19.11.2012 Views

Interleukin-33 Induces Expression of Adhesion Molecules and ...

Interleukin-33 Induces Expression of Adhesion Molecules and ...

Interleukin-33 Induces Expression of Adhesion Molecules and ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Interleukin</strong>-<strong>33</strong> <strong>Induces</strong> <strong>Expression</strong> <strong>of</strong> <strong>Adhesion</strong> <strong>Molecules</strong> <strong>and</strong> Inflammatory<br />

Activation in Human Endothelial Cells <strong>and</strong> in Human Atherosclerotic Plaques<br />

Svitlana Demyanets, Viktoria Konya, Stefan P. Kastl, Christoph Kaun, Sabine<br />

Rauscher, Alex<strong>and</strong>er Niessner, Richard Pentz, Stefan Pfaffenberger, Kathrin Rychli,<br />

Christ<strong>of</strong> E. Lemberger, Rainer de Martin, Akos Heinemann, Ihor Huk, Marion Gröger,<br />

Gerald Maurer, Kurt Huber <strong>and</strong> Johann Wojta<br />

Arterioscler Thromb Vasc Biol 2011, 31:2080-2089: originally published online July<br />

7, 2011<br />

doi: 10.1161/ATVBAHA.111.231431<br />

Arteriosclerosis, Thrombosis, <strong>and</strong> Vascular Biology is published by the American Heart Association.<br />

7272 Greenville Avenue, Dallas, TX 72514<br />

Copyright © 2011 American Heart Association. All rights reserved. Print ISSN: 1079-5642. Online<br />

ISSN: 1524-4636<br />

The online version <strong>of</strong> this article, along with updated information <strong>and</strong> services, is<br />

located on the World Wide Web at:<br />

http://atvb.ahajournals.org/content/31/9/2080<br />

Subscriptions: Information about subscribing to Arteriosclerosis, Thrombosis, <strong>and</strong> Vascular<br />

Biology is online at<br />

http://atvb.ahajournals.org//subscriptions/<br />

Permissions: Permissions & Rights Desk, Lippincott Williams & Wilkins, a division <strong>of</strong> Wolters<br />

Kluwer Health, 351 West Camden Street, Baltimore, MD 21202-2436. Phone: 410-528-4050. Fax:<br />

410-528-8550. E-mail:<br />

journalpermissions@lww.com<br />

Reprints: Information about reprints can be found online at<br />

http://www.lww.com/reprints<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


Data Supplement (unedited) at:<br />

http://atvb.ahajournals.org/http://atvb.ahajournals.org/content/suppl/2011/07/07/ATVBAHA.111.23143<br />

1.DC1.html<br />

Subscriptions: Information about subscribing to Arteriosclerosis, Thrombosis, <strong>and</strong> Vascular<br />

Biology is online at<br />

http://atvb.ahajournals.org//subscriptions/<br />

Permissions: Permissions & Rights Desk, Lippincott Williams & Wilkins, a division <strong>of</strong> Wolters<br />

Kluwer Health, 351 West Camden Street, Baltimore, MD 21202-2436. Phone: 410-528-4050. Fax:<br />

410-528-8550. E-mail:<br />

journalpermissions@lww.com<br />

Reprints: Information about reprints can be found online at<br />

http://www.lww.com/reprints<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


<strong>Interleukin</strong>-<strong>33</strong> <strong>Induces</strong> <strong>Expression</strong> <strong>of</strong> <strong>Adhesion</strong> <strong>Molecules</strong><br />

<strong>and</strong> Inflammatory Activation in Human Endothelial Cells<br />

<strong>and</strong> in Human Atherosclerotic Plaques<br />

Svitlana Demyanets, Viktoria Konya, Stefan P. Kastl, Christoph Kaun, Sabine Rauscher,<br />

Alex<strong>and</strong>er Niessner, Richard Pentz, Stefan Pfaffenberger, Kathrin Rychli, Christ<strong>of</strong> E. Lemberger,<br />

Rainer de Martin, Akos Heinemann, Ihor Huk, Marion Gröger, Gerald Maurer,<br />

Kurt Huber, Johann Wojta<br />

Objective—<strong>Interleukin</strong> (IL)-<strong>33</strong> is the most recently described member <strong>of</strong> the IL-1 family <strong>of</strong> cytokines <strong>and</strong> it is a lig<strong>and</strong> <strong>of</strong><br />

the ST2 receptor. While the effects <strong>of</strong> IL-<strong>33</strong> on the immune system have been extensively studied, the properties <strong>of</strong> this<br />

cytokine in the cardiovascular system are much less investigated.<br />

Methods/Results—We show here that IL-<strong>33</strong> promoted the adhesion <strong>of</strong> human leukocytes to monolayers <strong>of</strong> human<br />

endothelial cells <strong>and</strong> robustly increased vascular cell adhesion molecule-1, intercellular adhesion molecule-1,<br />

endothelial selectin, <strong>and</strong> monocyte chemoattractant protein-1 protein production <strong>and</strong> mRNA expression in human<br />

coronary artery <strong>and</strong> human umbilical vein endothelial cells in vitro as well as in human explanted atherosclerotic<br />

plaques ex vivo. ST2-fusion protein, but not IL-1 receptor antagonist, abolished these effects. IL-<strong>33</strong> induced<br />

translocation <strong>of</strong> nuclear factor-�B p50 <strong>and</strong> p65 subunits to the nucleus in human coronary artery endothelial cells<br />

<strong>and</strong> human umbilical vein endothelial cells <strong>and</strong> overexpression <strong>of</strong> dominant negative form <strong>of</strong> I�B kinase 2 or I�B�<br />

in human umbilical vein endothelial cells abolished IL-<strong>33</strong>-induced adhesion molecules <strong>and</strong> monocyte chemoattractant<br />

protein-1 mRNA expression. We detected IL-<strong>33</strong> <strong>and</strong> ST2 on both protein <strong>and</strong> mRNA level in human<br />

carotid atherosclerotic plaques.<br />

Conclusion—We hypothesize that IL-<strong>33</strong> may contribute to early events in endothelial activation characteristic for the<br />

development <strong>of</strong> atherosclerotic lesions in the vessel wall, by promoting adhesion molecules <strong>and</strong> proinflammatory<br />

cytokine expression in the endothelium. (Arterioscler Thromb Vasc Biol. 2011;31:2080-2089.)<br />

Key Words: leukocyte adhesion � endothelial cells � IL-<strong>33</strong> � ST2 � atherosclerosis<br />

Cardiovascular disease is the leading cause <strong>of</strong> death in<br />

Western societies. 1 Among cardiovascular pathologies<br />

atherosclerosis is thought to be the principal contributor<br />

to cardiovascular morbidity <strong>and</strong> mortality. Atherosclerosis<br />

is now generally thought to be a chronic<br />

inflammatory disorder. 2–4 Leukocyte trafficking from<br />

bloodstream to tissue is important for rapid leukocyte<br />

accumulation at sites <strong>of</strong> inflammatory response or tissue<br />

injury. Thus it is evident that leukocyte extravasation is<br />

considered a key event in the pathogenesis <strong>of</strong> atherosclerosis.<br />

In fact, endothelial dysfunction, a hallmark in the<br />

early development <strong>of</strong> atherosclerosis is, besides impaired<br />

vasomotor function <strong>of</strong> the vessel wall, characterized by<br />

increased adhesiveness <strong>of</strong> the activated or injured endo-<br />

thelium for leukocytes at the site <strong>of</strong> developing <strong>and</strong><br />

progressing atherosclerotic lesions. 5 The process <strong>of</strong> leukocyte<br />

extravasation comprises a complex multistep cascade<br />

that is orchestrated by a tightly coordinated sequence <strong>of</strong><br />

adhesive interactions <strong>of</strong> the leukocytes with vessel wall<br />

endothelial cells. Endothelial cells express an array <strong>of</strong><br />

adhesion molecules that control processes such as leukocyte<br />

rolling along <strong>and</strong> attachment to the endothelium <strong>and</strong><br />

transmigration <strong>of</strong> leukocytes into areas <strong>of</strong> inflammation. 6<br />

These leukocyte-endothelial interactions require the regulated<br />

expression <strong>of</strong> various adhesion molecules by endothelial<br />

cells such as intercellular adhesion molecule-1<br />

(ICAM-1), vascular cell AM-1 (VCAM-1) <strong>and</strong> endothelial<br />

selectin (E-selectin). 7 Several studies have demonstrated<br />

Received on: August 13, 2010; final version accepted on: June 25, 2011.<br />

From the Department <strong>of</strong> Internal Medicine II (S.D., S.P.K., C.K., A.N., R.P., S.P., K.R.,G.M., J.W.), Medical University <strong>of</strong> Vienna, Vienna, Austria,<br />

Institute <strong>of</strong> Experimental <strong>and</strong> Clinical Pharmacology (V.K., A.H.), Medical University <strong>of</strong> Graz, Graz, Austria, Ludwig Boltzmann Cluster for<br />

Cardiovascular Research (S.P.K., J.W.), Vienna, Austria, Skin <strong>and</strong> Endothelium Research Division (SERD), Department <strong>of</strong> Dermatology (S.R., M.G.),<br />

Medical University <strong>of</strong> Vienna, Vienna, Austria, Core Facility Imaging (S.R., M.G.), Medical University <strong>of</strong> Vienna, Vienna, Austria, Department <strong>of</strong><br />

Vascular Biology <strong>and</strong> Thrombosis Research (C.E.L., R.d.M.), Medical University <strong>of</strong> Vienna, Vienna, Austria, Department <strong>of</strong> Surgery (I.H.), Medical<br />

University <strong>of</strong> Vienna, Vienna, Austria, 3rd Medical Department for Cardiology <strong>and</strong> Emergency Medicine (K.H.), Wilhelminen Hospital, Vienna, Austria.<br />

Correspondence to Johann Wojta, Department <strong>of</strong> Internal Medicine II, Medical University <strong>of</strong> Vienna, Waehringer Guertel 18–20, A-1090 Vienna,<br />

Austria. E-mail johann.wojta@meduniwien.ac.at<br />

© 2011 American Heart Association, Inc.<br />

Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.111.231431<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek 2080der<br />

MedUniWien (IX0000096057) on September 2, 2011


that these endothelial cell adhesion molecules are upregulated<br />

by inflammatory mediators such as interleukin-1<br />

(IL-1) or tumor necrosis factor-� (TNF-�) <strong>and</strong> that their<br />

expression is increased in atherosclerotic lesions. 8–11 In<br />

addition to this process, chemokines such as monocyte<br />

chemoattractant protein-1 (MCP-1) attract activated leukocytes<br />

to the inflammatory site. 12<br />

<strong>Interleukin</strong>-<strong>33</strong> (IL-<strong>33</strong>) is the most recently described<br />

member <strong>of</strong> the IL-1-family <strong>of</strong> cytokines, <strong>and</strong> it is a lig<strong>and</strong><br />

<strong>of</strong> the ST2 receptor. 13 According to the present knowledge,<br />

it is suggested that IL-<strong>33</strong> is specifically released during<br />

necrotic cell death, which is thought to be associated with<br />

tissue damage during infection or trauma, but kept intracellular<br />

during apoptosis. 14,15 Because <strong>of</strong> these properties,<br />

IL-<strong>33</strong> was proposed to act as “alarmin,” as an endogenous<br />

“danger” signal to alert the immune system after infection<br />

or injury. 14,16 Consequently, most studies investigating the<br />

biological role <strong>of</strong> IL-<strong>33</strong> focus on its immunomodulatory<br />

functions. Thus, it was shown that IL-<strong>33</strong> is involved in<br />

polarization <strong>of</strong> T-cells toward the T helper type 2 (Th2)<br />

cell phenotype as well as in activation <strong>of</strong> mast cells,<br />

basophils, eosinophils, <strong>and</strong> natural killer cells. 13,17–19 IL-<strong>33</strong><br />

enhances adhesion, survival, <strong>and</strong> cytokine production <strong>of</strong><br />

human mast cells, eosinophils, <strong>and</strong> basophils. 19–22 Furthermore,<br />

IL-<strong>33</strong> was shown to be involved in the modulation <strong>of</strong><br />

inflammation as it can promote rheumatic <strong>and</strong> airway<br />

inflammatory diseases, anaphylactic shock, <strong>and</strong> inflammatory<br />

<strong>and</strong> fibrotic disorders <strong>of</strong> the gastro-intestinal tract. 23,24<br />

While the effects <strong>of</strong> IL-<strong>33</strong> on the immune system have<br />

been extensively studied, 18,19 the properties <strong>of</strong> this cytokine<br />

in the cardiovascular system are much less investigated.<br />

25,26 IL-<strong>33</strong> <strong>and</strong> ST2 receptor are expressed in human<br />

vein endothelial cells <strong>and</strong> in coronary artery endothelium,<br />

as well as in the thoracic aorta <strong>of</strong> apolipoprotein<br />

E-deficient (ApoE �/� ) mice. 27–29 In such mice, IL-<strong>33</strong> was<br />

also shown to inhibit the progression <strong>of</strong> high-fat dietinduced<br />

atherosclerosis <strong>and</strong> the accumulation <strong>of</strong> foam cells<br />

in the lesion. 28,30 In human endothelial cells, however, IL-<strong>33</strong><br />

was shown to induce inflammatory activation as evidenced by<br />

increased vascular permeability, the increased production <strong>of</strong><br />

inflammatory cytokines, <strong>and</strong> the stimulation <strong>of</strong> angiogenesis. 27,31<br />

