<<

Surfactant Protein A Activation of Atypical ζ in IκB-α-Dependent Anti-Inflammatory Immune Regulation

This information is current as Christina Moulakakis, Stefanie Adam, Ulrike Seitzer, Andra of September 29, 2021. B. Schromm, Michael Leitges and Cordula Stamme J Immunol 2007; 179:4480-4491; ; doi: 10.4049/jimmunol.179.7.4480 http://www.jimmunol.org/content/179/7/4480 Downloaded from

References This article cites 61 articles, 32 of which you can access for free at: http://www.jimmunol.org/content/179/7/4480.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online.

• Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists

• Fast Publication! 4 weeks from acceptance to publication

by guest on September 29, 2021 *average

Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2007 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology

Surfactant Protein A Activation of Atypical Protein Kinase C ␨ in I␬B-␣-Dependent Anti-Inflammatory Immune Regulation1

Christina Moulakakis,* Stefanie Adam,* Ulrike Seitzer,† Andra B. Schromm,‡ Michael Leitges,§ and Cordula Stamme2*¶

The pulmonary collectin surfactant protein (SP)-A has a pivotal role in anti-inflammatory modulation of lung immunity. The mechanisms underlying SP-A-mediated inhibition of LPS-induced NF-␬B activation in vivo and in vitro are only partially un- derstood. We previously demonstrated that SP-A stabilizes I␬B-␣, the primary regulator of NF-␬B, in alveolar macrophages (AM) both constitutively and in the presence of LPS. In this study, we show that in AM and PBMC from I␬B-␣ knockout/I␬B-␤ knockin mice, SP-A fails to inhibit LPS-induced TNF-␣ production and p65 nuclear translocation, confirming a critical role for I␬B-␣ in SP-A-mediated LPS inhibition. We identify atypical (a) protein kinase C (PKC) ␨ as a pivotal upstream regulator of SP-A- ؊ ؊ mediated I␬B-␣/NF-␬B pathway modulation deduced from blocking experiments and confirmed by using AM from PKC␨ / Downloaded from mice. SP-A transiently triggers aPKCThr410/403 phosphorylation, aPKC kinase activity, and translocation in primary rat AM. Coimmunoprecipitation experiments reveal that SP-A induces aPKC/p65 binding under constitutive conditions. Together the data indicate that anti-inflammatory macrophage activation via I␬B-␣ by SP-A critically depends on PKC␨ activity, and thus attribute a novel, stimulus-specific signaling function to PKC␨ in SP-A-modulated pulmonary immune response. The Journal of Immu- nology, 2007, 179: 4480–4491. http://www.jimmunol.org/ ulmonary surfactant is a lipid-protein complex that con- Alveolar macrophages (AM), normally accounting for ϳ95% of stitutes the alveolar liquid layer of the lung. The unique airspace leukocytes, are a unique class of professional phagocytes P composition of surfactant facilitates the functional combi- that represent the major effector cells of the pulmonary innate im- nation of different biological effects such as preventing alveolar mune system (6). SP-A directly interacts with AM through binding collapsing at expiration and immunomodulating innate pulmonary to cell surface receptors, resulting in modulation of chemotaxis, host defense responses (1, 2). The latter function is primarily me- phagocytosis, and modified pro- or anti-inflammatory immune re- diated by surfactant proteins (SP)3-A and SP-D belonging to the sponses (7). collectin family. SP-A, the most abundant pulmonary collectin, LPS, the main component of the outer leaflet of the outer by guest on September 29, 2021 binds and aggregates a variety of microorganisms and enhances membrane of Gram-negative bacteria, substantially contributes their phagocytosis and killing both in vivo and in vitro (3). SP-A- to the pathophysiology of the most frequent lung diseases, in- deficient mice exhibit delayed microbial clearance and higher lev- cluding pneumonia, acute lung injury, and acute respiratory dis- els of bronchoalveolar inflammatory mediators after intratracheal tress syndrome (8). Whereas LPS recognition benefits the host inoculation with a variety of clinically relevant pathogens (4), as by sensing bacteria and mobilizing defense mechanisms, an ex- well as to isolated LPS (5). aggerated response to LPS contributes to the development of a local or systemic septic shock syndrome. The interaction of picomolar concentrations of LPS with a complex in- cluding TLR4, MD-2, LPS-binding protein, and CD14 on host *Department of Immunochemistry and Biochemical Microbiology, Division of Cel- cells initiates the sequential activation of multiple signaling lular Pneumology, Research Center Borstel, Leibniz Center for Medicine and Bio- ␬ science, Borstel, Germany; †Department of Immunology and Cell Biology, Division pathways, including NF- B, a central regulator of LPS-medi- of Veterinary Infection Biology and Immunology, Research Center Borstel, Leibniz ated cell activation (9–11). ‡ Center for Medicine and Bioscience, Borstel, Germany; Emmy-Noether Group Im- The most prominent NF-␬B heterodimer p65/p50 is sequestered munobiophysics, Research Center Borstel, Leibniz Center for Medicine and Bio- science, Borstel, Germany; §Biotechnolgy Centre of Oslo, University of Oslo, Oslo, in the cytoplasm by a family of inhibitory proteins, among Norway; and ¶Department of Anesthesiology, University Hospital of Lu¨beck, Lu¨- which I␬B-␣ is the best characterized and assumed to function beck, Germany as the primary regulator of NF-␬B in both stimulated and rest- Received for publication February 15, 2007. Accepted for publication July 30, 2007. ing cells (12, 13). In response to many stimuli including LPS, The costs of publication of this article were defrayed in part by the payment of page ␬ ␣ ␬ charges. This article must therefore be hereby marked advertisement in accordance I B- becomes degraded via ubiquitination in an I B kinase with 18 U.S.C. Section 1734 solely to indicate this fact. (IKK)-dependent manner (12, 14, 15). The removal of I␬B-␣ 1 This work was supported by the Deutsche Forschungsgemeinschaft 609/1-3 and allows the nuclear translocation of NF-␬B and subsequently the 609/1-4 (to C.S.) and 621/2-2 (to A.B.S.). transcription of downstream target genes, including I␬B-␣ itself 2 Address correspondence and reprint requests to Dr. Cordula Stamme, Research (16). Secondary clues point to an I␬B-independent pathway, Center Borstel, Parkallee 22, D-23845 Borstel, Germany. E-mail address: ␬ [email protected] and include the phosphorylation of p65 to activate NF- B- dependent gene transcription (17). 3 Abbreviations used in this paper: SP, surfactant protein; AKBI, I␬B-␣ knockout/ I␬B-␤ knockin; AM, alveolar macrophage; aPKC, atypical protein kinase C; cPKC, SP-A has been shown to inhibit LPS-induced TNF-␣ production classical protein kinase C; DAG, diacylglycerol; HI, heat inactivated; IKK, I␬B ki- (18–21), inducible NO synthase protein expression (22), NF-␬B ac- nase; PKC, protein kinase C; ps, pseudosubstrate; wt, wild type. tivity (20, 23, 24), and, upon diverse stimuli, NADPH oxidase (25) in Copyright © 2007 by The American Association of Immunologists, Inc. 0022-1767/07/$2.00 immunocompetent cells. We recently demonstrated that SP-A exerts www.jimmunol.org The Journal of Immunology 4481

its anti-inflammatory effects on LPS-challenged AM via a mechanism 37°C in a 5% CO2 atmosphere. Then the medium was changed and the involving a SP-A-mediated direct modulation of the basal and LPS- cells were treated with SP-A (20–60 ␮g/ml) and/or LPS (10–100 ng/ml) coupled I␬B-␣ turnover in these cells (24). In that study, SP-A in- for indicated times at 37°C in the presence of 0.2% heat-inactivated (HI) ␬ ␣ FCS. In separate experiments, AM were treated with a panel of kinase creased I B- protein expression in a dose- and time-dependent man- inhibitors to determine their possible effect on I␬B-␣ protein expression as ner without inducing I␬B-␣ phosphorylation or I␬B-␣ mRNA levels follows: Go¨-6976 (5 ␮M), Go¨-6850 (5 nM), chelerythrine chloride (6 and both basal and in the presence of LPS. However, the signaling path- 12 ␮M), or aPKCps peptides (2, 5, and 10 ␮M) were used to inhibit distinct ways controlling the SP-A-mediated attenuation of LPS-induced subsets of cPKC, novel PKC, or aPKCs. Wortmannin (50 nM) was used as PI3K inhibitor, and (30 ␮M) to inhibit protein kinase CKII. proinflammatory signals via I␬B-␣ remain unknown. A role for individual members of the protein kinase C (PKC) Nuclear protein extraction ␬ ␣ family in both SP-A immune modulation (26) and I B- turnover After treatment, cells were scraped off with 500 ␮l of cold PBS and spun (27) has been suggested previously. PKC family members are ex- at 4,500 ϫ g, 5 min, 4°C. The resulting pellet was resuspended in 400 ␮l

