Glucocorticoid repression of inflammatory gene expression shows differential responsiveness by transactivation- and transrepression-dependent mechanisms

Elizabeth M King, Joanna E Chivers, Christopher F Rider, Anne Minnich, Mark A Giembycz, Robert Newton, Elizabeth M King, Joanna E Chivers, Christopher F Rider, Anne Minnich, Mark A Giembycz, Robert Newton

Abstract

Binding of glucocorticoid to the glucocorticoid receptor (GR/NR3C1) may repress inflammatory gene transcription via direct, protein synthesis-independent processes (transrepression), or by activating transcription (transactivation) of multiple anti-inflammatory/repressive factors. Using human pulmonary A549 cells, we showed that 34 out of 39 IL-1β-inducible mRNAs were repressed to varying degrees by the synthetic glucocorticoid, dexamethasone. Whilst these repressive effects were GR-dependent, they did not correlate with either the magnitude of IL-1β-inducibility or the NF-κB-dependence of the inflammatory genes. This suggests that induction by IL-1β and repression by dexamethasone are independent events. Roles for transactivation were investigated using the protein synthesis inhibitor, cycloheximide. However, cycloheximide reduced the IL-1β-dependent expression of 13 mRNAs, which, along with the 5 not showing repression by dexamethasone, were not analysed further. Of the remaining 21 inflammatory mRNAs, cycloheximide significantly attenuated the dexamethasone-dependent repression of 11 mRNAs that also showed a marked time-dependence to their repression. Such effects are consistent with repression occurring via the de novo synthesis of a new product, or products, which subsequently cause repression (i.e., repression via a transactivation mechanism). Conversely, 10 mRNAs showed completely cycloheximide-independent, and time-independent, repression by dexamethasone. This is consistent with direct GR transrepression. Importantly, the inflammatory mRNAs showing attenuated repression by dexamethasone in the presence of cycloheximide also showed a significantly greater extent of repression and a higher potency to dexamethasone compared to those mRNAs showing cycloheximide-independent repression. This suggests that the repression of inflammatory mRNAs by GR transactivation-dependent mechanisms accounts for the greatest levels of repression and the most potent repression by dexamethasone. In conclusion, our data indicate roles for both transrepression and transactivation in the glucocorticoid-dependent repression of inflammatory gene expression. However, transactivation appears to account for the more potent and efficacious mechanism of repression by glucocorticoids on these IL-1β-induced genes.

Conflict of interest statement

Competing Interests: The authors have read the journal’s policy and have the following conflicts: This study was partly supported by funding from AstraZeneca and GlaxoSmithKline. AM is currently employed by Bristol-Myers Squibb and while at Aventis Pharmaceuticals (Bridgewater, NJ) was responsible for conducting the microarray profiling. There are no further patents, products in development or marketed products to declare. This does not alter the authors’ adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Figure 1. Effect of IL-1β and dexamethasone…
Figure 1. Effect of IL-1β and dexamethasone on inflammatory gene expression. A.
A549 cells were either not stimulated (NS) or treated with IL-1β (1 ng/ml), dexamethasone (dex) (1 µM) or a combination of the two for 1, 2, 6 and 18 h. Cells were then harvested for RNA and real-time PCR was carried out for the indicated genes and GAPDH. Data (n = 3), normalised to GAPDH are expressed as either log2 fold over NS at 1 h (left panel) or as percentage of IL-1β (right hand panel) and plotted as means. Genes are grouped based on expression patterns: ‘Early-phase’ genes are those which have a peak of expression at 1 or 2 h (top two graphs); ‘Late-phase’ genes have a peak of expression at 6 h or later with less than 50% of that peak expression observed at 1 or 2 h (bottom two graphs); and ‘Intermediate’ genes are those that fall into neither of the above categories (middle two graphs). B. Effect of dexamethasone from right hand panel of A is plotted at each time point for all genes repressed by dexamethasone. Significance was tested using one-way ANOVA with a Bonferroni post-test and is indicated: *, P<0.05; ***, P<0.001.
