatment using normal saline. Mice were sacrificed on d 1, 3, and 7 after treatment. Temporal bone samples were fixed for histological, cellular, and molecular analyses. Ear washing fluid (EWF) was collected for culture and analyses of pro-inflammatory cytokines and inflammatory cells. SP-A-mediated bacterial aggregation and killing and phagocytosis by macrophages were studied in vitro. SP-A expression was detected in the ME and Eustachian tube mucosa of WT mice but not KO mice. After infection, KO mice showed more severe inflammation evidenced by increased ME mucosal thickness and inflammatory cell infiltration and higher NF-κB activation compared to WT mice. The levels of IL-6 and IL-1β in the EWF of infected KO mice were higher compared to infected WT mice on d 1. Our studies demonstrated that SP-A mediated NTHi aggregation and killing and enhanced bacterial phagocytosis by macrophages in vitro and modulated inflammation of the ME in otitis media in vivo.
... SP-A can also block binding of ligands to TLR2 and TLR4 receptors [122] and it is able to regulate TLRs expression in human macrophages [157]. In another studies SP-A decreased release of TNF-α from LPS-stimulated rat and human AM [110,158]. SP-D has a key regulatory role, because SP-D deficient mice exhibit constitutive activation of AM [159]. However, in contrast to SP-A, SP-D was shown to slightly enhance TNF-α production in LPS-stimulated AM [160]. ...
Article
Full-text available
The main function of the lungs is oxygen transport from the atmosphere into the blood circulation, while it is necessary to keep the pulmonary tissue relatively free of pathogens. This is a difficult task because the respiratory system is constantly exposed to harmful substances entering the lungs by inhalation or via the blood stream. Individual types of lung cells are equipped with the mechanisms that maintain pulmonary homeostasis. Because of the clinical significance of acute respiratory distress syndrome (ARDS) the article refers to the physiological role of alveolar epithelial cells type I and II, endothelial cells, alveolar macrophages, and fibroblasts. However, all these cells can be damaged by lipopolysaccharide (LPS) which can reach the airspaces as the major component of the outer membrane of Gram-negative bacteria, and lead to local and systemic inflammation and toxicity. We also highlight a negative effect of LPS on lung cells related to alveolar-capillary barrier and their response to LPS exposure. Additionally, we describe the molecular mechanism of LPS signal transduction pathway in lung cells.
... The latter is also regulated by the binding of SP-A to the cell surface of AM via its CRD domain. SP-A inhibits and stimulates at the same time inflammatory responses of AM reducing the LPS-induced TNF-α production or inducing an opsonin-like mechanism involved in phagocytosis of pathogens and inhaled nanoparticles[45,48].This opsonization model was assessed by Ruge et al. using magnetite nanoparticles (mNPs) coated with different materials to modulate their surface properties. Here, SP-A binding induced the formation of mNP aggregates, thereby promoting subsequent scavenging by phagocytic cells. ...
Article
Pulmonary surfactant (PS) has been extensively studied because of its primary role in mammalian breathing. The deposition of this surface-active material at the alveolar air-water interface is essential to lower surface tension, thus avoiding alveolar collapse during expiration. In addition, PS is involved in host defense, facilitating the clearance of potentially harmful particulates. PS has a unique composition, including 92% of lipids and 8% of surfactant proteins (SPs) by mass. Although they constitute the minor fraction, SPs to a large extent orchestrate PS-related functions. PS contains four surfactant proteins (SPs) that can be structurally and functionally divided in two groups, i.e. the large hydrophilic SP-A and SP-D and the smaller hydrophobic SP-B and SP-C. The former belong to the family of collectins and are involved in opsonization processes, thus promoting uptake of pathogens and (nano)particles by phagocytic cell types. The latter SPs regulate interfacial surfactant adsorption dynamics, facilitating (phospho)lipid transfer and membrane fusion processes. In the context of pulmonary drug delivery, the exploitation of PS as a carrier to promote drug spreading along the alveolar interface is gaining interest. In addition, recent studies investigated the interaction of PS with drug-loaded nanoparticles (nanomedicines) following pulmonary administration, which strongly influences their biological fate, drug delivery efficiency and toxicological profile. Interestingly, the specific biophysical mode-of-action of the four SPs affect the drug delivery process of nanomedicines both on the extra-and intracellular level, modulating pulmonary distribution, cell targeting and intracellular delivery. This knowledge can be harnessed to exploit SPs for the design of unique and bio-inspired drug delivery strategies.