pressed in many different cell types, in which they regulate a wide of ice-cold buffer A (10 mM Tris, 5 mM MgCl2,10mMKCl,1mM variety of cellular processes such as cell differentiation, cytoskel- ethyleneglycol-bis-(2-aminoethyl ether)-N,N,NЈ,NЈ-tetraacetic acid, 0.3 M ␤ etal remodeling, and gene expression in response to diverse stimuli sucrose, 1 mM DTT, 10 mM -glycerol phosphate, 0.5 mM PMSF, and 1.5 ␮l of protease inhibitor mixture (Complete; Roche)) and incubated on ice (28, 29). Based on sequence homology and the mechanism of their for 15 min; 25 ␮l of 10% Nonidet P-40 was then added and vortexed for regulation, individual members of the PKC family are subdivided 10 s. The nuclei were pelleted by centrifugation at 4,500 ϫ g, 5 min, 4°C. ␣ ␤ ␤ The supernatant was taken to represent the cytosolic fraction. The pellet into three classes as follows: classical PKCs (cPKC: , I, II, and ϩ ␮ ␥) are diacylglycerol (DAG) and Ca2 dependent, whereas novel was resuspended in 30 l of buffer B (20 mM Tris, 5 mM MgCl2, 320 mM KCl, 0.2 mM ethyleneglycol-bis-(2-aminoethyl ether)-N,N,NЈ,NЈ-tetraace- ␦ ␧ ␩ ␪ Downloaded from PKCs ( , , , and ) are responsive to DAG, but are insensitive tic acid, 25% glycerol, 1 mM DTT, and the mixture of protease inhibitors 2ϩ to Ca . The atypical (a) PKCs (␨ and ␭/␫) do not bind to DAG and mentioned above) and incubated on ice for 15 min, followed by centrifu- are not responsive to Ca2ϩ (30, 31). gation at 16,100 ϫ g, 10 min, 4°C. The supernatant containing the nuclear In the present study, we confirmed the role of I␬B-␣ in SP-A fraction was transferred to a new vial. Cytosolic cell fractions (30–40 ␮gof ␬ ␣ ␮ anti-inflammatory effects by using I␬B-␣ knockout/I␬B-␤ knockin protein) were immunoblotted for I B- . Nuclear extracts (2 g of protein) of the cells were analyzed by EMSA for NF-␬B DNA-binding activity. (AKBI) AM and PBMC. We further investigated the role of PKC

in SP-A-specific anti-inflammatory signaling in primary AM and Isolation and stimulation of mouse AM and PBMC http://www.jimmunol.org/ identified the aPKC␨ isoform as a central regulator of SP-A-me- Gene-deficient mice and respective controls were killed by i.p. injection diated I␬B-␣/NF-␬B modulation. of pentobarbital, followed by exsanguination by cardiac puncture for PBMC isolation. The lungs were lavaged with 1 ml of PBS containing Materials and Methods 0.2 mM EDTA. AM were plated at a density of 2 ϫ 105/ml in 24-well plates (Nunc) in the presence of 0.2% HI-FCS. PBMC were isolated by Mice and reagents density gradient using Ficoll-Histopaque-1083 (Sigma-Aldrich). Hepa- Primary cells were obtained from pathogen-free male Sprague-Dawley rats rinized blood was mixed with an equal amount of HBSS and centrifuged Ϫ Ϫ ϫ (Charles River Laboratories), from AKBI mice (32), or from PKC␨ / at 420 g. The interphase layer containing the PBMC fraction was mice generated as described before (33). SV129 and CD1 wild-type (wt) collected and washed twice with HBSS and once in serum-free RPMI ϫ 6 control mice were from Charles River Laboratories. Animal care and ex- 1640. Cells were plated at a density of 1 10 /ml in 96-well plates. by guest on September 29, 2021 ␮ periments were conducted according to protocols approved by the AM and PBMC were left untreated or treated with SP-A (20–60 g/ml) Schleswig-Holstein Ministry of Environment, Nature, and Forestation. All for 1 h and/or stimulated with LPS (0.1–100 ng/ml) for4hat37°C in mice used were between 6 and 12 wk of age and were maintained at the the presence of 0.2 and 10% HI-FCS, respectively. After stimulation, ϫ Research Center Borstel animal facility under specific pathogen-free cells were centrifuged at 200 g, and cell-free supernatants were col- ␣ conditions. lected for TNF- determination. In separate experiments, AM were left ␮ The smooth LPS from Salmonella friedenau strain H909 was extracted untreated or treated with SP-A (40 g/ml) for 1 h, and/or stimulated by the phenol/water method, purified, lyophilized, and transformed into the with LPS (100 ng/ml) for1hat37°C in the presence of 0.2% HI-FCS. ␮ triethylamine salt form. RPMI 1640 medium, DMEM, and Dulbecco’s PBS Cytosolic cell fractions (30–40 g of protein/lane) were immunoblot- ␬ ␣ ␮ were from Invitrogen Life Technologies. Ham’s-F12 medium and FCS ted for I B- . Nuclear extracts (2 g of protein/lane) of the cells were ␬ were from BioWhittaker. Poly(dI/dC) was purchased from Pharmacia. analyzed by EMSA for NF- B DNA-binding activity. ␥ 32 [ - P]ATP was supplied by Hartmann Analytics; T4 polynucleotide ki- TNF-␣ ELISA nase was purchased from Roche. Rabbit polyclonal anti-I␬B-␣, anti-PKC␨ (in the manuscript referred to as anti-aPKC), anti-p65, and HRP-conju- TNF-␣ was determined in pooled cell-free supernatants of stimulated gated goat anti-rabbit IgG were from Santa Cruz Biotechnology; a specific AKBI and wt control cells by sandwich DuoSet ELISA using goat anti- Ab against PKC␨ was generated as described (34); anti-phospho PKC␨/␭ mouse TNF-␣ Ab and biotinylated goat anti-mouse TNF-␣ Ab (R&D Sys- (Thr410/403) Ab was from Cell Signaling Technology. Go¨-6850 was from tems), according to the manufacturer’s protocol. Calbiochem; chelerythrine chloride was from Tocris Bioscience; Go¨-6976 was from Alexis Biochemicals; apigenin and wortmannin were from Sig- Immunoprecipitation ma-Aldrich; and aPKC pseudosubstrate (ps) peptide was from BioSource Rat AM (2 ϫ 106/well) were stimulated for the indicated times with SP-A, International. Myelin basic protein and ATP/ mixture were from LPS, or, for some experiments, the complement protein C1q that shares Upstate Biotechnology; C1q was from Advanced Research Technologies. All structural and functional homology with SP-A. After culture, cells were other reagents (except as noted) were obtained from Sigma-Aldrich. lysed on ice for 30 min in 500 ␮l of lysis buffer (50 mM Tris-HCl (pH 8.0), SP-A purification 150 mM NaCl, 1% Nonidet P-40). The lysates were spun at 9300 ϫ g for 15 min, and the supernatants were precleared by adding protein A-agarose Human SP-A was purified from the bronchoalveolar lavage of patients with (50 ␮l) and incubated at 4°C for 45 min, followed by centrifugation at alveolar proteinosis, as described in detail (35). Briefly, the lavage fluid 9300 ϫ g for 10 min. The precleared supernatant was incubated with anti- was treated with butanol to extract SP-A, and the resulting pellet was aPKC Ab, anti-p65 Ab, control IgG, or no Ab for2hat4°C, after which sequentially solubilized in octylglucoside and 5 mM Tris (pH 7.4). SP-A 50 ␮l of protein A-agarose was added for2hat4°Cwith gentle rotation. was treated with polymyxin B agarose beads to reduce endotoxin contam- The immune complexes were collected by centrifugation at 9300 ϫ g for ination. SP-A preparations were tested for the presence of bacterial endo- 5 min at 4°C, washed three times with cold lysis buffer, and released by toxin using a Limulus amebocyte lysate assay (BioWhittaker); all SP-A boiling with 5ϫ sample buffer. Samples were used to determine kinase preparations used contained Ͻ0.2 pg endotoxin/␮g SP-A. activity or for Western analysis. Stimulation of rat AM Western analysis AM were isolated, as described previously (22). Cells were plated at 0.8 ϫ Western analysis was performed on cytosolic extracts, membrane fractions, 106/500 ␮l in 24-well plates (Nunc) and allowed to attach for 90 min at and immunoprecipitated samples from rat and mouse cells. Experiments 4482 SP-A-ACTIVATED PKC␨ IN I␬B-␣ STABILIZATION Downloaded from http://www.jimmunol.org/ by guest on September 29, 2021

FIGURE 1. SP-A fails to inhibit LPS-induced activation of AKBI cells. Pooled PBMC or AM from either three to four AKBI (A and C) or three wt mice (B and D) were left untreated or treated with 20–60 ␮g/ml SP-A (37°C, 1 h), and then exposed either to medium, or to 0.1–100 ng/ml LPS. Cell-free supernatants were harvested after 4 h for the determination of TNF-␣ by ELISA. The data shown are mean Ϯ SE of three independent p Ͻ 0.01; †, p Ͻ 0.001 (vs ,ءء ;p Ͻ 0.05 ,ء .experiments. Statistical analysis was performed using a two-way ANOVA with a Bonferroni posttest LPS-induced TNF-␣ release in the absence of SP-A). SP-A fails to inhibit LPS-induced p65 nuclear translocation in AKBI AM. E, AM from wt (a–d) or AKBI (e–h) mice were left untreated or treated with 40 ␮g/ml SP-A (37°C, 1 h), exposed to medium, or to 100 ng/ml LPS (1 h), and then analyzed by confocal microscopy. An Ab against NF-␬B p65 was detected by goat anti-rabbit IgG Ab conjugated to Alexa Fluor 488 (green), and cell nuclei were counterstained with propidium iodide (red). Overlays of single stainings are shown. Images are representative of three independent experiments. p Ͻ 0.001 ,ءء .F, Pixel densitometry of NF-␬B p65 was quantified and statistically analyzed with one-way ANOVA and Newman-Keuls post hoc test (vs control); #, p Ͻ 0.001 (vs LPS).