Figure 2. Effect of dexamethasone on inflammatory…
Figure 2. Effect of dexamethasone on inflammatory gene expression.
A. The effect of dexamethasone (1 µM) is shown on the induction of inflammatory genes by IL-1β (1 ng/ml) at 6 h. Data from Figures S2 and S3 were combined and the effect of dexamethasone expressed as a percentage of IL-1β for each gene. 100% indicates no effect of dexamethasone. Data (n = 9) are plotted as means ± SE. Genes are listed by descending efficacy to repression by dexamethasone. Statistical analysis was performed by non-parametric paired t-test. * P<0.05; ** P<0.01. B. Relationship between induction by IL-1β and repression by dexamethasone. The effect of dexamethasone (1 µM) expressed as a percentage of IL-1β is plotted against the fold induction of each gene. Data are derived from Figure 2A and Figure S2. Linear regression was performed using GraphPad Prizm software. C. Repression by dexamethasone is shown for the most highly sensitive gene (IL-1β), two genes showing intermediate sensitivity (EFNA1 & IFIT3 isoform 2) and the lowest sensitivity gene (CFB) (that showed significant repression by 1 µM dexamethasone). Actual EC50 values are indicated. D. Relationship between the effect of 1 µM dexamethasone (efficacy) and the sensitivity (log EC50) of the repression by dexamethasone for individual genes. The effect of dexamethasone (1 µM) expressed as percentage of IL-1β is plotted against the log EC50 for the repression of each gene. Data are derived from Figure 2A and Figure S3 respectively. Linear regression was performed using GraphPad Prizm software.
Figure 3. Effect of ORG34517 and GR-specific…
Figure 3. Effect of ORG34517 and GR-specific siRNA on repression of inflammatory gene expression by dexamethasone. A.
A549 cells stably transfected with a 2×GRE reporter were incubated with the indicated concentrations of ORG34517 for 30 min prior to stimulation with increasing concentrations of dexamethasone (Dex) as indicated for 6 h. Cells were then harvested for luciferase assay. Data (n = 4–5) are expressed as a percentage of 1 µM dexamethasone and plotted as means ± SE. B. A549 cells were incubated with either lamin- (control) or GR-specific siRNA for 24 h prior to harvesting for western blot analysis of lamin A/C, GR and GAPDH. Following densitometric analysis, data (n = 5), normalised to GAPDH and expressed as percentage of NS are plotted as means ± SE. Significance, relative to lamin siRNA treated cells, using ANOVA with a Dunnet’s post-test, is indicated: ns, not significant; ***, P<0.001. Representative blots are shown. C. A549 cells stably transfected with a 2×GRE reporter were incubated with lamin- (control) or GR-specific siRNA for 24 h prior to stimulation with increasing concentrations of dexamethasone (Dex) as indicated. After 6 h, cells were harvested for luciferase assay. Data (n = 4–5) are expressed as percentage of 1 µM dexamethasone and plotted as means ± SE. D. A549 cells were either not treated or incubated with either ORG34517 for 30 min (left panel of each graph), or lamin- (control) or GR-specific siRNA for 24 h (right panel of each graph) prior to being either not stimulated (data not shown), or treated with IL-1β (1 ng/ml) in the absence (data not shown) or presence of either 0.1 or 1 µM dexamethasone (Dex) for 6 h. Cells were then harvested for real-time PCR analysis of the indicated genes and GAPDH. Data (n = 5–6) normalised to GAPDH and expressed as percentage of IL-1β are plotted as means ± SE. Left panel of each graph: significance relative to IL-1β+Dex was tested using a paired t-test and is indicated: *, P<0.05; **, P<0.01; ***, P<0.001. Right panel of each graph: significance relative to IL-1β+Dex+Lamin siRNA was tested using ANOVA with a Dunnett’s post test (see also Table S4) and is indicated: *, P<0.05; **, P<0.01; ***, P<0.001.
