An inhaled dose of budesonide induces genes involved in transcription and signaling in the human airways: enhancement of anti- and proinflammatory effector genes

Richard Leigh, Mahmoud M Mostafa, Elizabeth M King, Christopher F Rider, Suharsh Shah, Curtis Dumonceaux, Suzanne L Traves, Andrew McWhae, Tyler Kolisnik, Cora Kooi, Donna M Slater, Margaret M Kelly, Mark Bieda, Anna Miller-Larsson, Robert Newton, Richard Leigh, Mahmoud M Mostafa, Elizabeth M King, Christopher F Rider, Suharsh Shah, Curtis Dumonceaux, Suzanne L Traves, Andrew McWhae, Tyler Kolisnik, Cora Kooi, Donna M Slater, Margaret M Kelly, Mark Bieda, Anna Miller-Larsson, Robert Newton

Abstract

Although inhaled glucocorticoids, or corticosteroids (ICS), are generally effective in asthma, understanding their anti-inflammatory actions in vivo remains incomplete. To characterize glucocorticoid-induced modulation of gene expression in the human airways, we performed a randomized placebo-controlled crossover study in healthy male volunteers. Six hours after placebo or budesonide inhalation, whole blood, bronchial brushings, and endobronchial biopsies were collected. Microarray analysis of biopsy RNA, using stringent (≥2-fold, 5% false discovery rate) or less stringent (≥1.25-fold, P ≤0.05) criteria, identified 46 and 588 budesonide-induced genes, respectively. Approximately two third of these genes are transcriptional regulators (KLF9, PER1, TSC22D3, ZBTB16), receptors (CD163, CNR1, CXCR4, LIFR, TLR2), or signaling genes (DUSP1, NFKBIA, RGS1, RGS2, ZFP36). Listed genes were qPCR verified. Expression of anti-inflammatory and other potentially beneficial genes is therefore confirmed and consistent with gene ontology (GO) terms for negative regulation of transcription and gene expression. However, GO terms for transcription, signaling, metabolism, proliferation, inflammatory responses, and cell movement were also associated with the budesonide-induced genes. The most enriched functional cluster indicates positive regulation of proliferation, locomotion, movement, and migration. Moreover, comparison with the budesonide-induced expression profile in primary human airway epithelial cells shows considerable cell type specificity. In conclusion, increased expression of multiple genes, including the transcriptional repressor, ZBTB16, that reduce inflammatory signaling and gene expression, occurs in the airways and blood and may contribute to the therapeutic efficacy of ICS. This provides a previously lacking insight into the in vivo effects of ICS and should promote strategies to improve glucocorticoid efficacy in inflammatory diseases.

Keywords: Anti‐inflammatory; asthma; corticosteroid; gene expression; transactivation.

Figures

Figure 1
Figure 1
Gene expression in bronchial biopsies following budesonide inhalation. RNA was prepared from biopsy samples taken 5–6 h after placebo (Plac) or budesonide (1600 μg) (Bud) inhalation. (A) RNA was subjected to expression profiling using Affymetrix PrimeView microarrays. Analysis (n = 11 individuals) was performed with Partek Genomics Suite (v6.6). Resultant fold expression and P values were used to generate a volcano plot. Probe sets with coloring (red = 109 increased probe sets, blue = 35 repressed probe sets) indicate fold ≥2 or ≤0.5 with  0.05. (B) i, Budesonide‐induced probe sets meeting the expression criteria; ≥2‐fold, FDR 0.05, or ≥1.25‐fold,  0.05, were analyzed by the DAVID gene ID conversion tool to give: 46 (Table 2) and 588 upregulated genes (Table S5), respectively. ii, Budesonide‐repressed probe sets (≤0.5‐fold,  0.05) were analyzed by the DAVID gene ID conversion tool to give 28 repressed genes (Table 2). Using gene ontology (GO) and NCBI Gene information, genes were assigned to one of six groups: 1, Transcriptional control; 2, Membrane receptors (Receptors); 3, Signaling and regulation of signaling (Signaling); 4, Metabolic and metabolism (Metabolic); 5, Other function; or 6, Not assigned, where insufficient data were available to assign a particular function or activity and were plotted as pie charts. (C) Real‐time PCR was performed for the indicated genes. Data from the 12 individuals are expressed as the gene of interest/GAPDH and are plotted (arbitrary units) as means ±SE. Significance was assessed by Wilcoxon signed rank test. **  0.01, ***  0.001.