using the inhibitors above were performed under four different condi- and transferred to nitrocellulose membrane. The membranes were then tions, as follows: 1) in the absence of SP-A and LPS (basal); 2) in the incubated with anti-I␬B-␣, anti-aPKC, anti-PKCThr410/403, anti-p65, or presence of SP-A (40 ␮g/ml, 1 h) (constitutive); 3) in the presence of ␤-actin (mouse monoclonal) at a 1/700, 1/200, 1/700, 1/200, and 1/1000 LPS (10 ng/ml, 30 min) (induced); and 4) in the presence of SP-A (40 dilution, respectively. Goat anti-rabbit IgG-HRP or rabbit anti-mouse ␮g/ml, 1 h) before LPS (10 ng/ml, 30 min) (modulated). After treat- IgG-peroxidase conjugate served as secondary Abs. Immunoreactive ment, cytosolic fractions were assayed for protein content by the bicin- proteins were visualized using the ECL Western blotting detection sys- choninic acid reagent (Pierce Biotechnology), separated on SDS-PAGE, tem (Amersham). The Journal of Immunology 4483

FIGURE 2. An aPKC is involved in I␬B-␣ sta- bilization. A–D, Representative cytosolic I␬B-␣ and ␤-actin Western blots. A, Primary rat AM were left untreated or treated with 5 ␮MGo¨-6976, 5 nM Go¨- 6850, 6 and 12 ␮M chelerythrine chloride, 50 nM wortmannin (37°C, 30 min), or 30 ␮M apigenin (37°C, 1 h). Equal amounts (30–40 ␮g of protein) of the cytosolic fractions were subjected to SDS- PAGE and immunoblotted for I␬B-␣ or ␤-actin (A– D). Analysis of band intensities was performed us- ing Optimas software (Media Cybernetics). Data are expressed as arbitrary units (a.u.) (mean Ϯ SE) from four to five independent experiments. B, AM were left untreated or preincubated (37°C, 30 or 60 min) with the inhibitors, as in A, and were then exposed to 40 ␮g/ml SP-A (1 h). Data are expressed as per- centage of I␬B-␣ protein expression in the absence of SP-A (dotted line, 100%) (mean Ϯ SE) from four to five independent experiments. C, AM were left Downloaded from untreated or treated with the inhibitors, as in A, and were then exposed to 10 ng/ml LPS (30 min). Data are expressed as a.u. (mean Ϯ SE) from four to five independent experiments. D, AM were left un- treated or treated with SP-A, as in B, and were then exposed to 10 ng/ml LPS (30 min). Data are ex- pressed as percentage of I␬B-␣ protein expression http://www.jimmunol.org/ in the absence of SP-A (dotted line, 100%) (mean Ϯ SE) from four to five independent experiments. Data in A and C were analyzed by one-way ANOVA, followed by Newman-Keuls post hoc test; data in B and D were analyzed by paired Student’s t test when ,ء .expressed as percentage of the control response p Ͻ 0.05 (vs I␬B-␣ expression in the absence of SP-A). by guest on September 29, 2021

In vitro aPKC kinase assay Total cell lysates were then centrifuged at 100,000 ϫ g for 30 min at 4°C to separate cytosolic from particulate fractions. The resulting pel- ␮ Immunoprecipitated aPKC was subjected to the kinase assay in 100 l lets were extracted in 150 ␮l of hypotonic fractionation buffer contain- ␤ of reaction mixture (20 mM MOPS (pH 7.2), 25 mM -glycerol phos- ing 0.5% Triton X-100 and centrifuged at 16,000 ϫ g for 20 min at 4°C. phate, 1 mM DTT, 5 mM EGTA, 1 mM Na3VO4, 75 mM MgCl2, 0.5 The resulting supernatant was taken to represent the membrane fraction. ␮ ␥ 32 ␮ mM ATP, 100 Ci of [ - P]ATP) and 10 l of myelin basic protein Equal amount of membrane protein for each sample (30–40 ␮g) was substrate at 37°C with gentle shaking. Reaction was stopped by adding separated by SDS-PAGE and blotted with anti-aPKC Ab. 50 ␮l of Laemmli buffer. Immunocomplexes were released from aga- rose beads by boiling and centrifugation. The supernatant was trans- ferred to a new vial and subjected to 12% SDS-PAGE. The gel was analyzed with a PhosphorImager (Amersham Biosciences) to quantitate Immunofluorescence microscopy band intensities. AM were plated at a density of 1 ϫ 105 cells on 8-well Lab Tek II chamber slides (Nunc). Cells were allowed to attach for 90 min at 37°C in a 5% CO ␬ 2 NF- B activation assay atmosphere in the presence of 0.2% HI-FCS. After incubation with the Ϫ After exposing the cells to the experimental conditions, nuclear extracts indicated stimuli, cells were fixed with ice-cold ( 20°C) methanol, washed were prepared and analyzed, as described above. The activity of NF-␬Bin with PBS, and permeabilized with 0.25% Triton X-100. After a repeated the nuclear extracts was determined by EMSA. NF-␬B oligonucleotides washing, the cells were blocked with 10% BSA/PBS, and incubated with ␨ were end labeled with [␥-32P]ATP using T4 kinase. Two micrograms of isoform-specific anti-PKC Ab, anti-p65 Ab, or control IgG at a 1/250, crude nuclear extract was incubated for 20 min in binding buffer containing 1/50, or 1/100 dilution, respectively. Goat anti-rabbit IgG conjugated to 50 ␮g of poly(dI/dC)/ml with 7.5 fmol of the 32P-labeled oligonucleotides Alexa Fluor 488 served as secondary Ab. Cell nuclei were counterstained encoding the consensus NF-␬B site 5Ј-AGCTCAGAGGGGACTTTC with propidium iodide. Samples were analyzed using a Leica TCS SP CGAGAGAGC-3Ј. Samples were separated by electrophoresis in 5% poly- confocal laser scanning microscope (Leica Microsystems). All images acrylamide gels for2hat180V,after which gels were analyzed with a were acquired under identical settings with Leica TCSNT software and PhosphorImager. assembled using Adobe Photoshop 6.0. An average of 120 (Fig. 1F)or100 (Fig. 5, C and E) cells was counted per condition and experiment, which Assay for aPKC membrane translocation was repeated three times. Rat AM were treated with SP-A (40 ␮g/ml) for the times indicated. Incubation was stopped with ice-cold PBS, and the cells were resus- Statistical analysis pended in 300 ␮l of hypotonic fractionation buffer (10 mM Tris (pH 7.4), 4.5 mM EDTA, 2.5 mM EGTA, 2.3 mM 2-ME, 1 mM PMSF, 10 Data were statistically analyzed, as indicated in the figure legends using ␮g/ml aprotinin, 10 ␮g/ml leupeptin, 10 ␮g/ml pepstatin, and 0.1 mM GraphPad Prism (version 4.0; GraphPad). Values were considered signif- Ͻ Na3VO4), incubated for 20 min at 4°C while rotating, and sonicated. icant when p 0.05. Data are presented as SEM. 4484 SP-A-ACTIVATED PKC␨ IN I␬B-␣ STABILIZATION

Results SP-A fails to inhibit LPS-induced TNF-␣ release by AKBI cells To confirm our previous findings on the role of I␬B-␣ in anti- inflammatory activity of SP-A, in this study, we used primary im- mune cells from I␬B-␣-deficient mice. In AKBI mice, the inte- grated I␬B-␤ gene is under the control of the I␬B-␣ promoter, and at the same time a null mutation in I␬B-␣ is introduced (32). Un- like I␬B-␣-deficient mice, which are postnatal lethal (36), AKBI mice have a normal phenotype, and NF-␬B induction by PMA or TNF-␣ in AKBI mouse fibroblasts and thymocytes is identical compared with wt cells (32). In support of this notion, AKBI mice reveal a functional redundancy of I␬B-␣ and I␬B-␤ upon LPS challenge in vivo (37). In line with these data, we show in this study that PBMC (Fig. 1A) and AM (Fig. 1C) from AKBI mice released similar amounts of TNF-␣ in response to LPS (0.1–100 ng/ml) compared with wt cells (Fig. 1, B and D). However, whereas SP-A (20–60 ␮g/ml) significantly ( p Ͻ 0.05 to p Ͻ 0.001) and in a dose-dependent manner inhibited LPS-induced Downloaded from TNF-␣ release by wt cells (Fig. 1, B and D), SP-A failed to inhibit LPS-induced TNF-␣ release by PBMC (Fig. 1A) or AM (Fig. 1C) from AKBI mice. These data confirm that SP-A-mediated inhibi- tion of LPS-induced TNF-␣ production in PBMC and AM criti- cally depends on the presence of I␬B-␣, and that I␬B-␤ could not compensate the lack of I␬B-␣ under these conditions. http://www.jimmunol.org/

