Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms...

12
HORMONES - CYTOKINES - SIGNALING Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA SAURA,CARLOS ZARAGOZA,MANUELA IAZ-CAZORLA,OCTAVIO HERN´ ANDEZ-PERERA,EUDORA ENG, CHARLES J. LOWENSTEIN,DOLORES EREZ-SALA, and SANTIAGO LAMAS Departamento de Estructura y Funcio ´n de Proteı ´nas, Centro de Investigaciones Biolo ´gicas, Consejo Superior de Investigaciones Cientı ´ficas, Instituto “Reina Sofı ´a” de Investigaciones Nefrolo ´gicas, Madrid, Spain, and Nephrology and Cardiology Divisions, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells. In previous studies we reported that stimulation of rat mesangial cells (RMC) with lipopoly- saccharide (LPS) 1 tumor necrosis factor alpha (TNF-a) (L/T) elicits inducible nitric oxide synthase (NOS2) mRNA expression, which is inhibited by dexamethasone (DX). We have now analyzed the mecha- nisms responsible for this inhibitory effect. Dexamethasone had no destabilizing effect on NOS2 mRNA. Transfection of RMC with several luciferase reporter constructs from the 59 flanking regulatory region of the rat NOS2 gene established the importance of the NF-kB site in the transcriptional activation of the NOS2 gene. DNA mobility shift assays showed activation by L/T of the NF-kB complex in a time-dependent manner. Dexamethasone specifically inhibited this activation in a process dependent on the glucocorticoid receptor and with a markedly greater effect when it was added prior to L/T. Dexamethasone increased the expression of the IkB-a transcript and protein in the cytoplasm. While treatment of RMC with L/T induced the transient decrement of cytoplas- mic p65 levels and its appearance in the nucleus, preincubation with DX prevented this effect. Co-immunoprecipitation and immunocytochemical studies demonstrated that IkB-a is associated with p65 in the cytoplasm of RMC after treatment with DX and L/T. These results prove that inhibition of NF-kB-mediated transcription is a crucial mechanism by which DX inhibits NOS2 expression, and that this occurs by increasing cytoplasmic IkB-a levels and sequestering the activating subunits of NF-kB in the cytoplasm. The need for previous induction of IkB-a could provide a molecular explanation for the limited efficacy of these agents in the therapy of septic shock. Nitric oxide (NO) is a labile free radical with varied biological functions, including neurotransmission, vasodilation and partici- pation in the nonadaptive immune response, [1]. Its synthesis occurs through the oxidation of one of the guanidino groups of L-arginine and also yields L-citrulline as a co-product. The group of enzymes that catalyze this reaction are termed nitric oxide synthases (EC 1.14.13.39). Three isoforms, named NOS1 (neuro- nal or nNOS), NOS2 (inducible or iNOS) and NOS3 (endothelial or ecNOS) sharing in common functional and structural proper- ties, have been identified. In terms of the regulation of their expression these enzymes are, nevertheless, markedly different [2]. NOS2 is an enzyme involved in the high output synthesis of NO in cells or tissues that have been exposed to extracellular stimuli. In mammals, the synthesis of NO by this enzyme is believed to be critical in pathophysiological conditions such as inflammation and septic shock [3]. In inflammatory scenarios NO may serve defense functions, contributing to the killing of infec- tive intracellular organisms or tumor cells. In septic shock, NO may contribute to the hemodynamic imbalance and high mortality of its final stages, a concept supported by studies in NOS2- deficient mutant mice [4, 5]. Thus, knowledge of the pharmaco- logical regulation of NOS2 expression becomes important in order to design strategies to maintain NO synthesis at non- harmful levels. NOS2 is mainly regulated at the level of transcrip- tion [6]. A fairly large number of substances have been described to up- or down-regulate NOS2 mRNA expression either by acting on mRNA stability or on the transcription rate [7]. Among these substances, glucocorticoids were one of the first to be shown to down-regulate NO synthesis and NOS2 expression [8, 9]. The recent characterization of the 59 flanking regulatory region of the NOS2 gene permits the exploration of the precise molecular mechanisms of the glucocorticoid effects. In the promoter regions characterized so far in the NOS2 genes, no sequences similar to the glucocorticoid responsive element have been described [10 – 12]. Hence, the molecular pathways by which these agents exert their effect on NOS2 transcription remain incompletely under- stood. Mesangial cells have well characterized physiological functions in the kidney glomeruli [13], and are the main players in their inflammatory response. They have been shown to synthesize NO under several stimuli [14, 15], and possible sources and roles of this mediator in glomerulonephritis has been brought up in the past several years [16]. In this study, we investigated the potential mechanisms for the glucocorticoid inhibition of NOS2 expression in rat mesangial cells. Our results offer experimental evidence to support the concept that glucocorticoids suppress NOS2 expres- sion in a process where their interaction with the NF-kB family of proteins is an essential step to inactivate transcription of the NOS2 gene. 1 M. Saura and C. Zaragoza contributed equally to this article. Key words: Nitric oxide synthase, mesangial cells, transcription, glucocor- ticoids, NF-kB. Received for publication February 3, 1997 and in revised form August 5, 1997 Accepted for publication August 6, 1997 © 1998 by the International Society of Nephrology Kidney International, Vol. 53 (1998), pp. 38 – 49 38

Transcript of Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms...

Page 1: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

HORMONES - CYTOKINES - SIGNALING

Involvement of transcriptional mechanisms in the inhibition ofNOS2 expression by dexamethasone in rat mesangial cells

MARTA SAURA, CARLOS ZARAGOZA, MANUELA DIAZ-CAZORLA, OCTAVIO HERNANDEZ-PERERA, EUDORA ENG,CHARLES J. LOWENSTEIN, DOLORES PEREZ-SALA, and SANTIAGO LAMAS

Departamento de Estructura y Funcion de Proteınas, Centro de Investigaciones Biologicas, Consejo Superior de InvestigacionesCientıficas, Instituto “Reina Sofıa” de Investigaciones Nefrologicas, Madrid, Spain, and Nephrology and Cardiology Divisions, JohnsHopkins University School of Medicine, Baltimore, Maryland, USA

Involvement of transcriptional mechanisms in the inhibition of NOS2expression by dexamethasone in rat mesangial cells. In previous studieswe reported that stimulation of rat mesangial cells (RMC) with lipopoly-saccharide (LPS) 1 tumor necrosis factor alpha (TNF-a) (L/T) elicitsinducible nitric oxide synthase (NOS2) mRNA expression, which isinhibited by dexamethasone (DX). We have now analyzed the mecha-nisms responsible for this inhibitory effect. Dexamethasone had nodestabilizing effect on NOS2 mRNA. Transfection of RMC with severalluciferase reporter constructs from the 59 flanking regulatory region of therat NOS2 gene established the importance of the NF-kB site in thetranscriptional activation of the NOS2 gene. DNA mobility shift assaysshowed activation by L/T of the NF-kB complex in a time-dependentmanner. Dexamethasone specifically inhibited this activation in a processdependent on the glucocorticoid receptor and with a markedly greatereffect when it was added prior to L/T. Dexamethasone increased theexpression of the IkB-a transcript and protein in the cytoplasm. Whiletreatment of RMC with L/T induced the transient decrement of cytoplas-mic p65 levels and its appearance in the nucleus, preincubation with DXprevented this effect. Co-immunoprecipitation and immunocytochemicalstudies demonstrated that IkB-a is associated with p65 in the cytoplasm ofRMC after treatment with DX and L/T. These results prove that inhibitionof NF-kB-mediated transcription is a crucial mechanism by which DXinhibits NOS2 expression, and that this occurs by increasing cytoplasmicIkB-a levels and sequestering the activating subunits of NF-kB in thecytoplasm. The need for previous induction of IkB-a could provide amolecular explanation for the limited efficacy of these agents in thetherapy of septic shock.