Article
The objective of this study was to investigate the expression and the role of surfactant protein A (SP-A) in the middle ear (ME) mucosa in response to bacterial infection in a rat model. Otitis media (OM) was induced by surgical inoculation of non-typeable Haemophilus influenza (NTHi) into the ME cavity of Sprague-Dawley rats. The rats were divided into an NTHi-induced OM group and a phosphate-buffered saline-injected control group. The NTHi-induced OM and control groups were subdivided into sets of 6 rats, one for each of the 6 time points (0, 1, 2, 4, 7, and 14 days post-inoculation), at which point the rats were euthanized after inoculation. The concentrations of SP-A in the ME effusion were determined by an enzyme-linked immunosorbent assay (ELISA). Tissue expression of SP-A, interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) in infected ME mucosa was assessed by immunohistochemical staining. For mRNA expression quantification, RNA was extracted from the ME mucosa and SP-A expression was monitored and compared between the control and OM groups using quantitative polymerase chain reaction (PCR). Expression of IL-1β IL-6, and TNF-α in the ME mucosa was also evaluated. SP-A expression was evaluated in the effusion of pediatric OM patients (70 ears) who received ventilation-tube insertion by ELISA. SP-A was detected in normal rat ME mucosa before bacterial inoculation. SP-A expression was up-regulated in the NTHi-induced OM group (p = 0.046). Immunohistochemical staining revealed increased SP-A expression on post-inoculation day 1, 2, and 4 in the OM group. Expression of proinflammatory cytokines (IL-1β IL-6, and TNF-α) in the ME also increased significantly on post-inoculation day 1, 2, and 4 in the OM group. It correlated with changes in SP-A expression. Expression of SP-A was also identified in the ME effusion of humans. SP-A exists in the ME of the rat and was up-regulated in the ME of NTHi-induced OM. Expression of IL-1β IL-6, and TNF-α was increased in the ME of the bacteria-induced OM in the rat model. The results suggest that SP-A may play a significant role in the early phase of OM induction and subsequent recovery from it.
Article
Full-text available
Surfactant protein A (SP-A) has been implicated in the regulation of pulmonary host defense and inflammatory events. We analyzed the impact of SP-A on the Candida albicans-induced cytokine response in human alveolar macrophages (AM) and its precursor cells, the monocytes, which rapidly expand the alveolar mononuclear phagocyte pool under inflammatory conditions. Both recombinant human SP-A and natural canine SP-A were employed. SP-A dose-dependently down-regulated the proinflammatory cytokine response of AM and monocytes to both viable and nonviable Candida, including TNF-α, IL-1β, macrophage inflammatory protein-1α, and monocyte chemoattractant protein-1. In contrast, SP-A did not affect the baseline liberation of these cytokines. The release of the antiinflammatory cytokines IL-1 receptor antagonist and IL-6 was not inhibited by SP-A under baseline conditions and in response to fungal challenge. The inhibitory effect of SP-A on proinflammatory cytokine release was retained upon reassembly of the apoprotein with natural surfactant lipids and in the presence of serum constituents, for mimicry of plasma leakage into the alveolar space. It was not reproduced by the homologous proteins complement component C1q and type IV collagen. It was independent of Candida-SP-A binding and phagocyte C1q receptor occupancy, but apparently demanded SP-A internalization by the mononuclear phagocytes, effecting down-regulation of proinflammatory cytokine synthesis at the transcriptional level. We conclude that SP-A limits excessive proinflammatory cytokine release in AM and monocytes confronted with fungal challenge in the alveolar compartment. These data lend further credit to an important physiological role of SP-A in regulating alveolar host defense and inflammation.