SP-A fails to inhibit LPS-induced p65 nuclear translocation in AKBI AM Because p65 nuclear translocation is tightly linked to NF-␬B- dependent TNF-␣ gene induction (15), we next investigated the effect of SP-A on LPS-induced p65 localization in AKBI and wt AM by confocal microscopy. LPS-induced p65 nuclear accu- FIGURE 3. aPKC inhibition suppresses SP-A-mediated I␬B-␣ stabili- mulation in wt AM (Fig. 1Ec) was almost completely prevented zation. A–D, Representative cytosolic I␬B-␣ and actin Western blots. A, ␮ (60 Ϯ 5%) by pretreatment of the cells with SP-A (Fig. 1, Ed Primary rat AM were left untreated or treated with 2–10 M aPKCps by guest on September 29, 2021 and F). In contrast LPS-induced p65 nuclear accumulation in (37°C, 1 h) (control). Cytosolic cell fractions were immunoblotted for I␬B-␣ or ␤-actin. Densitometric results are in arbitrary units (a.u.) (mean Ϯ AKBI AM (Fig. 1Eg) was not inhibitable by SP-A (Fig. 1, Eh SE) from five independent experiments. B, AM were left untreated or and F). As expected, comparable amounts of nuclear p65 are treated as in A and were then exposed to 40 ␮g/ml SP-A (1 h). Densito- seen under resting or SP-A conditions in wt and AKBI AM. metric results are expressed as percentage of I␬B-␣ protein expression in Together the data suggest that SP-A-mediated inhibition of the absence of SP-A (control, dotted line) (mean Ϯ SE) from five inde- LPS-induced p65 nuclear translocation critically depends on the pendent experiments. C, AM were left untreated or treated as in A and were presence of I␬B-␣. then exposed to 10 ng/ml LPS (30 min). Densitometric results are in a.u. (mean Ϯ SE) from five independent experiments. D, AM were left un- An aPKC is involved in I␬B-␣ stabilization treated or treated with SP-A as in B and were then exposed to 10 ng/ml LPS (30 min). Data are expressed as percentage of I␬B-␣ protein expression in To elucidate the underlying molecular mechanisms of SP-A-me- the absence of SP-A (control, dotted line) (mean Ϯ SE) from five inde- diated immune protection, we focused on identifying the upstream pendent experiments. Data in A and C were analyzed by one-way ANOVA, regulators involved in the stabilization of I␬B-␣ by SP-A. Because followed by Newman-Keuls post hoc test; data in B and D were analyzed members of the PKC family have been shown to be involved in by paired Student’s t test when expressed as percentage of the control .(p Ͻ 0.02 (vs control ,ءء ;p Ͻ 0.05 ,ء .I␬B-␣ turnover (27) and SP-A immune functions (26), we used a response panel of PKC inhibitors with different specificity to test the role of PKC in SP-A-mediated I␬B-␣ stabilization in primary rat AM. Experiments were performed under four different conditions as follows: 1) distinct inhibitors alone (basal, Fig. 2A); 2) inhibitors Go¨-6976. The effect of SP-A on I␬B-␣ stabilization was still sig- plus SP-A (40 ␮g/ml) (constitutive, Fig. 2B); 3) inhibitors plus nificant (149 Ϯ 12% of control) in the presence of Go¨-6976 (Fig. LPS (10 ng/ml) (induced, Fig. 2C); and 4) inhibitors plus SP-A (40 2B), but was abolished in the presence of LPS (Fig. 2D). Whereas ␮g/ml), followed by LPS (10 ng/ml) (modulated, Fig. 2D). Cyto- treatment of AM with Go¨-6850, which inhibits the cPKCs ␣ and ␥ solic I␬B-␣ protein expression was determined by Western as well as the DAG-sensitive but Ca2ϩ-insensitive novel PKC iso- analysis. forms ␦ and ␧, did not inhibit the SP-A-mediated stabilization of Neither of the inhibitors had a significant effect on I␬B-␣ protein I␬B-␣ (Fig. 2B), it inhibited SP-A’s effect in the presence of LPS expression under basal conditions (Fig. 2A). In line with our pre- (Fig. 2D). In sharp contrast, treatment of the cells with cheleryth- vious data (24), SP-A significantly increased I␬B-␣ protein ex- rine chloride, an inhibitor of Ca2ϩ- and DAG-dependent and pression by 210 Ϯ 44% ( p Ͻ 0.05) under constitutive conditions DAG-independent PKCs including the DAG-insensitive atypical (Fig. 2B) and by 137 Ϯ 10% ( p Ͻ 0.05) in the presence of LPS isoforms ␨ and ␭/␫, abolished the stabilizing effect of SP-A on ␣ ␤ ␤ ␥ ␬ ␣ (Fig. 2D). cPKC isoforms ( , I, II, ), which are diacylglycerol I B- both constitutively and in the presence of LPS, suggesting (DAG) sensitive and Ca2ϩ responsive, are specifically inhibited by that aPKCs might be involved in SP-A effects (Fig. 2, B and D). The Journal of Immunology 4485 Downloaded from

FIGURE 4. SP-A stimulates aPKCThr410/403 phosphorylation and kinase activity. A, AM were incubated with 20 ␮g/ml SP-A at 37°C for 1, 3, 5, or 10 min. aPKC was immunoprecipitated from whole cell lysates. The presence of aPKCThr410/403 was detected by immunoblotting with the corresponding Ab. Anti-aPKC Ab was used to evaluate the aPKC loads. Anti-rabbit IgG Ab was used as a control to show specificity for aPKC. Blots are representative of

three independent experiments. B, AM were treated with 20 ␮g/ml SP-A for 0, 5, 10, 30, or 60 min. aPKC was immunoprecipitated from whole cell lysates. http://www.jimmunol.org/ The presence of aPKCThr410/403 was detected by immunoblotting with the corresponding Ab. Densitometric results (mean Ϯ SE) are given as percentage of the zero time point (control) from four independent experiments. C, AM were left untreated or treated with 20 ␮g/ml SP-A for the times indicated. aPKC was immunoprecipitated from cell lysates and subjected to an in vitro kinase assay using myelin basic protein as a substrate. aPKC kinase activity was determined, as described in Materials and Methods. The phosphorylated proteins were run on a 12% SDS-PAGE gel. D, Densitometrical analysis of four independent experiments. The SP-A structural homologue C1q (20 ␮g, 1 h) was included as control protein. Data are expressed as percentage of aPKC activity at the corresponding time point (control). E, AM were left untreated (lane 1), or treated with 40 ␮g/ml SP-A (lane 2) or 100 ng/ml LPS (lane 3), or pretreated with 10 ␮M aPKCps peptides before the addition of 40 ␮g/ml SP-A (lane 4). Cells were subjected to an in vitro aPKC kinase assay. The gel is representative of three independent experiments. Data were analyzed by a one-way ANOVA with a Dunnett’s post hoc test, or with paired Student’s t .(p Ͻ 0.05 (vs control ,ء .test by guest on September 29, 2021

Both SP-A and LPS can activate PI3K in human macrophages 3D). Treatment of the cells with aPKCps at 10 ␮M resulted in (38) and in THP-1 cells (39), respectively. In the present study, a significant inhibition of I␬B-␣ by SP-A (Fig. 3B). Interest- the PI3K inhibitor wortmannin significantly reduced I␬B-␣ ex- ingly, in the presence of LPS, aPKCps pretreatment at any con- pression in the presence of LPS (Fig. 2C). Wortmannin abro- centration resulted in a significant inhibition ( p Ͻ 0.02) of gated SP-A’s effect on I␬B-␣ stabilization both constitutively I␬B-␣ protein levels by SP-A (Fig. 3D) when compared with and in the presence of LPS (Fig. 2, B and D). In mouse em- the corresponding aPKCps concentration in the absence of bryonic fibroblasts, CKII is critically involved in the basal turn- SP-A. As expected, LPS-induced NF-␬B activity in AM was over of I␬B-␣ (12). Apigenin, a selective CKII inhibitor, in- abolished after pretreatment with aPKCps (data not shown). hibited I␬B-␣ stabilization by SP-A constitutively and in the Taken together, these results suggest that an aPKC isoform is presence of LPS (Fig. 2, B and D). involved in I␬B-␣ stabilization by SP-A both constitutively and Because, among aPKCs, PKC␨ has been shown to modulate in the presence of LPS. I␬B-␣ turnover (27) and IKK/NF-␬B activation in both nuclear and whole lung extracts (33), we wanted to elucidate the role of 410/403 aPKC isoforms in SP-A’s AM immunomodulation more precisely. SP-A stimulates aPKCThr phosphorylation and kinase Therefore, we used the isoform-specific cell-permeable inhibi- activity tory myristoylated peptide derived from the ps motif of aPKCs. To investigate whether SP-A can stimulate aPKC activation, the aPKCps mimics the substrate and maintains aPKC in its non- phosphorylation status of aPKC was determined. To activate active form. Of note, the ps peptides are not totally specific, aPKC, phosphorylation of the activation loop consensus threo- because the ps sequences (SIYRRGARRWRKL) are identical nine residue Thr410 by PI3K-dependent PDK1 is substantial in both isoforms PKC␨ and PKC␭/␫ (40). (41). In fact, treatment of AM with SP-A significantly increased aPKCThr410/403 phosphorylation after 1–10 min (Fig. 4A)of ␬ ␣ aPKC inhibition suppresses SP-A-mediated I B- stabilization incubation and then declined to baseline (Fig. 4B). Again, cytosolic I␬B-␣ protein in AM was determined under We then investigated whether the observed SP-A-mediated in- four different conditions (basal, in the presence of SP-A, or crease in aPKCThr410/403 phosphorylation correlates with an in- LPS, or both). Treatment of the cells with aPKCps (2–10 ␮M) creased kinase activity. SP-A, but not C1q, a structural homologue did not significantly affect I␬B-␣ protein level under basal (Fig. of SP-A, stimulates aPKC activity in a time-dependent manner, 3A) or LPS (Fig. 3C) conditions. Compared with basal condi- reaching a maximum at1h(p Ͻ 0.05) (Fig. 4, C and D). SP-A- tions, SP-A significantly increased I␬B-␣ protein expression induced aPKC activity (Fig. 4E, lane 2) was comparable to that both constitutively (Fig. 3B) and in the presence of LPS (Fig. induced by LPS (Fig. 4E, lane 3), used as a positive control (42). 4486 SP-A-ACTIVATED PKC␨ IN I␬B-␣ STABILIZATION Downloaded from http://www.jimmunol.org/ by guest on September 29, 2021