Figure 4. Effect of IκBαΔN on inflammatory…
Figure 4. Effect of IκBαΔN on inflammatory gene expression.
A. A549 cells were infected with the NF-κB-dependent reporter, Ad5-NF-κB-luc, in the presence of Ad5-IκBαΔN (0.1–100 MOI) or Ad5-null. After 36 h, cells were stimulated with IL-1β (1 ng/ml) for 6 h before harvesting for luciferase assay and western blot analysis of IκBα and GAPDH. Data (n = 2) expressed as percentage of IL-1β are plotted as mean ± SE. Representative blots are shown. B. A549 cells were infected with 100 MOI of Ad5-IκBαΔN or Ad5-null for 24 h prior to stimulation with IL-1β for 6 h. Cells were harvested for western blot analysis of IκBα and GAPDH. Blots representative of 4 experiments are shown. C. Cells from B were harvested for RNA and SYBR green real-time PCR carried out for GAPDH and the indicated genes. Data (n = 4) normalised to GAPDH and expressed as percentage of IL-1β are plotted as mean ± S.E. Significance relative to IL-1β was tested using AVOVA with a Dunnett’s post-test. * P<0.05; ** P<0.01; *** P<0.001. Genes not significantly repressed by dexamethasone (from Figure 2A) are highlighted in bold font. D. A549 cells infected with the NF-κB-dependent reporter, Ad5-NF-κB-luc, for 36 h were incubated with various concentrations of dexamethasone, as indicated, for 1 h prior to stimulation with IL-1β. After 6 h cells were harvested for luciferase assay. Data (n = 5) expressed as percentage of IL-1β are plotted as mean ± SE.
Figure 5. Effect of CHX on dexamethasone-dependent…
Figure 5. Effect of CHX on dexamethasone-dependent repression of inflammatory gene expression.
A549 cells were treated with IL-1β (1 ng/ml) (not shown), or IL-1β and dexamethasone (dex) (1 µM) in the absence or presence of cycloheximide (CHX) (100 µg/ml) as indicated, for 4 h. Cells were then harvested for RNA and real-time PCR carried out for the indicated genes and GAPDH. Data (n = 4) normalised to GAPDH and plotted as percentage of IL-1β or IL-1β+CHX are expressed as mean ± S.E. Significance, relative to IL-1β+dex, using a paired t-test is indicated; *, P<0.05; **, P<0.01; ***, P<0.001. A. Full reversal (significantly different from IL-1β+dex and from IL-1β+CHX), B. partial reversal (significantly different from only IL-1β+dex) and C. no reversal (not significantly different from IL-1β+dex) of dexamethasone-dependent repression by CHX.
Figure 6. Relationship between the sensitivity and…
Figure 6. Relationship between the sensitivity and potency of repression by dexamethasone and the reversal by cycloheximide in A549 cells. A.
Data showing the effect of 1 µM dexamethasone (as % IL-1β) plotted against the EC50 for repression of each target mRNA by dexamethasone (i.e. Figure 2D), were overlaid with the effect of cycloheximide (CHX) on that repression (as shown in Figure 5). The logEC50 values for repression by dexamethasone of mRNAs showing: B, full reversal, or; C, full+partial reversal of this repression by cycloheximide were compared with the logEC50 values for mRNAs showing no reversal of dexamethasone-dependent repression by cycloheximide. Likewise, the effect of dexamethasone at 1 µM, expressed as a percentage of IL-1β, was compared for mRNAs showing: D, full reversal, or; E, full+partial reversal of this repression by cycloheximide were compared with mRNAs showing no reversal of dexamethasone-dependent repression by cycloheximide. Statistical analyses were performed by unpaired t-test. * P<0.05, *** P<0.001. F. Data showing the effect of 1 µM dexamethasone at 1, 2, 6 and 18 h (from Figure 1) are plotted for the group of genes showing reversal of repression by cycloheximide (left graph) and the group of genes showing no reversal (right graph). Statistical analysis was performed using ANOVA with a Bonferroni post-test and is indicated: *, P<0.05; **, P<0.01; ***, P<0.001.

