[go: up one dir, main page]

Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2009 Dec 29;107(1):320–325. doi: 10.1073/pnas.0909361107

Pulmonary surfactant phosphatidylglycerol inhibits respiratory syncytial virus–induced inflammation and infection

Mari Numata a, Hong Wei Chu a, Azzeddine Dakhama b, Dennis R Voelker a,1
PMCID: PMC2806703  PMID: 20080799

Abstract

Respiratory syncytial virus (RSV) is the most common cause of hospitalization for respiratory tract infection in young children. It is also a significant cause of morbidity and mortality in elderly individuals and in persons with asthma and chronic obstructive pulmonary disease. Currently, no reliable vaccine or simple RSV antiviral therapy is available. Recently, we determined that the minor pulmonary surfactant phospholipid, palmitoyl-oleoyl-phosphatidylglycerol (POPG), could markedly attenuate inflammatory responses induced by lipopolysaccharide through direct interactions with the Toll-like receptor 4 (TLR4) interacting proteins CD14 and MD-2. CD14 and TLR4 have been implicated in the host response to RSV. Treatment of bronchial epithelial cells with POPG significantly inhibited interleukin-6 and -8 production, as well as the cytopathic effects induced by RSV. The phospholipid bound RSV with high affinity and inhibited viral attachment to HEp2 cells. POPG blocked viral plaque formation in vitro by 4 log units, and markedly suppressed the expansion of plaques from cells preinfected with the virus. Administration of POPG to mice, concomitant with viral infection, almost completely eliminated the recovery of virus from the lungs at 3 and 5 days after infection, and abrogated IFN-γ (IFN-γ) production and the enhanced expression of surfactant protein D (SP-D). These findings demonstrate an important approach to prevention and treatment of RSV infections using exogenous administration of a specific surfactant phospholipid.

Keywords: antiviral, innate immunity, respiratory epithelium


Respiratory syncytial virus (RSV) is an important pathogen that infects 98% of children within the first 2 years of life, and also causes serious disease in elderly individuals and persons with chronic lung disease. In the 1980s, an estimated 100,000 children were hospitalized annually with RSV infection in the United States (1). Although RSV is commonly considered a pediatric disease, it is also highlighted as an opportunistic pathogen (2), with infections producing a mortality rate of 30–100% in immunosuppressed individuals (1). There is growing appreciation that RSV is an important pathogen in elderly and high-risk patients, and a cause of acute exacerbations of asthma (3, 4) and chronic obstructive pulmonary disease (COPD) (5). Over the period 1999–2003, RSV was responsible for hospitalization rates of 10.6% for pneumonia, 11.4% for COPD, 5.4% for congestive heart failure, and 7.2% for asthma (6).

No vaccine is currently available for prevention of RSV infection. Several vaccine candidates have not only proved to be ineffective, but have also been shown to lead to vaccine-enhanced disease (7, 8). Inhibitors directed against the RSV fusion protein (F protein) were abandoned partly because of the frequency of resistant mutations mapping to the F gene (9). A monoclonal antibody against F protein, Palivizumab, has restricted application and it is recommended for prophylactic use during the RSV season, for high-risk infants (1). Currently the only therapy for an active RSV infection is the aerosolized nucleotide analog ribavirin, but its use is limited because a beneficial effect on clinical outcome remains unproved (10). Experimentation with siRNAs directed against RV has produced promising results (11, 12), although the mechanisms of epithelial cell uptake of the inhibitor remain unknown (13, 14). In the context of the aforementioned therapeutic problems, a good understanding of pathogenesis of RSV disease is key to developing novel therapies. TLR4 and CD14 are well-established molecules of innate immunity that function in the recognition and response to lipopolysaccharide (LPS) (15). Unexpectedly, the RSV F protein appears to stimulate innate immunity through TLR4 and CD14, although the mechanism of action is poorly understood (16).

Recently, we found that POPG inhibits proinflamamtory cytokine production induced by LPS in vitro and in vivo through direct interactions with CD 14 and MD2 (17). Additional studies have identified phosphatidylglycerol (PG) as an antagonist of LPS binding protein (LBP), and CD14 (1820). Although PG constitutes about 10% of surfactant phospholipids, the high concentration of lipid in the extracellular surfactant layer within the alveolus, results in PG levels as high as 3.5 mg/mL This extraordinary level of PG is not found in any other tissue or mucosal surface in mammals. In human pulmonary surfactant, POPG is the most abundant molecular species present within the PG class of phospholipid. Recent findings now indicate that selected surfactant lipids and proteins may complement each other in the homeostatic suppression of inflammation in the lung (17, 21, 22).

We have undertaken a comprehensive examination of the role of POPG in regulating multiple aspects of innate immunity. As part of this endeavor we focused upon the interrelationships between CD14, POPG, and RSV. The goals of this study were to determine whether POPG did the following: (i) suppressed the inflammatory response of epithelial cells to RSV, (ii) altered the cytopathic effects elicited by the virus, (iii) directly interacted with RSV, (iv) altered the infectivity of the virus, and (v) acted in vivo to alleviate the effects of viral infection. Our findings demonstrate that POPG is a potent antiviral agent against RSV that is active in vitro and in vivo.

Results

POPG Inhibits RSV-Elicited Cytokine Production in Human Epithelial Cells.