FIGURE 5. SP-A favors PKC␨ plasma membrane translocation. A, AM were left untreated or treated with 40 ␮g/ml SP-A for the times indicated. Cell membrane isolation was performed, as described in Materials and Methods. Cytosolic and membrane fractions were subjected to SDS-PAGE and immunoblotted for aPKC. Data shown are representative of three experiments. B, AM adhered to chamber slides at 1 ϫ 105 cells/well and were left untreated and then exposed to medium or treated with 40 ␮g/ml SP-A (1 h), and were then further either exposed to 100 ng/ml LPS (1 h) or treated with 100 ng/ml LPS (1 h) alone. Slides were blocked and incubated with an isotype-specific Ab (control IgG; a), or a specific Ab against PKC␨ (b–e) that was detected by goat anti-rabbit IgG Ab conjugated to Alexa Fluor 488. f–j, Cell nuclei were counterstained with propidium iodide (PI). Cells were visualized by confocal microscopy. k–o, Overlay of single stainings. Images shown are representative of three independent experiments with similar results. C, Pixel density of cell-membrane PKC␨ was quantified from total PKC␨ and statistically analyzed by one-way p Ͻ 0.001 (vs control); #, p Ͻ 0.001 (vs LPS). D, AM were left untreated or treated with 40 ␮g/ml ,ءء .ANOVA and Newman-Keuls post hoc test SP-A (1 h), and then exposed to medium, or to 100 ng/ml LPS (1 h) and further treated as described for B. a–e, Cells were incubated with an isotype-specific Ab (control IgG; a), or an Ab against NF-␬B p65 (b–e) that was detected by goat anti-rabbit IgG Ab conjugated to Alexa Fluor 488. f–j, Cell nuclei were counterstained with PI; k–o, overlay of single stainings. Images shown are representative of three independent experiments. E, Pixel density of cell membrane was quantified from total NF-␬B p65 and statistically analyzed by one-way ANOVA and Newman-Keuls post hoc p Ͻ 0.001 (vs control); #, p Ͻ 0.001 (vs LPS). SP-A enhances PKC/p65 coimmunoprecipitation under resting conditions. F, AM were left ,ءء .test untreated or treated with 40 or 60 ␮g/ml SP-A (1 h). NF-␬B p65 was immunoprecipitated from whole cell lysates, as described in Materials and Methods. Anti-rabbit IgG or no Ab was used as a control. aPKC was detected by immunoblotting with the corresponding Ab. Anti-p65 Abs were used to evaluate p65 loads. G, Densitometric results of three independent experiments. Data are expressed as arbitrary units (a.u.) (mean Ϯ SE). Data .(p Ͻ 0.05 (vs untreated ,ء .were analyzed by one-way ANOVA with DunnettЈs posttest

Incubation of AM with aPKCps peptides (Fig. 4E, lane 4) abol- translocation by two approaches, i.e., membrane fractionation and ished the aPKC activity in the presence of SP-A. Together, the confocal microscopy. results support the idea that SP-A mediates I␬B-␣ stabilization in ␨ primary AM via activation of aPKC. SP-A favors the accumulation of PKC in the plasma membrane Even though phosphorylation at Thr410 facilitates kinase activ- Cell fractionation demonstrated that aPKC membrane translo- ity, it is not sufficient to provide full activity. A rapid autophos- cation is stimulated by SP-A in a time-dependent manner, phorylation is necessary for a concomitant translocation of PKC␨ reaching a maximum at 1 h (Fig. 5A). Using confocal micros- (43, 44). Therefore, we examined the effect of SP-A on aPKC copy, we confirmed that SP-A, compared with basal conditions, The Journal of Immunology 4487 Downloaded from http://www.jimmunol.org/ by guest on September 29, 2021

FIGURE 5. (continued)

favors a translocation of PKC␨ to the plasma membrane in pri- ence of LPS, favored both PKC␨ and p65 membrane mary AM, as detected by an isoform-specific Ab (Fig. 5B, l and localization, the effect of SP-A on PKC␨/p65 interaction was m). In contrast, LPS stimulation of AM induced a translocation examined next. SP-A, significantly and in a concentration-de- of PKC␨ (Fig. 5Bn) toward the nucleus. Pretreatment of the pendent manner, increased PKC␨/p65 coimmunoprecipitation cells with SP-A, however, largely reduced LPS-mediated nu- under constitutive conditions (Fig. 5, F and G). Because the clear translocation of PKC␨ (Fig. 5Bo). SP-A-enhanced interaction obviously has no NF-␬B trans-ac- tivating potency, we hypothesized that this effect of SP-A di- ␨ SP-A modulates p65 localization and p65/PKC rectly or indirectly prevents p65 nuclear translocation. coimmunoprecipitation ␨ ␬ ␣ Stimulus-induced PKC␨ associates with and phosphorylates PKC is essential for SP-A-mediated I B- stabilization and ␬ trans-activating p65 (33, 45). We next asked whether SP-A- inhibition of LPS-induced NF- B activity favored PKC␨ membrane localization is associated with the dis- To establish the selective role of PKC␨ in mediating the SP-A tribution of p65. We confirmed our initial observation in wt effect described, we used mice deficient for the PKC␨ isoform (33). mouse AM (Fig. 1E) and in rat AM (Fig. 5D), and indeed re- Whereas PKC␭/␫Ϫ/Ϫ mice are embryonic lethal (46), PKC␨Ϫ/Ϫ vealed that p65 distribution strongly parallels that of PKC␨ mice are grossly normal, but exhibit an impairment of IKK (Fig. 5B) under SP-A or LPS conditions (Fig. 5, B, Dc, Dd, and activation in whole lung extracts after LPS challenge (33). In E). Furthermore, as in wt mouse AM, pretreatment of the cells freshly isolated AM from PKC␨Ϫ/Ϫ mice, however, NF-␬Bac- with SP-A almost abolished LPS-induced p65 nuclear translo- tivation was apparent both basal and LPS induced. SP-A failed cation and induced an accumulation of p65 at the plasma mem- to inhibit basal and only slightly reduced LPS-induced NF-␬B brane (Fig. 5, De and E). Because SP-A, alone or in the pres- DNA binding compared with wt AM (Fig. 6, A and B, lower 4488 SP-A-ACTIVATED PKC␨ IN I␬B-␣ STABILIZATION

FIGURE 6. PKC␨ is vital for SP- A-mediated I␬B-␣ stabilization and inhibition of LPS-induced NF-␬B ac- tivity. A and B, Pooled AM from ei- ther four wt (A) or four PKC␨-defi- cient mice (B) were left untreated or treated with 40 ␮g/ml SP-A (1 h), and then exposed to either medium or 100 ng/ml LPS (1 h). Cytosolic cell frac- tions were immunoblotted for I␬B-␣ (upper panel). Nuclear extracts of the cells were analyzed by EMSA for NF-␬B DNA-binding activity Downloaded from (middle panel). Data shown are rep- resentative of three independent ex- periments. Densitometric results are expressed as arbitrary units (a.u.). Data were analyzed by one-way ANOVA with a post hoc Newman- /p Ͻ http://www.jimmunol.org ,ءء ;p Ͻ 0.01 ,ء .Keuls test 0,001 (vs control); #, p Ͻ 0.01; ##, p Ͻ 0.001 (vs LPS). by guest on September 29, 2021