In this article, we provide evidence for yet another aspect<br />

<strong>of</strong> inflammatory activation <strong>of</strong> human endothelial cells by<br />

IL-<strong>33</strong> by showing that this cytokine, which we found to be<br />

expressed in human atherosclerotic tissue, stimulates adhesion<br />

<strong>of</strong> leukocytes to the endothelium under both static <strong>and</strong><br />

flow conditions <strong>and</strong> upregulates the expression <strong>of</strong> the adhesion<br />

molecules ICAM-1, VCAM-1, <strong>and</strong> E-selectin <strong>and</strong> <strong>of</strong> the<br />

chemokine MCP-1 in human endothelial cells in vitro.<br />

Furthermore, we demonstrate that the latter effects <strong>of</strong> IL-<strong>33</strong><br />

are also operative in explanted human atherosclerotic plaque<br />

tissue ex vivo.<br />

Methods<br />

Cell Culture<br />

Human umbilical vein endothelial cells (HUVEC) were isolated<br />

from fresh umbilical cords. Human coronary artery endothelial<br />

cells (HCAEC) were isolated from pieces <strong>of</strong> coronary arteries<br />

obtained from patients undergoing heart transplantation. Such<br />

Demyanets et al IL-<strong>33</strong> <strong>Induces</strong> <strong>Adhesion</strong> <strong>Molecules</strong> in Endothelial Cells 2081<br />

endothelial cells were isolated by mild collagenase treatment,<br />

characterized <strong>and</strong> cultivated as described. 32 For some experiments,<br />

HCAEC (Lonza, Verviers, Belgium) were used; cells were<br />

maintained in EGM-2 MV Bullet kit medium (Lonza) with 5%<br />

fetal calf serum (Lonza).<br />

Isolation <strong>of</strong> Human Polymorph-Nuclear<br />

Leukocytes <strong>and</strong> Monocytes<br />

Human polymorph-nuclear leukocytes (PMNL; 99% neutrophils<br />

<strong>and</strong> less than 1% eosinophils) were isolated from heparinized<br />

(100 U/mL) peripheral venous blood <strong>of</strong> healthy donors as<br />

described. <strong>33</strong> For details, please see supplemental material,<br />

available online at http://atvb.ahajournals.org. Human monocytes<br />

were isolated from peripheral blood mononuclear cells by negative<br />

magnetic selection according to manufacturer’s instructions<br />

(Monocyte Isolation Kit II, MACS, Miltenyi Biotec, Bergisch<br />

Gladbach, Germany). The purity <strong>of</strong> the monocyte preparation<br />

was 97%.<br />

<strong>Adhesion</strong> Assay for PMNL <strong>and</strong> Monocytes Under<br />

Static <strong>and</strong> Flow Conditions<br />

PMNL adhesion to HCAEC or HUVEC under static conditions was<br />

measured as described. <strong>33</strong> PMNL or monocyte adhesion to HCAEC<br />

under flow conditions was performed using Venaflux platform<br />

(Cellix, Dublin, Irel<strong>and</strong>). 34–36 For details, please see online supplemental<br />

material.<br />

Tissue Sampling<br />

Atherosclerotic plaques were collected from 35 patients undergoing<br />

carotid endarterectomy. All subjects were Caucasian <strong>and</strong> did not<br />

suffer from acute infection or autoimmune or neoplastic disease. For<br />

details, please see online supplemental material. The study has been<br />

reviewed <strong>and</strong> approved by the Ethic Committee <strong>of</strong> the Medical<br />

University <strong>of</strong> Vienna, Austria, <strong>and</strong> all study subjects gave informed<br />

consent.<br />

Treatment <strong>of</strong> Cells<br />

HCAEC <strong>and</strong> HUVEC were incubated in minimum essential<br />

medium (M199; Sigma) containing 1.25% fetal calf serum<br />

(Lonza) without or with recombinant human IL-<strong>33</strong> (R&D Systems,<br />

Minneapolis, MN) at concentrations between 100 ng/mL<br />

<strong>and</strong> 0.01 ng/mL for time periods between 1 <strong>and</strong> 48 hours. For<br />

soluble receptor inhibition experiments, IL-<strong>33</strong> (1 ng/mL) was<br />

incubated with recombinant human ST2/IL-1 R4 Fc chimera<br />

(sST2-Fc; 5 �g/mL, R&D Systems) for 15 minutes at 37°C before<br />

addition to the cells for 24 hours, as described previously. 37 In<br />

addition, recombinant human immunoglobulin G 1 Fc (IgG 1-Fc; 5<br />

�g/mL, R&D Systems) was used as an isotype control. In order to<br />

determine whether the action <strong>of</strong> IL-<strong>33</strong> is dependent on IL-1 effect,<br />

HUVEC were cultured for 6 hours in the presence <strong>of</strong> IL-<strong>33</strong> (100<br />

ng/mL) or recombinant human IL-1� (200 U/mL; R&D Systems)<br />

with or without IL-1 receptor antagonist (IL-1Ra) (10 �g/mL;<br />

R&D Systems). This concentration <strong>of</strong> IL-1Ra was shown previously<br />

to inhibit IL-1�-induced ICAM-1 <strong>and</strong> VCAM-1 expression<br />

in HUVEC. 38 In additional experiments, the cells were preincubated<br />

for 1 hour with the mitogen-activated protein/extracellular<br />

signal-regulated kinase (MEK) inhibitor U0126 (Promega, Madison,<br />

WI), or the phosphatidylinositol 3-kinase (PI3K) inhibitor<br />

LY-294002 (Calbiochem/Merck, Darmstadt, Germany) at the<br />

indicated concentrations that were similar to concentrations used<br />

by us <strong>and</strong> others in in vitro studies with endothelial cells. 39–42<br />

Thereafter, the cells were treated with IL-<strong>33</strong> at a concentration <strong>of</strong><br />

100 ng/mL for 24 hours (for ICAM-1, VCAM-1, MCP-1 measurement)<br />

or 4 hours (for E-selectin determination). TNF-�<br />

(Roche Diagnostics, Indianapolis, IN) at 10 pM was used as a<br />

positive control in our experiments. The culture supernatants were<br />

collected followed by removal <strong>of</strong> cell debris by centrifugation <strong>and</strong><br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


2082 Arterioscler Thromb Vasc Biol September 2011<br />

stored at �80°C until used. The total cell number was counted<br />

with a hemocytometer after trypsinization.<br />

Flow Cytometry<br />

ICAM-1, VCAM-1, <strong>and</strong> E-selectin expressions at the cell surface<br />

were measured by means <strong>of</strong> flow cytometry (FACS Canto II, Becton<br />

Dickinson, Franklin Lakes, NJ). For details, please see online<br />

supplemental material.<br />

Antigen Determination<br />

MCP-1, IL-6, <strong>and</strong> IL-8 antigen in cell culture supernatants or in<br />

supernatants from stimulated explanted atherosclerotic plaque tissue<br />

was measured by specific ELISAs using monoclonal antibodies (all<br />

from Bender MedSystems, Vienna, Austria).<br />

Total RNA Purification <strong>and</strong> cDNA Preparation<br />

mRNA was isolated using High Pure RNA Tissue Kit (Roche).<br />

Reverse transcription was performed using Transcriptor First Str<strong>and</strong><br />

cDNA Synthesis Kit (Roche). For details, please see online supplemental<br />

material.<br />

RealTime Polymerase Chain Reaction<br />

RealTime-PCR was performed using LightCycler� TaqMan� Master<br />

(Roche) according to the manufacturer’s instructions. For details,<br />

please see online supplemental material.<br />

Nuclear Extraction <strong>and</strong> Analysis <strong>of</strong><br />

NF-�B/DNA Binding<br />

Preparation <strong>of</strong> nuclear extracts was performed using a Nuclear<br />

Extract Kit (Active Motif, Rixensart, Belgium) according to the<br />

manufacturer’s instructions. For details, please see online supplemental<br />

material.<br />

NF-�B Translocation Staining<br />

HCAEC were treated with fresh M199 containing 1.25% fetal calf<br />

serum without or with 1, 10, or 100 ng/mL IL-<strong>33</strong> for 1 hour <strong>and</strong><br />

staining for NF-�B p50 <strong>and</strong> p65 subunits was performed. For details,<br />

please see online supplemental material.<br />

Immun<strong>of</strong>luorescence Analysis <strong>of</strong> IL-<strong>33</strong> <strong>and</strong> ST2 in<br />

Human Atherosclerotic Tissue<br />

Human carotid endatherectomy tissues were fixed in 4% formaldehyde<br />

<strong>and</strong> embedded in paraffin. Sections (5 �m) were deparaffinized<br />

according to st<strong>and</strong>ard procedure <strong>and</strong> then boiled for<br />

antigen retrieval in citrate buffer (DAKO North America, Inc,<br />

CA). The following primary antibodies were used: mouse monoclonal<br />

antibody anti-IL<strong>33</strong> (clone Nessy-1, 1:1000 dilution; Alexis<br />

Biochemicals, Enzo Life Sciences AG, Lausen, Switzerl<strong>and</strong>),<br />

rabbit polyclonal antibody anti-ST2 (IL1RL1) (1:100 dilution;<br />

Sigma), <strong>and</strong> rabbit polyclonal antibody anti-von Willebr<strong>and</strong><br />

factor (1:500 dilution; Dako). For details, please see online<br />

supplemental material.<br />

Adenoviral Infection<br />

HUVEC were infected with adenoviral vectors for overexpression<br />

<strong>of</strong> I�B� (AdV-I�B�) or for overexpression <strong>of</strong> a mutant dominant<br />

negative I�B kinase 2 (AdV-dnIKK2), respectively, as described<br />

previously. 43,44 For details, please see online supplemental<br />

material.<br />

Statistical Analysis<br />

ANOVA followed by Bonferroni-Holm multiple comparisons correction<br />

was carried out for experiments having more than 2 experimental<br />

groups. Dunnett’s post-hoc test was used to compare treated<br />

groups with the reference untreated group. Normally distributed data<br />

were analyzed with t tests in case <strong>of</strong> 2 groups. Mean concentrations<br />

<strong>of</strong> the respective protein for each plaque before <strong>and</strong> after stimulation<br />

with IL-<strong>33</strong> were compared using Wilcoxon signed-rank test for<br />

nonparametric distribution. For mRNA correlation, a Spearman<br />

correlation for nonparametric variables was calculated using SPSS<br />

16.0 (SPSS, Chicago, IL). Values are expressed as mean�SD.<br />

Values <strong>of</strong> P�0.05 were considered significant.<br />

Results<br />

IL-<strong>33</strong> Promotes <strong>Adhesion</strong> <strong>of</strong> Human Leukocytes to<br />

the Monolayer <strong>of</strong> Endothelial Cells<br />

When HCAEC were pretreated with 100 ng/mL IL-<strong>33</strong> for 4<br />

hours, an increase in the number <strong>of</strong> PMNL adhering to the<br />

endothelial cell monolayer was seen already 5, 15, <strong>and</strong>, more<br />

prominently, 30 minutes after the addition <strong>of</strong> PMNL as<br />

compared to untreated control (Figure 1A <strong>and</strong> 1B). Similar<br />

results were also observed with HUVEC (control 5 minutes<br />

2.4�1.7; IL-<strong>33</strong> 5 minutes 8.4�3.7, P�0.05; control 15<br />

minutes 4.8�2.3; IL-<strong>33</strong> 15 minutes 14.2�5.9, P�0.05;<br />

control 30 minutes 5.2�2.6; IL-<strong>33</strong> 30 minutes 42.4�15.7,<br />

P�0.05; values represent numbers <strong>of</strong> PMNL per 5�10 5<br />

square (sq) microns <strong>and</strong> are given as mean�SD <strong>of</strong> 5<br />

pictures, respectively).<br />

Under flow conditions (0.5 dyne/cm 2 for 2 minutes <strong>and</strong> 20<br />

seconds at 37°C), IL-<strong>33</strong> at concentrations <strong>of</strong> 10 ng/mL <strong>and</strong> 100<br />

ng/mL induced statistically significant (P�0.05) adhesion <strong>of</strong><br />

both PMNL <strong>and</strong> monocytes to monolayers <strong>of</strong> HCAEC (Figure<br />

1C <strong>and</strong> 1D). TNF-� at 10 pM, used as a positive control, also<br />

significantly increased adhesion <strong>of</strong> both PMNL (control 8�4,<br />

TNF-� 107�26, P�0.05) <strong>and</strong> monocytes (control 4�2,<br />

TNF-� 10�1, P�0.05) to HCAEC under flow condition. To<br />

address which adhesion molecules might mediate the PMNL<br />

adhesion, we also performed additional experiments with<br />

blocking antibodies against E-selectin, ICAM-1, <strong>and</strong><br />

VCAM-1 alone, or with all three blocking antibodies together.<br />

E-selectin <strong>and</strong> VCAM-1 antibodies significantly<br />

(P�0.05) reduced PMNL adhesion to HCAEC monolayers<br />

which had been activated by 10 ng/mL <strong>of</strong> IL-<strong>33</strong> (Figure 1E<br />