We first examined the effects of POPG on cytokine production (IL-6 and IL-8) induced by RSV in bronchial epithelial cells. Normal human bronchial epithelial cells (NHBE) were pretreated with POPG for 1 h, followed by infection with RSV at a multiplicity of 2/cell. The untreated and RSV infected cultures were examined at 48 h and 72 h after infection. As shown in Fig. 1, control cultures secreted low levels of IL-6 and IL-8 into the culture medium at 48 h after sham infection, whereas the RSV infected cells increased production of these cytokines by 5- to10-fold. Cultures of NHBE that were treated with RSV and POPG showed dramatically suppressed levels of both IL-6 and IL-8 that were not different from those in uninfected cells. The full range of variability among different preparations of NHBE cells is shown in Fig. S1. Nearly identical findings were also obtained with the BEAS2B cell line (Fig. S2). Control experiments monitoring epithelial cell protein synthesis and retention of responsiveness to the TLR5 ligand, flagellin, revealed that POPG treatment was not pleiotropically inhibitory to metabolism or signaling processes (Fig. S3). These findings clearly demonstrate that POPG is a potent suppressor of the inflammatory cytokine production, which accompanies viral challenge.

Fig. 1.

Fig. 1.

POPG suppresses RSV induced IL-6 and IL-8 production by epithelial cells. IL-6 and IL-8 production by the NHBE cells was measured by ELISA after sham treatment (CONL) or infection with virus (RSV) for 48 h, in either the absence or presence of 200 μg/mL POPG, or palmitoyl-oleoyl-phosphatidylcholine (+POPG or +POPC). The viral multiplicity of infection was 2. Additional control experiments included cells treated with phospholipid alone (POPG, POPC). Values shown are means ± SE for three independent experiments. *P < 0.01. The average IL-6 production upon RSV challenge was 745 ± 298 pg/mL. The average IL-8 production was 11,344 ± 3,209 pg/mL.

POPG Prevents Cell Death Caused by RSV Infection in Bronchial Epithelium.

RSV infection induces syncytia formation and cell death in epithelial cells, and we next examined the ability of POPG to interfere with the cytopathic effects (CPE) of the virus. At 48 h after infection, RSV produced prominent contraction of the cells within monolayers of NHBE cells, and numerous synctytia became apparent. None of this CPE was evident in either control cultures or infected cultures treated with POPG. At 72 h after viral infection, the cultures treated with RSV contained predominantly lysed cells, large syncytia, and scattered islands of cells adherent to the substratum as shown in the micrographs in Fig. 2. In contrast, the RSV infected cultures treated with POPG showed a healthy cell monolayer that was indistinguishable from cultures of uninfected cells. POPG treatment alone produced no significant alteration in the appearance of the cells. From these findings, we conclude that POPG can play a major role in protecting the primary NHBE cells from infection by RSV and subsequent cell death. POPG also protected BEAS2B cells from RSV infection (Fig. S4).

Fig. 2.

Fig. 2.

POPG prevents the cytopathic effects of RSV upon epithelial cells. NHBE cells were either sham treated (CONL) or challenged with virus (RSV) for 72 h, in either the absence or presence of 200 μg/mL phospholipids (+POPG, +POPC) as indicated. The viral multiplicity was 2. Additional conditions exposed the cells to phospholipids alone (POPG, POPC). Bar inset in the CONL panels corresponds to 50 μm.

POPG Specifically Attenuates Proinflammatory Cytokine Production Induced by RSV.

Previous studies have provided evidence that the antagonism of LPS activation through LBP/CD14/MD2/TLR4 is dependent upon the molecular class and species of phospholipid used as an antagonist (1720). We examined the molecular specificity of the POPG effect on RSV-elicited cytokine production by comparing the effects of POPG with a phospholipid counterpart containing a choline polar moiety, palmitoyl-oleoyl-phosphatidylcholine (POPC). These data are presented in Fig. 1 and show that POPC fails to attenuate IL-6 and IL-8 production by NHBE. In addition, POPC is ineffective at suppressing the CPE induced by RSV in NHBE cells as shown in Fig. 2. Thus the actions we observe for POPG are specific to this class of phospholipid, as compared with the most abundant class of phospholipid present in pulmonary surfactant. This result parallels the actions of POPG and POPC upon the CD14/MD2/TLR4 complex (17).

RSV Binds CD14 AND POPG.

CD14 and TLR4 have been implicated in regulation of the innate immune response to RSV, but the mechanism of this process is not clearly understood (16). We undertook experiments to examine the binding interactions between the virus, CD14, and POPG. The data presented in Fig. 3A demonstrate direct physical interactions between RSV and CD14. This binding interaction is concentration dependent and saturable. The addition of POPG inhibits the RSV-CD14 interaction in a concentration dependent reaction as shown in Fig. 3B. The maximum inhibition of binding occurs at ≈200 μg/mL POPG. An examination of the direct physical interaction between RSV and POPG is shown in Fig. 3C. The virus binds to POPG in a concentration-dependent and saturable reaction. In contrast to POPG, the lipid POPC is a weak-binding ligand for the virus that interacts with RSV nonspecifically. The consequences of the interactions of POPG with RSV upon epithelial cell responses are shown in Fig. 3D, and demonstrate the lipid concentration dependent inhibition of IL-8 production from BEAS2B cells elicited by the virus. The apparent IC50 of the POPG upon IL-8 production is ≈50 μg/mL, and this value correlates well with the apparent IC50 of the lipid for disrupting viral binding to CD14 (Fig. 3B). From these data, we conclude that one important mode of action of POPG is its direct interaction with the virus, which can interfere with viral recognition of specific cell surface ligands required for viral infection and induction of inflammation.