References

    1. Barnes PJ (2006) Corticosteroids: the drugs to beat. Eur J Pharmacol 533: 2–14.
    1. Newton R, Leigh R, Giembycz MA (2010) Pharmacological strategies for improving the efficacy and therapeutic ratio of glucocorticoids in inflammatory lung diseases. Pharmacol Ther 125: 286–327.
    1. Newton R (2000) Molecular mechanisms of glucocorticoid action: what is important? Thorax 55: 603–613.
    1. Rhen T, Cidlowski JA (2005) Antiinflammatory action of glucocorticoids–new mechanisms for old drugs. N Engl J Med 353: 1711–1723.
    1. De Bosscher K, Vanden Berghe W, Haegeman G (2003) The interplay between the glucocorticoid receptor and nuclear factor-kappaB or activator protein-1: molecular mechanisms for gene repression. Endocr Rev 24: 488–522.
    1. Clark AR, Belvisi MG (2012) Maps and legends: the quest for dissociated ligands of the glucocorticoid receptor. Pharmacol Ther 134: 54–67.
    1. Ito K, Barnes PJ, Adcock IM (2000) Glucocorticoid receptor recruitment of histone deacetylase 2 inhibits interleukin-1beta-induced histone H4 acetylation on lysines 8 and 12. Mol Cell Biol 20: 6891–6903.
    1. Barnes PJ (2011) Glucocorticosteroids: current and future directions. Br J Pharmacol 163: 29–43.
    1. Ray A, Prefontaine KE (1994) Physical association and functional antagonism between the p65 subunit of transcription factor NF-kappa B and the glucocorticoid receptor. Proc Natl Acad Sci U S A 91: 752–756.
    1. Nissen RM, Yamamoto KR (2000) The glucocorticoid receptor inhibits NFkappaB by interfering with serine-2 phosphorylation of the RNA polymerase II carboxy-terminal domain. Genes Dev 14: 2314–2329.
    1. Luecke HF, Yamamoto KR (2005) The glucocorticoid receptor blocks P-TEFb recruitment by NFkappaB to effect promoter-specific transcriptional repression. Genes Dev 19: 1116–1127.
    1. Auphan N, DiDonato JA, Rosette C, Helmberg A, Karin M (1995) Immunosuppression by glucocorticoids: inhibition of NF-kappa B activity through induction of I kappa B synthesis. Science 270: 286–290.
    1. Scheinman RI, Cogswell PC, Lofquist AK, Baldwin AS Jr (1995) Role of transcriptional activation of I kappa B alpha in mediation of immunosuppression by glucocorticoids. Science 270: 283–286.
    1. Brostjan C, Anrather J, Csizmadia V, Stroka D, Soares M, et al. (1996) Glucocorticoid-mediated repression of NFkappaB activity in endothelial cells does not involve induction of IkappaBalpha synthesis. J Biol Chem 271: 19612–19616.
    1. Ray KP, Farrow S, Daly M, Talabot F, Searle N (1997) Induction of the E-selectin promoter by interleukin 1 and tumour necrosis factor alpha, and inhibition by glucocorticoids. Biochem J 328: 707–715.
    1. Newton R, Hart LA, Stevens DA, Bergmann M, Donnelly LE, et al. (1998) Effect of dexamethasone on interleukin-1beta-(IL-1beta)-induced nuclear factor-kappaB (NF-kappaB) and kappaB-dependent transcription in epithelial cells. Eur J Biochem 254: 81–89.
    1. Heck S, Bender K, Kullmann M, Gottlicher M, Herrlich P, et al. (1997) I kappaB alpha-independent downregulation of NF-kappaB activity by glucocorticoid receptor. EMBO J 16: 4698–4707.