<strong>and</strong> 1F). ICAM-1 antibody also reduced IL-<strong>33</strong>–mediated<br />

PMNL adhesion, however, to a lesser extent than E-selectin<br />

or VCAM-1 antibodies (Figure 1E <strong>and</strong> 1F). If all 3 antibodies<br />

were applied simultaneously, IL-<strong>33</strong>-induced PMNL adhesion<br />

was reduced to the level <strong>of</strong> untreated endothelial cells (Figure<br />

1E <strong>and</strong> 1F). Please see, also, respective movies in the<br />

supplemental data.<br />

IL-<strong>33</strong> Stimulates VCAM-1, ICAM-1, E-Selectin,<br />

<strong>and</strong> MCP-1 <strong>Expression</strong> in Human Endothelial<br />

Cells via ST2 Receptor <strong>and</strong> in IL-1-, MEK-, <strong>and</strong><br />

PI3K-Independent Manner<br />

When HUVEC were treated with IL-<strong>33</strong> at concentrations from<br />

0.01 to 100 ng/mL for 4, 16, 24, <strong>and</strong> 48 hours, a concentrationdependent<br />

upregulation <strong>of</strong> ICAM-1, VCAM-1, <strong>and</strong> MCP-1<br />

protein production was observed at all time points (Figure 2A, B,<br />

D). E-selectin expression was also significantly increased in<br />

these cells, however, only after 4 hours <strong>of</strong> incubation (Figure<br />

2C). TNF-� at 10 pM induced upregulation <strong>of</strong> these adhesion<br />

molecules <strong>and</strong> MCP-1 with similar kinetics in HUVEC<br />

(ICAM-1 [mean fluorescence intensity, MFI], control: 4 hours,<br />

185�9; 16 hours, 239�3; 24 hours, 209�14; 48 hours, 169�9;<br />

TNF-�: 4 hours, 638�107; 16 hours, 2594�42; 24 hours,<br />

2028�160; 48 hours, 1061�49. VCAM-1 [MFI], control: 4<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


Figure 1. <strong>Interleukin</strong> (IL)-<strong>33</strong> promotes adhesion <strong>of</strong> human leukocytes<br />

to monolayers <strong>of</strong> endothelial cells. A, Confluent monolayers <strong>of</strong> human<br />

coronary artery endothelial cells (HCAEC) were preincubated for 4<br />

hours with IL-<strong>33</strong> (100 ng/mL). HBSS (1.0 mL) containing 1�10 6 /mL<br />

PMNL was then added to the endothelial cell monolayer for 5, 15, <strong>and</strong><br />

30 minutes. Respective photomicrographs are shown. B, <strong>Adhesion</strong> <strong>of</strong><br />

polymorph-nuclear leukocytes (PMNL) to monolayers <strong>of</strong> HCAEC was<br />

determined as described in “Methods.” Experiments were performed<br />

twice. Values represent numbers <strong>of</strong> PMNL per 5�10 5 square (sq)<br />

microns <strong>and</strong> are given as mean�SD <strong>of</strong> 10 pictures, respectively. Original<br />

magnification �100. *P�0.05 compared to control. C, HCAEC<br />

were grown on VenaEC biochips <strong>and</strong> preincubated for 4 hours with<br />

IL-<strong>33</strong> (10 or 100 ng/mL). Endothelial monolayers were then superfused<br />

with suspensions <strong>of</strong> 3x10 6 /mL PMNL or monocytes at 0.5<br />

dyne/cm 2 for 2 minutes <strong>and</strong> 20 seconds at 37°C using Venaflux<br />

Nanopump. Part <strong>of</strong> the respective photomicrographs is shown. D,<br />

<strong>Adhesion</strong> <strong>of</strong> PMNL or monocytes to monolayers <strong>of</strong> HCAEC was<br />

determined as described in “Methods.” Experiments were performed<br />

4 times, <strong>and</strong> 10 images were made at each experiment. Values represent<br />

numbers <strong>of</strong> PMNL or monocytes per 5�10 5 sq microns <strong>and</strong> are<br />

given as mean�SD <strong>of</strong> 40 pictures, respectively. Original magnification<br />

�100. *P�0.05 compared to control for PMNL. §P�0.05 compared<br />

to control for monocytes. E, Endothelial cell layers were preincubated<br />

for 4 hours with or without 10 ng/mL <strong>of</strong> IL-<strong>33</strong>. Afterward, the cells<br />

were incubated with blocking antibodies for E-selectin, vascular cell<br />

adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1<br />

(ICAM-1) (10 �g/mL each), or all three antibodies together (5 �g/mL<br />

each), or with an isotype matched control antibody at the same concentrations<br />

for 15 minutes at 37°C followed by superfusion with suspensions<br />

<strong>of</strong> 3�10 6 /mL PMNL at 0.5 dyne/cm 2 for 2 minutes <strong>and</strong> 20<br />

seconds at 37°C. Part <strong>of</strong> the respective photomicrographs is shown.<br />

F, <strong>Adhesion</strong> <strong>of</strong> PMNL to monolayers <strong>of</strong> HCAEC was determined as<br />

described in “Methods.” Experiments were performed 4 times, <strong>and</strong> 10<br />

photos were made at each experiment. Values represent numbers <strong>of</strong><br />

PMNL per 5�10 5 sq microns <strong>and</strong> are given as mean�SD <strong>of</strong> 40 pictures,<br />

respectively. Original magnification �100. *P�0.05 compared to<br />

control. §P�0.05 compared to IL-<strong>33</strong> pretreated cells.<br />

Demyanets et al IL-<strong>33</strong> <strong>Induces</strong> <strong>Adhesion</strong> <strong>Molecules</strong> in Endothelial Cells 2083<br />

Figure 2. Effects <strong>of</strong> interleukin (IL)-<strong>33</strong> on intercellular adhesion<br />

molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-<br />

1), endothelial selectin (E-selectin) <strong>and</strong> monocyte chemoattractant<br />

protein-1 (MCP-1) protein production by HUVEC <strong>and</strong><br />

HCAEC. Confluent monolayers <strong>of</strong> HUVEC were incubated for 4<br />

(224), 16 (�), 24 (X), or 48 hours (�) in the absence or presence<br />

<strong>of</strong> IL-<strong>33</strong> from 0.01 ng/mL to 100 ng/mL. Confluent monolayers<br />

<strong>of</strong> HCAEC were incubated for 4 hours (▫) in the absence or<br />

presence <strong>of</strong> IL-<strong>33</strong> from 0.01 ng/mL to 100 ng/mL. A, E, ICAM-1,<br />

B, F, VCAM-1, <strong>and</strong> C, G, E-selectin expression at the cell surface<br />

was measured by means <strong>of</strong> flow cytometry as described in<br />

“Methods.” D, H, MCP-1 protein was determined in conditioned<br />

media by ELISA as described in Methods.” Each experiment<br />

was performed in triplicates. Values are given in mean fluorescence<br />

intensity (MFI) (A–C, E–G) or in pg/10 4 cells (D, H) <strong>and</strong><br />

represent mean values�SD <strong>of</strong> 3 different experiments. The overall<br />

ANOVA comparing different concentrations for each tested<br />

molecule, <strong>and</strong> time point remained significant after correction<br />

for 6 comparisons (ICAM-1, VCAM-1, E-selectin, MCP-1, IL-6,<br />

IL-8). *P�0.05 compared to control 4 hours; $P�0.05 compared<br />

to control 16 hours; §P�0.05 compared to control 24 hours;<br />

#P�0.05 compared to control 48 hours.<br />

hours, 97�6; 16 hours, 102�6; 24 hours, 91�9; 48 hours,<br />

110�3; TNF-�: 4 hours, 505�109; 16 hours, 412�50; 24<br />

hours, 256�2; 48 hours 169�13. E-selectin [MFI], control: 4<br />

hours, 116�5; 16 hours, 117�2; 24 hours 109�4; 48 hours,<br />

109�5; TNF-�: 4 hours, 1441�318; 16 hours, 124�14; 24<br />

hours, 103�3; 48 hours, 139�6. MCP-1 [pg/10 4 cells], control:<br />

4 hours, 715�59; 16 hours 1039�119; 24 hours 1458�102; 48<br />

hours 3988�507; TNF-� 4 hours, 11 760�981; 16 hours,<br />

54 094�7437; 24 hours, 67 730�1463; 48 hours, 85 696�<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


2084 Arterioscler Thromb Vasc Biol September 2011<br />

8594). In agreement with published data, IL-6 <strong>and</strong> IL-8 protein<br />

production was also increased under IL-<strong>33</strong> treatment in both<br />

endothelial cell types (data not shown). 31<br />

Also in HCAEC, IL-<strong>33</strong> treatment dose-dependently increased<br />

ICAM-1 (Figure 2E), VCAM-1 (Figure 2F),<br />

E-selectin (Figure 2G), <strong>and</strong> MCP-1 protein (Figure 2H) after<br />

4 hours <strong>of</strong> incubation.<br />

As IL-<strong>33</strong> was shown to exert its effects by binding to<br />

cell surface T1/ST2, 13 we examined whether incubation<br />

with the soluble extracellular domain <strong>of</strong> ST2 coupled to the<br />

Fc fragment <strong>of</strong> human IgG 1 (sST2-Fc) could interfere with<br />

the stimulatory effect <strong>of</strong> IL-<strong>33</strong>. Indeed, sST2-Fc, but not<br />

IgG 1-Fc used as an isotype control, significantly inhibited<br />

the production <strong>of</strong> ICAM-1, VCAM-1, E-selectin, <strong>and</strong><br />

MCP-1 protein induced by IL-<strong>33</strong> in human endothelial<br />

cells (Supplemental Figure I). sST2-Fc does not affect<br />

basal production <strong>of</strong> any <strong>of</strong> these proteins (Supplemental<br />

Figure I).<br />

IL-<strong>33</strong> at a concentration <strong>of</strong> 100 ng/mL also robustly<br />

upregulated mRNA specific for ICAM-1, VCAM-1,<br />

E-selectin <strong>and</strong> MCP-1 in HUVEC between 1 hour <strong>and</strong> 9<br />

hours <strong>of</strong> incubation with maximal up-regulation between 3<br />

<strong>and</strong> 6 hours (please see Supplemental Figure II). Similarly,<br />

levels <strong>of</strong> mRNA specific for ICAM-1, VCAM-1,<br />

E-selectin, <strong>and</strong> MCP-1 were increased also in HCAEC<br />

after 9 hours <strong>of</strong> incubation with 100 ng/mL IL-<strong>33</strong><br />

12.0�5.1-fold, 17.9�4.1-fold, 246.2�51.9-fold, <strong>and</strong><br />

9.4�3.7-fold, respectively.<br />

We tested also for the effect <strong>of</strong> IL-<strong>33</strong> treatment on IL-1<br />

expression <strong>and</strong> found that IL-<strong>33</strong> also significantly upregulated<br />

IL-1 mRNA expression in both HUVEC <strong>and</strong> HCAEC<br />

(Supplemental Table I). Maximal upregulation <strong>of</strong> IL-1<br />

mRNA was achieved at 6 hours in HUVEC <strong>and</strong> at 3 hours in<br />

HCAEC (Supplemental Table I). Since ICAM-1, VCAM-1,<br />

E-selectin, <strong>and</strong> MCP-1 production may also depend on the<br />

autocrine action <strong>of</strong> IL-1, we determined the role <strong>of</strong> endogenous<br />

IL-1 in the production <strong>of</strong> these biomolecules by IL-<strong>33</strong>.<br />

We found no inhibitory effect <strong>of</strong> IL-1Ra (10 �g/mL) on<br />

IL-<strong>33</strong>-induced ICAM-1, VCAM-1, E-selectin, <strong>and</strong> MCP-1<br />

mRNA expression in HUVEC. In contrast, when used at the<br />

same concentration IL-1Ra completely inhibited IL-1�induced<br />

mRNA expression <strong>of</strong> these proteins (Supplemental<br />

Table II).<br />

As IL-<strong>33</strong> was previously shown to activate Erk1/2 31 <strong>and</strong><br />

PI3K/Akt 27 pathways in endothelial cells, we tested in additional<br />

experiments whether the MEK inhibitor U0126 or<br />

PI3K inhibitor LY-294002 could modify IL-<strong>33</strong>-induced<br />

ICAM-1, VCAM-1, E-selectin, or MCP-1 protein production.<br />

As shown in Supplemental Table III, neither U0126 at<br />

concentration 1 �mol/L nor LY-294002 at concentration<br />

5 �mol/L abrogated IL-<strong>33</strong>-induced ICAM-1, VCAM-1,<br />

E-selectin, or MCP-1 proteins.<br />

As IL-<strong>33</strong> is known to exert a Th2 polarizing effect, 13 we<br />

also investigated the production <strong>of</strong> the Th2 cytokines IL-4<br />