Fig. 3.

Fig. 3.

POPG binds RSV and inhibits viral interactions with CD14, and cell surface binding. (A) Binding of 1 μg/mL CD14 to RSV at different concentrations of virus, ranging from 0.05 to 0.5 × 107 plaque forming units/mL. Bound CD14 was detected by ELISA. (B) Inhibitory action of POPG upon RSV-CD14 interaction. Bound CD14 was detected by ELISA. (C) Binding data quantifying the interactions between RSV and POPG and POPC. Aliquots containing 1.25-nmol of phospholipid were adsorbed onto microtiter wells. Bound virus was detected by ELISA. (D) Concentration-dependent effects of POPG upon IL-8 production by BEAS2B cells challenged with the RSV at a multiplicity of infection of 1, in either the absence or presence of the indicated concentrations of POPG, added 30 min before virus. IL-8 present in culture supernatants after 48 h was quantified by ELISA. (E) Effects of POPG upon cell surface binding of RSV. Suspensions of HEp2 cells were incubated with RSV at a multiplicity of 50, for 10 min at 37°C, and then washed three times with PBS at 0°C. Subsequently, the cells were fixed and treated with a mouse monoclonal antibody against RSV. Phycoerythrin-conjugated antibody directed against mouse IgG was used for detection. A representative FACScan is shown in which the cells were not challenged with virus (NO RSV) or challenged with virus alone (RSV), or challenged with virus and phospholipids (RSV + POPG, RSV + POPC). (F) Summary of FACScan data from three independent experiments. *P < 0.01 for the RSV + POPG sample compared with RSV treatment alone.

We next examined whether POPG could effectively inhibit the direct binding of RSV to cell surfaces. For these experiments, HEp2 cells in suspension were used as targets for viral binding. HEp2 cells are routinely used in quantitative plaque assays for RSV (23). After the binding reaction, the cells were analyzed for attached virus by FACScan, using fluorescent antibody. The results presented in Fig. 3E demonstrate significant binding of the RSV to the cell surface in the absence of lipid as detected using FACScan. The addition of POPG markedly inhibits the association of the RSV with the cell surface. In contrast, POPC does not significantly alter the viral attachment to the cell surface. The data in Fig. 3F summarizes the findings from three independent experiments, with cell surface binding expressed as the mean fluorescence intensity. The data clearly demonstrate that 200 μg/mL POPG directly inhibits the binding of the virus to the cell surface by more than 80%, even at a viral multiplicity of infection of 50.

POPG Prevents Plaque Progression and Viral Spreading After an Infection Is Established.

The preceding findings provide strong evidence that POPG can effectively inhibit RSV infection and the subsequent inflammation. We next examined the efficacy of POPG as an antiviral agent after the establishment of a viral infection. In these experiments, monolayers of HEp2 cells were infected with RSV for 2 h to enable viral attachment and internalization. Subsequently, unattached viruses were removed, and the cells were overlayed with agar under standard conditions for a plaque assay (23), except that the agar medium composition was modified to contain either no lipid supplements, or 200–500 μg/mL POPG, or 500 μg/mL POPC. After 6 days of culture, viral plaques were quantified using Neutral Red staining as shown in Fig. 4A. Initially, cursory inspection of the Neutral Red stained plates showed the expected plaque number on RSV-infected plates, but no plaques on plates treated with RSV plus POPG. The monolayers exposed to RSV and POPC produced the same number of plaques as those treated with virus alone. However, careful inspection of the Neutral Red–stained plates from cultures treated with RSV plus POPG, revealed the presence of unusual appearing regions of the monolayer, which we refer to as indefinite plaques. In subsequent experiments, staining with anti-RSV antibody revealed that the usual complete plaques had an expected bullseye appearance, created by lysed cells surrounded by a perimeter of cells darkly staining for viral antigen, whereas the indefinite plaques appeared as minute foci of viral infection as shown in Fig. 4B. Higher magnification views of the different plaque types are shown in Fig. 4C. Quantification of plaques demonstrated that POPG treatment eliminated nearly all of the complete plaques, and produced indefinite plaques at 25–50% of the frequency of normal plaques as shown in Fig. 4D. These findings clearly show that POPG can act to suppress viral spreading in epithelial monolayers after an RSV infection has been established.

Fig. 4.

Fig. 4.

POPG prevents the progression of RSV infection in sheets of epithelial cells. (A) Monolayers of 1 × 106 HEp2 cells were infected with RSV at a viral multiplicity of 10−4 for 2 h. After the infection period, the washed monolayer was overlayed with 0.3% agarose prepared in tissue culture medium containing either no additons (RSV), or 200, or 500 μg/mL phospholipid (+POPG200, +POPG500, +POPC200). After 6 days the cultures were stained with neutral red and the plaque numbers quantified. (B) Plaques were detected by immunostaining using HRP-conjugated polyclonal antibody directed against RSV. (C) Microscopic views of individual plaques visualized with neutral red (micrographs 1 and 2) and HRP-conjugated antibody (micrographs 3 and 4). (D) Results from three independent experiments performed as described above. Complete plaques form obvious zones of clearing on plates stained with neutral red, and form plaques with bullseye morphology upon immunostaining. Indefinite plaques are difficult to detect on plates stained with neutral red, and upon immunostaining, form small dark regions, which lack the characteristic bullseye appearance associated with zones of lysis of complete plaques. The average plaque number for RSV treated cells was 150 ± 15, for n = 3. *P < 0.01.