Nitric oxide (NO) is a labile free radical with varied biologicalfunctions, including neurotransmission, vasodilation and partici-pation in the nonadaptive immune response, [1]. Its synthesisoccurs through the oxidation of one of the guanidino groups ofL-arginine and also yields L-citrulline as a co-product. The groupof enzymes that catalyze this reaction are termed nitric oxidesynthases (EC 1.14.13.39). Three isoforms, named NOS1 (neuro-nal or nNOS), NOS2 (inducible or iNOS) and NOS3 (endothelial

or ecNOS) sharing in common functional and structural proper-ties, have been identified. In terms of the regulation of theirexpression these enzymes are, nevertheless, markedly different[2]. NOS2 is an enzyme involved in the high output synthesis ofNO in cells or tissues that have been exposed to extracellularstimuli. In mammals, the synthesis of NO by this enzyme isbelieved to be critical in pathophysiological conditions such asinflammation and septic shock [3]. In inflammatory scenarios NOmay serve defense functions, contributing to the killing of infec-tive intracellular organisms or tumor cells. In septic shock, NOmay contribute to the hemodynamic imbalance and high mortalityof its final stages, a concept supported by studies in NOS2-deficient mutant mice [4, 5]. Thus, knowledge of the pharmaco-logical regulation of NOS2 expression becomes important inorder to design strategies to maintain NO synthesis at non-harmful levels. NOS2 is mainly regulated at the level of transcrip-tion [6]. A fairly large number of substances have been describedto up- or down-regulate NOS2 mRNA expression either by actingon mRNA stability or on the transcription rate [7]. Among thesesubstances, glucocorticoids were one of the first to be shown todown-regulate NO synthesis and NOS2 expression [8, 9]. Therecent characterization of the 59 flanking regulatory region of theNOS2 gene permits the exploration of the precise molecularmechanisms of the glucocorticoid effects. In the promoter regionscharacterized so far in the NOS2 genes, no sequences similar tothe glucocorticoid responsive element have been described [10–12]. Hence, the molecular pathways by which these agents exerttheir effect on NOS2 transcription remain incompletely under-stood.

Mesangial cells have well characterized physiological functionsin the kidney glomeruli [13], and are the main players in theirinflammatory response. They have been shown to synthesize NOunder several stimuli [14, 15], and possible sources and roles ofthis mediator in glomerulonephritis has been brought up in thepast several years [16]. In this study, we investigated the potentialmechanisms for the glucocorticoid inhibition of NOS2 expressionin rat mesangial cells. Our results offer experimental evidence tosupport the concept that glucocorticoids suppress NOS2 expres-sion in a process where their interaction with the NF-kB family ofproteins is an essential step to inactivate transcription of theNOS2 gene.

1 M. Saura and C. Zaragoza contributed equally to this article.

Key words: Nitric oxide synthase, mesangial cells, transcription, glucocor-ticoids, NF-kB.

Received for publication February 3, 1997and in revised form August 5, 1997Accepted for publication August 6, 1997

© 1998 by the International Society of Nephrology

Kidney International, Vol. 53 (1998), pp. 38–49

38

Page 2: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

METHODS

Reagents

Recombinant human tumor necrosis factor (rHuTNF-a, 9.8 3106 U/mg) was a gift from Knoll Pharmaceuticals (Whipanny, NJ,USA); cell culture media, fetal calf serum and media supplementswere from Gibco (Paisley, Scotland, UK). Taq polymerase wasfrom Promega (Madison, WI, USA), and dNTPs from GibcoBRL; deoxycytidine 59-triphosphate (a-32P; 3000 Ci/mmol) wasfrom Amersham (Buckinghamshire, UK); RU-486 (Mifepristone)was a gift from Roussel-UCLAF (Romainville, France). All otherreagents, including lipopolysaccharide (LPS) E. coli serotype0127:B8, dexamethasone and pyrrolidine dithiocarbamate(PDTC), were from Sigma Chemical (St. Louis, MO, USA).

Polyclonal antibodies against the human c-Rel protein and N-termini of human p65 and p50 proteins were kindly provided byDr. Nancy Rice (National Cancer Institute, Bethesda, MD, USA).The antibody against IkB-a was from Santa Cruz Biotechnologies(Santa Cruz, CA, USA).

Plasmids

Plasmid kB (33) con A-LUC contained three copies of the kBmotif from the HIV enhancer with no enhancer element of theconalbumin promoter [17]. RSV-b GAL was an expression vectorof b-galactosidase under control of the Rous sarcoma viruspromoter. The plasmid containing the cDNA for IkB-a was a giftof M.T. Dıaz-Meco and J. Moscat (Centro de Biologıa Molecular,Madrid, Spain).

Cell culture

Rat mesangial cells were characterized as described [18]. Cellswere used between passages 2 and 10. They were cultured in 100mm plates at 37°C with 5% CO2 and fed every 48 hours withRPMI 1640 supplemented with 10% fetal calf serum, L-glutamine(2 mM), penicillin (50 U/ml), and streptomycin (50 mg/ml).Potential toxicity of reagents on RMC was ruled out by trypanblue exclusion (. 90% of cells were viable) and measurements ofLDH release to the cell culture media.

RNA isolation and Northern blot analysis

Total cellular RNA was isolated from RMC with the guani-dinium thiocyanate-phenol-chloroform method [19], size-frac-tionated by electrophoresis (10 mg/lane) through denaturing 1%agarose-0.66 M formaldehyde gels, transferred to Hybond hybrid-ization transfer membranes (Amersham, UK), and UV cross-linked before hybridization as previously described [20]. A 699 bpfragment corresponding to the NOS2 cDNA was obtained fromtotal RNA of stimulated RMC, using reverse transcriptase fol-lowed by polymerase chain reaction (RT-PCR). The followingprimers were used for amplification: sense: 59 GAGAGATCCGATTTAGAGTCT 39; antisense: 59 GCAGATTCTGCTGGGA-TTTCA 39. Polymerase chain reaction was performed for 35cycles, each consisting of one minute at 94°C, one minute at 55°and one minute at 72°C. This 699 bp fragment was submitted tomanual and automatic sequencing, and its nucleotide sequence isavailable at the Genbank database (Accession number U48829).For Northern analysis, it was labeled with [a-32P]dCTP andrandom hexamers, using a commercial kit (Boehringer Mann-heim, Germany). For the Northern blot analysis of IkB-a, we useda BamHI/HindIII 1.4 kb fragment of the rat IkB-a cDNA labeled

as above. Hybridization was performed at 42°C for 12 to 16 hours,membranes were then washed at final stringency conditions of55°C, 0.2 3 SSC, 0.1% SDS and exposed with XAR Kodak film,using an intensifying screen. Blots were rehybridized with ab-actin cDNA probe [21]. Densitometric analysis was performedon an Apple scanner using Image 1.55 (NIH, Bethesda, MD,USA). Results are expressed in arbitrary units as the ratio ofNOS2/b-actin expression.

Cloning of 5*-flanking region of rat NOS2

To isolate a 59 flanking region fragment from the rat NOS2gene, PCR was performed using the following set of primers:sense (iNOSRP1): 59 CCAGAGAGCTCCGTGCCCAG 39; anti-sense (iNOSRP2): 59 GGAAGC TTGTGGGGCCAGAGTCT-CA 39. iNOSRP1 (20-mer) was selected from the reportedmacrophage murine regulatory region [11] (positions 2338 to2319). Sequence of the iNOSRP2 primer was based on theprevious cloned rat iNOS cDNA [22, 23], which is identical to thehomologous murine gene. A HindIII restriction site was added tothe 59 region of the iNOSRP2 oligomer. A 388 bp PCR fragmentamplified from rat genomic DNA with the above-cited oligomers,hybridized with the 1749 bp reported 59 murine NOS2 regulatoryregion. The fragment was cloned into a luciferase reporterplasmid (PGL-3 basic; Promega, Madison, WI, USA) (380-piNOS-LUC) and several clones were sequenced manually onboth strands with the dideoxy protocol (Sequenase version 2.0DNA sequencing kit; Amersham) and with a Perkin-Elmer 377automatic sequencer. Sequence analysis and search for transcrip-tion factors binding sites were done using the Findpatternsalgorithm from the database of the Genetics Computer Group(Madison, WI, USA). An additional 727 bp fragment from the59 flanking region of the rat iNOS gene was cloned by PCRusing the same antisense primer (iNOSRP2) and a sense primer(iNOSRP3): 59 CCACCAGACCTAGGTAGGGT 39 located atthe 2710 to 2693 positions of the rat iNOS promoter [24]. A SacIrestriction site was added in order to clone the fragment in thePGL-3 plasmid (720-piNOS-LUC). The 720 bp rat iNOS pro-moter fragment containing the mutated NF-kB site was con-structed using PCR. An antisense primer carrying the mutation inthe NF-kB motif (iNOSRPmut.): 59 ATAACTGTTCCCAAAG-GGAGAGTAACCAGTTATCGATGAA TTC 39 was used withthe sense iNOSRP3 described above to amplify a 700 bp fragmentthat was digested with SacI/PstI and inserted upstream of theTATA box in the original -700-iNOS-PGL-3 construct (720(NF-kB mut)-piNOS-LUC).

