Glucocorticoids limit acute lung inflammation in concert with inflammatory stimuli by induction of SphK1

Sabine Vettorazzi, Constantin Bode, Lien Dejager, Lucien Frappart, Ekaterina Shelest, Carina Klaßen, Alpaslan Tasdogan, Holger M Reichardt, Claude Libert, Marion Schneider, Falk Weih, N Henriette Uhlenhaut, Jean-Pierre David, Markus Gräler, Anna Kleiman, Jan P Tuckermann, Sabine Vettorazzi, Constantin Bode, Lien Dejager, Lucien Frappart, Ekaterina Shelest, Carina Klaßen, Alpaslan Tasdogan, Holger M Reichardt, Claude Libert, Marion Schneider, Falk Weih, N Henriette Uhlenhaut, Jean-Pierre David, Markus Gräler, Anna Kleiman, Jan P Tuckermann

Abstract

Acute lung injury (ALI) is a severe inflammatory disease for which no specific treatment exists. As glucocorticoids have potent immunosuppressive effects, their application in ALI is currently being tested in clinical trials. However, the benefits of this type of regimen remain unclear. Here we identify a mechanism of glucocorticoid action that challenges the long-standing dogma of cytokine repression by the glucocorticoid receptor. Contrarily, synergistic gene induction of sphingosine kinase 1 (SphK1) by glucocorticoids and pro-inflammatory stimuli via the glucocorticoid receptor in macrophages increases circulating sphingosine 1-phosphate levels, which proves essential for the inhibition of inflammation. Chemical or genetic inhibition of SphK1 abrogates the therapeutic effects of glucocorticoids. Inflammatory p38 MAPK- and mitogen- and stress-activated protein kinase 1 (MSK1)-dependent pathways cooperate with glucocorticoids to upregulate SphK1 expression. Our findings support a critical role for SphK1 induction in the suppression of lung inflammation by glucocorticoids, and therefore provide rationales for effective anti-inflammatory therapies.

Figures

Figure 1. GCs inhibit lung inflammation by…
Figure 1. GCs inhibit lung inflammation by the GR in myeloid cells requiring an intact GR dimerization interface.
(a) GRflox and GRLysMCre mice were injected with vehicle (Co), LPS and OA (hereafter, the LPS and OA co-treatment will be designated as ‘LPS') or LPS and Dex (LPS+Dex). After 18 h, EB was injected i.v., and the mice were killed 30 min later. EB accumulation in the lung was determined after perfusion with PBS+5 mM EDTA. (b) Representative haematoxylin/eosin (H&E) staining of lung sections (scale bars, 0.1 mm), and (c) histological scores of GRflox and GRLysMCre mice treated as described in a. (d) GRflox and GRLysMCre mice were treated as described in a. Cell numbers were determined in the BAL 18 h after the indicated treatments. (e) GRdim and wild-type (WT) littermate mice were injected with vehicle (Co), LPS or LPS+Dex. After 24 h, EB was injected i.v. and 30 min later the mice were killed. EB accumulation in the lung was quantified as described in a. (f) Representative H&E staining of lung sections (scale bars, 0.1 mm) and (g) histological scores of lungs of littermate WT and GRdim mice from the experiment described in e. (h) GRdim and WT mice were treated as described in e. Cell numbers were determined in the BAL 24 h after the indicated treatments. Results are presented as mean±s.e.m. Number of biological replicates: (a) (6–10), (c) (18–20), (d) (7–13), (e) (4–8), (g) (12–16) and (h) (7–11). In a, data are from three independent biological experiments; in c, data are from four independent biological experiments; in d, data are from two independent biological experiments; in e, data are from two independent biological experiments; in g, data are from three independent biological experiments out of four; and in h, data are from two independent biological experiments. Statistical analysis was performed by one-way analysis of variance. *P<0.05, **P<0.01, ***P<0.001; ND, not detectable; NS, not significant.
Figure 2. S1P signalling increased by Dex…
Figure 2. S1P signalling increased by Dex or S1PR1 agonist reduces ALI in GRLysMCre mice.