In additional experiments, we also quantified the suppression of plaque formation in vitro by treating HEp2 cells with virus in the presence or absence of 200 μg/mL POPG. The infection of HEp2 cells with RSV alone produced 3.2 ± 0.6 × 107 plaques. When the same viral inoculum was administered in the presence of POPG, only 3.1 ± 0.1 × 103 plaques were obtained. Thus, POPG effectively prevents the in vitro infection of epithelial cells by 4 log units.

Intranasal Administration of POPG Inhibits RSV Infection in Mice.

The potency of POPG as an anti-RSV agent was next examined using a mouse model of viral infection. BALB/c mice were inoculated with RSV intranasally, either in the absence, or presence of 75–150 μg of POPG. At 3 and 5 days after the infection, the animals were euthanized and the lungs were harvested and processed for RSV burden, quantified by plaque assay. Comparisons were made between groups that were uinfected, RSV infected, RSV infected plus POPG treated, inactivated RSV infected, and POPG treated alone, groups. The data obtained after 5 days of viral infection are presented in Fig. 5A. The results provide clear evidence that POPG suppresses in vivo infection in the mouse by a factor of 1,700, when compared with animals treated with RSV alone (RSV infection = 5355 ± 693 plaques; RSV infection + POPG = 3.1 ± 1.2 plaques). No plaques were obtained from uninfected animals, or animals treated with UV-inactivated RSV or POPG alone. Similar results were obtained after 3 days of infection, except the number of viral plaques produced with RSV treatment (822 ± 115 plaques per left lung; n = 3) were significantly lower than those obtained after 5 days of infection. Figure 5B shows the effects of viral infection upon the influx of inflammatory cells into the lung, recovered in the BALF. Control mice contain low levels of lymphocytes and neutrophils in lavage but their percentage increases 3- to 20-fold after viral infection. In addition, the total cells recovered from the lavage of infected animals is increased an average of 2.1-fold. The treatment of RSV-infected animals with POPG completely attenuates the increase in lymphocytes and neutrophils recovered in lavage, elicited by virus. With RSV infection, IFN-γ levels increase from undetectable levels to nearly 8 ng/mL of lavage, at 5 days after infection; and this antiviral response is completely abrogated by treatment with POPG (Fig. 5C). SP-D levels also increase 3.5-fold in response to RSV infection, and this response is also completely inhibited by POPG treatment as shown in Fig. 5D. The effects of RSV and POPG upon lung histopathology were also evaluated and are presented in Fig. 6. The histopathology scores are shown in Fig. 6A. Control mice had a pathology score of 2.7 ± 0.4, and RSV-infected mice had a score of 8.6 ± 0.6. Treatment of mice with RSV and POPG resulted in a pathology score of 3.4 ± 0.5, which is significantly different from RSV infection, but not different from control mice or animals treated with POPG alone. Representative tissue sections are shown in Fig. 6B and provide evidence that POPG also suppresses the appearance of inflammatory cellular infiltrates and pneumonia, which accompanies the viral infection. Microscopy also demonstrates that POPG installation into the lungs does not significantly alter the appearance or inflammatory status of the tissue.

Fig. 5.

Fig. 5.

POPG prevents RSV infection and inflammation in vivo. BALB/c mice were infected with 1 × 107 RSV in either the absence or presence of 150 μg of POPG, as indicated. Five days after infection, the animals were euthanized and tissues and fluids were analyzed to evaluate the progress of the infection. (A) Amount of virus present in the left lung was quantified using the plaque assay. (B) Presence of inflammatory cells was quantified using stained Cytospin preparations. (C) IFN-γ levels present in BALF were quantified by ELISA. (D) Levels of murine SP-D were quantified by ELISA. Values shown are means + SE for three independent experiments. In each experiment, the control and treatment groups contained five to eight mice. *P < 0.01 compared with corresponding RSV-treated groups; $$P < 0.001.

Fig. 6.

Fig. 6.

POPG reduces the histopathology elicited by RSV. (A) Paraffin sections (4-μm) were stained with H&E, analyzed by light microscopy, and assigned a histopathology score. The groups consisted of untreated mice (CONL), virus-infected mice (RSVPOPG-treated mice (POPG), and virus-plus-POPG–treated mice (RSV + POPG). (B) Representative micrographs from experiment. Values shown in A are means ± SE for three independent experiments. Each experiment contained 5–8 mice per group. *P < 0.01 for the comparison of RSV treatment with RSV + POPG.

Collectively, the data presented in Figs. 5 and 6 demonstrate that POPG can block RSV infection and replication in vivo, and protect the lungs from inflammatory cell infiltration and inflammatory cytokine production. These findings suggest that POPG may have significant potential for preventing RSV infections in vulnerable human populations, and treating infections after they become established.