Figure 2
Figure 2
Gene Ontology (GO) analysis of biopsy microarray data. (A) GO analysis of biological process: the 588 (≥1.25‐fold,  0.05) budesonide‐induced gene list was analyzed by DAVID functional annotation to produce gene clusters (≥2 genes/cluster) corresponding to 647 GO annotation terms. GO terms corresponding to biological process (GOTERM_BP_FAT and KEGG_PATHWAY) were extracted. Those significantly associated ( 0.05) with the gene list are plotted with the numbers of genes (as a percentage of the list total) for each term along with the fold enrichment for each term. GO terms with less than 2% of the total genes are not plotted unless significantly enriched (Benjamini ≤0.05). Many terms were excluded as being redundant or having wide meaning (Table S7). (B) Functional cluster analysis, using DAVID, of the 588 (≥1.25‐fold,  0.05) gene list produced 183 gene clusters. The top 20 clusters are listed (Table S8). The most highly enriched cluster, containing GO terms for cell proliferation and movement, is depicted. GO terms are represented by filled circles where size is proportional to the number of genes. Individual genes are color coded to indicate expression induced by budesonide. GO terms are linked to the respective genes by lines. qPCR was performed for four genes in the biopsy samples as shown in Figure 1C. (C) The 28 budesonide‐repressed genes (≤0.5‐fold,  0.05) (Table 2) were functionally annotated using DAVID as in A. Abbreviations are as follows: +ve, positive; ‐ve, negative, reg, regulation; proc, process; metab, metabolism; transn, transcription. A number of GO terms are further abbreviated: +ve reg nucleic acid metabolic process = GO:0045935~positive regulation of nucleobase, nucleoside, nucleotide, and nucleic acid metabolic process; +ve reg nucleic acid metabolic process = GO:0045934~negative regulation of nucleobase, nucleoside, nucleotide, and nucleic acid metabolic process; transmembrane receptor PTK pathway GO:0007169 ~transmembrane receptor protein tyrosine kinase signaling pathway; enzyme‐linked receptor pathway, GO:0007167~enzyme‐linked receptor protein signaling pathway.
Figure 3
Figure 3
Expression of glucocorticoid‐induced genes in human cells. (A) Human bronchial epithelial (HBE) cells, airway smooth muscle (ASM) cells, lung fibroblasts, and endothelial cells were either not treated or treated with dexamethasone (1 μmol/L) (Dex) or budesonide (Bud) at 0.3 μmol/L (or 0.1 μmol/L for HBE cells). RNA was subjected to qPCR for the indicated genes and GAPDH. Where threshold cycle (CT) values were ≥34, then the gene was deemed not to be expressed. Data, n = 7–8 individuals (HBE), n = 8 individuals (ASM), n = 7 individuals (fibroblasts), n = 6 individuals (endothelial), expressed as the gene of interest/GAPDH, are plotted as log2‐fold relative to not treated as means ±SE. Significance was assessed by paired analysis of variance (ANOVA) (Dunn's Multiple Comparison Test). * < 0.05, ** < 0.01, *** < 0.001. (B) Primary HBE cells from six individuals were not treated or treated with budesonide (0.1 μmol/L). RNA was prepared and microarray analysis performed using Affymetrix Primeview arrays. Analysis was performed with Partek Genomics Suite (v6.6) and resultant fold expression and P values were used to generate a volcano plot (Fig. S6). Budesonide‐induced and ‐repressed probe sets meeting the criteria; i, ≥2‐fold,  0.05, or ii, ≤0.5‐fold,  0.05, were analyzed by the DAVID gene ID conversion tool to give: i, 84 upregulated and ii, 114 downregulated genes, respectively (Table S10). Using gene ontology (GO) and NCBI gene information, genes were assigned to one of six groups: 1, Transcriptional control; 2, Membrane receptors (Receptors); 3, Signaling and regulation of signaling (Signaling); 4, Metabolic and metabolism (Metabolic); 5, Other function; or 6, Not assigned, where insufficient data were available to assign a particular function or activity, and were plotted as pie charts.