panel). Compared with wt AM, SP-A-mediated I␬B-␣ stabili- ␬B-dependent gene expression in the basal state of cell activation zation was almost abolished in PKC␨Ϫ/ϪAM (Fig. 6, A and B, and for making p65/I␬B complexes responsive to diverse stimuli. upper panel). Surprisingly, in the presence of LPS, I␬B-␣ was We previously suggested that SP-A inhibits LPS-induced NF-␬B enhanced in PKC␨Ϫ/Ϫ AM compared with wt AM, and this activation by posttranscriptionally slowing the basal turnover of effect was abolished when cells had been pretreated with SP-A I␬B-␣ in primary AM (24). However, the in vivo proof and the (Fig. 6B). Taken together, these data indicate that PKC␨, con- intracellular signaling pathways involved have not yet been stitutively and in the presence of LPS is required for SP-A- defined. mediated anti-inflammatory signaling in AM. The present study confirms that I␬B-␣ is critically involved in SP-A anti-inflammatory effects. In AKBI mice, the integrated Discussion I␬B-␤ gene is under the control of the I␬B-␣ promoter, and at the Emerging evidence demonstrates a pivotal role of the pulmonary same time a null mutation in I␬B-␣ is introduced (32). AKBI mice collectin SP-A in anti-inflammatory immunomodulation of innate have a normal phenotype and reveal a functional redundancy of immune responses of the lung to LPS (1, 2). Airway inflammation I␬B-␣ and I␬B-␤ upon LPS challenge in vivo (37). In line with associated with local or systemic LPS release from Gram-negative this, we found that LPS-induced TNF-␣ release by AKBI cells was bacteria is still a major cause of life-threatening pulmonary dis- comparable to that by wt cells, confirming the described in vivo eases (8). Efficient negative signaling cascades to prevent auto- redundancy of I␬B-␣ and I␬B-␤ in response to LPS (37). How- toxic mediator release by AM, the major effector cell of the pul- ever, whereas SP-A significantly inhibited LPS-induced TNF-␣ monary innate immune system, in response to LPS, and, in release (Fig. 1, B and D) and p65 nuclear translocation in wt cells particular, the modulation of NF-␬B activation threshold by SP-A (Fig. 1Ed), it failed to inhibit TNF-␣ production (Fig. 1, A and C) have been studied intensively in recent years. Inhibition of LPS- as well as p65 nuclear accumulation in AKBI cells (Fig. 1Eh). induced NF-␬B activity by SP-A has been suggested to occur via These data strongly suggest that I␬B-␤ could not compensate the direct interaction of SP-A with components of the LPS receptor lack of I␬B-␣ in SP-A-mediated inhibition of LPS-induced cell complex, including LPS-binding protein, CD14, TLR-4, and activation. MD-2, but also independently of LPS-specific signal transduction The present study further identifies aPKC␨ as an essential up- pathways (19–21, 23, 24, 47, 48). Intracellularly, the pivotal role stream mediator of I␬B-␣/NF-␬B regulation by SP-A in primary of I␬B-␣ in NF-␬B regulation has recently been highlighted by cells. In PKC␨Ϫ/Ϫ AM, SP-A-mediated I␬B-␣ stabilization was Tergaonkar et al. (13), demonstrating that I␬B-␣ (as well as ␤ and almost completely abrogated (Fig. 6B), strongly supporting our ␧) is essential for both preventing NF-␬B DNA binding and NF- experiments using the inhibitory aPKCps peptides in rat AM The Journal of Immunology 4489

FIGURE 7. A model implicating PKC␨ in SP-A- mediated I␬B-␣ stabilization, leading to inhibited NF-␬B-induced cell activation. A, In resting AM, SP-A-activated PKC␨ is implicated as a p65-inter- acting kinase that reduces macrophage-inflamma- tory responsiveness by promoting I␬B-␣ stabiliza- tion via retarded degradation (broken arrow), subsequently altering the threshold for NF-␬B acti- vation. B, Classical LPS-induced NF-␬B activation pathway. LPS-induced PKC␨ leads to I␬B-␣ phos- phorylation, ubiquitination, and degradation (bold arrow) via IKK, allowing p65 nuclear translocation and NF-␬B activation. Downloaded from

(Fig. 3, A and B). The combined data confirm that the PKC␨ iso- SP-A, directly or indirectly, prevents p65 nuclear translocation.

form is critically involved in SP-A-mediated I␬B-␣ stabilization Confocal and cell fractionation analysis of p65 localization in rat http://www.jimmunol.org/ and suggest that the inhibition or the lack of PKC␨ potentiates the AM revealed a substantial inhibition of LPS-induced p65 nuclear rate of I␬B-␣ turnover in resting AM. translocation by SP-A. Based on the combined results, we propose In contrast to the well-characterized I␬B-␣ turnover under stim- a model in which SP-A-mediated PKC␨ activation stabilizes I␬B-␣ ulus-induced conditions, little is known about the mechanisms that by preventing p65 nuclear translocation in primary AM. regulate the basal turnover of I␬B-␣. In mouse embryonic fibro- A previous study (33) showed that NF-␬B DNA-binding activ- blasts, an efficient basal turnover of I␬B-␣ requires the C-terminal ity is reduced in nuclear lung extracts of PKC␨Ϫ/Ϫ mice treated CKII phosphorylation (Ser293), whereas IKK phosphorylation with LPS or IL-1 i.p. In whole lung extracts, the lack of PKC␨ (Ser32 and Ser36) plays no role under basal conditions (12). Even resulted in an impairment of IKK activation upon LPS stimulation, though I␬B-␣ is not a direct substrate of PKC␨ (49), PKC␨-asso- suggesting a substantial role of PKC␨ in the control of stimulus- by guest on September 29, 2021 ciated CKII preferentially phosphorylates Ser293 compared with induced IKK/NF-␬B signaling cascade in the pulmonary compart- nonassociated CKII in transfected NIH3T3 cells, thereby poten- ment (33). In the present study, I␬B-␣ expression in response to tially accelerating the basal turnover of I␬B-␣ (27). In the present LPS was decreased in wt AM (Fig. 6A), as expected. Surprisingly, study, however, the inhibition or the lack of PKC␨ accelerated LPS enhanced I␬B-␣ in PKC␨Ϫ/Ϫ AM (Fig. 6B). Possible expla- I␬B-␣ turnover under basal conditions in AM. In addition, apige- nations might include that the lack of PKC␨ impairs the LPS- nin, a selective CKII inhibitor, abolished the SP-A-mediated sta- induced IKK activation and the subsequent I␬B-␣ degradation, as bilization of I␬B-␣, suggesting that in AM both PKC␨ and CKII previously suggested (33). Alternatively, the I␬B-␣ increase ob- activity contribute to this process. served in PKC␨Ϫ/Ϫ AM could result from de novo transcription One central route controlling I␬B-␣ turnover is the proteasome- and might be referred to a compensatory increase of PKC␭/␫. How- mediated degradation of ubiquitinated I␬B-␣. PKC␨ has been ever, Western blot analysis of lung extracts from wt and PKC␨- shown to influence proteasome-mediated protein degradation (50, deficient mice for PKC␨ and PKC␭/␫ protein expression with spe- 51). However, when we investigated the effect of SP-A on I␬B-␣ cific Abs revealed that the level of PKC␭/␫ is not affected by the ubiquitination in AM, SP-A enhanced the presence of ubiquitin- loss of PKC␨ (55), rendering the latter explanation unlikely. The conjugated I␬B-␣ (and other proteins) alone and in combination LPS-mediated I␬B-␣ increase in PKC␨Ϫ/Ϫ AM was abolished by with a proteasome inhibitor and/or aPKCps, suggesting that SP- pretreatment of the cells with SP-A, proposing that SP-A not only A-activated PKC␨ does not stabilize I␬B-␣ by inhibiting its deg- fails to stabilize I␬B-␣, but provides a signal that triggers I␬B-␣ radation via the ubiquitin-dependent pathway (data not shown). degradation, presumably by activating IKK in a PKC␨-indepen- The finding that this PKC isoenzyme is involved in SP-A-me- dent way. Of note, in PKC␨Ϫ/Ϫ AM, SP-A failed to inhibit basal diated anti-inflammation was initially surprising because LPS- or and LPS-induced NF-␬B activation compared with wt cells, sug- cytokine-induced PKC␨ activity was previously shown to induce gesting that the down-modulation of constitutive and LPS-induced NF-␬B activation by distinct mechanisms, as follows: first, NF-␬B activity by SP-A is dependent on PKC␨ (Fig. 6B). through its activation of IKK (52, 53), and second, by phosphor- The mechanisms underlying PKC␨ activation are not completely ylating p65 (45, 54). Besides PKC␨, kinases implicated in p65 understood and display substantial differences for different cell phosphorylation include CKII, protein kinase A, IKK2, Akt, p38, types, stimuli, and incubation conditions (30). PKC␨ can be acti- p42, and p44 (17). Among these kinases, only PKC␨ has been vated in vitro by phosphatidylinositol-3,4,5-triphosphat (42, 56), shown to directly interact with p65 (33) and phosphorylates p65 at phosphatidic acid (57), ceramide, and arachidonic acid (58, 59), Ser311 in TNF-␣-stimulated mouse fibroblasts (45). In the present and/or by direct interaction with binding proteins (60). To activate study, SP-A enhanced the interaction of PKC␨ with p65 in primary PKC␨, phosphorylation of the activation loop consensus threonine AM under constitutive conditions (Fig. 5D). Because SP-A alone residue Thr410 by PI3K-induced phosphoinositide-dependent ki- has no NF-␬B trans-activating potency, we thus hypothesized that nase 1 is substantial (41). Besides different activation mechanisms, 4490 SP-A-ACTIVATED PKC␨ IN I␬B-␣ STABILIZATION