Discussion

The function of PG in surfactant has long been enigmatic; but our recent work (17) and the data in this report provide compelling evidence that this lipid plays an important role in regulating innate immunity and RSV infection. The lipid suppresses virus-induced activation of epithelial cells and release of the inflammatory cytokines IL-6 and IL-8. A major mechanism of action of POPG is direct binding to RSV and inhibition of the interaction of the virus with the epithelial cell surface. The inhibitory effect of the POPG upon viral attachment and internalization in HEp2 cells in vitro, reduces plaque formation by 4 log units. A few studies have shown that RSV induces expression of TLR4 in epithelial cells and suggest that acquisition of responsiveness to LPS by the epithelium enhances inflammation by the virus through responses to bacterial ligands (24). However, our in vitro studies show direct responses to the virus potently increase IL-6 and IL-8 production independent of the presence of bacterial ligands for TLR4. Thus the suppression of inflammation by POPG appears to be a direct effect upon the virus and cellular receptors for the intact pathogen and its constituents.

The efficacy of PG as an antiviral agent also extends to the process of viral spreading after an infection is established. The action of PG upon RSV prevents the cell to cell spreading of the virus through sheets of epithelial cells. This finding suggests that POPG treatment of human subjects could be effective after the presence of RSV as a pathogen is diagnosed. The in vivo mouse studies show significant attenuation of the viral infection with simultaneous administration of the RSV and lipid. These in vivo results predict that the prophylactic administration of POPG by inhalation, to high-risk individuals, may be a simple and economical means for preventing infections.

The application of POPG as an antiviral agent presents some distinct advantages to other antiviral and anti-inflammatory agents. Foremost, POPG is a natural constituent of human surfactant, and it has already been administered to adults and neonates in a variety of clinical settings as part of a more complex mixture that contains multiple surfactant components (2527). In addition, POPG is a low-molecular-weight lipid and as such, is extremely unlikely to induce any type of acquired immune response. Our previous studies show that the physical form in which POPG is delivered to cells challenged with microbial products, has important effects upon the anti-inflammatory activity (and by inference the antiviral activity) of the lipid. The data demonstrate that segregated pools of PG (i.e., not randomly mixed with other surfactant components) are necessary for effectively blocking engagement of TLR4-dependent processes. Indeed, it is likely that the random mixing of PG with other surfactant constituents has obscured its potency as both an anti-inflammatory and anti-RSV agent in previous studies (2527). The cost and chemical stability of POPG also make it an attractive candidate for antiviral treatment in both developed and undeveloped countries.

In summary, our findings provide clear evidence that POPG is a potent suppressor of RSV infection and the inflammatory host response to the virus. The data suggest that POPG is an important lead compound for a new family of pharmacological agents for suppression of CD14/MD2/TLR4-mediated inflammatory events and RSV infection, by direct administration through the airways.

Methods

Virus Preparation and Titration.

The human RSV A2 strain VR-1540 was obtained from American Tissue Culture Collection. Virus stocks were prepared under endotoxin-free conditions. The virus was grown in HEp2 cell monolayers using Dulbecco’s Modified Eagle’s Medium/Ham’s F-12 Medium (DMEM/F12: GIBCO) plus 5% growth bovine serum (GBS, HyClone) and purified using methods previously described (28, 29). Viral titers and growth were determined by quantitative plaque assays (23). RSV replication and plaque formation were also determined by immunostaining for viral antigens using HRP-conjugated polyclonal goat anti-human RSV antibody (AbD Serotec).

Bronchial Epithelial Cell Tissue Culture, Infection, and Surfactant Phospholipid Treatments.

Beas2B cells were obtained from ATCC and cultured using LHC9 medium (Lonza). Normal human primary bronchial epithelial cells (NHBE) were obtained by bronchoscopy and airway brushing of normal, nonsmoking volunteers with no history of lung disease or current medications (30, 31), or from surgical specimens of tracheas and mainstem bronchi, as previously described (31). All human tissue was acquired under protocols approved by the National Jewish Health Institutional Review Board. Cell culture and characterization was performed as described previously (31). NHBE cells were seeded into 24-well plates and grown until 80% confluent, before exposure to RSV. Cells were infected at a viral multiplicity of 1.5–2.0, and incubated for 72 h. Phospholipids were obtained from AVANTI and liposomes were prepared as previously described (17). For examination of phospholipid inhibition of RSV infection, the cells were preincubated with POPG and POPC liposomes (200 μg/mL) for 1 h and subsequently infected with virus in the presence of liposomes for up to 72 h. IL-6 and IL-8 production in culture supernatants were assayed by ELISA (eBioscience, Biosource). Previous studies have demonstrated that NHBE cells and BEAS2B cell are positive for expression of TLR4, CD14, and MD2 by either Western blotting or PCR (3234); and HEp2 cells are positive for CD14 expression (35, 36).

Binding of RSV to CD14 and Phospholipid.

RSV viral stocks were diluted into 50 μl PBS (PBS, pH 7.4) and coated overnight in 96-well, half-area plates (Corning) at 4°C. The wells were washed with 20 mM Tris buffer (pH 7.4) containing 150 mM NaCl, 5 mM CaCl2 (buffer A) and blocked with buffer A + 5% BSA (Sigma) for 2 h, at 37°C. Recombinant human CD14 protein (R&D Systems, 1 μg/mL) was added and incubated at 37°Cfor 1 h. Subsequently the wells were washed with buffer A and labeled with monoclonal anti-human CD14 antibody (R&D Systems) diluted 1:1,500 in buffer A + 5% BSA at 37°C for 1 h, followed by the incubation with HRP-labeled anti-mouse IgG (1:2,000) for 1 h. After washing the wells with buffer A, the bound antibody was detected with o-phenylenediamine as substrate and measuring absorbance at 490 nm. In competitive binding reactions, varying concentrations of POPG in buffer A + 5% BSA were added to wells immediately before the addition of CD14 (1 μg/mL).