Figure 4
Figure 4
Comparison of budesonide‐modulated gene expression in bronchial biopsies with human bronchial epithelial cells in culture. (A) Fold change (budesonide/not treated) for all probe sets in human bronchial epithelial cells (HBE) treated with budesonide (Bud) (0.1 μmol/L) (data from Fig. 3B) was plotted against the fold change (budesonide/placebo) obtained with the bronchial biopsies (data from Fig. 1A). (B) Heat maps representing the data in panel A are shown for: i) genes (≥2‐fold or ≤0.5‐fold, P ≤ 0.05) ranked by fold (budesonide/placebo) in the biopsies; and for ii) genes (≥2‐fold or ≤0.5‐fold, P ≤ 0.05) by fold (budesonide/not treated) in the human bronchial epithelial cells. Genes with multiple probe sets are represented by the probe set with the greatest absolute value (modulus). Genes with probe sets showing ≥2‐fold and ≤0.5‐fold for each ranked set are shown. Repressed genes in panel ii) are listed in Table S10. (C) qPCR was performed for TLR2 in the biopsy samples as described in Figure 1C.
Figure 5
Figure 5
Elevated serum budesonide and budesonide‐induced gene expression in blood. (A) Blood was collected 5–6 h post placebo (Plac) or budesonide (Bud) (1600 μg) inhalation, immediately prior to bronchoscopy. Serum was prepared and samples analyzed by LC‐MS/MS to determine budesonide concentration. Data (n = 12 individuals) are plotted as mean ± SE. (B) RNA was prepared from whole blood taken 5–6 h post placebo or budesonide inhalation. Data, from all 12 individuals, are expressed as the gene of interest/GAPDH and are plotted as means ± SE. All genes investigated by qPCR in Figures 1, 2, 3 were analyzed, but only those showing significant changes are shown. Significance was assessed by Wilcoxon signed rank test. *  0.05, ** P ≤ 0.01, *** P ≤ 0.001.

References

    1. Ayroldi E, Riccardi C (2009). Glucocorticoid‐induced leucine zipper (GILZ): a new important mediator of glucocorticoid action. FASEB J 23: 3649–3658.
    1. Baida G, Bhalla P, Kirsanov K, Lesovaya E, Yakubovskaya M, Yuen K, et al. (2015). REDD1 Functions at the Crossroads Between the Therapeutic and Adverse Effects of Topical Glucocorticoids. EMBO Mol Med 7: 42–58.
    1. Bateman ED, Hurd SS, Barnes PJ, Bousquet J, Drazen JM, Fitzgerald M, et al. (2008). Global strategy for asthma management and prevention: GINA executive summary. Eur Respir J 31: 143–178.
    1. Boldt HB, Conover CA (2007). Pregnancy‐associated plasma protein‐A (PAPP‐A): a local regulator of IGF bioavailability through cleavage of IGFBPs. Growth Horm IGF Res 17: 10–18.
    1. Brooks SA, Blackshear PJ (2013). Tristetraprolin (TTP): interactions With MRNA and proteins, and current thoughts on mechanisms of action. Biochim Biophys Acta 1829: 666–679.
    1. Carpenter S, Carlson T, Dellacasagrande J, Garcia A, Gibbons S, Hertzog P, et al. (2009). TRIL, a functional component of the TLR4 signaling complex, highly expressed in brain. J Immunol 183: 3989–3995.
    1. Carpenter S, Wochal P, Dunne A, O'Neill LA (2011). Toll‐like receptor 3 (TLR3) signaling requires TLR4 interactor with leucine‐rich repeats (TRIL). J Biol Chem 286: 38795–38804.