Article
Full-text available
Rodent studies indicate that marijuana smoke can adversely affect the antimicrobial function of pulmonary alveolar macrophages (PAM). To evaluate whether marijuana smoke similarly affects human PAM, we compared phagocytosis, fungistatic/fungicidal activity, and superoxide anion (O2-) production of PAM recovered from marijuana smokers (MS), tobacco smokers (TS), marijuana and tobacco smokers (MTS), and nonsmokers (NS). Although PAM from the four groups were equally capable of ingesting Candida albicans, the macrophages from smokers differed from normal PAM in their ability to restrict intracellular yeast germination (ungerminated candida in smokers' PAM = 68 +/- 3% [46] versus NS = 54 +/- 6% [17], mean +/- SEM [n], p = 0.022). Despite heightened fungistatic activity, PAM from MS and TS destroyed significantly fewer intracellular yeast (28 +/- 2 and 29 +/- 2%, respectively) after 4 h than did macrophages recovered from NS (40 +/- 4%, p less than 0.05). Both basal and stimulated (dihydrocytochalasin B + opsonized zymosan or phorbol myristate acetate) O2- production were similar in PAM from MS and NS, but significantly increased in PAM from TS (p less than 0.05). Our findings indicate that marijuana smoking does not alter the phagocytic behavior or the respiratory burst of human PAM, but marijuana smoking does decrease the ability of human PAM to destroy ingested Candida albicans. These findings contrast with the effects of tobacco smoking, which not only decreases the fungicidal activity of human PAM but also increases their respiratory burst.
Article
Surfactant protein A (SP-A) has been implicated in the regulation of pulmonary host defense and inflammatory events. We analyzed the impact of SP-A on the Candida albicans-induced cytokine response in human alveolar macrophages (AM) and its precursor cells, the monocytes, which rapidly expand the alveolar mononuclear phagocyte pool under inflammatory conditions. Both recombinant human SP-A and natural canine SP-A were employed. SP-A dose-dependently down-regulated the proinflammatory cytokine response of AM and monocytes to both viable and nonviable Candida, including TNF-α, IL-1β, macrophage inflammatory protein-1α, and monocyte chemoattractant protein-1. In contrast, SP-A did not affect the baseline liberation of these cytokines. The release of the antiinflammatory cytokines IL-1 receptor antagonist and IL-6 was not inhibited by SP-A under baseline conditions and in response to fungal challenge. The inhibitory effect of SP-A on proinflammatory cytokine release was retained upon reassembly of the apoprotein with natural surfactant lipids and in the presence of serum constituents, for mimicry of plasma leakage into the alveolar space. It was not reproduced by the homologous proteins complement component C1q and type IV collagen. It was independent of Candida-SP-A binding and phagocyte C1q receptor occupancy, but apparently demanded SP-A internalization by the mononuclear phagocytes, effecting down-regulation of proinflammatory cytokine synthesis at the transcriptional level. We conclude that SP-A limits excessive proinflammatory cytokine release in AM and monocytes confronted with fungal challenge in the alveolar compartment. These data lend further credit to an important physiological role of SP-A in regulating alveolar host defense and inflammation.
Article
Respiratory infection with Pneumocystis carinii (PC) is the most frequent serious opportunistic infection in the clinical setting of acquired immunodeficiency syndrome (AIDS). The factors responsible for the predisposition of human immunodeficiency virus (HIV)-infected patients for PC infection are not fully understood. We postulated that changes in the alveolar lining material (ALM) could play a role in the pathogenesis of PC infection in AIDS. We have compared constituents of ALM in bronchoalveolar lavage fluid from normal, nonsmoking volunteers with that of HIV-infected patients with pneumonia. Using an ELISA, we found that surfactant protein A (SP-A) was markedly elevated in the pneumonia patients. Mean SP-A values for the normal nonsmoking individuals (n = 21) were 1.50 +/- 0.25 micrograms/ml (mean +/- SEM). SP-A levels in the HIV-infected patients (n = 22) were significantly elevated (p less than 0.01) with a mean of 5.23 +/- 0.54 micrograms/ml. This increase was greatest in the patients with mor
Article
The capacity of alveolar macrophages and peripheral blood monocytes from patients with non-small cell lung cancer to develop tumoricidal function after in vitro stimulation with different macrophage activators was investigated. Alveolar macrophages were found to be impaired in their ability to develop cytotoxic activity compared with either the peripheral blood monocytes from the same patients or alveolar macrophages from patients with nonmalignant lung disorders. This result was observed consistently under diverse culture conditions and with different macrophage activators including gamma-interferon (gamma-IFN), granulocyte-macrophage colony-stimulating factor (GM-CSF), phorbol myristate acetate, or endotoxin. The impairment in tumoricidal function observed in alveolar macrophages was not associated with reduced target cell binding compared to peripheral blood monocytes. Alveolar macrophages from patients with lung cancer were found to secrete significantly greater amounts of tumor necrosis factor (TNF) and interleukin-1 (IL-1) than either peripheral blood monocytes from the same patients or alveolar macrophages from the patients with nonmalignant disorders. These results are consistent with either different regulatory pathways for cytotoxicity and cytokine secretion in the alveolar macrophages of patients with lung cancer or diversity in the subpopulations of cells responsible for these functions.