<strong>and</strong> IL-5 <strong>and</strong> the T regulatory cytokine IL-10 45 in human<br />

endothelial cells after treatment with IL-<strong>33</strong>. As shown in<br />

Supplemental Figure III, IL-<strong>33</strong> at 100 ng/mL did not modulate<br />

the production <strong>of</strong> IL-4, IL-5, or IL-10 in HCAEC at any<br />

time point tested (4, 16, <strong>and</strong> 24 hours).<br />

IL-<strong>33</strong> <strong>Induces</strong> NF-�B Nuclear Translocation <strong>and</strong><br />

IL-<strong>33</strong> Effects on Endothelial Cells<br />

are NF-�B-Dependent<br />

As can be seen in Figure 3, IL-<strong>33</strong> at 100 ng/mL induced<br />

statistically significant (P�0.05) nuclear translocation <strong>of</strong><br />

NF-�B subunits p50 (panel A) <strong>and</strong> p65 (panel B) in human<br />

endothelial cells 15, 30, <strong>and</strong> 60 minutes after incubation. In<br />

HCAEC, IL-<strong>33</strong> at 1 ng/mL (panel D), 10 ng/mL (panel E),<br />

<strong>and</strong> 100 ng/mL (panel F) induced nuclear translocation <strong>of</strong> p50<br />

<strong>and</strong> p65 as compared to untreated cells (panel C). Adenoviral<br />

overexpression <strong>of</strong> dnIKK2 or I�B� in HUVEC abolished the<br />

IL-<strong>33</strong>-induced mRNA expression specific for ICAM-1,<br />

VCAM-1, E-selectin, <strong>and</strong> MCP-1 as compared to control<br />

adenovirus (AdV-GFP) (Table).<br />

IL-<strong>33</strong> <strong>and</strong> ST2 Protein <strong>and</strong> mRNA Are Expressed<br />

in Human Atherosclerotic Plaque <strong>and</strong><br />

Recombinant IL-<strong>33</strong> <strong>Induces</strong> a Proadhesive <strong>and</strong><br />

Proinflammatory State in Explanted<br />

Atherosclerotic Plaque Tissue<br />

As shown on Figure 4 using fluorescence immunohistochemistry,<br />

we detect both IL-<strong>33</strong> <strong>and</strong> ST2 protein in human carotid<br />

endatherectomy specimens. IL-<strong>33</strong> protein is expressed by<br />

endothelial cells as shown by colocalization with von Willebr<strong>and</strong><br />

factor (Figure 4A). Nuclear IL-<strong>33</strong> expression <strong>and</strong><br />

membrane-bound ST2 expression was found on the same<br />

cells (Figure 4B <strong>and</strong> 4C). Using RealTime-PCR, we detected<br />

also mRNA specific for IL-<strong>33</strong> <strong>and</strong> ST2 in human carotid<br />

plaques samples. Moreover, we found a positive correlation<br />

between IL-<strong>33</strong> mRNA <strong>and</strong> ST2 mRNA expression (r�0.566;<br />

P�0.044; Figure 4D) in atherosclerotic tissue. In order to<br />

determine whether the effect <strong>of</strong> IL-<strong>33</strong> on the expression <strong>of</strong><br />

adhesion molecules <strong>and</strong> MCP-1 seen in vitro is reproducible<br />

in an ex vivo situation, we treated explanted human carotid<br />

atherosclerotic plaques with IL-<strong>33</strong> for 24 hours (n�12) to<br />

determine MCP-1, IL-6, <strong>and</strong> IL-8 protein, or for 6 hours<br />

(n�7) to assess ICAM-1, VCAM-1, E-selectin, MCP-1, IL-6,<br />

<strong>and</strong> IL-8 mRNA. As shown in Figure 5, IL-<strong>33</strong> significantly<br />

increased ICAM-1 mRNA (P�0.018), VCAM-1 mRNA<br />

(P�0.028), E-selectin mRNA (P�0.018), MCP-1 mRNA<br />

(P�0.018), <strong>and</strong> IL-6 mRNA (P�0.018) expression as well as<br />

MCP-1 protein (P�0.031), IL-6 protein (P�0.00006) <strong>and</strong><br />

IL-8 protein (P�0.003). IL-8 mRNA level also increased.<br />

However, the difference did not reach statistical significance<br />

(P�0.063, data not shown).<br />

Discussion<br />

Leukocyte interaction with vascular endothelial cells is a<br />

pivotal event in the inflammatory response characteristic for<br />

many pathologies including atherosclerosis. 6 Circulating leukocytes<br />

adhere poorly to healthy endothelium under physiological<br />

conditions. When the endothelium becomes activated,<br />

however, it expresses adhesion molecules that bind cognate<br />

lig<strong>and</strong>s on leukocytes. This switch from a resting, antiadhesive<br />

to an inflammatory activated, adhesive endothelium is a<br />

key event in the development <strong>of</strong> endothelial dysfunction,<br />

which is considered a hallmark in the early pathogenesis <strong>of</strong><br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


Figure 3. <strong>Interleukin</strong> (IL)-<strong>33</strong> induces NF-�B p50 <strong>and</strong> p65 subunit nuclear translocation in human coronary artery <strong>and</strong> umbilical vein endothelial<br />

cells. Confluent monolayers <strong>of</strong> human umbilical vein endothelial cells were incubated for 15, 30, or 60 minutes in the absence<br />

or presence <strong>of</strong> IL-<strong>33</strong> at 100 ng/mL. Preparation <strong>of</strong> nuclear extracts <strong>and</strong> quantification <strong>of</strong> (A) p50 <strong>and</strong> (B) p65 NF-�B subunits were performed<br />

as described in “Methods.” Each experiment was performed in triplicates. Values are given as OD492 nm <strong>and</strong> represent mean<br />

values�SD <strong>of</strong> 2 different experiments. C–F, Confluent monolayers <strong>of</strong> human coronary artery endothelial cells were incubated for 1 hour<br />

in the absence (C) or presence <strong>of</strong> IL-<strong>33</strong> at 1 (D), 10 (E), or 100 ng/mL (F). p50 (in green), p65 (in red) <strong>and</strong> nuclei (in blue) staining was<br />

performed as described in “Methods.” Original magnification �1000. A representative experiment is shown. Experiments were performed<br />

2 times.<br />

atherosclerosis. Selectins are involved in the rolling <strong>and</strong><br />

tethering <strong>of</strong> leukocytes on the vascular wall. ICAM-1 <strong>and</strong><br />

VCAM-1 induce firm adhesion <strong>of</strong> inflammatory cells at the<br />

vascular surface. 5,7 Chemokines such as MCP-1 provide a<br />

Table. AdV-dnIKK2 <strong>and</strong> AdV-I�B� Inhibited <strong>Interleukin</strong>-<strong>33</strong>-Induced<br />

ICAM-1, VCAM-1, E-Selectin, <strong>and</strong> MCP-1 mRNA <strong>Expression</strong>s<br />

in HUVEC<br />

ICAM-1 VCAM-1 E-Selectin MCP-1<br />

No virus 3.4�0.7 9.2�3.8 17.1�0.9 2.5�0.3<br />

AdV-GFP 5.2�1.0 10.1�1.5 17.2�5.4 2.5�0.01<br />

AdV-dnIKK2 1.2�0.1 2.8�0.3 9.3�3.7 1.1�0.2<br />

AdV-I�B� 1.1�0.5 1.6�0.1 4.7�1.5 0.8�0.04<br />

HUVEC were left uninfected or were infected with a recombinant adenovirus<br />

expressing dnIKK2 (AdV-dnIKK2), I�B� (AdV-I�B�) or a control adenovirus<br />

(AdV-GFP) for 4–6 h. 48-h post infection cells were stimulated with<br />

<strong>Interleukin</strong>-<strong>33</strong> (100 ng/mL) for 6 h whereas control cells were left untreated.<br />

mRNA was prepared <strong>and</strong> RealTime-PCR with primers specific for intercellular<br />

adhesion molecule-1 (ICAM-1), vascular cell AM-1 (VCAM-1), endothelial<br />

selectin (E-selectin), monocyte chemoattractant protein-1 (MCP-1), <strong>and</strong> GAPDH<br />

was performed as described in “Methods.” Each experiment was performed in<br />

triplicates. <strong>Adhesion</strong> molecules <strong>and</strong> MCP-1 mRNA levels were normalized<br />

according to the respective GAPDH mRNA level. Values are given as x-fold <strong>of</strong><br />

control, which was set as 1 <strong>and</strong> represent mean values�SD <strong>of</strong> 2 different<br />

experiments. The overall ANOVA for each tested molecule remained significant<br />

after correction for 4 comparisons.<br />

Demyanets et al IL-<strong>33</strong> <strong>Induces</strong> <strong>Adhesion</strong> <strong>Molecules</strong> in Endothelial Cells 2085<br />

chemotactic stimulus to the adherent leukocytes, directing<br />

their diapedesis <strong>and</strong> migration into the vessel wall. 12 Classical<br />

inflammatory mediators such as IL-1 <strong>and</strong> TNF-� were shown<br />

to induce VCAM-1, ICAM-1, <strong>and</strong> E-selectin in human<br />

endothelial cells. 8,9<br />

Here, we show for the first time that the novel IL-1<br />

cytokine family member, IL-<strong>33</strong>, induces rapid adhesion <strong>of</strong><br />

leukocytes to monolayers <strong>of</strong> human endothelial cells isolated<br />

from coronary artery <strong>and</strong> umbilical vein. On further<br />

analysis, we could show upregulation <strong>of</strong> the production <strong>of</strong><br />

the adhesion molecules VCAM-1, ICAM-1, E-selectin,<br />

<strong>and</strong> the chemokine MCP-1 in these endothelial cells by<br />

IL-<strong>33</strong>. The upregulation <strong>of</strong> these adhesion molecules was<br />

concentration-dependent with a significant increase seen at<br />

concentrations between 1 ng/mL <strong>and</strong> 100 ng/mL <strong>of</strong> IL-<strong>33</strong>.<br />

In agreement with a recently published study, we found<br />

that IL-<strong>33</strong> also upregulated the inflammatory cytokines<br />

IL-6 <strong>and</strong> IL-8 in human endothelial cells. 31 In parallel with<br />

increased VCAM-1, ICAM-1, E-selectin, <strong>and</strong> MCP-1 protein<br />

production, IL-<strong>33</strong> caused a marked upregulation <strong>of</strong><br />

specific mRNA for VCAM-1, ICAM-1, E-selectin, <strong>and</strong><br />

MCP-1 in human coronary artery <strong>and</strong> umbilical vein<br />

endothelial cells. The effect was specific for IL-<strong>33</strong> because<br />

sST2-Fc but not IgG 1-Fc abrogated these IL-<strong>33</strong>-induced<br />

effects in endothelial cells. We also showed that IL-<strong>33</strong><br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


2086 Arterioscler Thromb Vasc Biol September 2011<br />

Figure 4. <strong>Expression</strong> <strong>of</strong> interleukin (IL)-<strong>33</strong> <strong>and</strong> ST2 in human<br />

atherosclerotic tissue. A, Staining for IL-<strong>33</strong> <strong>and</strong> von Willebr<strong>and</strong><br />

factor or (B, C) staining for IL-<strong>33</strong> <strong>and</strong> ST2 was performed as<br />

described in “Methods.” Original magnification �200 (A) or<br />

�630 (B, C). Representative pictures are shown. Staining was<br />

representative for 3 different donors. D, RNA was isolated from<br />

carotid endartherectomy lesions (n�13) <strong>and</strong> IL-<strong>33</strong> mRNA<br />

<strong>and</strong> ST2 mRNA <strong>and</strong> mRNA for GAPDH was determined by<br />

RealTime-PCR as described in “Methods.” mRNA levels <strong>of</strong> IL-<strong>33</strong><br />

<strong>and</strong> ST2 were correlated after adjustment for GAPDH.<br />

exerted direct proinflammatory properties on human endothelial<br />

cells without a requirement for intermediate autocrine<br />

or juxtacrine action <strong>of</strong> IL-1�, as the natural antagonist<br />

IL-1Ra, which inhibits IL-1� action by preventing its<br />

binding to specific receptors, did not inhibit IL-<strong>33</strong>-induced<br />

upregulation <strong>of</strong> ICAM-1, VCAM-1, E-selectin, or MCP-1<br />

specific mRNA in these cells. We also show that IL-<strong>33</strong><br />

induces nuclear translocation <strong>of</strong> NF-�B p50 <strong>and</strong> p65<br />

subunits in both types <strong>of</strong> endothelial cells suggesting that<br />

the effects <strong>of</strong> IL-<strong>33</strong> are mediated via the NF-�B pathway.<br />

This notion is further supported by our finding that the<br />

stimulatory effect <strong>of</strong> IL-<strong>33</strong> on adhesion molecule <strong>and</strong><br />