(a) GRdim and wild-type (WT) littermate control mice were treated as described in Fig. 1e. The S1P level in plasma (pmol ml−1) was determined by LC/MS/MS after 24 h. (b) GRflox and GRLysMCre mice were treated as described in Fig. 1a. The S1P level in plasma (pmol ml−1) was determined by LC/MS/MS after 18 h. (c) GRflox and GRLysMCre mice were treated as described in Fig. 1a, and an additional group of LPS-treated mice received SEW2781 (LPS+SEW). EB was injected i.v. 30 min before killing. Representative lungs are shown to demonstrate EB accumulation. (d) Quantification of EB accumulation of the experiment described in c determined as described in Fig. 1a. Results are presented as mean±s.e.m. Number of biological replicates: (a) (9), (b) (15–20) and (d) (9 or 10). Data in a are from two independent biological experiments, and data from b and d are from three independent biological experiments. Statistical analysis was performed by one-way analysis of variance. *P<0.05, **P<0.01 and ***P<0.001; NS, not significant.
Figure 3. SphK1 expression in myeloid cells…
Figure 3. SphK1 expression in myeloid cells is essential for the anti-inflammatory effects of GCs in ALI.
(a) SphK1flox and SphK1LysMCre mice were treated as described in Fig. 1a. The S1P level in plasma (pmol ml−1) was determined 24 h later by LC/MS/MS. (b) SphK1flox and SphK1LysMCre mice were treated as described in Fig. 1a, and lung specimens with accumulated EB are shown. (c) EB accumulation of isolated lungs was quantified as described in Fig. 1a. (d) Representative haematoxylin/eosin staining of lung sections from SphK1flox and SphK1LysMCre mice treated as described in Fig. 1a (scale bars, 0.1 mm). (e) Cell numbers were determined in the BAL 24 h after the indicated treatments. (f) IL-6 plasma levels (ng ml−1) in SphK1flox and SphK1LysMCre mice were determined by ELISA 5 h after LPS or LPS+Dex treatment. Results are presented as mean±s.e.m. Number of biological replicates: (a) (8–15), (c) (5–12), (e) (5–12) and (f) (5–12). In a, c and e, the data are from two independent biological experiments; and in f, the data are from one biological experiment. Statistical analysis was performed by one-way analysis of variance. *P<0.05, **P<0.01 and ***P<0.001; ND, not detectable; NS, not significant.
Figure 4. Dex and inflammatory stimuli synergistically…
Figure 4. Dex and inflammatory stimuli synergistically elevate SphK1 mRNA and activity dependent on the GR in macrophages.
(a) BMDMs derived from wild-type (WT) and GRdim mice were treated with vehicle (Co), LPS, Dex or LPS+Dex for 4 h. SphK1 mRNA expression was analysed by quantitative RT–PCR. (b) BMDMs derived from WT and GRdim mice were treated as described in a for 4 h. SphK1 activity was determined by the conversion of C17- and C18-sphingosine to S1P by LC/MS/MS. (ce) BMDMs derived from WT mice were treated for 2 h with vehicle (Co), (c) Pam3CSK4 (TLR1/2 agonist), (d) polyIC (TLR3 agonist), (e) TNFα, Dex, or co-treatment with (c) Pam3CSK4 and Dex (Pam+Dex), (d) polyIC and Dex (polyIC+Dex), (e) TNFα and Dex (TNF+Dex). SphK1 mRNA expression was analysed by quantitative RT–PCR. Results are presented as mean±s.e.m. Number of biological replicates: (a) (5 or 6), (b) (4–8), (c) (5–7), (d) (3–5) and (e) (6 or 7). Statistical analysis was performed by one-way analysis of variance. *P<0.05, **P<0.01 and ***P<0.001.
Figure 5. Dex and LPS treatment synergistically…
Figure 5. Dex and LPS treatment synergistically elevate SphK1 mRNA dependent on the p38 MAPK–MSK1 pathway.