    1. Sakai DD, Helms S, Carlstedt-Duke J, Gustafsson JA, Rottman FM, et al. (1988) Hormone-mediated repression: a negative glucocorticoid response element from the bovine prolactin gene. Genes Dev 2: 1144–1154.
    1. Drouin J, Trifiro MA, Plante RK, Nemer M, Eriksson P, et al. (1989) Glucocorticoid receptor binding to a specific DNA sequence is required for hormone-dependent repression of pro-opiomelanocortin gene transcription. Mol Cell Biol 9: 5305–5314.
    1. Bilodeau S, Vallette-Kasic S, Gauthier Y, Figarella-Branger D, Brue T, et al. (2006) Role of Brg1 and HDAC2 in GR trans-repression of the pituitary POMC gene and misexpression in Cushing disease. Genes Dev 20: 2871–2886.
    1. Wang JC, Derynck MK, Nonaka DF, Khodabakhsh DB, Haqq C, et al. (2004) Chromatin immunoprecipitation (ChIP) scanning identifies primary glucocorticoid receptor target genes. Proc Natl Acad Sci U S A 101: 15603–15608.
    1. So AY, Cooper SB, Feldman BJ, Manuchehri M, Yamamoto KR (2008) Conservation analysis predicts in vivo occupancy of glucocorticoid receptor-binding sequences at glucocorticoid-induced genes. Proc Natl Acad Sci U S A 105: 5745–5749.
    1. Reddy TE, Pauli F, Sprouse RO, Neff NF, Newberry KM, et al. (2009) Genomic determination of the glucocorticoid response reveals unexpected mechanisms of gene regulation. Genome Res 19: 2163–2171.
    1. Surjit M, Ganti KP, Mukherji A, Ye T, Hua G, et al. (2011) Widespread negative response elements mediate direct repression by agonist-liganded glucocorticoid receptor. Cell 145: 224–241.
    1. Stellato C (2004) Post-transcriptional and nongenomic effects of glucocorticoids. Proc Am Thorac Soc 1: 255–263.
    1. Clark AR (2007) Anti-inflammatory functions of glucocorticoid-induced genes. Mol Cell Endocrinol 275: 79–97.
    1. Newton R, Holden NS (2007) Separating transrepression and transactivation: a distressing divorce for the glucocorticoid receptor? Mol Pharmacol 72: 799–809.
    1. Clark AR, Martins JR, Tchen CR (2008) Role of dual specificity phosphatases in biological responses to glucocorticoids. J Biol Chem 283: 25765–25769.
    1. Ayroldi E, Riccardi C (2009) Glucocorticoid-induced leucine zipper (GILZ): a new important mediator of glucocorticoid action. FASEB J 23: 3649–3658.
    1. Kassel O, Sancono A, Kratzschmar J, Kreft B, Stassen M, et al. (2001) Glucocorticoids inhibit MAP kinase via increased expression and decreased degradation of MKP-1. EMBO J 20: 7108–7116.
    1. Lasa M, Abraham SM, Boucheron C, Saklatvala J, Clark AR (2002) Dexamethasone causes sustained expression of mitogen-activated protein kinase (MAPK) phosphatase 1 and phosphatase-mediated inhibition of MAPK p38. Mol Cell Biol 22: 7802–7811.
    1. Issa R, Xie S, Khorasani N, Sukkar M, Adcock IM, et al. (2007) Corticosteroid Inhibition of Growth-Related Oncogene Protein-{alpha} via Mitogen-Activated Kinase Phosphatase-1 in Airway Smooth Muscle Cells. J Immunol 178: 7366–7375.
    1. Kelly M, King E, Rider C, Gwozd C, Holden N, et al... (2011) Corticosteroid-induced gene expression in allergen-challenged asthmatic subjects taking inhaled budesonide. Br J Pharmacol.