MCP-1 expression is abolished by adenoviral overexpression<br />

<strong>of</strong> I�B <strong>and</strong> dnIKK in these cells. In agreement with<br />

our observations, IL-<strong>33</strong> has been shown to activate NF-�B<br />

in various other cell types such as mast cells, 13,20,46<br />

eosinophils, 47 basophils, 18 CD4 � T-cells 48 <strong>and</strong> rat neonatal<br />

cardiac myocytes <strong>and</strong> fibroblasts. 25 Although IL-<strong>33</strong> was<br />

previously shown to activate Erk1/2 <strong>and</strong> Akt pathways, 27,31<br />

MEK inhibitor U0126 or PI3K inhibitor LY-294002 did<br />

not abrogate induction by IL-<strong>33</strong> <strong>of</strong> any <strong>of</strong> the proteins<br />

tested in our study.<br />

Moreover, we found that IL-<strong>33</strong> <strong>and</strong> ST2 are expressed in<br />

human atherosclerotic carotid tissue on both protein <strong>and</strong><br />

mRNA level. Our study is the first that demonstrated expression<br />

<strong>of</strong> both IL-<strong>33</strong> <strong>and</strong> ST2 in human atherosclerotic tissue.<br />

IL-<strong>33</strong> protein is localized to endothelial cells in human<br />

atherosclerotic tissue. It should be emphasized that a previous<br />

study demonstrated increased IL-<strong>33</strong> expression in the atherosclerotic<br />

aorta <strong>of</strong> ApoE �/� mice fed a high-fat diet as<br />

compared to ApoE �/� mice fed a normal diet <strong>and</strong> to<br />

Figure 5. <strong>Interleukin</strong> (IL)-<strong>33</strong> induces a proadhesive <strong>and</strong> proinflammatory<br />

state in explanted atherosclerotic plaque tissue ex<br />

vivo. A–D, F, Fresh carotid endarterectomy samples (n�7) were<br />

incubated with or without IL-<strong>33</strong> at 100 ng/mL for 6 hours <strong>and</strong><br />

(A) intracellularl adhesion molecule-1 (ICAM-1), (B) vascular cell<br />

adhesion molecule-1 (VCAM-1), (C) endothelial selection<br />

(E-selectin), (D) monocyte chemoattractant protein-1 (MCP-1)<br />

<strong>and</strong> (F) IL-6 mRNA was determined by RealTime-PCR as<br />

described in “Methods.” E, G, H, Fresh carotid endarterectomy<br />

samples (n�12) were incubated with or without IL-<strong>33</strong> at 100<br />

ng/mL for 24 hours <strong>and</strong> E, MCP-1, G, IL-6, <strong>and</strong> H, IL-8 protein<br />

in supernatant was measured by specific ELISA as described in<br />

“Methods.” A–H, Individual values <strong>of</strong> the respective protein for<br />

each carotid endarterectomy sample represent the mean value<br />

<strong>of</strong> 3 independent measurements. Box plots show median (bold<br />

black lines), interquartile range (boxes), <strong>and</strong> values within 1.5<br />

interquartile ranges from the upper or lower edge <strong>of</strong> the box<br />

(whiskers). *P�0.05 compared to control.<br />

wild-type mice. 28 Here, we describe that in human atherosclerotic<br />

plaques nuclear IL-<strong>33</strong> <strong>and</strong> membrane-bound ST2<br />

protein are expressed by the same cells, namely by endothelial<br />

cells, <strong>and</strong> that IL-<strong>33</strong> mRNA significantly correlates with<br />

ST2 mRNA expression in carotid atherosclerotic tissue,<br />

suggesting that both proteins are highly coregulated in this<br />

tissue. Furthermore, ex vivo treatment <strong>of</strong> atherosclerotic<br />

tissue samples with IL-<strong>33</strong> increased the expression <strong>of</strong><br />

ICAM-1, VCAM-1, E-selectin, <strong>and</strong> MCP-1 in these tissue<br />

specimens.<br />

Since the discovery <strong>of</strong> IL-<strong>33</strong> in 2005, numerous immunomodulator<br />

effects <strong>of</strong> this cytokine were described in<br />

different cells. IL-<strong>33</strong> enhances adhesion <strong>and</strong> survival <strong>of</strong><br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


mast cells, eosinophils <strong>and</strong> basophils, as well as release <strong>of</strong><br />

different cytokines from these cells, <strong>and</strong> is a chemoattractant<br />

for Th2 cells in vitro. 19–22,49 Furthermore, it was<br />

shown recently that IL-<strong>33</strong> upregulated cell surface expression<br />

<strong>of</strong> the adhesion molecule ICAM-1 on eosinophils, but<br />

it suppressed that <strong>of</strong> ICAM-3 <strong>and</strong> L-selectin. 47 In an in<br />

vivo experimental sepsis model, IL-<strong>33</strong> treatment increased<br />

neutrophil influx into the peritoneal cavity <strong>and</strong> induced<br />

more efficient bacterial clearance, which was associated<br />

with reduced mortality. 50 In a murine model <strong>of</strong> collageninduced<br />

arthritis, IL-<strong>33</strong> treatment markedly exacerbated<br />

mononuclear <strong>and</strong> polymorphonuclear cell infiltration into<br />

the joint which was associated with disease progression. 51<br />

Taken together, IL-<strong>33</strong> appears to be an important immune<br />

regulator that plays a role in inflammation, especially in<br />

the rapid recruitment <strong>of</strong> certain effector cells into sites <strong>of</strong><br />

ongoing inflammation.<br />

Compared to these effects <strong>of</strong> IL-<strong>33</strong> on the immune<br />

system, data on its contribution to cardiovascular pathophysiology<br />

is scarce. In human endothelial cells, IL-<strong>33</strong><br />

induced inflammatory activation as evidenced by increased<br />

vascular permeability, increased production <strong>of</strong> inflammatory<br />

cytokines, <strong>and</strong> the stimulation <strong>of</strong> angiogenesis. 27,31 A<br />

possible role for IL-<strong>33</strong> in the development <strong>and</strong> progression<br />

<strong>of</strong> atherosclerosis is suggested but not well characterized<br />

yet. IL-<strong>33</strong> was shown to be expressed in cells that are<br />

known to be present in atherosclerotic lesions <strong>and</strong> to<br />

activate such cells. 2,3 For example, IL-<strong>33</strong> was found in<br />

coronary artery endothelium, 29 <strong>and</strong> was shown to induce<br />

the production <strong>of</strong> proinflammatory cytokines from human<br />

mast cells <strong>and</strong> T-cells <strong>and</strong> to enhance lipopolysaccharide<br />

(LPS)-induced TNF-�, IL-6, <strong>and</strong> IL-1� production from<br />

macrophages. 24,52,53 It should be noted that recently generated<br />

IL-<strong>33</strong>-deficient mice showed a substantially diminished<br />

LPS-induced systemic inflammatory response. 54 Furthermore,<br />

IL-<strong>33</strong> mRNA expression was found in human<br />

endothelial cells, coronary artery smooth muscle cells, <strong>and</strong><br />

peripheral blood monocytes <strong>and</strong> leukocytes. 13,28,29,55 In<br />

contrast to our data presented here, which suggest a<br />

detrimental contribution <strong>of</strong> IL-<strong>33</strong> to the early development<br />

<strong>of</strong> atherosclerosis, in ApoE �/� mice fed on a high-fat diet<br />

treatment with IL-<strong>33</strong> had reduced atherosclerotic lesions in<br />

the thoracic aorta via increased IL-5 <strong>and</strong> oxidized low<br />

density lipoprotein auto-antibody production as well as by<br />

decreased macrophage foam cell formation. 28,30 It should<br />

be noted, however, that in these latter studies, which<br />

showed antiatherosclerotic effects <strong>of</strong> long-term administration<br />

(6 weeks) <strong>of</strong> IL-<strong>33</strong>, 1 �g/injection <strong>of</strong> the cytokine<br />

was administered into ApoE �/� mice that already had<br />

developed atherosclerosis. 28,30 IL-<strong>33</strong> administration increased<br />

Th2 cytokines, IL-4, IL-5, <strong>and</strong> IL-13, <strong>and</strong> reduced<br />

the Th1 cytokine IFN-� by lymph node cells in vitro <strong>and</strong> in<br />

serum ex vivo. 28 The level <strong>of</strong> the Th17 cytokine IL-17 was<br />

not different in lymph node cells between IL-<strong>33</strong>- <strong>and</strong><br />

PBS-treated animals, <strong>and</strong> IL-17 <strong>and</strong> IL-10 serum levels<br />

were not detectable. Local cytokine expression in the<br />

vasculature ex vivo was not investigated in this study.<br />

There is strong evidence supporting a proatherogenic role<br />

<strong>of</strong> Th1 cells, whereas a possible contribution <strong>of</strong> the Th2<br />

Demyanets et al IL-<strong>33</strong> <strong>Induces</strong> <strong>Adhesion</strong> <strong>Molecules</strong> in Endothelial Cells 2087<br />

subset in atherosclerotic lesion formation is controversially<br />

discussed. 45,56 The role <strong>of</strong> the Th2 pathway in the<br />

development <strong>of</strong> atherosclerosis seems to be depending on<br />

the stage <strong>and</strong> site <strong>of</strong> the lesion, <strong>and</strong> on the experimental<br />

model. 57 Both anti- <strong>and</strong> proatherogenic effects <strong>of</strong> IL-4, another<br />

prototypical Th2 cytokine, <strong>and</strong> IL-17, a cytokine produced by<br />

Th17 cells, were shown in different studies using different<br />

experimental approaches. 57 It should be noted that Th2 cell<br />

induction is promoted in severe hyperlipidemia, 58 <strong>and</strong> under<br />

these conditions Th2 cytokines could be a target for IL-<strong>33</strong> as<br />

shown in this experimental mouse model <strong>of</strong> atherosclerosis. 28,30<br />

These contradictory findings could be also explained on<br />

the basis <strong>of</strong> different roles for IL-<strong>33</strong> <strong>and</strong> ST2 in distinct<br />

ST2-expressing cells. In support <strong>of</strong> that concept, IL-<strong>33</strong> is able<br />

to enhance the production <strong>of</strong> the Th1 cytokine IFN-� by<br />

natural killer cells <strong>and</strong> invariant cells, 22,59 which are found in<br />

atherosclerotic lesions <strong>and</strong> are thought to be involved in the<br />

pathogenesis <strong>of</strong> atherosclerosis. 60 It is <strong>of</strong> interest that in our<br />

study recombinant human IL-<strong>33</strong> at 100 ng/mL did not affect<br />

the production <strong>of</strong> the Th2 cytokines IL-4 <strong>and</strong> IL-5 <strong>and</strong> the T<br />

regulatory cytokine IL-10 in human coronary artery endothelial<br />

cells.<br />

Upregulation <strong>of</strong> adhesion molecules <strong>and</strong> chemokines in<br />

endothelial cells as shown here by us is, besides an impaired<br />

vasomotor function, a key event in endothelial dysfunction<br />

that is considered to be an early hallmark in the pathogenesis<br />

<strong>of</strong> atherosclerosis. 2,3 It should also be emphasized that in one<br />

<strong>of</strong> the articles mentioned above IL-<strong>33</strong> injection into the mice<br />

enhanced their IL-6 serum levels. 28 IL-6 has been shown to<br />

contribute to development <strong>of</strong> atherosclerotic lesions, <strong>and</strong> its<br />

serum levels correlate with poor prognosis in humans with<br />

cardiovascular disease <strong>and</strong> are an independent predictor <strong>of</strong><br />

sudden death in asymptomatic men. 61–63 Furthermore, it<br />

should be noted that IL-<strong>33</strong> was shown to be associated with<br />

other human inflammatory pathologies such as rheumatoid<br />

arthritis, systemic sclerosis, inflammatory bowel disease,<br />

chronic pancreatitis, asthma, psoriasis, <strong>and</strong> anaphylactic<br />

shock. 23,24 However, the mechanisms <strong>of</strong> the deleterious effects<br />

<strong>of</strong> IL-<strong>33</strong> in these disorders are not completely understood<br />

yet.<br />

In conclusion, we provide evidence for the first time that<br />

the latest member <strong>of</strong> the IL-1 cytokine family, the<br />

“alarmin” IL-<strong>33</strong>, 14,16 is present in human atherosclerotic<br />

lesion <strong>and</strong> stimulates the expression <strong>of</strong> the adhesion<br />

molecules ICAM-1, VCAM-1, E-selectin, <strong>and</strong> the chemokine<br />

MCP-1 in atherosclerotic tissue ex vivo <strong>and</strong> in human<br />

coronary artery <strong>and</strong> umbilical vein endothelial cells in an<br />

IL-1, MEK- <strong>and</strong> PI3K-independent but ST2- <strong>and</strong> NF-�B–<br />

dependent manner in vitro. Given the fact that the expression<br />

<strong>of</strong> ICAM-1, VCAM-1, E-selectin, <strong>and</strong> MCP-1 in<br />

atherosclerotic lesions has been reported to increase during<br />

atherogenesis <strong>and</strong> that this expression seems to be directly<br />

associated with plaque progression, 2,3,10,11 we hypothesize<br />

that IL-<strong>33</strong>, by promoting adhesion molecules <strong>and</strong> proinflammatory<br />

cytokine expression in the endothelium as well<br />

as adhesion <strong>of</strong> blood cells to the endothelium, may<br />

contribute to early events in endothelial dysfunction characteristic<br />

for the development <strong>of</strong> atherosclerotic lesions in<br />

the vessel wall.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


2088 Arterioscler Thromb Vasc Biol September 2011<br />

Sources <strong>of</strong> Funding<br />

This work was supported by a grant from the Austrian Science Fund<br />

to Svitlana Demyanets (FWF-Project number T445-B11) <strong>and</strong> to<br />