(a) ChIP of wild-type (WT) and GRdim BMDMs was conducted 4 h after the indicated treatment (Dex, LPS+Dex). Shown are the quantitative RT–PCR results with SphK1 primer. (b) Quantitative analysis of SphK1 mRNA expression in BMDMs treated for 2 h with vehicle (Co), LPS, Dex or LPS+Dex in the presence or absence of 10 μM SB203580 (p38 MAPK inhibitor). (c) BMDMs from WT and p38αMx1Cre mice were treated for 2 h with vehicle (Co), LPS, Dex or LPS+Dex, then SphK1 mRNA expression was analysed. (d) Quantitative analysis of SphK1 mRNA expression in BMDMs treated for 2 h with vehicle (Co), LPS, Dex or LPS+Dex in the presence or absence of 10 μM SB747651A (MSK1 inhibitor). (e) The direct (physical) and indirect (functional) association among the downregulated genes after MSK1 inhibition is achieved using the STRING 10.1 database, and finally the network is visualized using Cytoscape 3.1.0. Results are presented as mean±s.e.m. Number of technical replicates: (a) (3); number of biological replicates: (b) (3), (c) (3 or 4) and (d) (6–8). Statistical analysis was performed by one-way analysis of variance. **P<0.01 and ***P<0.001; NS, not significant.
Figure 6. GCs and LPS regulate SphK1…
Figure 6. GCs and LPS regulate SphK1 and S1P dependent on GR dimerization and the p38–MSK1 pathway in macrophages to limit ALI.
During inflammation, SphK1 is moderately expressed. This results in a low activation of S1PR1, an increase in lung endothelial barrier permeability and exacerbation of ALI (left). During GC treatment of ALI, SphK1 expression in macrophages is synergistically induced by the GR- and LPS-triggered signalling via activation of TLR4 p38 MAPK–MSK1. This leads to an elevation of S1P, which acts on S1PR1 to decrease lung endothelial barrier permeability. This enhanced S1P level is a prerequisite to limit ALI (right).

References

    1. Ware L. B. & Matthay M. A. The acute respiratory distress syndrome. N. Engl. J. Med. 342, 1334–1349 (2000).
    1. Bernard G. R. et al.. Report of the American-European Consensus conference on acute respiratory distress syndrome: definitions, mechanisms, relevant outcomes, and clinical trial coordination. Consensus Committee. J. Crit. Care. 9, 72–81 (1994).
    1. Herridge M. S. et al.. One-year outcomes in survivors of the acute respiratory distress syndrome. N. Engl. J. Med. 348, 683–693 (2003).
    1. Khilnani G. C. & Hadda V. Corticosteroids and ARDS: A review of treatment and prevention evidence. Lung India 28, 114–119 (2011).
    1. Tang B. M., Craig J. C., Eslick G. D., Seppelt I. & McLean A. S. Use of corticosteroids in acute lung injury and acute respiratory distress syndrome: a systematic review and meta-analysis. Crit. Care Med. 37, 1594–1603 (2009).
    1. Marik P. E., Meduri G. U., Rocco P. R. & Annane D. Glucocorticoid treatment in acute lung injury and acute respiratory distress syndrome. Crit. Care Clin. 27, 589–607 (2011).
    1. Meduri G. U. et al.. Methylprednisolone infusion in early severe ARDS: results of a randomized controlled trial. Chest 131, 954–963 (2007).
    1. Steinberg K. P. et al.. Efficacy and safety of corticosteroids for persistent acute respiratory distress syndrome. N. Engl. J. Med. 354, 1671–1684 (2006).
    1. Hubner S. & Tuckermann J. Molecular mechanisms of the glucocorticoid receptor in steroid therapy - lessons from transgenic mice. Biomol. Concepts 3, 241–253 (2012).
    1. De Bosscher K., Haegeman G. & Elewaut D. Targeting inflammation using selective glucocorticoid receptor modulators. Curr. Opin. Pharmacol. 10, 497–504 (2010).