For measurement of direct interactions between RSV and lipids, 1.25 nmol of the specified phospholipids in 20-μl aliquots of ethanol were added to 96-well half-area plates, and the solvent was evaporated using a warm-air blower. Nonspecific binding was blocked with bufferA + 5% BSA, and varying concentrations of RSV were added to the wells and incubated at 37°C for 1 h. After the wells were washed with buffer A, HRP-conjugated anti-human RSV antibody diluted in bufferA + 5% BSA (1:500) was added and incubated for 1 h at 37°C. The amount of bound RSV was detected as described above.

Binding of RSV to HEp2 Cells and Inhibition by Phospholipid.

Adherent HEp2 cells were detached from tissue culture plates by 10-min exposure to PBS containing 10 mM EDTA, at 0°C. HEp2 cells (3 × 105) in suspension were incubated with RSV at a multiplicity of 50/cell for 10 min at 37°C in either the presence or absence of 200 μg/mL phospholipid liposomes. Subsequently, the cells were diluted and washed three times with PBS at 4°C, by centrifugation. The cells were fixed overnight in 1% buffered paraformaldehyde, followed by washing three times with PBS. The fixed cells were labeled for 1 h at 4°C, with monoclonal mouse anti-human RSV antibody (AbD Serotec) diluted 1:100 with PBS + 5% BSA. Unbound primary antibody was removed by washing the wells three times with cold PBS. Next, the cells were incubated with phycoerythrin (PE) conjugated anti-mouse IgG (e-Bioscience) for 1 h at 4°C. The unbound fluorescent antibody was removed by washing the cells using centrifugation at 4°C in PBS. The cell associated fluorescence was determined using FACScan. Typically, 20,000 cells were used to quantify the fluorescence of the HEp2 cells. Graphic analysis of the fluorescence and calculation of mean fluorescence intensity was performed using Cell Quest software.

In Vivo Suppression of RSV Infection by POPG.

Female BALB/c mice, 6 weeks old, were obtained from Jackson Laboratory. The animals were anesthetized by i.p.injection of Avertin at a dose of 0.25 g/kg. The viral inoculation volume for each group was adjusted to 50 μL. Sham infections were performed using the same amount of medium plus PBS. We diluted virus with PBS in a total volume of 50 μl that contained 1 × 107 plaque forming units. Liposome POPG was likewise diluted in PBS, and each mouse received 75–150 μg of the lipid. RSV and POPG mixtures were prepared in PBS and inoculated intranasally into mice. Aliquots of RSV stocks were inactivated by exposure to 1,800 mJ of UV radiation in a Stratalinker UV cross-linker (UVP), for the purpose of eliminating viral infectivity without significantly altering the conformation of viral proteins, or inactivating endogenous mediators, as previously described (37). UV inactivation reduced RSV infectivity by 8 log units. At the end of the experiment, mice were euthanized by i.p.injection of 0.25 mL of Nembutal (10 mg/mL). Animal experiments followed all prescribed guidelines and were approved by the Institutional Animal Care and Use committee.

Measurement of Viral Infection and Inflammation.

To quantify viral titers in mice, we homogenized the left lungs in DMEM/F12 at 0°C, shortly after removal from the animals. Freshly prepared lung extracts were centrifuged at 2,000 × g for 15 min at 4°C, and serial dilutions of the supernatants were assayed for viral plaque formation.

For histopathology, lungs were fixed overnight in 10% formalin, washed with PBS, and subsequently dehydrated in 70% ethanol for 48 h. The lungs were paraffin embedded, and 4-μm sections were prepared and stained with H&E. The tissue was evaluated in a double-blinded fashion using light microscopy and a histopathologic inflammatory scoring system, as described previously (38). We collected bronchoalveolar lavage fluid (BALF) by intratracheal instillation of the lungs with 1.0 ml of saline, followed by recovery of the solution. Total recovery was typically 0.7–0.9 mL, and the cells were sedimented at 5,000 × g for 15 min at 4°C. The resultant cell pellet was resuspended in 150 μL of PBS, stained with Trypan Blue, and the total cell number was quantified using light microscopy. Differential cell counts were performed on cytospin preparations stained with Trypan Blue. The cell-free supernatants resulting from the centrifugation of BALF were stored at –80°C and used for cytokine measurements. IFN-γ was measured by ELISA using primary and HRP-conjugated secondary antibodies from BD Biosc0iences. SP-D was measured by ELISA using a polyclonal antibody for solid phase capture and an HRP-conjugated polyclonal antibody for detection. Rabbit polyclonal antibodies against mouse SP-D were generated from purified recombinant protein.

Statistical Analysis.

All results are expressed as mean ± SE. ANOVA was used to determine the level of difference between all groups. Groups were compared by unpaired Student’s t test. Differences are considered to be significant at P < 0.05.