    1. Chinenov Y, Coppo M, Gupte R, Sacta MA, Rogatsky I (2014). Glucocorticoid receptor coordinates transcription factor‐dominated regulatory network in macrophages. BMC Genom 15: 656.
    1. Chivers JE, Cambridge LM, Catley MC, Mak JC, Donnelly LE, Barnes PJ, et al. (2004). Differential effects of RU486 reveal distinct mechanisms for glucocorticoid repression of prostaglandin E release. Eur J Biochem 271: 4042–4052.
    1. Chivers JE, Gong W, King EM, Seybold J, Mak JC, Donnelly LE, 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. Clark AR, Belvisi MG (2012). Maps and legends: the quest for dissociated ligands of the glucocorticoid receptor. Pharmacol Ther 134: 54–67.
    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. Cockcroft DW, Killian DN, Mellon JJ, Hargreave FE (1977). Bronchial reactivity to inhaled histamine: a method and clinical survey. Clin Allergy 7: 235–243.
    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. Donnelly LE, Barnes PJ (2001). Expression of heme oxygenase in human airway epithelial cells. Am J Respir Cell Mol Biol 24: 295–303.
    1. Galon J, Franchimont D, Hiroi N, Frey G, Boettner A, Ehrhart‐Bornstein M, et al. (2002). Gene profiling reveals unknown enhancing and suppressive actions of glucocorticoids on immune cells. FASEB J 16: 61–71.
    1. van de Garde MD, Martinez FO, Melgert BN, Hylkema MN, Jonkers RE, Hamann J (2014). Chronic exposure to glucocorticoids shapes gene expression and modulates innate and adaptive activation pathways in macrophages with distinct changes in leukocyte attraction. J Immunol 192: 1196–1208.
    1. Gibbons DL, Abeler‐Dorner L, Raine T, Hwang IY, Jandke A, Wencker M, et al. (2011). cutting edge: regulator of G Protein signaling‐1 selectively regulates gut T cell trafficking and colitic potential. J Immunol 187: 2067–2071.
    1. Giembycz MA, Newton R (2014). How phosphodiesterase 4 inhibitors work in patients with chronic obstructive pulmonary disease of the severe, bronchitic, frequent exacerbator phenotype. Clin Chest Med 35: 203–217.
    1. Giembycz MA, Newton R (2015). Potential mechanisms to explain how LABAs and PDE4 inhibitors enhance the clinical efficacy of glucocorticoids in inflammatory lung diseases. F1000Prime Rep 7:16.
    1. Gratchev A, Kzhyshkowska J, Kannookadan S, Ochsenreiter M, Popova A, Yu X, et al. (2008). Activation of a TGF‐beta‐specific multistep gene expression program in mature macrophages requires glucocorticoid‐mediated surface expression of TGF‐beta receptor II. J Immunol 180: 6553–6565.
    1. Himes BE, Jiang X, Wagner P, Hu R, Wang Q, Klanderman B, et al. (2014). RNA‐Seq transcriptome profiling identifies CRISPLD2 As a glucocorticoid responsive gene that modulates cytokine function in airway smooth muscle cells. PLoS ONE 9: e99625.
    1. Holden NS, Bell MJ, Rider CF, King EM, Gaunt DD, Leigh R, et al. (2011). Beta2‐adrenoceptor agonist‐induced RGS2 expression is a genomic mechanism of bronchoprotection that is enhanced by glucocorticoids. Proc Natl Acad Sci U S A 108: 19713–19718.
    1. Holden NS, George T, Rider CF, Chandrasekhar A, Shah S, Kaur M, et al. (2014). Induction of regulator of G‐protein signaling 2 expression by long‐acting Beta2‐adrenoceptor agonists and glucocorticoids in human airway epithelial cells. J Pharmacol Exp Ther 348: 12–24.
    1. Huang DW, Sherman BT, Lempicki RA (2009). Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57.
    1. Joshi T, Johnson M, Newton R, Giembycz MA (2015). The long‐acting beta2 ‐adrenoceptor agonist, indacaterol, enhances glucocorticoid receptor‐mediated transcription in human airway epithelial cells in a gene‐ and agonist‐dependent manner. Br J Pharmacol 172: 2634–2653.
    1. Kanehisa M, Goto S, Sato Y, Kawashima M, Furumichi M, Tanabe M (2014). Data, information, knowledge and principle: back to metabolism in KEGG. Nucleic Acids Res 42: D199–D205.
    1. Kaur M, Chivers JE, Giembycz MA, Newton R (2008a). Long‐acting beta2‐adrenoceptor agonists synergistically enhance glucocorticoid‐dependent transcription in human airway epithelial and smooth muscle cells. Mol Pharmacol 73: 203–214.
    1. Kaur M, Holden NS, Wilson SM, Sukkar MB, Chung KF, Barnes PJ, et al. (2008b). Effect of beta2‐adrenoceptor agonists and other CAMP‐elevating agents on inflammatory gene expression in human ASM cells: a role for protein kinase A. Am J Physiol Lung Cell Mol Physiol 295: L505–L514.
    1. Kelly MM, King EM, Rider CF, Gwozd C, Holden NS, Eddleston J, et al. (2012). Corticosteroid‐induced gene expression in allergen‐challenged asthmatic subjects taking inhaled budesonide. Br J Pharmacol 165: 1737–1747.
    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. King EM, Chivers JE, Rider CF, Minnich A, Giembycz MA, Newton R (2013). Glucocorticoid repression of inflammatory gene expression shows differential responsiveness by transactivation‐ and transrepression‐dependent mechanisms. PLoS ONE 8: e53936.
    1. Luo W, Brouwer C (2013). Pathview: an R/bioconductor package for pathway‐based data integration and visualization. Bioinformatics 29: 1830–1831.
    1. Luo W, Friedman MS, Shedden K, Hankenson KD, Woolf PJ (2009). GAGE: generally applicable gene set enrichment for pathway analysis. BMC Bioinformatics 10: 161.
    1. Manetsch M, Ramsay EE, King EM, Seidel P, Che W, Ge Q, et al. (2012). Corticosteroids and beta(2)‐agonists upregulate mitogen‐activated protein kinase phosphatase 1: in vitro mechanisms. Br J Pharmacol 166: 2049–2059.
    1. Masoli M, Holt S, Weatherall M, Beasley R (2004). Dose‐response relationship of inhaled budesonide in adult asthma: a meta‐analysis. Eur Respir J 23: 552–558.
    1. Miller MR, Hankinson J, Brusasco V, Burgos F, Casaburi R, Coates A, et al. (2005). Standardisation of Spirometry. Eur Respir J 26: 319–338.
    1. Miyata M, Lee JY, Susuki‐Miyata S, Wang WY, Xu H, Kai H, et al. (2015). Glucocorticoids suppress inflammation via the upregulation of negative regulator IRAK‐M. Nat Commun 6: 6062.
    1. Newton R (2000). Molecular Mechanisms of Glucocorticoid Action: what Is Important? Thorax 55: 603–613.
    1. Newton R (2014). Anti‐inflammatory glucocorticoids: changing concepts. Eur J Pharmacol 724: 231–236.
    1. Newton R, Holden NS (2007). Separating transrepression and transactivation: a distressing divorce for the glucocorticoid receptor? Mol Pharmacol 72: 799–809.
    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. Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP, et al. (2014). The IUPHAR/BPS Guide to Pharmacology: an expert‐driven knowledgebase of drug targets and their ligands. Nucleic Acids Res 42: D1098–D1106.
    1. Reddy TE, Pauli F, Sprouse RO, Neff NF, Newberry KM, Garabedian MJ, et al. (2009). Genomic determination of the glucocorticoid response reveals unexpected mechanisms of gene regulation. Genome Res 19: 2163–2171.
    1. Sadler AJ, Rossello FJ, Yu L, Deane JA, Yuan X, Wang D, et al. (2015). BTB‐ZF transcriptional regulator plzf modifies chromatin to restrain inflammatory signaling programs. Proc Natl Acad Sci U S A 112: 1535–1540.
    1. Sasse SK, Mailloux CM, Barczak AJ, Wang Q, Altonsy MO, Jain MK, et al. (2013). The glucocorticoid receptor and KLF15 regulate gene expression dynamics and integrate signals through feed‐forward circuitry. Mol Cell Biol 33: 2104–2115.
    1. Schacke H, Docke WD, Asadullah K (2002). Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther 96: 23–43.
    1. Shah S, King EM, Chandrasekhar A, Newton R (2014). 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.
    1. Shah S, Mostafa MM, McWhae A, Traves SL, Newton R (2016). Negative feed‐forward control of tumor necrosis factor (TNF) by tristetraprolin (ZFP36) is limited by the mitogen‐activated protein kinase phosphatase, dual‐specificity phosphatase 1 (DUSP1): implications for regulation by glucocorticoids. J Biol Chem 291: 110–125.
    1. Staal B, Williams BO, Beier F, Vande Woude GF, Zhang YW (2014). Cartilage‐specific deletion of mig‐6 results in osteoarthritis‐like disorder with excessive articular chondrocyte proliferation. Proc Natl Acad Sci U S A 111: 2590–2595.
    1. Stellato C (2004). Post‐transcriptional and nongenomic effects of glucocorticoids. Proc Am Thorac Soc 1: 255–263.
    1. Surjit M, Ganti KP, Mukherji A, Ye T, Hua G, Metzger D, et al. (2011). Widespread negative response elements mediate direct repression by agonist‐liganded glucocorticoid receptor. Cell 145: 224–241.
    1. Uings IJ, Farrow SN (2005). A Pharmacological Approach to Enhancing the Therapeutic Index of Corticosteroids in Airway Inflammatory Disease. Curr Opin Pharmacol 5: 221–226.
    1. Wang JC, Derynck MK, Nonaka DF, Khodabakhsh DB, Haqq C, Yamamoto KR (2004). Chromatin Immunoprecipitation (ChIP) Scanning Identifies Primary Glucocorticoid Receptor Target Genes. Proc Natl Acad Sci U S A 101: 15603–15608.
    1. Wierzbicki T, Iqbal SM, Cuvelier SL, Awong G, Tibbles LA, Patel KD (2003). IL‐4 Primes Human Endothelial Cells for Secondary Responses to Histamine. J Leukoc Biol 74: 420–427.
    1. Woodruff PG, Boushey HA, Dolganov GM, Barker CS, Yang YH, Donnelly S, et al. (2007). Genome‐wide profiling identifies epithelial cell genes associated with asthma and with treatment response to corticosteroids. Proc Natl Acad Sci U S A 104: 15858–15863.
    1. Xie Y, Jiang H, Nguyen H, Jia S, Berro A, Panettieri RA Jr., et al. (2012). Regulator of G protein signaling 2 is a key modulator of airway hyperresponsiveness. J Allergy Clin Immunol 130: 968–976.
    1. Yick CY, Zwinderman AH, Kunst PW, Grunberg K, Mauad T, Fluiter K, et al. (2013). Glucocorticoid‐induced changes in gene expression of airway smooth muscle in patients with asthma. Am J Respir Crit Care Med 187: 1076–1084.
    1. Zhang YW, Vande Woude GF (2007). Mig‐6, signal transduction, stress response and cancer. Cell Cycle 6: 507–513.
    1. Zhang N, Truong‐Tran QA, Tancowny B, Harris KE, Schleimer RP (2007). Glucocorticoids enhance or spare innate immunity: effects in airway epithelium are mediated by CCAAT/enhancer binding proteins. J Immunol 179: 578–589.
    1. Zhang H, Sweezey NB, Kaplan F (2015). LGL1 modulates proliferation, apoptosis, and migration of human fetal lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 308: L391–L402.

Source: PubMed

3
Iratkozz fel