    1. Baschant U. & Tuckermann J. The role of the glucocorticoid receptor in inflammation and immunity. J. Steroid Biochem. Mol. Biol. 120, 69–75 (2010).
    1. Frijters R. et al.. Prednisolone-induced differential gene expression in mouse liver carrying wild type or a dimerization-defective glucocorticoid receptor. BMC Genomics 11, 359 (2010).
    1. Tuckermann J. P. et al.. Macrophages and neutrophils are the targets for immune suppression by glucocorticoids in contact allergy. J. Clin. Invest. 117, 1381–1390 (2007).
    1. Baschant U. et al.. Glucocorticoid therapy of antigen-induced arthritis depends on the dimerized glucocorticoid receptor in T cells. Proc. Natl Acad. Sci. USA 108, 19317–19322 (2011).
    1. Vandevyver S. et al.. Glucocorticoid receptor dimerization induces MKP1 to protect against TNF-induced inflammation. J. Clin. Invest. 122, 2130–2140 (2012).
    1. Kleiman A. et al.. Glucocorticoid receptor dimerization is required for survival in septic shock via suppression of interleukin-1 in macrophages. FASEB J. 26, 722–729 (2012).
    1. Bhattacharyya S., Brown D. E., Brewer J. A., Vogt S. K. & Muglia L. J. Macrophage glucocorticoid receptors regulate Toll-like receptor 4-mediated inflammatory responses by selective inhibition of p38 MAP kinase. Blood 109, 4313–4319 (2007).
    1. Patel H. B. et al.. The impact of endogenous annexin A1 on glucocorticoid control of inflammatory arthritis. Ann. Rheum. Dis. 71, 1872–1880 (2012).
    1. Mehta D. & Malik A. B. Signaling mechanisms regulating endothelial permeability. Physiol. Rev. 86, 279–367 (2006).
    1. Kohama T. et al.. Molecular cloning and functional characterization of murine sphingosine kinase. J. Biol. Chem. 273, 23722–23728 (1998).
    1. Garcia J. G. et al.. Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement. J. Clin. Invest. 108, 689–701 (2001).
    1. Camerer E. et al.. Sphingosine-1-phosphate in the plasma compartment regulates basal and inflammation-induced vascular leak in mice. J. Clin. Invest. 119, 1871–1879 (2009).
    1. McVerry B. J. et al.. Sphingosine 1-phosphate reduces vascular leak in murine and canine models of acute lung injury. Am. J. Respir. Crit. Care Med. 170, 987–993 (2004).
    1. Peng X. et al.. Protective effects of sphingosine 1-phosphate in murine endotoxin-induced inflammatory lung injury. Am. J. Respir. Crit. Care Med. 169, 1245–1251 (2004).
    1. Tauseef M. et al.. Activation of sphingosine kinase-1 reverses the increase in lung vascular permeability through sphingosine-1-phosphate receptor signaling in endothelial cells. Circ. Res. 103, 1164–1172 (2008).
    1. Bursten S. L. et al.. An increase in serum C18 unsaturated free fatty acids as a predictor of the development of acute respiratory distress syndrome. Crit. Care Med. 24, 1129–1136 (1996).
    1. Quinlan G. J., Lamb N. J., Evans T. W. & Gutteridge J. M. Plasma fatty acid changes and increased lipid peroxidation in patients with adult respiratory distress syndrome. Crit. Care Med. 24, 241–246 (1996).
    1. Matute-Bello G. et al.. An official American Thoracic Society workshop report: features and measurements of experimental acute lung injury in animals. Am. J. Respir. Cell Mol. Biol. 44, 725–738 (2011).
    1. Matute-Bello G., Frevert C. W. & Martin T. R. Animal models of acute lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 295, L379–L399 (2008).
    1. Schuster D. P. ARDS: clinical lessons from the oleic acid model of acute lung injury. Am. J. Respir. Crit. Care. Med. 149, 245–260 (1994).