distinct intracellular localizations may have a major impact on 10. Shimazu, R., S. Akashi, H. Ogata, Y. Nagai, K. Fukudome, K. Miyake, and cell-specific PKC␨ function (29). We show that SP-A enhanced M. Kimoto. 1999. MD-2, a molecule that confers lipopolysaccharide responsive- ness on Toll-like receptor 4. J. Exp. Med. 189: 1777–1782. 410/403 aPKCThr phosphorylation (Fig. 4, A and B), aPKC kinase 11. Akira, S., and K. Takeda. 2004. Toll-like receptor signalling. Nat. Rev. Immunol. activity (Fig. 4, C–E), and translocation (Fig. 5, A and B) in pri- 4: 499–511. 12. Pando, M. P., and I. M. Verma. 2000. Signal-dependent and -independent deg- mary rat AM. In line with previous work (60), we found that LPS radation of free and NF-␬B-bound I␬B␣. J. Biol. Chem. 275: 21278–21286. treatment of AM favored a nuclear accumulation of PKC␨, in con- 13. Tergaonkar, V., R. G. Correa, M. Ikawa, and I. M. Verma. 2005. Distinct roles trast to the cell membrane accumulation of the kinase in the pres- of I␬B proteins in regulating constitutive NF-␬B activity. Nat. Cell Biol. 7: 921–923. ence of SP-A. However, pretreatment of the cells with SP-A 14. Brown, K., S. Gerstberger, L. Carlson, G. Franzoso, and U. Siebenlist. 1995. largely prevented LPS-induced subcellular localization of the ki- Control of I␬B-␣ proteolysis by site-specific, signal-induced phosphorylation. nase (Fig. 6B), the biological role of which remains to be estab- Science 267: 1485–1488. 15. Ghosh, S., and M. Karin. 2002. Missing pieces in the NF-␬B puzzle. Cell 109 lished. Interestingly, recent data suggest that LPS-induced and (Suppl. 1): S81–S96. PI3K-dependent PKC␨ activation can be modified by anti- 16. Brown, K., S. Park, T. Kanno, G. Franzoso, and U. Siebenlist. 1993. Mutual ␬ ␬ ␣ inflammatory mediators. As shown in RAW 264.7 cells, LPS-in- regulation of the transcriptional activator NF- B and its inhibitor, I B- . Proc. Natl. Acad. Sci. USA 90: 2532–2536. duced PKC␨ activation is potentiated by the lipid mediator 15- 17. Chen, L. F., and W. C. Greene. 2004. Shaping the nuclear action of NF-␬B. Nat. ⌬ Rev. Mol. Cell Biol. 5: 392–401. deoxy- (12, 14)-PGJ2, resulting in an inhibition of IKK and ␬ 18. McIntosh, J. C., S. Mervin-Blake, E. Conner, and J. R. Wright. 1996. Surfactant NF- B activity (61). protein A protects growing cells and reduces TNF-␣ activity from LPS-stimu- Fig. 7 depicts a model implicating SP-A-induced PKC␨ as a lated macrophages. Am. J. Physiol. 271: L310–L319. p65-interacting kinase that reduces macrophage-inflammatory re- 19. Sano, H., H. Sohma, T. Muta, S. Nomura, D. R. Voelker, and Y. Kuroki. 1999. Pulmonary surfactant protein A modulates the cellular response to smooth and ␬ ␣ Downloaded from sponsiveness by promoting I B- stabilization under resting con- rough lipopolysaccharides by interaction with CD14. J. Immunol. 163: 387–395. ditions, subsequently leading to inhibition of LPS-induced NF-␬B 20. Gardai, S. J., Y. Q. Xiao, M. Dickinson, J. A. Nick, D. R. Voelker, K. E. Greene, activation. However, the mechanisms of PKC␨ activation as well and P. M. Henson. 2003. By binding SIRP␣ or calreticulin/CD91, lung collectins ␨ ␬ ␣ act as dual function surveillance molecules to suppress or enhance inflammation. as the PKC effectors operative in I B- -dependent anti- Cell 115: 13–23. inflammatory modulation by SP-A in the resting AM were not 21. Garcia-Verdugo, I., F. Sanchez-Barbero, K. Soldau, P. S. Tobias, and C. Casals. identified in the present study, and will be the focus of future 2005. Interaction of SP-A (surfactant protein A) with bacterial rough lipopoly- saccharide (Re-LPS), and effects of SP-A on the binding of Re-LPS to CD14 and

efforts. LPS-binding protein. Biochem. J. 391: 115–124. http://www.jimmunol.org/ In summary, the evidence presented in this work indicates that 22. Stamme, C., E. Walsh, and J. R. Wright. 2000. Surfactant protein A differentially ␬ ␣ regulates IFN-␥- and LPS-induced nitrite production by rat alveolar macro- anti-inflammatory macrophage activation via I B- by SP-A crit- phages. Am. J. Respir. Cell Mol. Biol. 23: 772–779. ically involves PKC␨ activity. In addition, the data imply a possi- 23. Alcorn, J. F., and J. R. Wright. 2004. Surfactant protein A inhibits alveolar mac- ble role of PKC␨ as a positive or negative regulator in I␬B-␣/ rophage cytokine production by CD14-independent pathway. Am. J. Physiol. 286: ␬ L129–L136. NF- B signaling pathways in a stimulus-specific manner in the 24. Wu, Y., S. Adam, L. Hamann, H. Heine, A. J. Ulmer, U. Buwitt-Beckmann, and lung. Because uncontrolled NF-␬B activation may well be delete- C. Stamme. 2004. Accumulation of inhibitory ␬B-␣ as a mechanism contributing rious, tonic activation of PKC␨ by SP-A may serve as a physio- to the anti-inflammatory effects of surfactant protein-A. Am. J. Respir. Cell Mol. ␣ Biol. 31: 587–594. logical brake on induction of TNF- in AM localized to sites of 25. Crowther, J. E., V. K. Kutala, P. Kuppusamy, J. S. Ferguson, A. A. Beharka, pulmonary inflammation. Further studies on the role of SP-A im- J. L. Zweier, F. X. McCormack, and L. S. Schlesinger. 2004. Pulmonary surfac- by guest on September 29, 2021 munoregulatory mechanisms unique to the lung will potentially tant protein A inhibits macrophage reactive oxygen intermediate production in response to stimuli by reducing NADPH oxidase activity. J. Immunol. 172: lead to specific therapeutic strategies in inflammatory and infec- 6866–6874. tious human lung diseases. 26. Schagat, T. L., M. J. Tino, and J. R. Wright. 1999. Regulation of protein phos- phorylation and pathogen phagocytosis by surfactant protein A. Infect. Immun. 67: 4693–4699. Acknowledgments 27. Bren, G. D., K. N. Pennington, and C. V. Paya. 2000. PKC-␨-associated CK2 participates in the turnover of free I␬B␣. J. Mol. Biol. 297: 1245–1258. We thank Professor John Engelhardt (University of Iowa, Iowa City, IA) 28. Moscat, J., and M. T. Diaz-Meco. 2000. The atypical protein kinase Cs: func- for generously providing the AKBI mice, Professor Karl Dalhoff (Univer- tional specificity mediated by specific protein adapters. EMBO Rep. 1: 399–403. sity of Lu¨beck, Lu¨beck, Germany) for providing lung lavage fluid from 29. Tan, S. L., and P. J. Parker. 2003. Emerging and diverse roles of protein kinase alveolar proteinosis patients, and Dr. Prim B. Singh (Research Center Bor- C in immune cell signalling. Biochem. J. 376: 545–552. ␨ ␨ stel, Borstel, Germany) for critical reading of the manuscript. 30. Hirai, T., and K. Chida. 2003. Protein kinase C (PKC ): activation mechanisms and cellular functions. J. Biochem. 133: 1–7. 31. Le Good, J. A., and D. N. Brindley. 2004. Molecular mechanisms regulating Disclosures protein kinase C␨ turnover and cellular transformation. Biochem. J. 378: 83–92. 32. Cheng, J. D., R. P. Ryseck, R. M. Attar, D. Dambach, and R. Bravo. 1998. The authors have no financial conflict of interest. Functional redundancy of the nuclear factor ␬B inhibitors I␬B␣ and I␬B␤. J. Exp. Med. 188: 1055–1062. 33. Leitges, M., L. Sanz, P. Martin, A. Duran, U. Braun, J. F. Garcia, F. Camacho, References M. T. Diaz-Meco, P. D. Rennert, and J. Moscat. 2001. Targeted disruption of the 1. McCormack, F. X., and J. A. Whitsett. 2002. The pulmonary collectins, SP-A and ␨PKC gene results in the impairment of the NF-␬B pathway. Mol. Cell 8: SP-D, orchestrate innate immunity in the lung. J. Clin. Invest. 109: 707–712. 771–780. 2. Wright, J. R. 2005. Immunoregulatory functions of surfactant proteins. Nat. Rev. 34. Kovac, J., H. Oster, and M. Leitges. 2007. Expression of the atypical protein Immunol. 5: 58–68. kinase C (aPKC) isoforms ␫/␭ and ␨ during mouse embryogenesis. Gene Expr. 3. Shepherd, V. L. 2002. Distinct roles for lung collectins in pulmonary host de- Patterns 7: 187–196. fense. Am. J. Respir. Cell Mol. Biol. 26: 257–260. 35. Wright, J. R., R. E. Wager, S. Hawgood, L. Dobbs, and J. A. Clements. 1987. ϭ 4. Levine, A. M., and J. A. Whitsett. 2001. Pulmonary collectins and innate host Surfactant apoprotein Mr 26,000–36,000 enhances uptake of liposomes by defense of the lung. Microbes Infect. 3: 161–166. type II cells. J. Biol. Chem. 262: 2888–2894. 5. Borron, P., J. C. McIntosh, T. R. Korfhagen, J. A. Whitsett, J. Taylor, and 36. Beg, A. A., W. C. Sha, R. T. Bronson, and D. Baltimore. 1995. Constitutive J. R. Wright. 2000. Surfactant-associated protein A inhibits LPS-induced cyto- NF-␬B activation, enhanced granulopoiesis, and neonatal lethality in I␬B␣-defi- kine and nitric oxide production in vivo. Am. J. Physiol. 278: L840–L847. cient mice. Genes Dev. 9: 2736–2746. 6. Monick, M. M., and G. W. Hunninghake. 2002. Activation of second messenger 37. Fan, C., Q. Li, Y. Zhang, X. Liu, M. Luo, D. Abbott, W. Zhou, and pathways in alveolar macrophages by endotoxin. Eur. Respir. J. 20: 210–222. J. F. Engelhardt. 2004. I␬B␣ and I␬B␤ possess injury context-specific functions 7. Van de Wetering, J. K., L. M. van Golde, and J. J. Batenburg. 2004. Collectins: that uniquely influence hepatic NF-␬B induction and inflammation. J. Clin. In- players of the innate immune system. Eur. J. Biochem. 271: 1229–1249. vest. 113: 746–755. 8. Piantadosi, C. A., and D. A. Schwartz. 2004. The acute respiratory distress syn- 38. Beharka, A. A., J. E. Crowther, F. X. McCormack, G. M. Denning, J. Lees, drome. Ann. Intern. Med. 141: 460–470. E. Tibesar, and L. S. Schlesinger. 2005. Pulmonary surfactant protein A activates 9. Poltorak, A., X. He, I. Smirnova, M. Y. Liu, H. C. Van, X. Du, D. Birdwell, a phosphatidylinositol 3-kinase/ signal transduction pathway in human E. Alejos, M. Silva, C. Galanos, et al. 1998. Defective LPS signaling in C3H/HeJ macrophages: participation in the up-regulation of mannose receptor activity. and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 282: 2085–2088. J. Immunol. 175: 2227–2236. The Journal of Immunology 4491