Akos Heinemann (P22521-B18), by the Start Funding Program <strong>of</strong><br />

the Medical University <strong>of</strong> Graz to Viktoria Konya (ASO109000101).<br />

Furthermore, the work was supported by the Association for the<br />

Promotion <strong>of</strong> Research in Arteriosclerosis, Thrombosis <strong>and</strong> Vascular<br />

Biology.<br />

None.<br />

Disclosures<br />

References<br />

1. Braunwald E. Shattuck lecture–cardiovascular medicine at the turn <strong>of</strong> the<br />

millennium: triumphs, concerns, <strong>and</strong> opportunities. N Engl J Med. 1997;<br />

<strong>33</strong>7:1360–1369.<br />

2. Ross R. Atherosclerosis–an inflammatory disease. N Engl J Med. 1999;<br />

340:115–126.<br />

3. Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874.<br />

4. Finn AV, Nakano M, Narula J, Kolodgie FD, Virmani R. Concept <strong>of</strong><br />

vulnerable/unstable plaque. Arterioscler Thromb Vasc Biol. 2010;30:<br />

1282–1292.<br />

5. Wagner DD, Frenette PS. The vessel wall <strong>and</strong> its interactions. Blood.<br />

2008;111:5271–5281.<br />

6. Langer HF, Chavakis T. Leukocyte-endothelial interactions in inflammation.<br />

J Cell Mol Med. 2009;13:1211–1220.<br />

7. Galkina E, Ley K. Vascular adhesion molecules in atherosclerosis.<br />

Arterioscler Thromb Vasc Biol. 2007;27:2292–2301.<br />

8. Libby P, Sukhova G, Lee RT, Galis ZS. Cytokines regulate vascular<br />

functions related to stability <strong>of</strong> the atherosclerotic plaque. J Cardiovasc<br />

Pharmacol. 1995;25 Suppl 2:S9–S12.<br />

9. Raab M, Daxecker H, Markovic S, Karimi A, Griesmacher A, Mueller<br />

MM. Variation <strong>of</strong> adhesion molecule expression on human umbilical vein<br />

endothelial cells upon multiple cytokine application. Clin Chim Acta.<br />

2002;321:11–16.<br />

10. O’Brien KD, McDonald TO, Chait A, Allen MD, Alpers CE. Neovascular<br />

expression <strong>of</strong> E-selectin, intercellular adhesion molecule-1, <strong>and</strong> vascular<br />

cell adhesion molecule-1 in human atherosclerosis <strong>and</strong> their relation to<br />

intimal leukocyte content. Circulation. 1996;93:672–682.<br />

11. van der Wal AC, Das PK, Tigges AJ, Becker AE. <strong>Adhesion</strong> molecules on<br />

the endothelium <strong>and</strong> mononuclear cells in human atherosclerotic lesions.<br />

Am J Pathol. 1992;141:1427–14<strong>33</strong>.<br />

12. Schober A. Chemokines in vascular dysfunction <strong>and</strong> remodeling.<br />

Arterioscler Thromb Vasc Biol. 2008;28:1950–1959.<br />

13. Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McClanahan TK,<br />

Zurawski G, Moshrefi M, Qin J, Li X, Gorman DM, Bazan JF, Kastelein<br />

RA. IL-<strong>33</strong>, an interleukin-1-like cytokine that signals via the IL-1<br />

receptor-related protein ST2 <strong>and</strong> induces T helper type 2-associated<br />

cytokines. Immunity. 2005;23:479–490.<br />

14. Lamkanfi M, Dixit VM. IL-<strong>33</strong> raises alarm. Immunity. 2009;31:5–7.<br />

15. Luthi AU, Cullen SP, McNeela EA, Duriez PJ, Afonina IS, Sheridan C,<br />

Brumatti G, Taylor RC, Kersse K, V<strong>and</strong>enabeele P, Lavelle EC, Martin<br />

SJ. Suppression <strong>of</strong> interleukin-<strong>33</strong> bioactivity through proteolysis by apoptotic<br />

caspases. Immunity. 2009;31:84–98.<br />

16. Moussion C, Ortega N, Girard JP. The IL-1-like cytokine IL-<strong>33</strong> is constitutively<br />

expressed in the nucleus <strong>of</strong> endothelial cells <strong>and</strong> epithelial cells<br />

in vivo: a novel ‘alarmin’? PLoS One. 2008;3:e<strong>33</strong>31.<br />

17. Drube S, Heink S, Walter S, Lohn T, Grusser M, Gerbaulet A, Berod L,<br />

Schons J, Dudeck A, Freitag J, Grotha S, Reich D, Rudeschko O,<br />

Norgauer J, Hartmann K, Roers A, Kamradt T. The receptor tyrosine<br />

kinase c-Kit controls IL-<strong>33</strong> receptor signaling in mast cells. Blood. 2010;<br />

115:3899–3906.<br />

18. Pecaric-Petkovic T, Didichenko SA, Kaempfer S, Spiegl N, Dahinden<br />

CA. Human basophils <strong>and</strong> eosinophils are the direct target leukocytes <strong>of</strong><br />

the novel IL-1 family member IL-<strong>33</strong>. Blood. 2009;113:1526–1534.<br />

19. Valent P. <strong>Interleukin</strong>-<strong>33</strong>: a regulator <strong>of</strong> basophils. Blood. 2009;113:<br />

1396–1397.<br />

20. Iikura M, Suto H, Kajiwara N, Oboki K, Ohno T, Okayama Y, Saito H,<br />

Galli SJ, Nakae S. IL-<strong>33</strong> can promote survival, adhesion <strong>and</strong> cytokine<br />

production in human mast cells. Lab Invest. 2007;87:971–978.<br />

21. Suzukawa M, Koketsu R, Iikura M, Nakae S, Matsumoto K, Nagase H,<br />

Saito H, Matsushima K, Ohta K, Yamamoto K, Yamaguchi M.<br />

<strong>Interleukin</strong>-<strong>33</strong> enhances adhesion, CD11b expression <strong>and</strong> survival in<br />

human eosinophils. Lab Invest. 2008;88:1245–1253.<br />

22. Smithgall MD, Comeau MR, Yoon BR, Kaufman D, Armitage R, Smith<br />

DE. IL-<strong>33</strong> amplifies both Th1- <strong>and</strong> Th2-type responses through its<br />

activity on human basophils, allergen-reactive Th2 cells, iNKT <strong>and</strong> NK<br />

cells. Int Immunol. 2008;20:1019–1030.<br />

23. Liew FY, Pitman NI, McInnes IB. Disease-associated functions <strong>of</strong><br />

IL-<strong>33</strong>: the new kid in the IL-1 family. Nat Rev Immunol. 2010;10:<br />

103–110.<br />

24. Oboki K, Ohno T, Kajiwara N, Saito H, Nakae S. IL-<strong>33</strong> <strong>and</strong> IL-<strong>33</strong><br />

receptors in host defense <strong>and</strong> diseases. Allergol Int. 2010;59:143–160.<br />

25. Sanada S, Hakuno D, Higgins LJ, Schreiter ER, McKenzie AN, Lee RT.<br />

IL-<strong>33</strong> <strong>and</strong> ST2 comprise a critical biomechanically induced <strong>and</strong> cardioprotective<br />

signaling system. J Clin Invest. 2007;117:1538–1549.<br />

26. Seki K, Sanada S, Kudinova AY, Steinhauser ML, H<strong>and</strong>a V, Gannon J,<br />

Lee RT. <strong>Interleukin</strong>-<strong>33</strong> prevents apoptosis <strong>and</strong> improves survival after<br />

experimental myocardial infarction through ST2 signaling. Circ Heart<br />

Fail. 2009;2:684–691.<br />

27. Choi YS, Choi HJ, Min JK, Pyun BJ, Maeng YS, Park H, Kim J, Kim<br />

YM, Kwon YG. <strong>Interleukin</strong>-<strong>33</strong> induces angiogenesis <strong>and</strong> vascular permeability<br />

through ST2/TRAF6-mediated endothelial nitric oxide production.<br />

Blood. 2009;114:3117–3126.<br />

28. Miller AM, Xu D, Asquith DL, Denby L, Li Y, Sattar N, Baker AH,<br />

McInnes IB, Liew FY. IL-<strong>33</strong> reduces the development <strong>of</strong> atherosclerosis.<br />

J Exp Med. 2008;205:<strong>33</strong>9–346.<br />

29. Bartunek J, Delrue L, Van Durme F, Muller O, Casselman F, De Wiest<br />

B, Croes R, Verstreken S, Goethals M, de Raedt H, Sarma J, Joseph L,<br />

V<strong>and</strong>erheyden M, Weinberg EO. Nonmyocardial production <strong>of</strong> ST2<br />

protein in human hypertrophy <strong>and</strong> failure is related to diastolic load. JAm<br />

Coll Cardiol. 2008;52:2166–2174.<br />

30. McLaren JE, Michael DR, Salter RC, Ashlin TG, Calder CJ, Miller AM,<br />

Liew FY, Ramji DP. IL-<strong>33</strong> reduces macrophage foam cell formation.<br />

J Immunol. 2010;185:1222–1229.<br />

31. Aoki S, Hayakawa M, Ozaki H, Takezako N, Obata H, Ibaraki N, Tsuru<br />

T, Tominaga S, Yanagisawa K. ST2 gene expression is proliferationdependent<br />

<strong>and</strong> its lig<strong>and</strong>, IL-<strong>33</strong>, induces inflammatory reaction in endothelial<br />

cells. Mol Cell Biochem. 2010;<strong>33</strong>5:75–81.<br />

32. Wojta J, Zoellner H, Gallicchio M, Hamilton JA, McGrath K. Gammainterferon<br />

counteracts interleukin-1 alpha stimulated expression <strong>of</strong><br />

urokinase-type plasminogen activator in human endothelial cells in vitro.<br />

Biochem Biophys Res Commun. 1992;188:463–469.<br />

<strong>33</strong>. Zhang WJ, Hufnagl P, Binder BR, Wojta J. Antiinflammatory activity<br />

<strong>of</strong> astragaloside IV is mediated by inhibition <strong>of</strong> NF-kappaB activation<br />

<strong>and</strong> adhesion molecule expression. Thromb Haemost. 2003;90:<br />

904–914.<br />

34. Philipose S, Konya V, Sreckovic I, Marsche G, Lippe IT, Peskar BA,<br />

Heinemann A, Schuligoi R. The prostagl<strong>and</strong>in E2 receptor EP4 is<br />

expressed by human platelets <strong>and</strong> potently inhibits platelet aggregation<br />

<strong>and</strong> thrombus formation. Arterioscler Thromb Vasc Biol. 2010;30:<br />

2416–2423.<br />

35. Choi EY, Chavakis E, Czabanka MA, Langer HF, Fraemohs L, Economopoulou<br />

M, Kundu RK, Orl<strong>and</strong>i A, Zheng YY, Prieto DA, Ballantyne<br />

CM, Constant SL, Aird WC, Papayannopoulou T, Gahmberg<br />

CG, Udey MC, Vajkoczy P, Quertermous T, Dimmeler S, Weber C,<br />

Chavakis T. Del-1, an endogenous leukocyte-endothelial adhesion<br />

inhibitor, limits inflammatory cell recruitment. Science. 2008;322:<br />

1101–1104.<br />

36. Konya V, Philipose S, Balint Z, Olschewski A, Marsche G, Sturm EM,<br />

Schicho R, Peskar BA, Schuligoi R, Heinemann A. Interaction <strong>of</strong> eosinophils<br />

with endothelial cells is modulated by prostagl<strong>and</strong>in EP4 receptors.<br />

Eur J Immunol. 2011.<br />

37. Palmer G, Lipsky BP, Smithgall MD, Meininger D, Siu S, Talabot-Ayer<br />

D, Gabay C, Smith DE. The IL-1 receptor accessory protein (AcP) is<br />

required for IL-<strong>33</strong> signaling <strong>and</strong> soluble AcP enhances the ability <strong>of</strong><br />

soluble ST2 to inhibit IL-<strong>33</strong>. Cytokine. 2008;42:358–364.<br />

38. Kaplanski G, Marin V, Fabrigoule M, Boulay V, Benoliel AM, Bongr<strong>and</strong><br />

P, Kaplanski S, Farnarier C. Thrombin-activated human endothelial cells<br />

support monocyte adhesion in vitro following expression <strong>of</strong> intercellular<br />

adhesion molecule-1 (ICAM-1; CD54) <strong>and</strong> vascular cell adhesion<br />

molecule-1 (VCAM-1; CD106). Blood. 1998;92:1259–1267.<br />

39. Rychli K, Kaun C, Hohensinner PJ, Rega G, Pfaffenberger S, Vyskocil E,<br />

Breuss JM, Furnkranz A, Uhrin P, Zaujec J, Niessner A, Maurer G, Huber<br />

K, Wojta J. The inflammatory mediator oncostatin M induces angiopoietin<br />

2 expression in endothelial cells in vitro <strong>and</strong> in vivo. J Thromb<br />