Supplementary Material

Supporting Information

Acknowledgments

This work was supported by NIH grants PHL073907 and M01 RR00051, CTSA 1UL1 RR025780 from NCRR/HIH, and a Colorado Bioscience Discovery Grant.

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

This article contains supporting information online at www.pnas.org/cgi/content/full/0909361107/DCSupplemental.

References

  • 1.Hall CB. Respiratory syncytial virus and parainfluenza virus. N Engl J Med. 2001;344:1917–1928. doi: 10.1056/NEJM200106213442507. [DOI] [PubMed] [Google Scholar]
  • 2.Whimbey E, Ghosh S. Respiratory syncytial virus infections in immunocompromised adults. Curr Clin Top Infect Dis. 2000;20:232–255. [PubMed] [Google Scholar]
  • 3.Nicholson KG, Kent J, Ireland DC. Respiratory viruses and exacerbations of asthma in adults. Br Med J. 1993;307:982–986. doi: 10.1136/bmj.307.6910.982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Teichtahl H, Buckmaster N, Pertnikovs E. The incidence of respiratory tract infection in adults requiring hospitalization for asthma. Chest. 1997;112:591–596. doi: 10.1378/chest.112.3.591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wedzicha JA. Exacerbations: Etiology and pathophysiologic mechanisms. Chest. 2002;121(Suppl):136S–141S. doi: 10.1378/chest.121.5_suppl.136S. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Falsey AR, Hennessey PA, Formica MA, Cox C, Walsh EE. Respiratory syncytial virus infection in elderly and high-risk adults. N Engl J Med. 2005;352:1749–1759. doi: 10.1056/NEJMoa043951. [DOI] [PubMed] [Google Scholar]
  • 7.Polack FP, et al. A role for immune complexes in enhanced respiratory syncytial virus disease. J Exp Med. 2002;196:859–865. doi: 10.1084/jem.20020781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kim HW, et al. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am J Epidemiol. 1969;89:422–434. doi: 10.1093/oxfordjournals.aje.a120955. [DOI] [PubMed] [Google Scholar]
  • 9.Morton CJ, et al. Structural characterization of respiratory syncytial virus fusion inhibitor escape mutants: Homology model of the F protein and a syncytium formation assay. Virology. 2003;311:275–288. doi: 10.1016/s0042-6822(03)00115-6. [DOI] [PubMed] [Google Scholar]
  • 10.American Academy of Pediatrics Committee on Infectious Diseases and Committee of Fetus and Newborn. Prevention of respiratory syncytial virus infections: Indications for the use of palivizumab and update on the use of RSV-IGIV. Pediatrics. 1998;102:1211–1216. doi: 10.1542/peds.102.5.1211. [DOI] [PubMed] [Google Scholar]
  • 11.Bitko V, Musiyenko A, Shulyayeva O, Barik S. Inhibition of respiratory viruses by nasally administered siRNA. Nat Med. 2005;11:50–55. doi: 10.1038/nm1164. [DOI] [PubMed] [Google Scholar]
  • 12.Zhang G, et al. Crystal structure of Thermus aquaticus core RNA polymerase at 3.3 Å resolution. Cell. 1999;98:811–824. doi: 10.1016/s0092-8674(00)81515-9. [DOI] [PubMed] [Google Scholar]
  • 13.Dorsett Y, Tuschl T. siRNAs: Applications in functional genomics and potential as therapeutics. Nat Rev Drug Discov. 2004;3:318–329. doi: 10.1038/nrd1345. [DOI] [PubMed] [Google Scholar]
  • 14.Ketzinel-Gilad M, Shaul Y, Galun E. RNA interference for antiviral therapy. J Gene Med. 2006;8:933–950. doi: 10.1002/jgm.929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4:499–511. doi: 10.1038/nri1391. [DOI] [PubMed] [Google Scholar]
  • 16.Kurt-Jones EA, et al. Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nat Immunol. 2000;1:398–401. doi: 10.1038/80833. [DOI] [PubMed] [Google Scholar]
  • 17.Kuronuma K, et al. Anionic pulmonary surfactant phospholipids inhibit inflammatory responses from alveolar macrophages and U937 cells by binding the lipopolysaccharide-interacting proteins CD14 and MD-2. J Biol Chem. 2009;284:25488–25500. doi: 10.1074/jbc.M109.040832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hashimoto M, Asai Y, Ogawa T. Treponemal phospholipids inhibit innate immune responses induced by pathogen-associated molecular patterns. J Biol Chem. 2003;278:44205–44213. doi: 10.1074/jbc.M306735200. [DOI] [PubMed] [Google Scholar]
  • 19.Mueller M, Brandenburg K, Dedrick R, Schromm AB, Seydel U. Phospholipids inhibit lipopolysaccharide (LPS)-induced cell activation: A role for LPS-binding protein. J Immunol. 2005;174:1091–1096. doi: 10.4049/jimmunol.174.2.1091. [DOI] [PubMed] [Google Scholar]
  • 20.Wang PY, Kitchens RL, Munford RS. Phosphatidylinositides bind to plasma membrane CD14 and can prevent monocyte activation by bacterial lipopolysaccharide. J Biol Chem. 1998;273:24309–24313. doi: 10.1074/jbc.273.38.24309. [DOI] [PubMed] [Google Scholar]