    1. Mittelstadt PR, Ashwell JD (2001) Inhibition of AP-1 by the glucocorticoid-inducible protein GILZ. J Biol Chem 276: 29603–29610.
    1. Ayroldi E, Migliorati G, Bruscoli S, Marchetti C, Zollo O, et al. (2001) Modulation of T-cell activation by the glucocorticoid-induced leucine zipper factor via inhibition of nuclear factor kappaB. Blood 98: 743–753.
    1. Eddleston J, Herschbach J, Wagelie-Steffen AL, Christiansen SC, Zuraw BL (2007) The anti-inflammatory effect of glucocorticoids is mediated by glucocorticoid-induced leucine zipper in epithelial cells. J Allergy Clin Immunol 119: 115–122.
    1. Ayroldi E, Zollo O, Bastianelli A, Marchetti C, Agostini M, et al. (2007) GILZ mediates the antiproliferative activity of glucocorticoids by negative regulation of Ras signaling. J Clin Invest 117: 1605–1615.
    1. Kwon OJ, Au BT, Collins PD, Baraniuk JN, Adcock IM, et al. (1994) Inhibition of interleukin-8 expression by dexamethasone in human cultured airway epithelial cells. Immunology 81: 389–394.
    1. Newton R, Eddleston J, Haddad E, Hawisa S, Mak J, et al. (2002) Regulation of kinin receptors in airway epithelial cells by inflammatory cytokines and dexamethasone. Eur J Pharmacol 441: 193–202.
    1. Catley MC, Sukkar MB, Chung KF, Jaffee B, Liao SM, et al. (2006) Validation of the anti-inflammatory properties of small-molecule IkappaB Kinase (IKK)-2 inhibitors by comparison with adenoviral-mediated delivery of dominant-negative IKK1 and IKK2 in human airways smooth muscle. Mol Pharmacol 70: 697–705.
    1. King EM, Holden NS, Gong W, Rider CF, Newton R (2009) Inhibition of NF-kappaB-dependent transcription by MKP-1: transcriptional repression by glucocorticoids occurring via p38 MAPK. J Biol Chem 284: 26803–26815.
    1. Chivers JE, Gong W, King EM, Seybold J, Mak JC, et al. (2006) Analysis of the dissociated steroid, RU24858, does not exclude a role for inducible genes in the anti-inflammatory actions of glucocorticoids. Mol Pharmacol 70: 2084–2095.
    1. Kaur M, Chivers JE, Giembycz MA, Newton R (2008) Long-acting beta2-adrenoceptor agonists synergistically enhance glucocorticoid-dependent transcription in human airway epithelial and smooth muscle cells. Mol Pharmacol 73: 203–214.
    1. Peeters BW, Ruigt GS, Craighead M, Kitchener P (2008) Differential effects of the new glucocorticoid receptor antagonist ORG 34517 and RU486 (mifepristone) on glucocorticoid receptor nuclear translocation in the AtT20 cell line. Ann N Y Acad Sci 1148: 536–541.
    1. Holden NS, Gong W, King EM, Kaur M, Giembycz MA, et al. (2007) Potentiation of NF-kappaB-dependent transcription and inflammatory mediator release by histamine in human airway epithelial cells. Br J Pharmacol 152: 891–902.
    1. Catley MC, Chivers JE, Holden NS, Barnes PJ, Newton R (2005) Validation of IKK beta as therapeutic target in airway inflammatory disease by adenoviral-mediated delivery of dominant-negative IKK beta to pulmonary epithelial cells. Br J Pharmacol 145: 114–122.
    1. Schleimer RP (2004) Glucocorticoids suppress inflammation but spare innate immune responses in airway epithelium. Proc Am Thorac Soc 1: 222–230.
    1. Cornish AL, Campbell IK, McKenzie BS, Chatfield S, Wicks IP (2009) G-CSF and GM-CSF as therapeutic targets in rheumatoid arthritis. Nat Rev Rheumatol 5: 554–559.