39. Guha, M., and N. Mackman. 2002. The phosphatidylinositol 3-kinase-Akt path- 51. Hernandez-Pigeon, H., A. Quillet-Mary, T. Louat, A. Schambourg, O. Humbert, way limits lipopolysaccharide activation of signaling pathways and expression of J. Selves, B. Salles, G. Laurent, and D. Lautier. 2005. hMutS␣ is protected from inflammatory mediators in human monocytic cells. J. Biol. Chem. 277: ubiquitin-proteasome-dependent degradation by atypical protein kinase C␨ phos- 32124–32132. phorylation. J. Mol. Biol. 348: 63–74. 40. Standaert, M. L., L. Galloway, P. Karnam, G. Bandyopadhyay, J. Moscat, and 52. Diaz-Meco, M. T., M. J. Lallena, A. Monjas, S. Frutos, and J. Moscat. 1999. R. V. Farese. 1997. Protein kinase C-␨ as a downstream effector of phosphati- Inactivation of the inhibitory ␬B protein kinase/nuclear factor ␬B pathway by dylinositol 3-kinase during insulin stimulation in rat adipocytes: potential role in Par-4 expression potentiates tumor necrosis factor ␣-induced apoptosis. J. Biol. glucose transport. J. Biol. Chem. 272: 30075–30082. Chem. 274: 19606–19612. 41. Le Good, J. A., W. H. Ziegler, D. B. Parekh, D. R. Alessi, P. Cohen, and 53. Lallena, M. J., M. T. Diaz-Meco, G. Bren, C. V. Paya, and J. Moscat. 1999. P. J. Parker. 1998. Protein kinase C isotypes controlled by phosphoinositide 3-ki- Activation of I␬B kinase ␤ by protein kinase C isoforms. Mol. Cell. Biol. 19: nase through the protein kinase PDK1. Science 281: 2042–2045. 2180–2188. 42. Herrera-Velit, P., K. L. Knutson, and N. E. Reiner. 1997. Phosphatidylinositol 54. Anrather, J., V. Csizmadia, M. P. Soares, and H. Winkler. 1999. Regulation of ␬ ras 3-kinase-dependent activation of protein kinase C-␨ in bacterial lipopolysaccha- NF- B RelA phosphorylation and transcriptional activity by p21 and protein ␨ ride-treated human monocytes. J. Biol. Chem. 272: 16445–16452. kinase C in primary endothelial cells. J. Biol. Chem. 274: 13594–13603. 43. Chou, M. M., W. Hou, J. Johnson, L. K. Graham, M. H. Lee, C. S. Chen, 55. Martin, P., R. Villares, S. Rodriguez-Mascarenhas, A. Zaballos, M. Leitges, A. C. Newton, B. S. Schaffhausen, and A. Toker. 1998. Regulation of protein J. Kovac, I. Sizing, P. Rennert, G. Marquez, A. Martinez, et al. 2005. Control of ␨ kinase C␨ by PI 3-kinase and PDK-1. Curr. Biol. 8: 1069–1077. T helper 2 cell function and allergic airway inflammation by PKC . Proc. Natl. Acad. Sci. USA 102: 9866–9871. 44. Parekh, D. B., W. Ziegler, and P. J. Parker. 2000. Multiple pathways control 56. Nakanishi, H., K. A. Brewer, and J. H. Exton. 1993. Activation of the ␨ isozyme protein kinase C phosphorylation. EMBO J. 19: 496–503. of protein kinase C by phosphatidylinositol 3,4,5-trisphosphate. J. Biol. Chem. 45. Duran, A., M. T. Diaz-Meco, and J. Moscat. 2003. Essential role of RelA Ser311 268: 13–16. phosphorylation by ␨PKC in NF-␬B transcriptional activation. EMBO J. 22: 57. Limatola, C., D. Schaap, W. H. Moolenaar, and W. J. van Blitterswijk. 1994. 3910–3918. Phosphatidic acid activation of protein kinase C-␨ overexpressed in COS cells: 46. Soloff, R. S., C. Katayama, M. Y. Lin, J. R. Feramisco, and S. M. Hedrick. 2004. comparison with other protein kinase C isotypes and other acidic lipids. Biochem. ␭ Targeted deletion of protein kinase C reveals a distribution of functions between J. 304: 1001–1008. the two atypical protein kinase C isoforms. J. Immunol. 173: 3250–3260. 58. Muller, G., M. Ayoub, P. Storz, J. Rennecke, D. Fabbro, and K. Pfizenmaier. Downloaded from 47. Sano, H., H. Chiba, D. Iwaki, H. Sohma, D. R. Voelker, and Y. Kuroki. 2000. 1995. PKC␨ is a molecular switch in signal transduction of TNF-␣, bifunctionally Surfactant proteins A and D bind CD14 by different mechanisms. J. Biol. Chem. regulated by ceramide and arachidonic acid. EMBO J. 14: 1961–1969. 275: 22442–22451. 59. Procyk, K. J., M. R. Rippo, R. Testi, F. Hofmann, P. J. Parker, and M. Baccarini. 48. Stamme, C., M. Muller, L. Hamann, T. Gutsmann, and U. Seydel. 2002. Surfac- 2000. Lipopolysaccharide induces jun N-terminal kinase activation in macro- tant protein A inhibits lipopolysaccharide-induced immune cell activation by pre- phages by a novel Cdc42/Rac-independent pathway involving sequential activa- venting the interaction of lipopolysaccharide with lipopolysaccharide-binding tion of protein kinase C␨ and phosphatidylcholine-dependent phospholipase C. protein. Am. J. Respir. Cell Mol. Biol. 27: 353–360. Blood 96: 2592–2598.

49. Janosch, P., M. Schellerer, T. Seitz, P. Reim, M. Eulitz, M. Brielmeier, W. Kolch, 60. Jaken, S., and P. J. Parker. 2000. Protein kinase C binding partners. BioEssays 22: http://www.jimmunol.org/ J. M. Sedivy, and H. Mischak. 1996. Characterization of I␬B kinases: I␬B-␣ is 245–254. not phosphorylated by Raf-1 or protein kinase C isozymes, but is a casein kinase 61. Castrillo, A., P. G. Traves, P. Martin-Sanz, S. Parkinson, P. J. Parker, and II substrate. J. Biol. Chem. 271: 13868–13874. L. Bosca. 2003. Potentiation of protein kinase C␨ activity by 15-deoxy-␦(12,14)- 50. Scott, M. T., A. Ingram, and K. L. Ball. 2002. PDK1-dependent activation of prostaglandin J2 induces an imbalance between mitogen-activated protein kinases atypical PKC leads to degradation of the p21 tumor modifier protein. EMBO J. and NF-␬B that promotes apoptosis in macrophages. Mol. Cell. Biol. 23: 21: 6771–6780. 1196–1208. by guest on September 29, 2021