Haemost. 2010;8:596–604.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


40. Jain S, Gabunia K, Kelemen SE, Panetti TS, Autieri MV. The antiinflammatory<br />

cytokine interleukin 19 is expressed by <strong>and</strong> angiogenic for<br />

human endothelial cells. Arterioscler Thromb Vasc Biol. 2011;31:<br />

167–175.<br />

41. Hatipoglu OF, Hirohata S, Cilek MZ, Ogawa H, Miyoshi T, Obika M,<br />

Demircan K, Shinohata R, Kusachi S, Ninomiya Y. ADAMTS1 is a<br />

unique hypoxic early response gene expressed by endothelial cells. J Biol<br />

Chem. 2009;284:16325–16<strong>33</strong>3.<br />

42. Hermann C, Assmus B, Urbich C, Zeiher AM, Dimmeler S. Insulinmediated<br />

stimulation <strong>of</strong> protein kinase Akt: A potent survival signaling<br />

cascade for endothelial cells. Arterioscler Thromb Vasc Biol. 2000;20:<br />

402–409.<br />

43. Oitzinger W, H<strong>of</strong>er-Warbinek R, Schmid JA, Koshelnick Y, Binder BR,<br />

de Martin R. Adenovirus-mediated expression <strong>of</strong> a mutant IkappaB<br />

kinase 2 inhibits the response <strong>of</strong> endothelial cells to inflammatory stimuli.<br />

Blood. 2001;97:1611–1617.<br />

44. Wrighton CJ, H<strong>of</strong>er-Warbinek R, Moll T, Eytner R, Bach FH, de Martin<br />

R. Inhibition <strong>of</strong> endothelial cell activation by adenovirus-mediated<br />

expression <strong>of</strong> I kappa B alpha, an inhibitor <strong>of</strong> the transcription factor<br />

NF-kappa B. J Exp Med. 1996;183:1013–1022.<br />

45. Hansson GK, Libby P. The immune response in atherosclerosis: a<br />

double-edged sword. Nat Rev Immunol. 2006;6:508–519.<br />

46. Pushparaj PN, Tay HK, H’Ng SC, Pitman N, Xu D, McKenzie A, Liew<br />

FY, Melendez AJ. The cytokine interleukin-<strong>33</strong> mediates anaphylactic<br />

shock. Proc Natl Acad Sci U S A. 2009;106:9773–9778.<br />

47. Chow JY, Wong CK, Cheung PF, Lam CW. Intracellular signaling<br />

mechanisms regulating the activation <strong>of</strong> human eosinophils by the novel<br />

Th2 cytokine IL-<strong>33</strong>: implications for allergic inflammation. Cell Mol<br />

Immunol. 2010;7:26–34.<br />

48. Kurowska-Stolarska M, Kewin P, Murphy G, Russo RC, Stolarski B,<br />

Garcia CC, Komai-Koma M, Pitman N, Li Y, Niedbala W, McKenzie<br />

AN, Teixeira MM, Liew FY, Xu D. IL-<strong>33</strong> induces antigen-specific IL-5�<br />

T cells <strong>and</strong> promotes allergic-induced airway inflammation independent<br />

<strong>of</strong> IL-4. J Immunol. 2008;181:4780–4790.<br />

49. Komai-Koma M, Xu D, Li Y, McKenzie AN, McInnes IB, Liew FY.<br />

IL-<strong>33</strong> is a chemoattractant for human Th2 cells. Eur J Immunol. 2007;<br />

37:2779–2786.<br />

50. Alves-Filho JC, Sonego F, Souto FO, Freitas A, Verri WA Jr,<br />

Auxiliadora-Martins M, Basile-Filho A, McKenzie AN, Xu D, Cunha FQ,<br />

Liew FY. <strong>Interleukin</strong>-<strong>33</strong> attenuates sepsis by enhancing neutrophil influx<br />

to the site <strong>of</strong> infection. Nat Med. 2010;16:708–712.<br />

51. Xu D, Jiang HR, Kewin P, Li Y, Mu R, Fraser AR, Pitman N, Kurowska-<br />

Stolarska M, McKenzie AN, McInnes IB, Liew FY. IL-<strong>33</strong> exacerbates<br />

antigen-induced arthritis by activating mast cells. Proc Natl Acad Sci<br />

USA. 2008;105:10913–10918.<br />

Demyanets et al IL-<strong>33</strong> <strong>Induces</strong> <strong>Adhesion</strong> <strong>Molecules</strong> in Endothelial Cells 2089<br />

52. Espinassous Q, Garcia-de-Paco E, Garcia-Verdugo I, Synguelakis M, von<br />

Aulock S, Sallenave JM, McKenzie AN, Kanellopoulos J. IL-<strong>33</strong> enhances<br />

lipopolysaccharide-induced inflammatory cytokine production from<br />

mouse macrophages by regulating lipopolysaccharide receptor complex.<br />

J Immunol. 2009;183:1446–1455.<br />

53. Ohno T, Oboki K, Morita H, Kajiwara N, Arae K, Tanaka S, Ikeda M,<br />

Iikura M, Akiyama T, Inoue J, Matsumoto K, Sudo K, Azuma M,<br />

Okumura K, Kamradt T, Saito H, Nakae S. Paracrine IL-<strong>33</strong> stimulation<br />

enhances lipopolysaccharide-mediated macrophage activation. PLoS<br />

One. 2011;6:e18404.<br />

54. Oboki K, Ohno T, Kajiwara N, Arae K, Morita H, Ishii A, Nambu A, Abe<br />

T, Kiyonari H, Matsumoto K, Sudo K, Okumura K, Saito H, Nakae S.<br />

IL-<strong>33</strong> is a crucial amplifier <strong>of</strong> innate rather than acquired immunity. Proc<br />

Natl Acad Sci U S A. 2010;107:18581–18586.<br />

55. Nile CJ, Barksby E, Jitprasertwong P, Preshaw PM, Taylor JJ. <strong>Expression</strong><br />

<strong>and</strong> regulation <strong>of</strong> interleukin-<strong>33</strong> in human monocytes. Immunology. 2010;<br />

130:172–180.<br />

56. Lahoute C, Herbin O, Mallat Z, Tedgui A. Adaptive immunity in atherosclerosis:<br />

mechanisms <strong>and</strong> future therapeutic targets. Nat Rev Cardiol.<br />

2011;8:348–358.<br />

57. Ait-Oufella H, Taleb S, Mallat Z, Tedgui A. Recent advances on the role<br />

<strong>of</strong> cytokines in atherosclerosis. Arterioscler Thromb Vasc Biol. 2011;31:<br />

969–979.<br />

58. Zhou X, Paulsson G, Stemme S, Hansson GK. Hypercholesterolemia is<br />

associated with a T helper (Th) 1/Th2 switch <strong>of</strong> the autoimmune response<br />

in atherosclerotic apo E-knockout mice. J Clin Invest. 1998;101:<br />

1717–1725.<br />

59. Bourgeois E, Van LP, Samson M, Diem S, Barra A, Roga S, Gombert JM,<br />

Schneider E, Dy M, Gourdy P, Girard JP, Herbelin A. The pro-Th2<br />

cytokine IL-<strong>33</strong> directly interacts with invariant NKT <strong>and</strong> NK cells to<br />

induce IFN-gamma production. Eur J Immunol. 2009;39:1046–1055.<br />

60. Perrins CJ, Bobryshev YV. Current advances in underst<strong>and</strong>ing <strong>of</strong> immunopathology<br />

<strong>of</strong> atherosclerosis. Virchows Arch. 2011;458:117–123.<br />

61. Huber SA, Sakkinen P, Conze D, Hardin N, Tracy R. <strong>Interleukin</strong>-6<br />

exacerbates early atherosclerosis in mice. Arterioscler Thromb Vasc Biol.<br />

1999;19:2364–2367.<br />

62. Rauchhaus M, Doehner W, Francis DP, Davos C, Kemp M, Liebenthal C,<br />

Niebauer J, Hooper J, Volk HD, Coats AJ, Anker SD. Plasma cytokine<br />

parameters <strong>and</strong> mortality in patients with chronic heart failure. Circulation.<br />

2000;102:3060–3067.<br />

63. Empana JP, Jouven X, Canoui-Poitrine F, Luc G, Tafflet M, Haas B,<br />

Arveiler D, Ferrieres J, Ruidavets JB, Montaye M, Yarnell J, Morange P,<br />

Kee F, Evans A, Amouyel P, Ducimetiere P. C-reactive protein, interleukin<br />

6, fibrinogen <strong>and</strong> risk <strong>of</strong> sudden death in European middle-aged<br />

men: the PRIME study. Arterioscler Thromb Vasc Biol. 2010;30:<br />

2047–2052.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


Supplemental Material<br />

Demyanets et al.<br />

� Supplemental Methods<br />

� Supplemental Tables I, II, III<br />

� Supplemental Figures I, II, III<br />

Supplemental Methods<br />

Isolation <strong>of</strong> human polymorph-nuclear leukocytes (PMNL)<br />

Human polymorph-nuclear leukocytes (PMNL; 99% neutrophils, less than 1% eosinophils)<br />

were isolated from heparinized (100 U/mL) peripheral venous blood <strong>of</strong> healthy donors as<br />

described. 1 Red blood cells were sedimented with 6% dextran (Pharmacia, Uppsala, Sweden)<br />

in Hank’s balanced salt solution (HBSS; Sigma, St. Louis, MO, USA) for 60 min at room<br />

temperature, leukocyte-rich plasma was collected <strong>and</strong> PMNL were then isolated by<br />

centrifugation, washed <strong>and</strong> resuspended to 1x10 6 cells/mL in RPMI-1640 (Sigma) with 10<br />

mmol/L HEPES (Sigma).<br />

<strong>Adhesion</strong> assay under static conditions<br />

PMNL adhesion to HCAEC or HUVEC was measured as described. 1 Briefly, endothelial cells<br />

were plated in gelatine coated 24-well plates <strong>and</strong> allowed to grow to confluence. The medium<br />

was removed <strong>and</strong> cells were then incubated with 1.0 mL/well <strong>of</strong> minimum essential medium<br />

(M199; Sigma) containing 20% fetal calf serum (FCS, Lonza, Verviers, Belgium) with or<br />

without recombinant human (rh) IL-<strong>33</strong> (R&D Systems; Minneapolis, MN). After 4 h <strong>of</strong><br />

incubation the culture supernatant <strong>of</strong> such treated cells was removed <strong>and</strong> the cells were gently<br />

washed three times with M199. 1.0 mL <strong>of</strong> HBSS containing 1 x 10 6 PMNL was then added to<br />

the endothelial cell monolayer in each well. The binding phase <strong>of</strong> the assay was performed at<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


37°C in a 5% CO2 atmosphere for 5, 15 <strong>and</strong> 30 min. Thereafter, the wells were washed with<br />

HBSS (1.0 mL/well) three times <strong>and</strong> all wells were examined under microscope in order to<br />

determine whether loss <strong>of</strong> endothelial cells had occurred during incubation or washing. An<br />

Olympus IMT 2 microscope was used for light microscopy with 10x lens with numeric<br />

apertures <strong>of</strong> 0.30. A Canon EOS-400 camera was used to acquire images.<br />

<strong>Adhesion</strong> assay under flow conditions<br />

HCAEC (4.3×10 5 /substrate) were grown on VenaEC biochips (Cellix, Dublin, Irel<strong>and</strong>). After<br />

reaching confluence (usually within two days) endothelial layers were incubated with fresh<br />

M199 (Sigma) containing 20% FCS (Lonza) with or without rh IL-<strong>33</strong> (10 or 100 ng/mL) for 4<br />

h. Endothelial monolayers were superfused with suspensions <strong>of</strong> 3x10 6 /mL PMNL or<br />

monocytes at 0.5 dyne/cm 2 for 2 min <strong>and</strong> 20 seconds at 37°C in the OKOLAB H201-T1<br />

heated cage. PMNL <strong>and</strong> monocytes were preconditioned in endothelial cells media for 5 min<br />

at 37°C prior to the flow experiment. In some experiments the endothelial layers were pre-<br />

incubated with blocking antibodies for E-selectin, VCAM-1, ICAM-1 (10 µg/mL each; R&D<br />

Systems) or all three antibodies together (5 µg/mL each) or with an isotype matched control<br />

antibody at the same concentrations for 15 min at 37°C. Cell adhesion was monitored by<br />

phase contrast on a Zeiss Axiovert 40 CFL microscope <strong>and</strong> a Zeiss A-Plan 10x/0.25 Ph1 lens,<br />

using Hamamatsu ORCA-03G digital camera <strong>and</strong> VenaFlux s<strong>of</strong>tware (Cellix). Computerized<br />

image analysis was performed by DucoCell analysis s<strong>of</strong>tware (Cellix), where adherent cells<br />

were quantified on each single image. 2-4<br />

Tissue sampling<br />

Atherosclerotic plaques were collected from 35 patients undergoing carotid endarterectomy.<br />