    1. Commins SP, Borish L, Steinke JW (2010) Immunologic messenger molecules: cytokines, interferons, and chemokines. J Allergy Clin Immunol 125: S53–S72.
    1. Vereecke L, Beyaert R, van Loo G (2011) Genetic relationships between A20/TNFAIP3, chronic inflammation and autoimmune disease. Biochem Soc Trans 39: 1086–1091.
    1. Fukai T, Ushio-Fukai M (2011) Superoxide dismutases: role in redox signaling, vascular function, and diseases. Antioxid Redox Signal 15: 1583–1606.
    1. Honda K, Taniguchi T (2006) IRFs: master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors. Nat Rev Immunol 6: 644–658.
    1. Taube C, Thurman JM, Takeda K, Joetham A, Miyahara N, et al. (2006) Factor B of the alternative complement pathway regulates development of airway hyperresponsiveness and inflammation. Proc Natl Acad Sci U S A 103: 8084–8089.
    1. Bartholomew WR, Shanahan TC (1990) Complement components and receptors: deficiencies and disease associations. Immunol Ser 52: 33–51.
    1. Wang Y, Zhang JJ, Dai W, Lei KY, Pike JW (1997) Dexamethasone potently enhances phorbol ester-induced IL-1beta gene expression and nuclear factor NF-kappaB activation. J Immunol 159: 534–537.
    1. Hofmann TG, Schmitz ML (2002) The promoter context determines mutual repression or synergism between NF-kappaB and the glucocorticoid receptor. Biol Chem 383: 1947–1951.
    1. Webster JC, Huber RM, Hanson RL, Collier PM, Haws TF, et al. (2002) Dexamethasone and tumor necrosis factor-alpha act together to induce the cellular inhibitor of apoptosis-2 gene and prevent apoptosis in a variety of cell types. Endocrinology 143: 3866–3874.
    1. Sukkar MB, Issa R, Xie S, Oltmanns U, Newton R, et al. (2004) Fractalkine/CX3CL1 production by human airway smooth muscle cells: induction by IFN-gamma and TNF-alpha and regulation by TGF-beta and corticosteroids. Am J Physiol Lung Cell Mol Physiol 287: L1230–L1240.
    1. King EM, Kaur M, Gong W, Rider CF, Holden NS, et al. (2009) Regulation of tristetraprolin expression by interleukin-1beta and dexamethasone in human pulmonary epithelial cells: roles for nuclear factor-kappaB and p38 mitogen-activated protein kinase. J Pharmacol Exp Ther 330: 575–585.
    1. Quante T, Ng YC, Ramsay EE, Henness S, Allen JC, et al. (2008) Corticosteroids reduce IL-6 in ASM cells via up-regulation of MKP-1. Am J Respir Cell Mol Biol 39: 208–217.
    1. Turpeinen T, Nieminen R, Moilanen E, Korhonen R (2010) Mitogen-activated protein kinase phosphatase-1 negatively regulates the expression of interleukin-6, interleukin-8, and cyclooxygenase-2 in A549 human lung epithelial cells. J Pharmacol Exp Ther 333: 310–318.
    1. Newton R, King EM, Gong W, Rider CF, Staples KJ, et al. (2010) Glucocorticoids inhibit IL-1beta-induced GM-CSF expression at multiple levels: roles for the ERK pathway and repression by MKP-1. Biochem J 427: 113–124.
    1. Ito K, Yamamura S, Essilfie-Quaye S, Cosio B, Ito M, et al. (2006) Histone deacetylase 2-mediated deacetylation of the glucocorticoid receptor enables NF-kappaB suppression. J Exp Med 203: 7–13.
    1. Diefenbacher M, Sekula S, Heilbock C, Maier JV, Litfin M, et al. (2008) Restriction to Fos family members of Trip6-dependent coactivation and glucocorticoid receptor-dependent trans-repression of activator protein-1. Mol Endocrinol 22: 1767–1780.

Source: PubMed

3
Subskrybuj