    1. Annane D. Corticosteroids for severe sepsis: an evidence-based guide for physicians. Ann. Intensive Care 1, 7 (2011).
    1. Matute-Bello G. et al.. Fas (CD95) induces alveolar epithelial cell apoptosis in vivo: implications for acute pulmonary inflammation. Am. J. Pathol. 158, 153–161 (2001).
    1. Caton M. L., Smith-Raska M. R. & Reizis B. Notch-RBP-J signaling controls the homeostasis of CD8- dendritic cells in the spleen. J. Exp. Med. 204, 1653–1664 (2007).
    1. McVerry B. J. & Garcia J. G. In vitro and in vivo modulation of vascular barrier integrity by sphingosine 1-phosphate: mechanistic insights. Cell. Signal. 17, 131–139 (2005).
    1. Boyle A. J., Mac Sweeney R. & McAuley D. F. Pharmacological treatments in ARDS; a state-of-the-art update. BMC Med. 11, 166 (2013).
    1. Zhao Y. et al.. Protection of LPS-induced murine acute lung injury by sphingosine-1-phosphate lyase suppression. Am. J. Respir. Cell Mol. Biol. 45, 426–435 (2011).
    1. Ble F. X. et al.. Activation of the lung S1P(1) receptor reduces allergen-induced plasma leakage in mice. Br. J. Pharmacol. 158, 1295–1301 (2009).
    1. Bode C. et al.. Erythrocytes serve as a reservoir for cellular and extracellular sphingosine 1-phosphate. J. Cell. Biochem. 109, 1232–1243 (2010).
    1. Alvarez S. E. et al.. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature 465, 1084–1088 (2010).
    1. Nayak D. et al.. Sphingosine kinase 1 regulates the expression of proinflammatory cytokines and nitric oxide in activated microglia. Neuroscience 166, 132–144 (2010).
    1. Wadgaonkar R. et al.. Differential regulation of sphingosine kinases 1 and 2 in lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 296, L603–L613 (2009).
    1. Schaper K. et al.. Sphingosine-1-phosphate exhibits anti-proliferative and anti-inflammatory effects in mouse models of psoriasis. J. Dermatol. Sci. 71, 29–36 (2013).
    1. Calabresi P. A. et al.. Safety and efficacy of fingolimod in patients with relapsing-remitting multiple sclerosis (FREEDOMS II): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Neurol. 13, 545–556 (2014).
    1. Chun J. & Hartung H. P. Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis. Clin. Neuropharmacol. 33, 91–101 (2010).
    1. Xiong Y. et al.. Sphingosine kinases are not required for inflammatory responses in macrophages. J. Biol. Chem. 288, 32563–32573 (2013).
    1. Nakagawa M. et al.. Glucocorticoid-induced granulocytosis: contribution of marrow release and demargination of intravascular granulocytes. Circulation 98, 2307–2313 (1998).
    1. Hammad S. M. et al.. Dual and distinct roles for sphingosine kinase 1 and sphingosine 1 phosphate in the response to inflammatory stimuli in RAW macrophages. Prostaglandins Other Lipid. Mediat. 85, 107–114 (2008).
    1. Wu W., Mosteller R. D. & Broek D. Sphingosine kinase protects lipopolysaccharide-activated macrophages from apoptosis. Mol. Cell. Biol. 24, 7359–7369 (2004).
    1. Forster A. et al.. Glucocorticoids protect renal mesangial cells from apoptosis by increasing cellular sphingosine-1-phosphate. Kidney Int. 77, 870–879 (2010).
    1. Nieuwenhuis B. et al.. Involvement of the ABC-transporter ABCC1 and the sphingosine 1-phosphate receptor subtype S1P(3) in the cytoprotection of human fibroblasts by the glucocorticoid dexamethasone. J. Mol. Med. 87, 645–657 (2009).
    1. Lim H. W. et al.. Genomic redistribution of GR monomers and dimers mediates transcriptional response to exogenous glucocorticoid in vivo. Genome Res. 25, 836–844 (2015).