Article
Induction of the expression of the mammalian heme oxygenase gene appears to be a general response to oxidant stress. In view of the role of glutathione in protecting cells against solar UVA radiation and other forms of oxidant stress, we have investigated the relationship between intracellular glutathione levels and the inducibility of the human heme oxygenase gene after treatment of populations of cultured skin fibroblasts with either UVA radiation or hydrogen peroxide. We observe a clear relationship between cellular glutathione status and both the constitutive and oxidant-inducible accumulation of heme oxygenase mRNA. Glutathione depletion may lead to enhanced gene expression either as a result of the potentiated accumulation of active oxygen intermediates or as a result of the direct influence of glutathione on a critical target involved in signal transduction.
Article
The characteristics of carbon monoxide (CO)-induced, endothelium-independent relaxation of rabbit aorta were compared with those of nitric oxide (NO)-induced and light-induced relaxation and endothelium-dependent relaxation mediated by endothelium-dependent relaxing factor (EDRF). CO was less than one thousandth as potent as NO as a relaxant. Various findings, including an increase in cyclic GMP associated with CO-induced relaxation, led to the conclusion that CO - like NO, EDRF and light - produces relaxation as a result of its stimulation of guanylate cyclase. LY 83583, which generates superoxide, was a potent, fast-acting inhibitor of acetylcholine-induced endothelium-dependent relaxation and NO-induced relaxation, and a fairly potent, moderately fast-acting inhibitor of photorelaxation, but only a very weak inhibitor of CO-induced relaxation. The ability of LY 83583 as well as hemoglobin to inhibit photorelaxation is consistent with the hypothesis that on radiation a photo-induced relaxing factor is formed which can stimulate guanylate cyclase and which can be inactivated by superoxide and by hemoglobin.
Article
The pulmonary surfactant-associated protein SP-A is encoded by presumably two different genes, resulting in slightly different amino acid sequences. Both gene products were expressed in Chinese hamster ovary cells. Their macromolecular structure differed significantly. SP-A alpha 3 exhibited a much higher amount of tetrameric to hexameric structures than SP-A alpha 2, for which dimeric structures predominate. These differences may be caused by the higher expression rates of SP-A alpha 3 presumably due to the presence of introns in the sequence. The occurrence of irregular disulfide links between individual oligomeric SP-A molecules composed of alpha 3-chains together with the demonstrated presence of both gene products in natural human SP-A suggest that the subunits of SP-A are heterotrimers of one alpha 2- and two alpha 3-chains.
Article
Respiratory infection with Pneumocystis carinii (PC) is the most frequent serious opportunistic infection in the clinical setting of acquired immunodeficiency syndrome (AIDS). The factors responsible for the predisposition of human immunodeficiency virus (HIV)-infected patients for PC infection are not fully understood. We postulated that changes in the alveolar lining material (ALM) could play a role in the pathogenesis of PC infection in AIDS. We have compared constituents of ALM in bronchoalveolar lavage fluid from normal, nonsmoking volunteers with that of HIV-infected patients with pneumonia. Using an ELISA, we found that surfactant protein A (SP-A) was markedly elevated in the pneumonia patients. Mean SP-A values for the normal nonsmoking individuals (n = 21) were 1.50 +/- 0.25 micrograms/ml (mean +/- SEM). SP-A levels in the HIV-infected patients (n = 22) were significantly elevated (p less than 0.01) with a mean of 5.23 +/- 0.54 micrograms/ml. This increase was greatest in the patients with more clinically severe pneumonia. The increase in SP-A did not appear to be pathogen-specific as it was also observed in cases of non-PC pneumonia. We also found that total protein levels were nearly five times higher in the HIV-infected pneumonia patients. These studies indicate that the protein component of the ALM is markedly different from normal in cases of HIV-associated PC and non-PC infection. Further investigation is needed to determine the mechanism of these alterations and their role, if any, in AIDS-related pneumonia.