Transient transfections and luciferase assay

In initial experiments RMC were transfected by a liposome-mediated method (DOTAP reagent, B.; Mannheim, Germany).Two 3 106 RMC were transfected with 380-piNOS-LUC andRSV-bGAL using a 5 mg DNA/10 mg liposome ratio during sixhours in serum-reduced medium. Sixteen hours after transfection,cells were treated with the corresponding reagents during 48hours. Plates were incubated with reporter lysis buffer (Promega,Madison, WI, USA) and cells were processed as recommended bythe manufacturer. Light output was measured during 10 secondswith an LKB-Wallac 1250 luminometer (Pharmacia, Uppsala,Sweden). Luciferase activity was normalized for variations intransfection efficency as determined by b-galactosidase activitymeasurements [25]. In subsequent experiments a dual-luciferase

Saura et al: Transcriptional effects of DX on NOS2 39

Page 3: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

reporter assay was used. In this procedure RMC (1 3 106 cells/60mm culture dish) were co-transfected by a liposome-mediatedmethod (lipofectin; Gibco BRL) with 380-piNOS-LUC, 720-piNOS-LUC or 720 (NF-kB mut)-piNOS-LUC and a secondreporter vector (pRL-SV40 vector containing the Renilla lucif-erase gene (Promega, Madison, WI, USA) using a 5 mg DNA/10mg liposome ratio during six hours in serum-reduced medium; 16hours after transfection, cells were treated with the correspondingreagents during 48 hours. Cellular lysates were prepared with 400ml of passive lysis buffer (Promega) and cells were processed asrecommended by the manufacturer. Light output from firefly andRenilla luciferases was measured over 10 seconds of integrationperiod of each luciferase reaction. Firefly luciferase activity wasnormalized for variations in transfection efficency as determinedby the Renilla luciferase activity measurements. Protein contentwas assayed by the Bradford method by using a BioRad dyereagent (Richmond, CA, USA). Transfections were done induplicate and each experiment was repeated at least three times.

Preparation of nuclear extracts

A modification of the method of Schreiber et al was used [26].Rat mesangial cells (RMC) were washed twice with PBS buffer,scraped in PBS and transferred to microcentrifuge tubes. Cellpellets were resuspended in 400 ml of buffer A containing: 10 mM

Hepes, pH 7.9, 10 mM KCl, 10 mM EDTA, 10 mM EGTA, 1 mM

dithiotreitol (DTT), 0.5 mM phenylmethylsulfonyl fluoride

(PMSF) and 1 mg/ml of leupeptin, antipain, trypsin inhibitor andpepstatin A. After 10 minutes at 4°C, NP-40 was added to reacha final concentration of 2%. Tubes were vigorously vortexed for 10seconds and nuclei sedimented by centrifugation for 30 seconds ina microfuge. Nuclear pellets were resuspended in 50 ml of bufferC: 20 mM Hepes, pH 7.9, 0.4 M NaCl, 1 mM EDTA,1 mM EGTA,1 mM DTT, 5 mM MgCl2, 25% glycerol, 1 mM PMSF and proteaseinhibitors at 1 mg/ml, by gently shaking at 4°C. After centrifuga-tion at 13,000 3 g for 15 minutes, nuclear debris was discardedand aliquots of the supernatant, containing the nuclear proteins,were stored at 280°C. Protein content was determined by theBradford method as described above.

Electrophoretic mobility shift assays (EMSAS)

Oligonucleotides were synthesized in a Beckman Oligo 1000 M

DNA synthesizer. The oligonucleotide sequence corresponding tothe consensus sequence for binding of NF-kB, based on the ratNOS2 promoter, was:

59 CCTACTGGGGACTCTCCCTTTG 3939 GGATGACCCCTGAGAGGGAAAC 59Primers were annealed by incubation for five minutes at 85°C in

phosphate buffer 10 mM, pH 7, and slowly cooled down to roomtemperature; 2 pmol of these primers were end-labeled using T4polynucleotide kinase (Promega) in the presence of 10 mCi of

Fig. 1. Effect of dexamethasone (DX) and RU-486 on NOS2 mRNA expression in ratmesangial cells (RMC). The RMC were treatedwith vehicle (CT); lipopolysaccharide (LPS; 10mg/ml, 8 hr) 1 TNF-a (L/T; 100 ng/ml, 8 hr);DX (1026 M, 8 hr); RU-486 (RU; 10 mM, 8 hr);DX 1 L/T and DX 1 L/T 1 RU-486. TotalRNA was extracted and hybridized with NOS2(upper panel) and b-actin (middle panel)probes as described. Shown in the lower panelis the densitometric analysis of NOS2 mRNAexpression after correction by the expression ofb-actin. The data are representative of threeindependent experiments. *P , 0.05 versuscontrol.

Saura et al: Transcriptional effects of DX on NOS240

Page 4: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

(32P)-g-ATP. For binding reaction 1 to 2 3 104 cpm of end-labeled oligonucleotide probes were incubated with 10 mg ofnuclear extract and 1 mg of poly(dI.dC) in binding buffer (4%glycerol, 1 mM MgCl2, 0.5 mM EDTA, 0,5 mM DTT, 50 mM NaCl,10 mM Tris-HCl, pH 7.5) at room temperature for 30 minutes.Protein-DNA complexes were separated by electrophoresis in a4% non-denaturing polyacrylamide gel and visualized by autora-diography.

For competition experiments 200-fold molar excess of compet-itor DNA was incubated in the mixture prior to the addition ofnuclear extracts. When supershift assays were carried out, poly-clonal antibodies against specific NF-kB subunits were added tothe extracts 10 minutes prior to the probe.

Western blot and immunoprecipitation assays

Total cellular proteins were isolated using standard methods[27]. In brief, cellular monolayers were washed with PBS andharvested in protein lysis buffer (1% Triton X-100, 10 mM

Tris/HCl, pH 7.6, 1 mM EDTA, 0.1% sodium deoxycholate, 500nM sodium orthovanadate, 50 mM NaF, 0.5 mM PMSF, and 1mg/ml of antipain and leupeptin). Lysates were centrifuged (1500g, 5 min) in a microfuge and the supernatant was collected.Protein concentration was determined by the Bradford method.Two hundred micrograms of total protein were separated in aSDS-polyacrylamide gel electrophoresis (10% for IkB-a and 8%for p65) and transferred to a PDVF membrane (Polyscreen;Dupont, Wilmington, DE, USA). For protein detection, blockedmembranes were incubated first, with anti-IkB-a or anti-p65antibodies in 3% BSA in PBS during one hour, washed andincubated with a secondary antibody. After extensive washing,ECL detection was performed according to the manufacturer’sprocedures (Amersham).

For the immunoprecipitation assays cytoplasmic extracts (150

to 200 mg) were pre-cleared with rat IgG non-immune antiseraand protein A/G sepharose (Santa Cruz Biotechnologies) for twohours at 4°C. After centrifugation (1500 g, 5 min, 4°C), thesupernatants were incubated with anti-human IkB-a antibody (1mg) or preimmune serum, overnight at 4°C. Complexes were thenincubated with protein A/G sepharose for four hours at 4°C withgentle rocking. Protein A/G sepharose immunocomplexes weresuccessively washed four times with low stringency buffer (100 mM

Hepes, pH 7.6, 250 mM NaCl, 5 mM EDTA, 0.25% Nonidet P-40).Proteins were eluted by incubation with 200 ml of elution buffer(20 mM Tris-HCl, pH 7.4, 50 mM NaCl, 1% SDS, 5 mM dithio-threitol) for 10 minutes at 95°C. Supernatants were diluted with200 ml of 20 mM Tris-HCl, pH 7.4, 50 mM NaCl, 1% Nonidet P-40and 1% sodium deoxycholate, divided into two aliquots andsubjected to a second round of immunoprecipitation using anti-P65 or anti-IkB-a antibodies and 20 ml of protein A/G agaroseduring four hours at 4°C. Specific immunocomplexes were washedthree times with RIPA buffer (phosphate buffer saline, 1%Nonidet P-40, 0.5% sodium deoxycholate and 0.1% SDS, 500 nM

sodium orthovanadate plus 0.5 mM PMSF, and 1 mg/ml of antipainand leupeptin) eluted in 13 protein sample buffer, separated bySDS-polyacrylamide gel electrophoresis and subjected to Westernblot as described above.

Immunocytochemistry

Rat mesangial cells were cultured for 24 hours in glass cover-slips and treated as described. Cells were fixed with cold methanol

Fig. 2. Effect of dexamethasone (DX) on NOS2 mRNA stability. Ratmesangial cells (RMC) were treated with LPS (10 mg/ml) 1 TNF-a (100ng/ml) (L/T) for 24 hours with or without DX (1026 M). After the additionof 10 mg/ml actinomycin D (ActD) total RNA was extracted at theindicated time points and hybridized to a 32P-labeled NOS2 probe asdescribed in the Methods section. The ratios of NOS2/28 S ribosomalmRNA levels for every time point after ActD addition were obtained bydensitometric analysis and are represented in arbitrary units with respectto the values obtained at zero time for every experimental condition. Thedata are means 6 SE from two independent experiments. Where error barsare not visible they are smaller than the data point. Symbols are: (E) L/T;(Œ) DX 1 L/T.

Fig. 3. Effect of dexamethasone (DX) and RU-486 on NOS2 transcrip-tion in rat mesangial cells (RMC) treated with LPS and TNF-a. RMCwere transfected with 388 bp (f; 2380-piNOS-LUC), 720 bp (o; 2720-piNOS-LUC) or 720 bp with NF-kB mutated (u; 2720(NF-kB mut)-piNOS-LUC) fragments of the 59 flanking regulatory region of rat NOS2gene. Transfected cells were stimulated with vehicle (CT), LPS (10 mg/ml)1 TNF-a (100 ng/ml), (L/T) or DX (1026 M) 1 L/T during 48 hours Afterassaying for luciferase activity and/or lacZ, the luciferase relative lightunits were normalized by lacZ or Renilla luciferase activity and proteinamount and the data were reported as relative luciferase units. Results aremeans 6 SEM of three independent experiments, each performed induplicate plates. *P , 0.05 versus CT, **P , 0.05 versus L/T treatment ofRMC transfected with –720-piNOS-LUC, #P , 0.05 versus CT and versusL/T treatment of RMC transfected with 2720-piNOS-LUC, ##P , 0.05versus L/T treatment of RMC transfected with 2380-piNOS-LUC.

Saura et al: Transcriptional effects of DX on NOS2 41

Page 5: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

during 10 minutes at 220°C. After three washes with PBS, theslides were blocked with 10% of normal rabbit serum in PBS plus2% bovine serum albumin (BSA). Cells were incubated for onehour with the primary antibody (anti-p65 diluted 1:500 in 3%BSA). After three washes with PBS, cells were incubated one hourwith a fluorescein isothiocianate-conjugated secondary antibody(Amersham) in the dark. The nonincorporated antibody waswashed with PBS and coverslips were mounted into slides with adrop of PermaFluor (Lipshaw Immunon, Pittsburgh, PA, USA).Slides were visualized with a fluorescence microscope.

Statistical analysis

Every experimental condition was duplicated within each ex-periment and each experiment was performed at least three times.Comparisons were made with analysis of variance followed byDunnett’s modification of the t-test, whenever comparisons weremade with a common control, and the unpaired two-tailedStudent’s t-test was used for other comparisons. This test was usedwhenever the number of total observations allowed to and afterperforming a Kolmogorov-Smirnov test to confirm the normaldistribution of the data. When the number of observations wasinsufficient to perform adequate parametric tests, non-parametrictests were employed (Kruskal-Wallis). The level of a statisticallysignificant difference was defined as P , 0.05.

RESULTS

Dexamethasone inhibition of LPS/TNF-a-induced-NOS2 mRNAexpression in rat mesangial cells involves the glucocorticoidreceptor

As we have previously reported, DX inhibits NOS2 mRNAexpression induced by LPS/TNF (L/T) in a concentration- andtime-dependent fashion [15]. In order to understand the mecha-nism of DX inhibition we evaluated the involvement of theglucocorticoid receptor (Gr) in this process. As shown in Figure 1,when rat mesangial cells were treated with DX, an 80 to 90%reduction of the L/T-induced NOS2 transcript levels was detected.Simultaneous addition of the glucocorticoid receptor antagonistRU-486 abolished the effect of DX, thus demonstrating theparticipation of Gr in the DX pathway. Neither DX nor RU-486alone induced the expression of NOS2.

Dexamethasone inhibits NOS2 mRNA expression by a directtranscriptional effect

In experiments designed to test the influence of DX on thestability of the NOS2 transcript elicited by a combination of LPSand TNF-a in RMC, we found that DX prolonged the half-life ofNOS2 mRNA slightly more than two hours (Fig. 2). These dataexcluded mRNA destabilization as a potential mechanism bywhich DX could be exerting its inhibitory effect on NOS2 mRNA

Fig. 4. Time course of NF-kB activation in rat mesangial cells induced with LPS1TNF-a. (A) Mesangial cells were incubated with LPS (10 mg/ml) 1TNF-a (100 ng/ml) for the indicated periods of time. Nuclear protein extracts were obtained as described in the Methods section and analyzed by EMSAusing the NF-kB binding oligonucleotide from the rat NOS2 59 flanking region as a probe. Data are representative of four independent experiments.(B) Supershift assays were carried out using the indicated antibodies against distinct proteins of the NF-kB protein family or no additions (CT).Antibodies were used at 1 mg/ml. Competition assays (200-fold excess of unlabeled oligonucleotide) and an unrelated competitor oligonucleotide wereused to ensure the specificity of the bands (not shown). The position of the NF-kB complexes and that of a nonspecific complex (NS) is indicated byarrows. Shown is a representative of three independent experiments.

Saura et al: Transcriptional effects of DX on NOS242

Page 6: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

expression and suggested a transcription-inhibitory effect of DX.To gain insight into the transcriptional regulation of rat NOS2 wecloned a 727 bp fragment from the 59 regulatory region of ratNOS2 obtained as specified in the experimental section. Thenucleotide sequence of this fragment (Genbank accession numberU61282) shares 98%, 89% and 64% identity with the reportedsequences of the corresponding regions from the rat [24], murine[10, 11] and human [12] promoters, respectively. Sequencescomplying for the consensus sequences of the TATA box, Oct-1,NF-IL-6, NF-kB and TNF-RE [28] all included in the 388 bpfragment, are 100% conserved in the rat sequence when com-pared to the murine promoter. The 727 bp fragment includedthree additional fully conserved g-IRE sites and a less wellconserved AP-1 site.

We next performed a functional characterization of this 59flanking region. In RMC transfected with 380-piNOS-LUC and720-piNOS-LUC, the combination of LPS and TNF-a induced asignificant increase in luciferase activity (Fig. 3). This increase wasmarkedly greater in the case of 720-piNOS-LUC. DX was able tocompletely prevent L/T-stimulated luciferase activity observed inthe case of 380-piNOS-LUC and significantly abrogated the effectof L/T on the promoter activity of the 720-piNOS-LUC construct(Fig. 3). This inhibitory effect of DX was prevented by theaddition of the glucocorticoid receptor antagonist, RU-486 (Fig.3). Transfection of RMC with a short construct of 97 nucleotideswhich included the TATA box but not the NF-kB element showedno response to LPS, TNF-a or their combination (N 5 2, data notshown).

To explore the potential involvement of NF-kB in LPS/TNF-induced activation, we carried out experiments with RMC trans-fected with the construct kB(x3)-con A-LUC. We observed thatLPS and TNF-a increased luciferase activity threefold in thesecells, and that this activation was markedly abrogated both by DXand PDTC, a well-characterized inhibitor of NF-kB activation(N 5 2, data not shown). When RMC were transfected with aplasmid where the NF-kB cis-regulatory sequence had beenmutated [720 (NF-kB mut)-piNOS-LUC], no increment in lucif-erase activity over basal levels was observed in either control orinduced RMC (Fig. 3). Taken together, these results indicate thatNF-kB activation is necessary for NOS2 transcription in RMC andthat this activation could be potentially modulated by DX. Thus,we investigated the role of DX in the pattern of NF-kB bindingactivity under the experimental conditions previously described.

Treatment of RMC with L/T promotes activation of NF-kB

Electrophoretic mobility shift assays (EMSAs), using oligonu-cleotide pairs with the kB sequence of the rat NOS2 promoter,indicated a clear increment in the binding activity of this tran-scription factor in RMC (Fig. 4A). This phenomenon could beobserved as early as 15 minutes after exposure of RMC to L/T andit reached its peak after six hours with a slight decline after eighthours, an observation fully consistent with the pattern of NOS2mRNA expression and NO2

2 accumulation previously described[15]. Competition with an excess of unlabeled oligonucleotidecompletely abolished the detection of the complex (Fig. 6), whilethe inclusion of an oligonucleotide containing the consensussequence for an unrelated transcription factor (SP-1) did notinterfere with formation of the complex (Fig. 6).

To identify the specific subunits involved in the formation of theactive NF-kB dimer in RMC treated with L/T, EMSAs were

performed in the presence of antibodies against the subunits p50,p65 and c-Rel, which have been shown to participate in theformation of the NF-kB dimer implicated in the activation of theNOS2 promoter [29, 30]. As shown in Figure 4B, incubation withanti-p65 or anti-p50 antibodies induced a clear supershift. Thiseffect was more intense when both antibodies were used together.However, the presence of an antibody directed against c-Relshifted only a minor proportion of the complex (Fig. 4B). Acombination of anti-c-Rel and anti-p50 induced a supershift thatwas significantly less evident than when the combination ofanti-p65 and anti-p50 was used. Therefore, our data suggest amajor role for the p50/p65 components in L/T-induced NF-kBactivation in RMC. Incubation with an antibody directed againstc-jun had no effect on the electrophoretic mobility of the complex.

Dexamethasone inhibits NF-kB activation in a time-dependentfashion through its interaction with the glucocorticoid receptor

As depicted in Figure 5, EMSAs performed in nuclear extractsfrom RMC stimulated with L/T for six hours clearly showed thatDX was able to inhibit NF-kB activation. This effect was partiallyreversed by RU-486, suggesting the need for the interactionbetween DX and the Gr for the inhibition to occur. Interestingly,DX-induced inhibition was clearly time-dependent (Fig. 6), beingmore intense when RMC had been pre-incubated for two hourswith the drug (63% inhibition) than when the latter was addedeither at the same time or two hours after the addition of L/T

Fig. 5. Lipopolysaccharide (LPS) 1 TNF-a-induced NF-kB-binding ac-tivity in rat mesangial cells (RMC) is repressed by dexamethasone (DX)and dependent on the glucocorticoid receptor. RMC were treated duringsix hours with: vehicle; L/T (LPS (10 mg/ml) 1 TNF-a (100 ng/ml); DX(1026 M); RU-486 (1026 M); DX 1 L/T; RU-486 1 L/T and RU-486 1 DX1 L/T as indicated. Abbreviations are in the legend to Figure 1. Nuclearextracts were obtained as described in the Methods section and 7 mg ofprotein were analyzed by EMSA using the NF-kB binding oligonucleotidefrom the rat NOS2 sequence as a probe. Results are representative ofthree independent experiments.

Saura et al: Transcriptional effects of DX on NOS2 43

Page 7: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

(42% and 25% inhibition, respectively). In contrast, PDTC had asignificant inhibitory effect (more than 70% inhibition) at alltime-points studied. These observations again show good corre-lation with the results on NOS2 mRNA levels and NO2

2 accu-mulation previously reported [15].

Dexamethasone induces the expression of IkB-a mRNA andprotein and promotes its association with p65 in the cytoplasmof rat mesangial cells treated with LPS and TNF-a

To gain insight into the mechanism of DX-mediated inhibitionof NF-kB activation, we explored whether the mRNA expressionof its inhibitory subunit, IkB, could be modified by the presence ofDX. As shown in Figure 7A, treatment of RMC with L/Tincreased IkB-a mRNA levels after an incubation period of 30minutes to one hour. However, when cells were treated with L/Tin the presence of DX, the IkB-a transcript was detectable asearly as 15 minutes. This effect of DX could be due to its abilityto induce IkB-a in RMC, as shown in Figure 7B.

After an appropriate stimulus, the IkB protein is known toundergo phosphorylation and subsequent proteolytic degradation,allowing the other two subunits of the NF-kB complex (that is,p50 and p65) to translocate to the nucleus where transcription ofthe target gene is activated [31]. Hence, we designed experimentsto address whether Dx is modulating this process, by increasingIkB levels and thus preventing NF-kB nuclear translocation. Asdepicted in Figure 8, treatment of RMC with L/T resulted indecreased IkB-a protein levels in the cytoplasm that was mostapparent after 30 minutes of stimulation (Fig. 8A, left panel) andcorrelated well with a significant increment of p65 protein levels in

the nucleus (Fig. 8B, left panel). When cells were pre-treated withDX the levels of IkB-a were consistently higher (Fig. 8A, rightpanel) and the appearance of nuclear p65 was significantlydelayed (Fig. 8B, right panel).

In this context it was of interest to determine if DX couldmodify the levels of IkB complexed with p65, as this could reflectmore precisely if DX-induced IkB transcript levels were involvedin hindering the translocation of NF-kB to the nucleus. For thispurpose we designed a protocol in which the association of theIkB-a and p65 subunits was evaluated by Western blot after twosubsequent co-immunoprecipitation steps using specific antibod-ies directed either against IkB-a or p65. Under control conditionsa detectable fraction of IkB-a appears associated with p65 (Fig. 9,upper panel). Treatment of RMC with L/T resulted in a markeddecrease in the amount of p65 which co-precipitates with IkB-a(Fig. 9, lower panel). Conversely, the amount of IkB-a that can beco-precipitated by the anti-p65 antibody is nearly undetectable(Fig. 9, upper panel). By contrast, treatment of RMC with DXinduced a significant increment in the amount of p65 whichappears associated with IkB-a in the cytoplasm (Fig. 9, lowerpanel). Also, almost all IkB-a can be co-immunoprecipitated bythe anti-p65 antibody. Pre-incubation with DX in cells treatedwith L/T results in the retention of p65 in the cytoplasm as aconsequence of its association with IkB-a (Fig. 9). This behavioris further illustrated in Figure 10 in which the subcellular local-ization of p65 is demonstrated by fluorescence microscopy. Undercontrol conditions most of the p65 immunoreactivity is localizedin the cytosol. Treatment of RMC with L/T induced the translo-cation of p65 to the nuclear compartment. This translocation was

Fig. 6. Effects of dexamethasone (DX) andpyrrolidine dithiocarbamate (PDTC) on NF-kBactivation. Rat mesangial cells (RMC) weretreated as follows for the indicated periods oftime (in hours): Vehicle; L/T (LPS (10 mg/ml)1 TNF-a (100 ng/ml)); DX (1026 M,); PDTC(100 mM); DX 1 L/T and PDTC 1 L/T.Abbreviations are in the legend of Figure 1.Nuclear extracts were analyzed by EMSA asdescribed above. Data are representative ofthree independent experiments. Competitionassays (200-fold excess of unlabeledoligonucleotide) and an unrelated competitoroligonucleotide (SP-1) were used to ensure thespecificity of the bands.

Saura et al: Transcriptional effects of DX on NOS244

Page 8: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

completely abrogated in the presence of DX. Furthermore,treatment of RMC with the Gr antagonist RU-486 completelyabolished the effect of DX, thus allowing the detection of the p65immunoreactive signal in the nucleus.

Taken together, results shown in Figures 7, 8, 9 and 10demonstrate that a fundamental mechanism by which DX inhibitsNF-kB-mediated transcriptional activation in RMC is by theup-regulation of the IkB-a mRNA and protein levels. As a

Fig. 7. Effect of dexamethasone (DX) on IkB-a mRNA expression in rat mesangial cells (RMC) stimulated with LPS 1 TNF-a. (A) RMC were treatedwith L/T (LPS (10 mg/ml) 1 TNF-a (100 ng/ml) with or without DX (1026 M) for the indicated time periods. Total RNA was extracted and 10 mganalyzed by Northern blot. The blot was probed first with MAD-3-IkB-a cDNA (upper panel). and reprobed with b-actin cDNA (middle panel). Shownin the lower panel is the densitometric analysis of the IkB-a expression time-course in the absence (open circles) and presence (black triangles) of DXafter correction by the expression of b-actin. The data are representative of two independent experiments. (B) Cells were treated with DX (1026 M) forthe indicated time periods. Total RNA was extracted and 10 mg analyzed by Northern blot. The blot was probed first with MAD-3-IkB-a cDNA (upperpanel). and reprobed with b-actin cDNA (middle panel). Shown in the lower panel is the densitometric analysis of IkB-a mRNA expression aftercorrection by the expression of b-actin. The data are representative of two independent experiments.

Fig. 8. Effects of dexamethasone (DX) on IkB-a protein levels and nuclear localization of p65 in rat mesangial cells (RMC). RMC were pretreatedwith DX (1026 M) for two hours and/or stimulated with LPS (10 mg/ml) 1 TNF-a (100 ng/ml) (L/T) for the indicated periods of time. Cytoplasmic (A)and nuclear (B) extracts were isolated and analyzed by immunoblotting with anti-IkB-a or anti-p65 antibodies, respectively. Data are representative oftwo independent experiments.

Saura et al: Transcriptional effects of DX on NOS2 45

Page 9: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

consequence of this increase, the NF-kB complex (p65 in theresults shown) is retained in the cytoplasm, a circumstance thatresults in very low levels of translocation into the cell nucleus andhence in markedly diminished transcriptional activation.

DISCUSSION

In this work we analyze some of the potential mechanismsresponsible for the inhibitory effect of DX on the expression ofNOS2 mRNA induced by LPS and TNF-a in RMC. Our resultsshow that DX modulates NOS2 gene expression by acting atmultiple levels. Whereas it slightly prolongs the NOS2 mRNAhalf-life, experiments in RMC transfected with promoter con-structs of the rat NOS2 gene clearly showed that DX inhibited thispromoter activity. Furthermore, we established the participationof the NF-kB transcription factor and its inhibitory subunit IkB ascrucial targets in the mechanism by which DX inhibits NOS2expression.

Dexamethasone inhibits the expression of many pro-inflamma-tory genes [32], and the molecular basis of this effect has beenrecently characterized. Research performed on the structure andfunction of the glucocorticoid receptor family identified generalmechanisms for the transcriptional regulation exerted by theseagents, which implied the participation of the HSP90-Gr complexand its activation after ligand binding [33]. Activation of Grresulted in its translocation to the nucleus where it bound tospecific cis-regulatory regions of various genes. However, analysisof the 59 flanking regulatory regions from several proinflamma-tory genes whose transcription was clearly regulated by DXshowed the absence of these sequences. Hence, alternative expla-nations supported by experimental data were proposed, includingthe interaction between Gr and other transcription factors such asAP-1 or NF-kB [32, 34–37], whose cognate cis-regulatory se-quences were present in the genes studied. Finally, another levelof complexity has been recently added, since a direct modulationof the activation of the paradigmatic proinflammatory transcrip-tion factor NF-kB by DX has been observed [31]. Thus, it appearsas if the effect of DX on gene expression may utilize one or morepathways depending upon the gene and the cell type or tissueinvolved, from which multiple levels of interaction may result, thefinal balance being the induction or repression of a particulargene.

Dexamethasone has inhibitory effects on NOS2 mRNA expres-

sion in several cell types [38, 39]. However, contradiction hasarisen regarding the potential molecular mechanisms of thisinhibition. In smooth muscle cells, DX modestly inhibited thetranscriptional rate of NOS2 mRNA synthesis while it doubled itshalf-life [39]. Availability of the promoter region of NOS2 hasmade it possible to extend the mechanistic investigation of NOS2expression. Two general concepts that have emerged from thefunctional analysis of these promoters include the profounddifferences in structure-function relationships between humanand murine cells [40] and the presence of two main regulatoryregions in the murine NOS2 promoter: one responsive to IFN-gthat could resemble an enhancer, and the other sensitive to LPS,closer to the transcriptional start site [10, 11]. In addition, thefunctional relevance of the cis-regulatory element NF-kB hasbeen analyzed in murine macrophages [29] and rat vascularsmooth-muscle cells [41]. Since the molecular characterization ofNOS2, several groups including ours have analyzed the effect ofDX on NOS2 protein and mRNA expression in rat mesangial cells[14, 15, 42]. The functional characterization of the 59 flankingregulatory region of NOS2 in cells of rat origin facilitates theunderstanding of the DX-mediated effects. In this study we showthat RMC transfected with two fragments of different length ofthis region, an NF-kB tandem and a fragment containing amutation for the NF-kB site, clearly establish that NF-kB is atranscription factor necessary, but not sufficient, for full promoteractivity. The interaction of other transcription factors with theregion of the promoter included in the construct of greater lengthmay actually enhance NF-kB-mediated transcriptional activationand, therefore, should be the object of further analysis. Also, thisset of results suggested an inhibitory effect of DX on theactivation of NF-kB. Accordingly, DX is able to inhibit NF-kBactivity in a process dependent on the Gr and the temporal courseof this phenomenon correlates closely with the inhibition of NOS2mRNA expression observed in previous studies [15]. In keepingwith these observations we have found that DX increases IkBmRNA. This effect, as supported by experiments looking at thetemporal course of appearance of IkB-a and p65 proteins incytoplasmic and nuclear extracts and co-immunoprecipitationassays of p65 and IkB-a plus immunocytological studies, results inthe persistent association of IkB-a with the activating subunits ofNF-kB, promoting its retention in the cytoplasm and henceimpeding its stimulus-dependent nuclear translocation. These

Fig. 9. Dexamethasone (DX) induces IkB-asynthesis and association with p65. Ratmesangial cells (RMC) were pretreated withDX (1026 M) for two hours and stimulated withL/T (LPS (10 mg/ml) 1 TNF-a (100 ng/ml)during four hours. Two hundred micrograms ofcytoplasmic extracts were precleared andimmunoprecipitated using either pre-immuneserum (PS) or anti-IkB-a antibodies andprotein A/G agarose. Immunocomplexes wereprecipitated, washed and eluted and thesupernatant was divided in two aliquots whichwere re-immunoprecipitated with either anti-p65 or anti-IkB-a antibodies.Immunocomplexes were separated by SDS-PAGE, transferred and analyzed byimmunoblotting. The data are representative ofthree different experiments. Abbreviations arein the legend to Figure 1.

Saura et al: Transcriptional effects of DX on NOS246

Page 10: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

results agree with data obtained in some cell lines [43, 44] but arein contrast with what has been described in endothelial cells or ratbrain, where the physical interaction between the glucocorticoidreceptor and NF-kB seems to be the fundamental mechanism ofthe glucocorticoid-mediated repression [45, 46]. Different resultshave been found in other studies looking at the mechanism of DXinhibition of NOS2 induction. In a transformed human cell line asimilar inhibition of NF-kB by DX was reported but with noapparent effect on IkB induction [47]. In RMC treated withIL-1b, transcriptional (including deactivation of NF-kB), post-transcriptional (mRNA stabilization) and post-translational ef-fects of DX were found [48]. This diversity of data emphasizes theimportance of characterizing each cell system with each particularstimulus, in order to identify potential pathways of therapeuticintervention in inflammatory processes.

In view of our study, it can be proposed that a main regulatorycheckpoint responsible for the inhibitory effect of DX on NOS2

expression takes place at the transcriptional level and that it issignificantly mediated through the induction of IkB-a. The factthat pretreatment of RMC with DX and the subsequent inductionof IkB-a expression has a stronger effect than co-treatment orpost-treatment with the drug is consistent with this contention,since higher intracellular levels of this inhibitory subunit wouldbetter prevent NF-kB activation, by sequestration of the complexin the cytoplasm.

It is well known that NO plays a critical role in the pathophys-iology of septic shock. The necessity of pretreating cells with DXin order to efficiently suppress NOS2 expression and NO synthe-sis, based on the above proposed mechanisms, might help toexplain the reported lack of effect of massive glucocorticoidadministration in the final stages of sepsis [49]. Future studiesshould then focus on obtaining therapeutic tools whose mainmechanism of action is distal to the establishment of intracellularsignals triggered by the proinflammatory stimuli.

Fig. 10. Dexamethasone (DX) treatmentretains p65 in the cytoplasm of L/T-stimulatedrat mesangial cells (RMC). RMC were platedinto coverslips and treated with vehicle (CT),DX (1026 M, 2 hr) and DX (1026 M, 2 hr) 1RU-486 (10 mM, 2 hr) (DX1RU) with orwithout LPS (10 mg/ml,1 hr) 1 TNF-a (100ng/ml, 1 hr) (L/T). After treatment, cells werefixed and the intracellular localization of p65was determined by indirect immunofluorescencewith an anti-p65 antibody. Microphotographsrepresent typical fields of RMC whereimmunofluorescent p65 is clearly visible.

Saura et al: Transcriptional effects of DX on NOS2 47

Page 11: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

ACKNOWLEDGMENTS

Part of this work was presented in abstract form at the InternationalConference on Biochemistry and Molecular Biology of Nitric Oxide,UCLA, Los Angeles, July 1996. This work was supported by DGICYTPB-93-0044 and Biomed 2 Concerted Action BMH4-CT96-0979. M.S. andC.Z. were supported by a fellowship from Fundacion Renal “Inigo Alvarezde Toledo” and are currently at the Division of Cardiology, Johns HopkinsSchool of Medicine, Baltimore, MD. We are grateful to Drs. Nancy Rice,M.T. Dıaz-Meco, L. Bosca and Roussel-UCLAF for valuable reagents.The authors also thank Drs. L. Bosca and J. Rey for helpful comments.

Reprint requests to Dr. Santiago Lamas, Centro de Investigaciones Bio-logicas/CSIC, Velazquez 144, 28006 Madrid, Spain.E-mail:[email protected]

APPENDIX

Abbreviations used in this article are: RMC, rat mesangial cells; DX,dexamethasone; LPS, lipopolysaccharide; TNFa, tumor necrosis factoralpha; L/T, lipopolysaccharide plus TNFa; NO, nitric oxide; NOS, nitricoxide synthase; NOS1, neuronal isoform of NOS; NOS2, inducible isoformof NOS; NOS3, endothelial isoform of NOS; rHuTNF, recombinanthuman tumor necrosis factor; a-32P, deoxycytidine 59-triphosphate; RU-486, Mifepristone; PDTC, pyrrolidine dithiocarbamate; RT-PCR, reversetranscriptase—polymerase chain reaction; EMSAS, electrophoretic mo-bility shift assays; BSA, bovine serum albumin; Gr, glucocorticoid recep-tor.

REFERENCES

1. NATHAN C: Nitric oxide as a secretory product of mammalian cells.FASEB J 6:3051–3064, 1992

2. LAMAS S, MICHEL T: Molecular biological features of nitric oxidesynthases, in Nitric Oxide and the Lung, edited by ZAPOL WM, BLOCHKD, New York, Marcel Dekker 1997, pp 59–73

3. SZABO C: Alterations in nitric oxide production in various forms ofcirculatory shock. New Horiz 3:2–32, 1995

4. MACMICKING JD, NATHAN C, HOM G, CHARTRAIN N, FLETCHER DS,TRUMBAUER M, STEVENS K, XIE Q-W, SOKOL K, HUTCHINSON N,CHEN H, MUDGETT JS: Altered responses to bacterial infection andendotoxic shock in mice lacking inducible nitric oxide synthase. Cell81:641–650, 1995 (published erratum appears in Cell 81:1170, 1995)

5. WEI XQ, CHARLES IG, SMITH A, URE J, FENG GJ, HUANG FP, XU D,MULLER W, MONCADA S, LIEW FY: Altered immune responses inmice lacking inducible nitric oxide synthase. Nature 375:408–411,1995

6. NATHAN C, XIE Q-W: Nitric oxide synthases: Roles, tolls and controls.Cell 78:915–918, 1994

7. BOGDAN C, ROLLINGHOFF M, VODOVOTZ Y, XIE Q-W, NATHAN C:Regulation of inducible nitric oxide synthase in macrophages bycytokines and microbial products, in Immunotherapy of Infections,edited by MASIHI N, New York-Basel-Hong Kong, Marcel Dekker,Inc., 1994, pp 37–54

8. KNOWLES RG, SALTER M, BROOKS SL, MONCADA S: Anti-inflamatoryglucocorticoids inhibit the induction by endotoxin of nitric oxidesynthase in the lung, liver and aorta of the rat. Biochem Biophys ResComm 172:1042–1048, 1990

9. DI ROSA M, RADOMSKI M, CARNUCCIO R, MONCADA S: Glucocorti-coids inhibit the induction of nitric oxide synthase in macrophages.Biochem Biophys Res Comm 172:1246–1252, 1990

10. XIE QW, WHISNANT R, NATHAN C: Promoter of the mouse geneencoding calcium-independent nitric oxide synthase confers inducibil-ity by interferon gamma and bacterial lipopolysaccharide. J Exp Med177:1779–1784, 1993

11. LOWENSTEIN CJ, ALLEY EW, RAVAL P, SNOWMAN AM, SNYDER SH,RUSSELL SW, MURPHY WJ: Macrophage nitric oxide synthase gene:Two upstream regions mediate the induction by interferon g andlipopolysaccharide. Proc Natl Acad Sci USA 90:9730–9734, 1993

12. CHARTRAIN NA, GELLER DA, KOTY PP, SITRIN NF, NUSSLER AK,HOFFMAN EP, BILLIAR TR, HUTCHINSON NI, MUDGETT JS: Molecularcloning, structure,and chromosomal localization of the human induc-ible nitric oxide synthase gene. J Biol Chem 269:6765–6772, 1994

13. MENE P, SIMONSON MS, DUNN MJ: Physiology of the mesangial cell.Physiol Rev 69:1347–1425, 1989

14. SHULTZ PJ, ARCHER SL, ROSENBERG ME: Inducible nitric oxidesynthase mRNA and activity in glomerular mesangial cells. Kidney Int46:683–689, 1994

15. SAURA M, LOPEZ S, RODRIGUEZ PUYOL M, RODRIGUEZ PUYOL D,LAMAS S: Regulation of inducible nitric oxide synthase expression inrat mesangial cells and isolated glomeruli. Kidney Int 47:500–509, 1995

16. RAIJ L, SHULTZ PJ: Endothelium-derived relaxing factor, nitric oxide:Effects on and production by mesangial cells and the glomerulus. J AmSoc Nephrol 3:1435–1441, 1993

17. LOZANO J, BERRA E, MUNICIO MM, DIAZ-MECO MT, DOMINGUEZ I,SANZ L, MOSCAT J: Protein kinase C z isoform is critical kB-dependent promoter activation by sphingomyelinase. J Biol Chem269:19200–19202, 1994

18. RODRIGUEZ-PUYOL D, LAMAS S, OLIVERA A, ORTEGA G, LOPEZ-FARRE A, HERNANDO L, LOPEZ-NOVOA JM: Actions of cyclosporin Aon cultured rat mesangial cells. Kidney Int 35:632–637, 1989

19. CHOMCZYNSKI P, SACCHI N: Single-step method of RNA isolation byguanidinium thiocyanate-phenol-chloroform extraction. Anal Bio-chem 162:156–159, 1987

20. LAMAS S, MICHEL T, COLLINS T, BRENNER BM, MARSDEN PA: Effectsof interferon gamma on nitric oxide synthase activity and endothelin-1production by vascular endothelial cells. J Clin Invest 90:879–887,1992

21. ALONSO S, MINTY A, BOURLET Y, BUCKINGHAM M: Comparison ofthree actin-coding sequences in the mouse: Evolutionary relationshipsbetween the actin genes of warm-blooded vertebrates. J Mol Evol23:11–22, 1986

22. NUNOKAWA Y, ISHIDA N, TANAKA S: Cloning of inducible nitric oxidesynthase in rat vascular smooth muscle cells. Biochem Biophys ResComm 191:89–94, 1993

23. GENG Y-J, ALMQVIST M, HANSSON GK: cDNA cloning and expressionof inducible nitric oxide synthase from rat vascular smooth musclecells. Biochim Biophys Acta 1218:421–424, 1994

24. EBERHARDT W, KUNZ D, HUMMEL R, PFEILSCHIFTER J: Molecularcloning of the rat inducible nitric oxide synthase gene promoter.Biochem Biophys Res Comm 223:752–756, 1996

25. KRESS C, VOGELS R, DE GRAAF W, BONNEROT C, MEIJLINK F,NICOLAS JF, DESCHAMPS J: Hox-2.3 upstream sequences mediate lacZexpression in intermediate mesoderm derivatives of transgenic mice.Development 109:775–786, 1990

26. SCHREIBER E, MATTHIAS P, MULLER MM, SCHAFFNER W: Rapiddetection of octamer binding proteins with ’mini-extracts’, preparedfrom a small number of cells. (abstract) Nucl Acids Res 17:6419, 1989

27. Current Protocols in Molecular Biology. Edited by AUSUBEL FM, BRENTR, KINGSTON RE, MOORE PE, SEIDMAN JG, SMITH JA, STRUHL K.New York, Wiley & Sons, 1991

28. FAISST S, MEYER S: Compilation of vertebrate-encoded transcriptionfactors. Nucl Acids Res 20:3–26, 1992

29. XIE Q-W, KASHIWABARA Y, NATHAN C: Role of the transcriptionfactor NF-kB/Rel in induction of nitric oxide synthase. J Biol Chem269:4705–4708, 1994

30. GOLDRING CEP, NARAYANAN R, LAGADEC P, JEANNIN JF: Transcrip-tional inhibition of the inducible nitric oxide synthase gene bycompetitive binding of NF-k/REL proteins. Biochem Biophys ResComm 209:73–79, 1995

31. BARNES PJ, KARIN M: Nuclear factor-kB–A pivotal transcriptionfactor in chronic inflammatory diseases. N Engl J Med 336:1066–1071,1997

32. BARNES PJ, ADCOCK I: Anti-inflammatory actions of steroids: Molec-ular mechanisms. TIPS Rev 14:436–441, 1993

33. PRATT WB: The role of heat-shock proteins in regulating the function,folding and trafficking of the glucocorticoid receptor. J Biol Chem268:21455–21458, 1993

34. MUKAIDA N, MORITA M, ISHIKAWA Y, RICE N, OKAMOTO S, KASA-HARA T, MATSUSHIMA K: Novel mechanism of glucocorticoid-medi-ated gene repression. J Biol Chem 269:16289–13295, 1994

35. RAY A, PREFONTAINE KE: Physical association and functionalantagonism between the P65 subunit of transcription factor NF-kBand the glucocorticoid receptor. Proc Natl Acad Sci USA 91:752–756, 1994

36. SCHEINMAN RI, GUALBERTO A, JEWELL CM, CIDLOWSKI JA, BALDWIN

Saura et al: Transcriptional effects of DX on NOS248

Page 12: Involvement of transcriptional mechanisms in the ... · Involvement of transcriptional mechanisms in the inhibition of NOS2 expression by dexamethasone in rat mesangial cells MARTA

AS JR: Characterization of mechanisms involved in transreppressionof NF-kB by activated glucocorticoid receptors. Mol Cell Biol 15:943–953, 1995

37. OHTSUKA T, KUBOTA A, HIRANO T, WATANABE K, YOSHIDA H,TSURUFUJI M, IIZUKA Y, KONISHI K, TSURUFUJI S: Glucocorticoid-mediated gene suppression of rat cytokine-induced neutrophil che-moattractant CINC/gro, a member of the interleukin-8 family,through impairment of NF-kB activation. J Biol Chem 271:1651–1659,1996

38. GELLER DA, NUSSLER AK, DI SILVIO M, LOWENSTEIN CJ, SHAPIRORA, WANG SC, SIMMONS RL, BILLIAR TR: Cytokines, endotoxin,and glucocorticoids regulate the expression of inducible nitricoxide synthase in hepatocytes. Proc Natl Acad Sci USA 90:522–526,1993

39. PERRELLA MA, YOSHIZUMI M, FEN Z, TSAI J, HSIEH C, KOUREMBA-NAS S, LEE M: Transforming growth factor-b 1, but not dexametha-sone, down-regulates nitric-oxide synthase mRNA after its inductionby interleukin-1b in rat smooth muscle cells. J Biol Chem 269:14595–14600, 1994

40. DE VERA ME, SHAPIRO RA, NUSSLER AK, MUDGETT JS, SIMMONSRL, MORRIS SM JR, BILLIAR TR, GELLER DA: Transcriptionalregulation of human inducible nitric oxide synthase (NOS2) gene bycytokines: Initial analysis of the human NOS2 promoter. Proc NatlAcad Sci USA 93:1054–1059, 1996

41. SPINK J, COHEN J, EVANS TJ: The cytokine responsive vascular smoothmuscle cell enhancer of inducible nitric oxide synthase. J Biol Chem270:29541–29547, 1995

42. KUNZ D, WALKER G, PFEILSCHIFTER J: Dexamethasone differentiallyaffects interleukin 1. b- and cyclic AMP-induced nitric oxide synthase

mRNA expression in renal mesangial cells. Biochem J 304:337–340,1994

43. SCHEINMAN RI, COGSWELL PC, LOFQUIST AK, BALDWIN AS JR: Roleof transcriptional activation of glucocorticoids. Science 270:283–86,1995

44. AUPHAN N, DIDONATO JA, ROSETTE C, HELMBERG A, KARIN M:Immunosuppression by glucocorticoids: Inhibition of NF-kB throughinduction of IkB synthesis. Science 270:286–290, 1995

45. BROSTJAN C, ANRATHER J, CSIZMADIA V, STROKA D, SOARES M,BACH FH, WINKLER H: Glucocorticoid-mediated repression of NFkBactivity in endothelial cells does not involve induction of IkB-asynthesis. J Biol Chem 271:19612–19616, 1996

46. TINO UNLAP M, JOPE RS: Dexamethasone attenuates NFkB DNAbinding activity without inducing IkB levels in rat brain in vivo. MolBrain Res 45:83–89, 1997

47. KLEINERT H, EUCHENHOFER C, IHRIG-BIEDERT I, FORSTERMANN U:Glucocorticoids inhibit the induction of nitric oxide synthase II bydown regulating cytokine-induced activity of transcription factornuclear factor-kB. Molec Pharmacol 49:15–21, 1996

48. KUNZ D, WALKER G, EBERHARDT W, PFEILSCHIFTER J: Molecularmechanisms of dexamethasone inhibition of nitric oxide synthaseexpression in interleukin 1 beta-stimulated mesangial cells: Evidencefor the involvement of transcriptional and posttranscriptional regula-tion. Proc Natl Acad Sci USA 93:255–259, 1996

49. BONE RC, FISHER CJ JR, CLEMMER TP, SLOTMAN GJ, METZ CA, BALK

RA, GROUP MSSS: A controlled clinical trial of high-dose methyl-prednisolone in the treatment of severe sepsis and septic shock.N Engl J Med 317:653–658, 1987

Saura et al: Transcriptional effects of DX on NOS2 49