    1. Shah S., King E. M., Chandrasekhar A. & Newton R. Roles for the mitogen-activated protein kinase (MAPK) phosphatase, DUSP1, in feedback control of inflammatory gene expression and repression by dexamethasone. J. Biol. Chem. 289, 13667–13679 (2014).
    1. Roux P. P. & Blenis J. ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol. Mol. Biol. Rev. 68, 320–344 (2004).
    1. Guma M. et al.. Antiinflammatory functions of p38 in mouse models of rheumatoid arthritis: advantages of targeting upstream kinases MKK-3 or MKK-6. Arthritis Rheum. 64, 2887–2895 (2012).
    1. Arcaroli J. et al.. Role of p38 MAP kinase in the development of acute lung injury. Clin. Immunol. 101, 211–219 (2001).
    1. Vermeulen L., Vanden Berghe W., Beck I. M., De Bosscher K. & Haegeman G. The versatile role of MSKs in transcriptional regulation. Trends Biochem. Sci. 34, 311–318 (2009).
    1. Sobue S. et al.. Implications of sphingosine kinase 1 expression level for the cellular sphingolipid rheostat: relevance as a marker for daunorubicin sensitivity of leukemia cells. Int. J. Hematol. 87, 266–275 (2008).
    1. Beck I. M. et al.. Mitogen- and stress-activated protein kinase 1 MSK1 regulates glucocorticoid response element promoter activity in a glucocorticoid concentration-dependent manner. Eur. J. Pharmacol. 715, 1–9 (2013).
    1. Beck I. M. et al.. Glucocorticoids and mitogen- and stress-activated protein kinase 1 inhibitors: possible partners in the combat against inflammation. Biochem. Pharmacol. 77, 1194–1205 (2009).
    1. Garcia J. G. Focusing on the flood: targeting functional polymorphisms in ALI permeability pathways. Am. J. Respir. Crit. Care Med. 183, 1287–1289 (2011).
    1. Sun X. et al.. Functional promoter variants in sphingosine 1-phosphate receptor 3 associate with susceptibility to sepsis-associated acute respiratory distress syndrome. Am. J. Physiol. Lung Cell. Mol. Physiol. 305, L467–L477 (2013).
    1. Reichardt H. M. et al.. DNA binding of the glucocorticoid receptor is not essential for survival. Cell 93, 531–541 (1998).
    1. Böhm C. et al.. The alpha-isoform of p38 MAPK specifically regulates arthritic bone loss. J.I Immunol. 183, 5938–5947 (2009).
    1. Engel F. B. et al.. p38 MAP kinase inhibition enables proliferation of adult mammalian cardiomyocytes. Genes Dev. 19, 1175–1187 (2005).
    1. Zhou Z., Kozlowski J. & Schuster D. P. Physiologic, biochemical, and imaging characterization of acute lung injury in mice. Am. J. Respir. Crit. Care Med. 172, 344–351 (2005).
    1. Vlasenko L. P. & Melendez A. J. A critical role for sphingosine kinase in anaphylatoxin-induced neutropenia, peritonitis, and cytokine production in vivo. J. Immunol. 174, 6456–6461 (2005).
    1. Sammani S. et al.. Differential effects of sphingosine 1-phosphate receptors on airway and vascular barrier function in the murine lung. Am. J. Respir. Cell Mol. Biol. 43, 394–402 (2010).
    1. Bode C. & Graler M. H. Quantification of sphingosine-1-phosphate and related sphingolipids by liquid chromatography coupled to tandem mass spectrometry. Methods Mol. Biol. 874, 33–44 (2012).
    1. Allende M. L. et al.. Mice deficient in sphingosine kinase 1 are rendered lymphopenic by FTY720. J. Biol. Chem. 279, 52487–52492 (2004).
    1. Uhlenhaut N. H. et al.. Insights into negative regulation by the glucocorticoid receptor from genome-wide profiling of inflammatory cistromes. Mol. Cell 49, 158–171 (2013).

Source: PubMed

3
Sottoscrivi