  • 21.Sano H, Kuroki Y. The lung collectins, SP-A and SP-D, modulate pulmonary innate immunity. Mol Immunol. 2005;42:279–287. doi: 10.1016/j.molimm.2004.07.014. [DOI] [PubMed] [Google Scholar]
  • 22.Wright JR. Immunoregulatory functions of surfactant proteins. Nat Rev Immunol. 2005;5:58–68. doi: 10.1038/nri1528. [DOI] [PubMed] [Google Scholar]
  • 23.McKimm-Breschkin JL. A simplified plaque assay for respiratory syncytial virus—direct visualization of plaques without immunostaining. J Virol Methods. 2004;120:113–117. doi: 10.1016/j.jviromet.2004.02.020. [DOI] [PubMed] [Google Scholar]
  • 24.Monick MM, et al. Respiratory syncytial virus up-regulates TLR4 and sensitizes airway epithelial cells to endotoxin. J Biol Chem. 2003;278:53035–53044. doi: 10.1074/jbc.M308093200. [DOI] [PubMed] [Google Scholar]
  • 25.Tibby SM, et al. Exogenous surfactant supplementation in infants with respiratory syncytial virus bronchiolitis. Am J Respir Crit Care Med. 2000;162:1251–1256. doi: 10.1164/ajrccm.162.4.9909004. [DOI] [PubMed] [Google Scholar]
  • 26.Luchetti M, et al. Multicenter, randomized, controlled study of porcine surfactant in severe respiratory syncytial virus-induced respiratory failure. Pediatr Crit Care Med. 2002;3:261–268. doi: 10.1097/00130478-200207000-00011. [DOI] [PubMed] [Google Scholar]
  • 27.Spragg RG, et al. Effect of recombinant surfactant protein C-based surfactant on the acute respiratory distress syndrome. N Engl J Med. 2004;351:884–892. doi: 10.1056/NEJMoa033181. [DOI] [PubMed] [Google Scholar]
  • 28.Dakhama A, et al. The enhancement or prevention of airway hyperresponsiveness during reinfection with respiratory syncytial virus is critically dependent on the age at first infection and IL-13 production. J Immunol. 2005;175:1876–1883. doi: 10.4049/jimmunol.175.3.1876. [DOI] [PubMed] [Google Scholar]
  • 29.LeVine AM, et al. Surfactant protein-d enhances phagocytosis and pulmonary clearance of respiratory syncytial virus. Am J Respir Cell Mol Biol. 2004;31:193–199. doi: 10.1165/rcmb.2003-0107OC. [DOI] [PubMed] [Google Scholar]
  • 30.Wark PA, et al. Asthmatic bronchial epithelial cells have a deficient innate immune response to infection with rhinovirus. J Exp Med. 2005;201:937–947. doi: 10.1084/jem.20041901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Uetani K, et al. Central role of double-stranded RNA-activated protein kinase in microbial induction of nitric oxide synthase. J Immunol. 2000;165:988–996. doi: 10.4049/jimmunol.165.2.988. [DOI] [PubMed] [Google Scholar]
  • 32.Guillot L, et al. Response of human pulmonary epithelial cells to lipopolysaccharide involves Toll-like receptor 4 (TLR4)-dependent signaling pathways: Evidence for an intracellular compartmentalization of TLR4. J Biol Chem. 2004;279:2712–2718. doi: 10.1074/jbc.M305790200. [DOI] [PubMed] [Google Scholar]
  • 33.Sha Q, Truong-Tran AQ, Plitt JR, Beck LA, Schleimer RP. Activation of airway epithelial cells by Toll-like receptor agonists. Am J Respir Cell Mol Biol. 2004;31:358–364. doi: 10.1165/rcmb.2003-0388OC. [DOI] [PubMed] [Google Scholar]
  • 34.Becker MN, Diamond G, Verghese MW, Randell SH. CD14-dependent lipopolysaccharide-induced beta-defensin-2 expression in human tracheobronchial epithelium. J Biol Chem. 2000;275:29731–29736. doi: 10.1074/jbc.M000184200. [DOI] [PubMed] [Google Scholar]
  • 35.El Ahmera OR, et al. Binding of bacteria to HEp-2 cells infected with influenza A virus. FEMS Immunol Med Microbiol. 1999;23:331–341. doi: 10.1111/j.1574-695X.1999.tb01255.x. [DOI] [PubMed] [Google Scholar]
  • 36.Raza MW, et al. Infection with respiratory syncytial virus enhances expression of native receptors for non-pilate Neisseria meningitidis on HEp-2 cells. FEMS Immunol Med Microbiol. 1999;23:115–124. doi: 10.1111/j.1574-695X.1999.tb01230.x. [DOI] [PubMed] [Google Scholar]
  • 37.Davis IC, Sullender WM, Hickman-Davis JM, Lindsey JR, Matalon S. Nucleotide-mediated inhibition of alevolar fluid clearance in BALB/c mice after respiratory syncytial virus infection. Am J Physiol Lung Cell Mol Physiol. 2004;286:L112–L120. doi: 10.1152/ajplung.00218.2003. [DOI] [PubMed] [Google Scholar]
  • 38.Cimolai N, Taylor GP, Mah D, Morrison BJ. Definition and application of a histopathological scoring scheme for an animal model of acute Mycoplasma pneumoniae pulmonary infection. Microbiol Immunol. 1992;36:465–478. doi: 10.1111/j.1348-0421.1992.tb02045.x. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supporting Information

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES