identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene...

8
Identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene expression Olivier Muller a , Sylvain Pradervand b , Stefan Berger c , Gabriel Centeno d , Aude Milet e , Pascal Nicod f , Thierry Pedrazzini g , François Tronche e , Günter Schütz c , Kenneth Chien b , Bernard C. Rossier d, , Dmitri Firsov d, a Cardiology Service, University Hospital, CHUV, CH-1011 Lausanne, Switzerland b Institute of Molecular Medicine, University of California at San Diego School of Medicine, La Jolla, CA 92093, USA c Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg, Germany d Department of Pharmacology and Toxicology, University of Lausanne, 27, Rue du Bugnon, CH-1005 Lausanne, Switzerland e CNRS UMR 7148, GMNC, Collège de France, Paris, France f Department of Internal Medicine, University Hospital, CHUV, CH-1011 Lausanne, Switzerland g Division of Hypertension, University Hospital, CHUV, CH-1011 Lausanne, Switzerland Received 8 June 2006; accepted 1 November 2006 Available online 14 December 2006 Abstract Corticosteroids (aldosterone, cortisol/corticosterone) exert direct functional effects on cardiomyocytes. However, gene networks activated by corticosteroids in cardiomyocytes, as well as the involvement of the mineralocorticoid receptor (MR) vs the glucocorticoid receptor (GR) in these effects, remain largely unknown. Here we characterized the corticosteroid-dependent transcriptome in primary culture of neonatal mouse cardiomyocytes treated with 10 6 M aldosterone, a concentration predicted to occupy both MR and GR. Serial analysis of gene expression revealed 101 aldosterone-regulated genes. The MR/GR specificity was characterized for one regulated transcript, namely ecto-ADP- ribosyltransferase-3 (Art3). Using cardiomyocytes from GR(null/null) or MR(null/null) mice we demonstrate that in GR(null/null) cardiomyocytes the response is abrogated, but it is fully maintained in MR(null/null) cardiomyocytes. We conclude that Art3 expression is regulated exclusively via the GR. Our study identifies a new set of corticosteroid-regulated genes in cardiomyocytes and demonstrates a new approach to studying the selectivity of MR- vs GR-dependent effects. © 2006 Elsevier Inc. All rights reserved. Keywords: Cardiomyocytes; Aldosterone; Glucocorticoids; Mineralocorticoid receptor; Glucocorticoid receptor The effects of adrenal corticosteroids (aldosterone, cortisol, or corticosterone in rodents) on the cardiovascular system have recently attracted considerable attention because of their possible involvement in the pathophysiology of heart disease. The RALES and EPHESUS clinical trials have shown that mineralocorticoid receptor (MR) antagonists added to a standard drug regimen substantially increase survival and decrease hospitalization in patients with heart failure [1,2]. Systemic glucocorticoids have also been recently recognized as a risk factor for heart failure [3]. In transgenic mouse studies, cardiomyocyte-specific downregulation of MR expression resulted in severe heart failure and cardiac fibrosis [4]. In the opposite experiment, when MR was overexpressed in cardio- myocytes, severe ventricular arrhythmia was observed as a main phenotype [5]. Together, these findings provide experimental evidence that the MR signaling pathway could be involved in heart disease. However, on the molecular level, the effects of corticosteroids on cardiomyocytes remain poorly understood. In vitro, corticosteroids have been shown to exert direct functional effects on cardiomyocytes. Whitehurst et al. showed that dexamethasone, a synthetic glucocorticoid, significantly increased the L-type Ca 2+ currents in neonatal rat cardiomyo- cytes [6]. Schulz et al. have shown that dexamethasone inhibits inducible nitric oxide synthase activity in adult rat Genomics 89 (2007) 370 377 www.elsevier.com/locate/ygeno Corresponding authors. Fax: +41 21 6925355. E-mail addresses: [email protected] (B.C. Rossier), [email protected] (D. Firsov). 0888-7543/$ - see front matter © 2006 Elsevier Inc. All rights reserved. doi:10.1016/j.ygeno.2006.11.001

Upload: olivier-muller

Post on 31-Oct-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene expression

7) 370–377www.elsevier.com/locate/ygeno

Genomics 89 (200

Identification of corticosteroid-regulated genes in cardiomyocytes by serialanalysis of gene expression

Olivier Muller a, Sylvain Pradervand b, Stefan Berger c, Gabriel Centeno d, Aude Milet e,Pascal Nicod f, Thierry Pedrazzini g, François Tronche e, Günter Schütz c, Kenneth Chien b,

Bernard C. Rossier d,⁎, Dmitri Firsov d,⁎

a Cardiology Service, University Hospital, CHUV, CH-1011 Lausanne, Switzerlandb Institute of Molecular Medicine, University of California at San Diego School of Medicine, La Jolla, CA 92093, USA

c Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg, Germanyd Department of Pharmacology and Toxicology, University of Lausanne, 27, Rue du Bugnon, CH-1005 Lausanne, Switzerland

e CNRS UMR 7148, GMNC, Collège de France, Paris, Francef Department of Internal Medicine, University Hospital, CHUV, CH-1011 Lausanne, Switzerland

g Division of Hypertension, University Hospital, CHUV, CH-1011 Lausanne, Switzerland

Received 8 June 2006; accepted 1 November 2006Available online 14 December 2006

Abstract

Corticosteroids (aldosterone, cortisol/corticosterone) exert direct functional effects on cardiomyocytes. However, gene networks activated bycorticosteroids in cardiomyocytes, as well as the involvement of the mineralocorticoid receptor (MR) vs the glucocorticoid receptor (GR) in theseeffects, remain largely unknown. Here we characterized the corticosteroid-dependent transcriptome in primary culture of neonatal mousecardiomyocytes treated with 10−6 M aldosterone, a concentration predicted to occupy both MR and GR. Serial analysis of gene expressionrevealed 101 aldosterone-regulated genes. The MR/GR specificity was characterized for one regulated transcript, namely ecto-ADP-ribosyltransferase-3 (Art3). Using cardiomyocytes from GR(null/null) or MR(null/null) mice we demonstrate that in GR(null/null) cardiomyocytesthe response is abrogated, but it is fully maintained in MR(null/null) cardiomyocytes. We conclude that Art3 expression is regulated exclusivelyvia the GR. Our study identifies a new set of corticosteroid-regulated genes in cardiomyocytes and demonstrates a new approach to studying theselectivity of MR- vs GR-dependent effects.© 2006 Elsevier Inc. All rights reserved.

Keywords: Cardiomyocytes; Aldosterone; Glucocorticoids; Mineralocorticoid receptor; Glucocorticoid receptor

The effects of adrenal corticosteroids (aldosterone, cortisol,or corticosterone in rodents) on the cardiovascular system haverecently attracted considerable attention because of theirpossible involvement in the pathophysiology of heart disease.The RALES and EPHESUS clinical trials have shown thatmineralocorticoid receptor (MR) antagonists added to astandard drug regimen substantially increase survival anddecrease hospitalization in patients with heart failure [1,2].Systemic glucocorticoids have also been recently recognized asa risk factor for heart failure [3]. In transgenic mouse studies,

⁎ Corresponding authors. Fax: +41 21 6925355.E-mail addresses: [email protected] (B.C. Rossier),

[email protected] (D. Firsov).

0888-7543/$ - see front matter © 2006 Elsevier Inc. All rights reserved.doi:10.1016/j.ygeno.2006.11.001

cardiomyocyte-specific downregulation of MR expressionresulted in severe heart failure and cardiac fibrosis [4]. In theopposite experiment, when MR was overexpressed in cardio-myocytes, severe ventricular arrhythmia was observed as a mainphenotype [5]. Together, these findings provide experimentalevidence that the MR signaling pathway could be involved inheart disease. However, on the molecular level, the effects ofcorticosteroids on cardiomyocytes remain poorly understood.

In vitro, corticosteroids have been shown to exert directfunctional effects on cardiomyocytes. Whitehurst et al. showedthat dexamethasone, a synthetic glucocorticoid, significantlyincreased the L-type Ca2+ currents in neonatal rat cardiomyo-cytes [6]. Schulz et al. have shown that dexamethasone inhibitsinducible nitric oxide synthase activity in adult rat

Page 2: Identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene expression

Table 1SAGE tags differentially expressed between Aldo and control SAGE libraries

SAGE tag Ctrl Aldo p Accession No. Gene

Induced tagsTACAAAAAAA 14 28 * NM_001013028.1 Expressed sequence AI597468CAGGAAAGCA 11 26 * NM_019657.2 Hydroxysteroid (17-β-dehydrogenase 12)CCGGGCAATG 10 25 * M_011817.1 Gadd45gCCAGCCCTGG 10 20 * NM_008963.1 Prostaglandin D2 synthase (brain) (Ptgds)TTTGGAAAAA 9 26 ** NM_027695.1 RIKEN cDNACAGGACCTGG 9 19 * NM_028677.3 RIKEN cDNACAGATGGTGA 8 20 * NM_009814.1 Calsequestrin 2 (Casq2)TATACAGTCG 7 19 * XM_485637.2 Ribosomal protein L7 (Rpl7)GGATTCAATA 6 19 ** NM_181728.1 ADP-ribosyltransferase 3 (Art3)TTCAATAAAT 6 19 ** XM_123229.4 Mortality factor 4-like 1 (Morf4l1)CAGCTCCTAA 5 18 * NM_028733.1 PKC and casein kinase substrate (Pacsin3)TCTAAAAAAA 5 17 * NM_145963.1 Potassium channel (Kcnj14)CCTAAAAAAA 5 16 * NM_011246.2 RAS guanyl-releasing protein 1 (Rasgrp1)CTGGCAGTGG 3 15 ** NM_007379.1 ATP-binding cassette Abca2TTCAATAAAT 3 11 * XM_123229.4 Mortality factor 4-like 1 (Morf4l1)AAATAAAGTT 3 10 * XM_484784.2 Sperm equatorial segment protein 1 (Spesp1)CTTTCTCAGT 3 10 * NM_007669.2 Cyclin-dependent kinase inhibitor 1A (P21)CAGTTGTCAA 2 10 * NM_008300.2 Heat shock protein 4 (Hspa4)CGGCAGAAGG 2 10 * NM_016666.1 AHR-interacting protein (Aip)TCCGTCCCTC 2 10 * NM_145385.1 Myeloid leukemia factor 2 (Mlf2)ACTGGGCCAT 2 9 * NM_025624.1 RIKEN cDNACCTTTGGGGT 2 9 * NM_010286.2 Tsc22d3CTGACAGTCT 1 16 **** XM_620813.1 Pol-like transcriptAGTGTACCGT 1 10 ** NM_026958.1 RIKEN cDNACTGACAATGA 1 10 ** NM_007759.1 Retinoic acid-binding protein II (Crabp2)GTTTCCCTTG 1 10 ** NM_011793.2 Barrier to autointegration factor 1 (Banf1)CAGGTCCGTG 1 9 * NM_011065.2 Period homolog 1 (Per1)CTGGAGTGAC 1 9 * NM_177809.2 Hypothetical proteinTGTAATTTCT 1 9 * NM_019771.1 Destrin (Dstn)AGTGTGATAG 1 8 * NM_025926.1 DnaJ (Hsp40) homolog (Dnajb4)CAAAGTTTAT 1 8 * XM_488509.2 Centrosomal protein 1 (Cep1)CCACCGCAGC 1 8 * NM_139059.1 Casein kinase 1, transcript variant 2TATCCTAGCC 1 8 * NM_029425.1 Suppressor of initiator codon mutationsCAAACTTAAC 1 7 * NM_013625.1 Platelet-activating factor acetylhydrolaseTGGTTGTACT 0 9 *** NM_009127.2 Stearoyl-coenzyme A desaturase 1 (Scd1)CCATCCCCTT 0 8 *** NM_175307.2 RIKEN cDNAGGAGTGAAAC 0 8 *** NM_175096.2 DNA segment, Chr 5TTGAACTTTC 0 8 *** XM_619223.1 Friend virus susceptibility 1 (Fv1)CTCACTGAGA 0 7 ** XM_356760.3 Similar to 60S ribosomal protein L17CACCTGCTTC 0 6 * NM_172701.1 δ-like 3 (Dll3), transcript variant 1TCTTCAGCAC 0 6 * NM_145558.1 Hydroxyacyl dehydrogenase (Hadhb)AAGGGGGAAA 0 5 * XM_619685.1 cDNA sequenceATCATGCTTC 0 5 * NM_009254.2 Serine proteinase inhibitor (Serpinb6c)CCCACCCCAT 0 5 * NM_021523.3 Huwe1CTCAAGAGAA 0 5 * NM_139297.2 UDP-glucose pyrophosphorylase 2 (Ugp2)CTGAAGAAAT 0 5 * NM_008053.1 Fragile X mental retardation gene 1 (Fxr1h)CTGCACCCGA 0 5 * NM_026366.1 RIKEN cDNACTGTGGATGA 0 5 * NM_016778.1 Bcl-2-related ovarian killer protein (Bok)CTTGGTAACT 0 5 * NM_206924.1 Jumping translocation breakpoint (Jtb)

Repressed tagsTGACGTGCCG 118 53 **** XM_619956.1 Glyceraldehyde-3-phosphate dehydrogenaseGGCTCAACAA 89 33 **** NM_026020.2 Ribosomal protein, large P2 (Rplp2)TGACCTGGCT 66 31 **** NM_025628.1 Cytochrome c oxidase, Cox6b1TGCGCTTCCA 50 18 **** XR_000348.1 H3 histone, family 3A (H3f3a)CAGCTCAGCC 48 21 *** NM_201353.1 Sema domain sequence (Sema5b)AGCTGAAGGT 42 21 * XM_357202.3 RIKEN cDNAACCGACCGCA 38 14 **** NM_008410.1 Integral membrane protein 2B (Itm2b)ACTCCTTTTG 36 17 ** NM_008617.2 Malate dehydrogenase 2 (Mdh2)CGCGAATGCT 33 14 ** NM_010885.2 NADH dehydrogenase (Ndufa2)GCCCTCATGC 31 15 * NM_023312.2 NADH dehydrogenase (Ndufa13)AGGCCTTTAG 29 8 **** NM_011179.2 Prosaposin (Psap)

(continued on next page)

371O. Muller et al. / Genomics 89 (2007) 370–377

Page 3: Identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene expression

Table 1 (continued)

SAGE tag Ctrl Aldo p Accession No. Gene

Repressed tagsCGGCGCCACA 28 8 *** NM_026305.1 Transcription elongation factor B (SIII)TGACACCCAA 26 11 * XM_128725.5 NADH dehydrogenase (Ndufv2)TTGCAAGGGG 24 9 ** NM_019770.2 RIKEN cDNATGACTCTGCC 21 6 *** NM_011652.2 Titin (Ttn)CACGACGGAC 21 8 * XM_488885.2 Hypothetical LOC433272AGCAAATATT 20 6 ** NM_026610.1 NADH dehydrogenase (Ndufb4)TGACCCCATG 20 7 ** NM_025939.2 Phosphoribosylaminoimidazole carboxylaseGCCAAGCGGA 18 7 ** NM_023202.2 NADH dehydrogenase (Ndufa7)AGGAGCTGGA 17 5 ** NM_181730.2 RNA polymerase 1-3 (Rpo1-3)AGGCAGAATA 16 4 ** NM_030229.2 Polymerase (RNA) III (DNA polypeptide H)TTGCCAGCCA 15 2 *** NM_025982.1 Sarcoma amplified sequence (Sas)AAAAGTTGAA 15 3 ** NM_016672.1 Dopa decarboxylase (Ddc)AGGCCTGAAC 15 4 * XM_485994.1 Protein phosphatase 1, catalytic subunitTGCATCGTGA 14 2 *** NM_011185.1 Proteasome subunit, β type 1 (Psmb1)ACCCTCAGGT 14 4 * NM_030225.3 Dihydrolipoamide S-succinyltransferaseTGAACACCCA 14 4 * NM_153526.2 Insulin-induced gene 1 (Insig1)CAAATCAGGA 13 3 * XM_485597.2 Mus musculus similar to DnaJ-like protein 2CACCGAAGGA 13 3 * XM_488877.1 LIM domain only 4 (Lmo4)GTGGCTCCCA 13 3 * NM_025396.1 6-Phosphogluconolactonase (Pgls)ATCAGTTAGA 12 3 * NM_144521.1 RIKEN cDNAATCAGTTAGA 12 3 * NM_011526.2 RIKEN cDNAACGGTTTTCT 11 1 ** NM_019760.1 Tumor differentially expressed 2 (Tde2)ACCAAAGGTA 11 2 * NM_021552.2 Similar to TGF-β-inducible nuclear proteinCCTCACATCC 10 1 * NM_026027.1 Prefoldin 1 (Pfdn1)CCCCCTTCCC 10 2 * NM_011670.1 Ubiquitin carboxy-terminal hydrolase L1CCCTTTCTCT 10 2 * NM_026914.1 RIKEN cDNAGCATTGCGCA 10 2 * NM_008948.1 Proteasome 26S subunit, ATPase 3 (Psmc3)GTGGGCTTCA 10 2 * NM_021607.2 Nicastrin (Ncstn)CAGTTTGCCT 9 1 * NM_029787.2 Diaphorase 1 (NADH) (Dia1)ACCCCCTTCC 8 1 * NM_019581.2 GTP binding protein 2 (Gtpbp2)TCAATTTTTT 8 1 * NM_011454.1 Serine proteinase inhibitor (Serpinb6b)TGGCCAACGG 8 1 * NM_180678.2 Glycyl-tRNA synthetase (Gars)TGACGCCGAG 7 0 * NM_008927.1 Mitogen-activated protein kinase kinase 1TGAGTTTTAT 7 0 * XM_130033.7 Rho GTPase-activating protein 21TGTCTTACCC 7 0 * NM_012010.2 Eukaryotic translation initiation factor 2AAGTGGTCAA 6 0 * NM_173751.3 IlvB (bacterial acetolactate synthase)-likeCCAGAGCCAG 6 0 * XM_203523.3 cDNA sequenceCGCTGTACTC 6 0 * NM_013917.1 Pituitary tumor-transforming 1 (Pttg1)CTCATGGATG 6 0 * NM_009722.1 Ca2+ transporting ATPase (Atp2a2)GCGCGGACGC 6 0 * NM_026938.1 RIKEN cDNATGACATAGTG 6 0 * NM_027959.1 Protein disulfide isomerase (Pdia6)

*p<0.05, **p<0.01, ***p<0.005, ****p<0.001.

372 O. Muller et al. / Genomics 89 (2007) 370–377

cardiomyocytes [7]. Aldosterone, the natural mineralocorticoid,was shown to modulate the activity of Cl−/HCO3

− and Na+/H+

transporters and of L-type Ca2+ channels in neonatal and adultrat cardiomyocytes, respectively [8,9].

Aldosterone and cortisol/corticosterone bind both the MR andthe glucocorticoid receptor (GR). Both hormones display higheraffinities for the MR than for the GR, whereas their respectiveaffinities for a given receptor are very similar [10]. In “classical”aldosterone target cells (i.e., kidney and colon), the MR isprotected from illicit occupation by glucocorticoids thanks to11β-hydroxysteroid dehydrogenase type II (11βHSD2), anenzyme that converts cortisol/corticosterone into inactive meta-bolites. Cardiomyocytes belong to the so-called “nonclassical”aldosterone target tissues that express both the MR and the GR,but not 11βHSD2. Thus, in cardiomyocytes, MR is not protectedfrom occupancy by glucocorticoids and is not aldosteroneselective. Taking into account that circulating cortisol/corticoster-

one levels are at least 100-fold higher than those of aldosterone,and that MR has the same affinity for aldosterone andglucocorticoids, cardiomyocyte MR may be permanently occu-pied by glucocorticoids. Thus, in cardiomyocytes, glucocorticoideffects could be mediated by both GR andMR. This fact makes itdifficult to attribute the observed functional effects to glucocorti-coids or aldosterone, on the one hand, and to GR or MRactivation, on the other hand.

To identify downstream target genes of different signalingpathways potentially involved in cardiac remodeling andhypertrophy, a serial analysis of gene expression (SAGE) wasperformed, using RNA from primary culture of neonatalC57BL/6 mouse cardiac myocytes stimulated for 4 h by LIF,neuregulin-1, aldosterone, and angiotensin II. We havepreviously described the results obtained from LIF stimulationwith the identification of the small proline-rich protein 1A as astress-inducible cardioprotective protein [11]. In the present

Page 4: Identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene expression

Fig. 2. Real-time quantitative RT-PCR analysis of Per1 mRNA expression. TotalRNAs were isolated from either untreated (control) or 10−6 M aldosterone-treated cardiomyocytes after the indicated periods of time. Quantification ofPer1 mRNA expression was performed using real-time quantitative RT-PCRanalysis. Per1 expression levels were normalized to those of actin. Each value isthe mean±SEM. The experiment was performed in triplicate.

373O. Muller et al. / Genomics 89 (2007) 370–377

report, we analyzed the effect of aldosterone on the samecellular model, performed during the same experimental day.Here, we identified a new set of 101 corticosteroid-regulatedgenes. One candidate gene, ecto-ADP-ribosyltransferase-3(Art3), was selected for further characterization. To discriminatebetween glucocorticoid/mineralocorticoid and GR/MR effectsin cardiomyocytes, we analyzed the changes in Art3 proteinexpression levels in response to the natural ligands (aldosteroneor corticosterone) in cardiomyocytes from wild-type, MR(null/null), or GR(null/null) animals, allowing unequivocal attribu-tion of the observed effect to the GR signaling pathway.

Results

Aldosterone-dependent transcriptome of neonatal mousecardiomyocytes

To characterize several major signaling pathways leading tocardiomyocyte remodeling, we constructed five SAGE librariesfrom neonatal mouse cardiomyocytes stimulated for 4 h withaldosterone, LIF, neuregulin-1, angiotensin II, or controlmedium (untreated) [11]. In the present study, we performedthe characterization and initial analysis of a SAGE libraryconstructed from aldosterone (10−6 M)-stimulated cells (AldoSAGE library). Importantly, at an aldosterone concentration of10−6 M, both MR and GR are occupied by the hormone. Thus,analysis of the Aldo SAGE library allowed us to characterizeboth MR- and GR-regulated genes. The assessment ofsignificant differences among control and Aldo SAGE libraries

Fig. 1. Validation of SAGE candidates by Northern blot hybridization.mRNAs were isolated from primary cultures of neonatal mouse cardiomyo-cytes either untreated (C) or treated for 4 h with 10−6 M aldosterone (A). Twoindependent experiments are presented for each condition. Blots were probedwith 32P-labeled Art3, Tsc22d3, or Pol-like transcript probes. The blot was alsoprobed with 32P-labeled α-myosin heavy chain (αMHC), to check for the equalloading of cardiomyocyte-originated RNAs. These experiments were performedat least four times for each of these transcripts and the results were alwayssimilar.

was performed by using Fisher's exact test (p<0.05). Suchanalysis allowed the identification of 101 transcripts differen-tially expressed between Aldo and control SAGE libraries (seeTable 1). To validate the SAGE data, Northern blot hybridizationwas performed on three induced genes, namely, Art3, the Pol-like transcript, and the glucocorticoid-induced leucine zipper(Tsc22d3) (Fig. 1). One gene, namely period homolog 1 (Per1),was validated using real-time quantitative RT-PCR analysis. Asshown in Fig. 2, Per1 mRNA levels were significantly increased

Fig. 3. (A) Characterization of anti-ART3 antibody. The new antibodyrecognizes two bands of ∼47 and ∼37 kDa (lane 1). To test for the specificityof the antibody, the blot was probed either with the preimmune serum (lane 2) orwith immune serum incubated in the presence of GST protein (lane 3) or Art3 –GST fusion protein (lane 4). (B) Time course of ART3 protein induction byaldosterone. Primary cultures of neonatal mouse cardiomyocytes were stimulatedby aldosterone (10−6 M) for the indicated times. Proteins were recovered directlyin the SDS –PAGE sample buffer and run through 10% SDS –PAGE. Thisexperiment was performed three times.

Page 5: Identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene expression

374 O. Muller et al. / Genomics 89 (2007) 370–377

at 4 h of aldosterone stimulation and remained equallyupregulated at 8 h of hormonal action.

Regulation of Art3 protein expression by 10−6 M aldosterone

Next, we addressed the question of aldosterone vs gluco-corticoids andMR vs GR selectivity of transcriptional activationof our identified transcripts. As an example, we performed thisanalysis for Art3, an enzyme responsible for mono-ADP-ribosylation of its target proteins [12]. We first developed ananti-Art3 polyclonal antibody raised against the C-terminal partof the Art3 protein. As shown in Fig. 3A, this antibodyspecifically recognizes two bands with apparent molecularmasses (Mr) of ∼47 and ∼37 kDa that were absent whenprobing was performed with the preimmune serum or when theantibody was in competition with the Art3-GST fusionprotein. The time-course analysis revealed that aldosteronestimulation of Art3 protein expression is delayed, compared toArt3 mRNA expression, and reaches its maximal levelsbetween 24 and 32 h of aldosterone stimulation (Fig. 3B). The32-h time point of stimulation with aldosterone/corticosteronewas chosen for all the following measurements of Art3 proteinexpression.

Corticosterone- versus aldosterone-dependent activation ofArt3 expression

The 10−6 M concentration of aldosterone used in theaforementioned experiments does not allow distinction betweenaldosterone- and glucocorticoid-stimulatory effects, on the onehand, or between MR- and GR-dependent effects, on the other

Fig. 4. Determination of the EC50 values for stimulation of Art3 protein expressioindicated doses of aldosterone or corticosterone for 32 h. Cardiomyocyte proteins werBio-Rad Phosphoimager system.

hand. Thus, in a first step toward the characterization ofhormonal and receptor specificity for Art3 expression regula-tion, we determined the effective concentrations of aldosteroneand corticosterone for half-maximal response (EC50) in primaryculture of wild-type mouse cardiomyocytes. As shown in Fig. 4,aldosterone and corticosterone stimulate Art3 protein expres-sion with EC50 values of 39±9 (n=9) and 23±8 nM (n=7),respectively. These experiments demonstrate that Art3 expres-sion can be stimulated by physiologically relevant doses ofcorticosterone but supraphysiological concentrations of aldos-terone are required to produce the same effect. Therefore, theseexperiments suggest that Art3 is a corticosterone-rather than analdosterone-regulated gene.

Art3 protein expression is stimulated exclusively via the GR

Since the previously mentioned experiments cannot excludethe involvement of MR in corticosterone- (or aldosterone-)mediated stimulation of Art3 expression, the requirement of MRand/or GR in the activation of Art3 expression was tested inmouse models selectively deprived of either MR or GR.Because mice lacking the GR die within a few hours after birthand mice lacking the MR die in the second week after birth[13,14], the experiments were performed on mouse fetusesdelivered by cesarean section on day 18.5 of gestation. Twominicultures of cardiomyocytes (2 wells of a 48-well plate)were prepared from one heart of each genotype: one minicultureserved as a control and the other was stimulated with saturatingaldosterone concentration (10−6 M). In a set of controlexperiments, the lack of MR expression and the presence ofGR in MR(null/null) mice, and the lack of GR expression and

n by aldosterone and corticosterone. Cardiomyocytes were stimulated with thee recovered and run in 10% SDS–PAGE.Western blots were quantified using the

Page 6: Identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene expression

Fig. 5. MR and GR expression in MR(null/null) and GR(null/null) mice.Ethidium bromide-stained gels containing RT-PCR amplification products of(A) GR or (B) MR mRNAs are shown. 35 PCR cycles were performed for allsamples. A single band of the expected size was obtained for both GR and MR(408 and 335 bp, respectively).

Fig. 6. MR vs GR specificity of aldosterone effect on Art3 protein expression.(A) Two minicultures of cardiomyocytes were prepared from one heart oflittermate mouse pups of each genotype: MR(+/+), MR(+/null), MR(null/null).One miniculture served as a control (C) and the other was stimulated withsaturating aldosterone (A) concentration (10−6 M). (B) Two minicultures ofcardiomyocytes were prepared from one heart of littermate mouse pups of eachgenotype: GR(+/+), GR(+/null), GR(null/null). One miniculture served as acontrol (C) and the other was stimulated with saturating aldosterone (A)concentration (10−6 M). These experiments were repeated four times withidentical results.

375O. Muller et al. / Genomics 89 (2007) 370–377

the presence of MR in GR(null/null) mice, were confirmed byRT-PCR analysis of corresponding mRNAs (Fig. 5). As shownin Fig. 6A, the activation of Art3 expression by aldosterone wasfully maintained in MR(+/null) and MR(null/null) mice. Incontrast, knockout mutation of GR in GR(null/null) miceresulted in a complete suppression of aldosterone activation ofArt3 expression (Fig. 6B). These experiments suggest thatcorticosteroids regulate Art3 expression exclusively via the GR.

Discussion

Corticosteroid-regulated genes in cardiomyocytes

In this study, we identified a new set of 101 corticosteroid-regulated transcripts in neonatal mouse cardiomyocytes.Analysis of regulated transcripts revealed that only a smallfraction of these were previously identified as aldosterone- orcorticosterone/cortisol-regulated genes. Expression of Art3[15], Tsc22d3 [16,17], Per1 [18], potassium channel Kcnj14[19], ribosomal protein L7 [20], stearoyl-coenzyme A desatur-ase 1 [21], cyclin-dependent kinase inhibitor p21 [22], anddiaphorase [23] has been shown to vary upon corticosteroidtreatment in different tissues and in different experimentalsettings. In two of these genes, namely Tsc22d3 and Per1, theglucocorticoid-responsive elements (GRE) have been charac-terized [24,25]. Among four genes validated in this study eitherby Northern hybridization or by quantitative real-time RT-PCR,Art3 was identified as a prednisolone-induced transcript indystrophic skeletal muscle in a mouse model of Duchennedisease [15]. Per1 gene expression was demonstrated to bestrongly upregulated by dexamethasone in fibroblasts [18].Tsc22d3 has been recently shown to be a corticosterone-upregulated transcript in rat cardiomyocytes [16]. Interestingly,the expression of Tsc22d3 was found to be significantly induced

by dexamethasone in thymocytes [26], but selectively upregu-lated by aldosterone in the kidney distal nephron [17]. The latterdemonstrates that the same transcript can be specificallyregulated by either glucocorticoids or aldosterone, in distincttarget tissue.

Here, using a combined pharmacological and transgenicapproach, we characterized the Art3 gene as being regulated byglucocorticoids through the activation of GR in cardiomyocytes.

Identification of MR- vs GR-mediated signaling pathways inclassical vs nonclassical aldosterone target cells

Serum corticosterone and aldosterone levels are regulated bydistinct physiological/pathophysiological stimuli, but follow, innormal situations, synchronous circadian secretion curves [27].Serum corticosterone concentration is at least 100-fold higherthan that of aldosterone and varies in concentration, ∼10–50 nM in the unbound form. Glucocorticoids (corticosterone/cortisol) and aldosterone exhibit a similar high affinity to MR(Kd∼0.5–2 nM) and a lower affinity to GR (Kd ~15–60 nM). Inclassical aldosterone target tissues (distal nephron, colon), theselectivity of MR to aldosterone is provided by the activity of11βHSD2, which converts corticosterone/cortisol into inactivemetabolites.

In cardiomyocytes expressing very low levels of 11β-HSD2activity, the role of aldosterone in MR activation remainsambiguous. Three hypotheses have been proposed to explainthe MR-mediated effects in cardiomyocytes. A first hypothesisproposes an endogenous synthesis of aldosterone in cardiactissues (cardiomyocytes, cardiofibroblast, or vessels). Accord-ing to this hypothesis, the local aldosterone production in theheart would be high enough to get tissue aldosterone concen-

Page 7: Identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene expression

376 O. Muller et al. / Genomics 89 (2007) 370–377

tration comparable to that of plasma glucocorticoids [28]. Thisinteresting “auto or paracrine” model of aldosterone action hasbeen recently questioned by a comprehensive study of Gomez-Sanchez et al. [29]. A new generation of highly specific anti-aldosterone and anti-corticosterone antibodies was used todemonstrate that cardiac aldosterone levels reflect directly thosemeasured in the general circulation. A second hypothesisproposes that cardiac MR becomes accessible for aldosteroneonly in pathophysiological situations, when circulating aldos-terone levels are higher than normal [30]. However, from thepharmacological viewpoint, it remains difficult to understandhow even elevated aldosterone levels can compete for MRbinding with glucocorticoids that are present at a 100-foldconcentration excess. A third hypothesis proposed by Funder[31], not yet supported by strong experimental evidence,implies that the tonic effect of glucocorticoids on MR couldbe modulated by changes in the intracellular redox state uponcertain pathophysiological circumstances. In cardiomyocytes,methodological factors make the analysis of MR vs GRspecificity much more difficult than in the kidney. There areno established cardiomyocyte cell lines that would allow adetailed analysis of MR and GR occupancy, as is possible inmCCD cells [32]. Indeed, the very small amount of material thatcan be obtained from primary cultures prevents such anapproach. Our method allows us to demonstrate as proof ofprinciple the transcriptional regulation of a specific genemediated by glucocorticoid activation of GR in cardiomyocytes.The existence of MR-mediated and aldosterone-mediatedtranscriptional effects in cardiomyocytes will require furthercharacterization of transcriptional regulation of identifiedcandidates by individual analysis of hormone and receptorselectivity.

Materials and methods

Primary culture of neonatal mouse cardiomyocytes

Hearts from newborn mice or from mouse fetuses (day 18.5 of gestation)were excised under sterile conditions and cut into four equal parts. Afterextensive washing, these parts were transferred into an Eppendorf tubecontaining 1 ml of Hanks’ medium (Gibco/Invitrogen), supplemented with0.5 mg/ml trypsin (USB). Incubation with trypsin lasted overnight with gentleagitation (700 rpm, Eppendorf Compact Thermomixer) at 4°C. At the end ofthe incubation with trypsin, tissue was allowed to settle for 1 min, and the500 μl trypsinization solution was removed and replaced by 500 μl of Lightmedium composed of DMEM/M-199 media (3/1, v/v), supplemented with1/100 times diluted (v/v) standard mix of penicillin/streptomycin/glutamine(Gibco/Invitrogen) and 10 mM Hepes (pH 7.3). Tubes were incubated for2.5 min with gentle agitation (1100 rpm, Eppendorf Compact Thermomixer) at37°C. Tissue was allowed to settle and medium was replaced by 250 μl ofHanks’ medium, supplemented with 300 U/ml collagenase type 2 (Worthing-ton Biochemical). Tubes were incubated for 1.5 min with gentle agitation(1100 rpm, Eppendorf Compact Thermomixer) at 37°C. Medium wasdiscarded and replaced by a new 250-μl aliquot of Hanks’/collagenasesolution. Tubes were incubated for 7 min at 37°C (1100 rpm, EppendorfCompact Thermomixer). Medium was removed and mixed with 250 μl ofComplete medium composed of the Light medium supplemented with 10%horse serum and 5% fetal calf serum (Gibco/Invitrogen). The collagenasedigestion step was performed two additional times. The three fractions ofcardiac cells were centrifuged separately at 750 g for 5 min at 4°C. Mediumwas discarded and cell pellets were pooled in 750 μl of Complete medium.

Cells were plated in a single well of a 24-well plate and incubated for 1 h15 min at 37°C (10% CO2). This step allows attachment of fibroblasts to thewell surface and their separation from other cells. This step was repeated once.The remaining cells were plated in 2 wells of a 24-well plate coated withgelatin and fibronectin. In the resulting culture, at least 85% of all cells werecardiomyocytes. The cardiomyocytes were actively beating throughout theexperimental period (3 days). The MR(null/null) and GR(null/null) neonatalmice were obtained by breeding of corresponding heterozygous mice bearingone null and one wild-type allele [33,34].

Serial analysis of gene expression

SAGE libraries were prepared with 20 μg of poly(A) mRNA as described[35]. A total of 250,350 tags were sequenced (63,445 tags for the control library,60,446 for the aldosterone library, 62,676 for the LIF library, 61,611 for theneuregulin library, and 62,618 for the AngII library). A total of 2927 differenttags, representing 2506 different genes, were found five or more times amongthe five libraries. This number represents the actual number of genes assessed fora significant up- or downregulation. Comparison between control and AldoSAGE libraries was performed by using Fisher's exact test. Two cut-offthresholds were used to identify the differentially expressed SAGE tags: (i)p<0.05 according to the Fisher test and (ii) a twofold change in tag numbersbetween the two libraries.

Development of an anti-Art3 antibody and western blotting

Rabbit polyclonal antibody was developed against a C-terminal peptide(D281MDN…NFEL370) of a mouse Art3 protein (NCBI Accession No.CAA69284) fused to a GST construct. The antibody was affinity purified andused at a 1:500 dilution. This antibody specifically recognizes a single band of∼37 kDa and a nonspecific band of ∼43 kDa. Proteins were electrophoresedthrough an 8–13% gradient SDS–PAGE and Western blotted, according to astandard procedure. Western blots were quantified using the Bio-RadPhosphoimager scan system.

Quantitative real-time PCR analysis of Per1 gene expression

Primary cultures of neonatal mouse cardiomyocytes were either untreated(control) or stimulated for 4 or 8 h with aldosterone (10− 6 M) Total RNA wasextracted and reverse transcribed using Superscript II reverse transcriptase(Invitrogen). Real-time quantitative PCR analysis was performed using theTaqMan system from Applied Biosystems. All reactions were performed intriplicate. All primer sets used in this study were from Applied Biosystems:TaqMan Gene Expression Assay PER1Mm00501813_m1 (for Per1) and mouseACTB (β-actin) Endogenous Control (for β-actin).

Qualitative RT-PCR analysis of GR and MR mRNA expression incardiomyocytes

Total RNA was extracted from cardiomyocytes originating from wild-type,GR(null/null), or MR(null/null) mice. RT-PCR was performed using Titan OneTube RT-PCR System (Roche). The sense and antisense primers designed toamplify a 408-bp fragment of GR mRNA were 5′-CCTTGGAGGTCA-GACCTGTT-3′ and 5′-CTTCTGATCCTGCTGCTGAG-3′, respectively. Thesense and antisense primers designed to amplify a 335-bp fragment of MRmRNA were 5′-CCAGTTCTCCGTTCTCTGTA-3′ and 5′-CTTGAGCAC-CAATCCGGTAG-3′, respectively. The amplification products were run on a2.5% agarose gel and stained with ethidium bromide.

Acknowledgments

This work was supported by Swiss National Fund forScientific Research Grants 3100AO-105592 (to D.F.) and 3100-061966 (to B.R.). Work in F.T.’s laboratory was supported bythe Ministère de l’Éducation Nationale, de la Recherche et de la

Page 8: Identification of corticosteroid-regulated genes in cardiomyocytes by serial analysis of gene expression

377O. Muller et al. / Genomics 89 (2007) 370–377

Technologie, the Fondation Recherche Médicale, and theCentre National de la Recherche Scientifique.

References

[1] B. Pitt, F. Zannad, W.J. Remme, R. Cody, A. Castaigne, A. Perez, et al.,The effect of spironolactone on morbidity and mortality in patients withsevere heart failure. Randomized Aldactone Evaluation Study Investiga-tors, N. Engl. J. Med. 341 (1999) 709–717.

[2] B. Pitt, W. Remme, F. Zannad, J. Neaton, F. Martinez, B. Roniker, et al.,Eplerenone, a selective aldosterone blocker, in patients with left ventriculardysfunction after myocardial infarction, N. Engl. J. Med. 348 (2003)1309–1321.

[3] P.C. Souverein, A. Berard, T.P. Van Staa, C. Cooper, A.C. Egberts, H.G.Leufkens, et al., Use of oral glucocorticoids and risk of cardiovascular andcerebrovascular disease in a population based case–control study, Heart 90(2004) 859–865.

[4] A.T. Beggah, B. Escoubet, S. Puttini, S. Cailmail, V. Delage, A. Ouvrard-Pascaud, et al., Reversible cardiac fibrosis and heart failure induced byconditional expression of an antisense mRNA of the mineralocorticoidreceptor in cardiomyocytes, Proc. Natl. Acad. Sci. U. S. A. 99 (2002)7160–7165.

[5] A. Ouvrard-Pascaud, Y. Sainte-Marie, J.P. Benitah, R. Perrier, C.Soukaseum, A.N. Cat, et al., Conditional mineralocorticoid receptorexpression in the heart leads to life-threatening arrhythmias, Circulation111 (2005) 3025–3033.

[6] R.M. Whitehurst Jr., M. Zhang, A. Bhattacharjee, M. Li, Dexamethasone-induced hypertrophy in rat neonatal cardiac myocytes involves an elevatedL-type Ca(2+)current, J. Mol. Cell. Cardiol. 31 (1999) 1551–1558.

[7] R. Schulz, E. Nava, S. Moncada, Induction and potential biologicalrelevance of a Ca(2+)-independent nitric oxide synthase in the myo-cardium, Br. J. Pharmacol. 105 (1992) 575–580.

[8] I. Korichneva, M. Puceat, E. Millanvoye-Van Brussel, G. Geraud, G.Vassort, Aldosterone modulates both the Na/H antiport and Cl/HCO3

exchanger in cultured neonatal rat cardiac cells, J. Mol. Cell. Cardiol. 27(1995) 2521–2528.

[9] J.P. Benitah, G. Vassort, Aldosterone upregulates Ca(2+) current in adultrat cardiomyocytes, Circ. Res. 85 (1999) 1139–1145.

[10] N. Farman, M.E. Rafestin-Oblin, Multiple aspects of mineralocorticoidselectivity, Am. J. Physiol. Renal. Physiol. 280 (2001) F181–F192.

[11] S. Pradervand, H. Yasukawa, O.G.Muller, H. Kjekshus, T. Nakamura, T.R.St Amand, et al., Small proline-rich protein 1A is a gp130 pathway- andstress-inducible cardioprotective protein, EMBO J. 23 (2004) 4517–4525.

[12] G. Glowacki, R. Braren, K. Firner, M. Nissen, M. Kuhl, P. Reche, et al.,The family of toxin-related ecto-ADP-ribosyltransferases in humans andthe mouse, Protein Sci. 11 (2002) 1657–1670.

[13] S. Berger, M. Bleich, W. Schmid, T.J. Cole, J. Peters, H. Watanabe, et al.,Mineralocorticoid receptor knockout mice: pathophysiology of Na+

metabolism, Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 9424–9429.[14] F. Tronche, C. Kellendonk, H.M. Reichardt, G. Schutz, Genetic dissection

of glucocorticoid receptor function in mice, Curr. Opin. Genet. Dev. 8(1998) 532–538.

[15] I. Fisher, D. Abraham, K. Bouri, E.P. Hoffman, F. Muntoni, J. Morgan,Prednisolone-induced changes in dystrophic skeletal muscle, FASEB J. 19(2005) 834–836.

[16] Q.M. Chen, D. Alexander, H. Sun, L. Xie, Y. Lin, J. Terrand, et al.,Corticosteroids inhibit cell death induced by doxorubicin in cardiomyo-cytes: induction of antiapoptosis, antioxidant, and detoxification genes,Mol. Pharmacol. 67 (2005) 1861–1873.

[17] O.G. Muller, R.G. Parnova, G. Centeno, B.C. Rossier, D. Firsov, J.D.Horisberger, Mineralocorticoid effects in the kidney: correlation between

alphaENaC, GILZ, and Sgk-1 mRNA expression and urinary excretion ofNa(+) and K(+), J. Am. Soc. Nephrol. 14 (2003) 1107–1115.

[18] A. Balsalobre, S.A. Brown, L. Marcacci, F. Tronche, C. Kellendonk, H.M.Reichardt, et al., Resetting of circadian time in peripheral tissues byglucocorticoid signaling, Science 289 (2000) 2344–2347.

[19] S.R. Pondugula, N.N. Raveendran, Z. Ergonul, Y. Deng, J. Chen, J.D.Sanneman, et al., Glucocorticoid regulation of genes in the amiloride-sensitive sodium transport pathway by semicircular canal duct epitheliumof neonatal rat, Physiol. Genomics 24 (2006) 114–123.

[20] E.R. James, V.M. Fresco, L.L. Robertson, Glucocorticoid-induced changesin the global gene expression of lens epithelial cells, J. Ocul. Pharmacol.Ther. 21 (2005) 11–27.

[21] C.M. Smas, L. Chen, L. Zhao, M.J. Latasa, H.S. Sul, Transcriptionalrepression of pref-1 by glucocorticoids promotes 3T3-L1 adipocytedifferentiation, J. Biol. Chem. 274 (1999) 12632–12641.

[22] F.Y. Wu, H. Chen, S.E. Wang, C.M. ApRhys, G. Liao, M. Fujimuro, et al.,CCAAT/enhancer binding protein alpha interacts with ZTA and mediatesZTA-induced p21(CIP-1) accumulation and G(1) cell cycle arrest duringthe Epstein-Barr virus lytic cycle, J. Virol. 77 (2003) 1481–1500.

[23] S. Gonzalez, F. Labombarda, M.C. Gonzalez Deniselle, F.E. Saravia, P.Roig, A.F. De Nicola, Glucocorticoid effects on Fos immunoreactivity andNADPH-diaphorase histochemical staining following spinal cord injury,Brain Res. 912 (2001) 144–153.

[24] J.C. Wang, M.K. Derynck, D.F. Nonaka, D.B. Khodabakhsh, C. Haqq,K.R. Yamamoto, Chromatin immunoprecipitation (ChIP) scanningidentifies primary glucocorticoid receptor target genes, Proc. Natl.Acad. Sci. U. S. A. 101 (2004) 15603–15608.

[25] S. Koyanagi, S. Okazawa, Y. Kuramoto, K. Ushijima, H. Shimeno, S.Soeda, et al., Chronic treatment with prednisolone represses the circadianoscillation of clock gene expression in mouse peripheral tissues, Mol.Endocrinol. 20 (2006) 573–583.

[26] F. D'Adamio, O. Zollo, R. Moraca, E. Ayroldi, S. Bruscoli, A. Bartoli,et al., A new dexamethasone-induced gene of the leucine zipper familyprotects T lymphocytes from TCR/CD3-activated cell death, Immunity7 (1997) 803–812.

[27] S. Hurwitz, R.J. Cohen, G.H. Williams, Diurnal variation of aldosterone andplasma renin activity: timing relation to melatonin and cortisol andconsistency after prolonged bed rest, J. Appl. Physiol. 96 (2004) 1406–1414.

[28] J.S. Silvestre, V. Robert, C. Heymes, B. Aupetit-Faisant, C. Mouas, J.M.Moalic, et al., Myocardial production of aldosterone and corticosterone inthe rat: physiological regulation, J. Biol. Chem. 273 (1998) 4883–4891.

[29] E.P. Gomez-Sanchez, N. Ahmad, D.G. Romero, C.E. Gomez-Sanchez,Origin of aldosterone in the rat heart, Endocrinology 145 (2004)4796–4802.

[30] J.W. Funder, Aldosterone action: fact, failure and the future, Clin. Exp.Pharmacol. Physiol. Suppl. 25 (1998) S47–S50.

[31] J.W. Funder, RALES, EPHESUS and redox, J. Steroid Biochem. Mol.Biol. 93 (2005) 121–125.

[32] H.P. Gaeggeler, E. Gonzalez-Rodriguez, N.F. Jaeger, D. Loffing-Cueni, R.Norregaard, J. Loffing, et al., Mineralocorticoid versus glucocorticoidreceptor occupancy mediating aldosterone-stimulated sodium transport ina novel renal cell line, J. Am. Soc. Nephrol. 16 (2005) 878–891.

[33] S. Berger, D.P. Wolfer, O. Selbach, H. Alter, G. Erdmann, H.M. Reichardt,et al., Loss of the limbic mineralocorticoid receptor impairs behavioralplasticity, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 195–200.

[34] F. Tronche, C. Kellendonk, O. Kretz, P. Gass, K. Anlag, P.C. Orban, et al.,Disruption of the glucocorticoid receptor gene in the nervous systemresults in reduced anxiety, Nat. Genet. 23 (1999) 99–103.

[35] M. Robert-Nicoud, M. Flahaut, J.M. Elalouf, M. Nicod, M. Salinas, M.Bens, et al., Transcriptome of a mouse kidney cortical collecting duct cellline: effects of aldosterone and vasopressin, Proc. Natl. Acad. Sci. U. S. A.98 (2001) 2712–2716.