All subjects were Caucasian <strong>and</strong> did not suffer from acute infection, autoimmune or<br />

neoplastic disease. The carotid endarterectomy samples for IL-<strong>33</strong> mRNA <strong>and</strong> ST2 mRNA<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


analysis (n=13, 85% male, mean age 70 years, 38% symptomatic) were snap-frozen in liquid<br />

nitrogen in the surgery room <strong>and</strong> stored at -80°C until RNA extraction. The carotid<br />

endarterectomy tissue for immun<strong>of</strong>luorescence analysis (n=3, 67% male, mean age 72 years,<br />

<strong>33</strong>% symptomatic) were fixed in 4% formalin <strong>and</strong> embedded in paraffin. The carotid<br />

endarterectomy samples (n=19), which were used for culture experiments, were collected in<br />

sterile tubes with M199 containing 5% FCS. These fresh tissues were cut into small pieces,<br />

<strong>and</strong> for each plaque, equal numbers <strong>of</strong> nonadjacent plaque pieces were r<strong>and</strong>omly distributed<br />

into wells <strong>of</strong> a 24-well plate filled with M199 that contained 5% FCS as described<br />

previously. 5 They were stimulated with 100 ng/mL rh IL-<strong>33</strong> for 24 h (n=12, 67% male, mean<br />

age 65 years, 42% symptomatic) for MCP-1, IL-6 <strong>and</strong> IL-8 protein determination or for 6 h<br />

(n=7, 100% male, mean age 69 years, 17% symptomatic) for ICAM-1, VCAM-1, E-selectin,<br />

MCP-1, IL-6 <strong>and</strong> IL-8 mRNA measurement. Stimulations were done in triplicates. After<br />

incubation, tissue was shock-frozen for RNA isolation or culture supernatants were collected<br />

for further analysis by ELISA.<br />

Flow cytometry<br />

ICAM-1, VCAM-1 <strong>and</strong> E-selectin expressions at the cell surface were measured by means <strong>of</strong><br />

flow cytometry (FACS Canto II, Becton Dickinson, Franklin Lakes, NJ, USA). HUVEC <strong>and</strong><br />

HCAEC were gently detached by detachment buffer (25 mM HEPES (Boehringer Mannheim<br />

GmbH, Germany), 10 mM EDTA (Pierce, Rockford, IL) in Dulbecco’s PBS without calcium<br />

<strong>and</strong> magnesium (PAA, Pasching, Austria)). Endothelial cells were incubated for 30 min at<br />

4°C in dark with primary antibodies against ICAM-1 FITC (mouse anti-human CD54;<br />

Beckman Coulter, Brea, CA, USA), VCAM-1 (PE-Cy TM 5 mouse anti-human CD106; BD<br />

Pharmingen, San Jose CA, USA) <strong>and</strong> E-selectin (PE mouse anti-human CD62E; BD<br />

Pharmingen) which were diluted 1:40 or with respective isotype-matched control antibodies<br />

in antibody diluents solution (DAKO North America, Inc., CA, USA; catalogue number<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


S3022). After the cells were washed once with PBS <strong>and</strong> were resuspended in fixative solution<br />

(FACS flow solution, distilled water, BD CellFix TM ), flow cytometric analysis was performed<br />

by FACS Diva s<strong>of</strong>tware (Becton Dickinson). For IL-4, IL-5, IL-10 determination supernatants<br />

were collected from HCAEC incubated in the absence or presence <strong>of</strong> 100 ng/ml IL-<strong>33</strong> for 4,<br />

16 <strong>and</strong> 24 h, respectively. Supernatants were processed per manufacturer`s instruction (BD<br />

Cytometric Bead Array, Human Th1/Th2 cytokine kit). In detail, 25µl conditioned<br />

supernatant was added to 25µl mixed beads (each specific for IL-4, IL-5, IL-10) <strong>and</strong> 25µl PE<br />

detection reagent. The mixture was incubated for 3 h at room temperature in the dark. The<br />

beads were washed <strong>and</strong> analyzed immediately by flow cytometry. Mean fluorescence<br />

intensities (MFI) for treated endothelial cells were compared to the MFI <strong>of</strong> unstimulated cells.<br />

Total RNA purification <strong>and</strong> cDNA preparation<br />

Cells were treated as described, supernatants were removed <strong>and</strong> total cellular RNA was<br />

isolated using High Pure RNA Isolation Kit (Roche, Basel, Switzerl<strong>and</strong>) according to the<br />

manufacturer’s instructions. For IL-<strong>33</strong> mRNA <strong>and</strong> ST2 mRNA measurement, three<br />

representative samples (each 25 mg <strong>of</strong> wt) were collected from each carotid artery<br />

lesion.Frozen tissue was homogenized using a ball mill (Retsch, Haan, Germany), <strong>and</strong> mRNA<br />

was isolated usingHigh Pure RNA Tissue Kit (Roche). Reverse transcription was performed<br />

using Transcriptor First Str<strong>and</strong> cDNA Synthesis Kit (Roche).<br />

RealTime Polymerase Chain Reaction<br />

RealTime-PCR was performed using LightCycler® TaqMan® Master (Roche) according to<br />

the manufacturer’s instructions. Primers were designed using the Roche Universal<br />

ProbeLibrary Assay Design Centre (http://www.universalprobelibrary.com/): GAPDH<br />

(forward primer: 5’-agccacatcgctcagacac-3’, reverse primer: 5’-gcccaatacgaccaaatcc-3’,<br />

UPLprobe #60; Amplicon Size [bp] 66) – ST2 (forward primer: 5’-ttgtcctaccattgacctctacaa-3’,<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


everse primer: 5’-gatccttgaagagcctgacaa-3’, UPLprobe #56; Amplicon Size [bp] 75) – IL-<strong>33</strong><br />

(forward primer: 5’-agcaaagtggaagaacacagc-3’, reverse primer: 5’-cttctttggccttctgttgg-3’,<br />

UPL probe #<strong>33</strong>, Amplicon Size [bp] 74) – VCAM-1 (forward primer: 5’-<br />

tgaatctaggaaattggaaaaagg-3’, reverse primer: 5’- tgaatctctggatccttaggaaa -3’, UPLprobe #39;<br />

Amplicon Size [bp] 69) – ICAM-1 (forward primer: 5’-ccttcctcaccgtgtactgg-3’, reverse<br />

primer: 5’-agcgtagggtaaggttcttgc-3’, UPLprobe #71; Amplicon Size [bp] 90) – E-<br />

selectin(forward primer: 5’ accagcccaggttgaatg-3’, reverse primer: 5’- ggttggacaaggctgtgc-3’,<br />

UPLprobe #86; Amplicon Size [bp] 89) –MCP-1 (forward primer: 5’-ttctgtgcctgctgctcat-3’,<br />

reverse primer: 5’-ggggcattgattgcatct-3’, UPLprobe #83; Amplicon Size [bp] 73) – IL-6<br />

(forward primer: 5’-gatgagtacaaaagtcctgatcca-3’, reverse primer: 5’-ctgcagccactggttctgt-3’,<br />

UPLprobe #40; Amplicon Size [bp] 130) – IL-8 (forward primer: 5’-agacagcagagcacacaagc-<br />

3’, reverse primer: 5’-atggttccttccggtggt-3’, UPLprobe #72; Amplicon Size [bp] 62). The<br />

amplification conditions consisted <strong>of</strong> an initial incubation at 95°C for 10 min, followed by 45<br />

cycles <strong>of</strong> 95°C for 10 sec, 63°C for 20 sec <strong>and</strong> 72°C for 6 sec <strong>and</strong> a final cooling to 40°C.<br />

Data was analysed using LightCycler S<strong>of</strong>tware Version 3.5 (Roche).<br />

Nuclear extraction <strong>and</strong> analysis <strong>of</strong> NF-κB/DNA binding<br />

Endothelial cells were incubated for 15, 30 or 60 min in 1.25% FCS with or without rh IL-<strong>33</strong><br />

at a concentration <strong>of</strong> 100 ng/mL. Preparation <strong>of</strong> nuclear extracts was performed using a<br />

Nuclear Extract Kit (Active Motif, Rixensart, Belgium) according to the manufacturer’s<br />

instructions. Quantitation <strong>of</strong> p50 <strong>and</strong> p65 NF-κB subunits in nuclear extracts <strong>of</strong> such treated<br />

cells was performed using the ELISA-based TransAM TM NF-κB Family kit (Active Motif,<br />

Rixensart, Belgium) as described previously. 6<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


NF-κB translocation staining<br />

HCAEC were seeded on Permanox chamber slides (Nunc Inc., Neparville, IL, USA).<br />

Confluent monolayers were treated with fresh M199 containing 1.25% FCS without or with 1,<br />

10 or 100 ng/mL IL-<strong>33</strong> for 1 h. Monolayers were washed with PBS <strong>and</strong> fixed with 3.7%<br />

paraformaldehyde. The cells were washed again <strong>and</strong> then permeabilized with 0.1% Triton X-<br />

100 (Sigma) in PBS for 20 min at room temperature, washed in PBS, <strong>and</strong> blocked with 5%<br />

bovine serum albumin (BSA; Sigma) in PBS for 30 min. Subsequently, cells were incubated<br />

for 1 h at room temperature with a goat polyclonal anti-human p50 antibody(Santa Cruz, CA,<br />

USA) at a dilution <strong>of</strong> 1:100 or goat IgG (R&D Systems) as an isotype control in diluent<br />

solution for primary antibody (DAKO, catalogue number S3022). After washing, a 1:200<br />

dilution <strong>of</strong> afluorescein (FITC)-conjugated mouse anti-goat antibody (Jackson<br />

ImmunoResearch Laboratories, West Grove, PA, USA)in diluent solution for secondary<br />

antibody (DAKO, catalogue number S0809) was incubated with the cells for 1 h at room<br />

temperature in the dark. After washing, the cells were incubated for 1 h with a rabbit<br />

polyclonal anti-human NF-κB p65 (Santa Cruz) at 1:100 or rabbit IgG (R&D Systems)<br />

followed by washing step <strong>and</strong> 1 h incubation with Texas Red dye-conjugated donkey anti-<br />

rabbit antibody (Jackson) at 1:200 in the dark.After final washes, cells were mounted using<br />

Vectashield mounting medium with 4',6-diamidino-2-phenylindole (DAPI;Vector<br />

Laboratories Inc., Burlingame, CA, USA) <strong>and</strong> sealed with nail polish. Fluorescence was<br />

assessed by microscopy with Zeiss Axio Imager.M2 with 100x lens (numeric aperture 0.9)<br />

with immersion oil (Imersol TM , Zeiss, Oberkochen, Germany) using AxioVision Rel. 4.8<br />

s<strong>of</strong>tware.<br />

Immun<strong>of</strong>luorescence analysis <strong>of</strong> IL-<strong>33</strong> <strong>and</strong> ST2 in human atherosclerotic tissue<br />

Primary antibodies, mouse monoclonal anti-IL<strong>33</strong> antibody (clone Nessy-1, 1:1000 dilution;<br />

Alexia Biochemicals, Enzo Life Sciences AG, Lausen, Switzerl<strong>and</strong>), rabbit polyclonal anti-<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


ST2 antibody (IL1RL1) (1:100 dilution; Sigma) <strong>and</strong> rabbit polyclonal antibody anti-von<br />

Willebr<strong>and</strong> factor (1:500 dilution; Dako), were incubated overnight at 4°C. After extensive<br />

washing in PBS, slides were incubated with secondary antibodies for 2 h at room temperature<br />

in the dark. Secondary antibodies were Alexa Fluor-488 goat anti-mouse IgG (Invitrogen-<br />

Molecular Probes), Alexa Fluor-546 goat anti-rabbit IgG (Invitrogen-Molecular Probes) <strong>and</strong><br />

Cy 5 goat anti-rabbit IgG (Jackson ImmunoResearch Laboratories). All antibodies were<br />

diluted in PBS containing 0.05% Tween-20, 3% BSA for blocking <strong>and</strong> 0.1% Triton X-100 for<br />

permeabilization. Nuclear counter staining was performed with DAPI (1µg/mL; Sigma) for 10<br />

min at room temperature. Tissue sections were analyzed with a confocal laser scanning<br />

microscope (LSM-700; Carl Zeiss) with 20x lens (numeric aperture 0.8) or 63x lens with oil<br />

(numeric aperture 1.4) using ZEN 2009 s<strong>of</strong>tware. Tissue sections <strong>of</strong> human normal tonsil or<br />

human colon from Crohn's disease patients, obtained from the Department <strong>of</strong> Pathology,<br />

Medical University <strong>of</strong> Vienna, Austria, were used as a positive control for IL-<strong>33</strong> or ST2<br />

staining, respectively. 7-10<br />

Adenoviral infection<br />

To study a potential NF-κB-dependent effect on endothelial cells upon IL-<strong>33</strong> stimulation,<br />

HUVEC were infected with adenoviral vectors for overexpression <strong>of</strong> IκBα (AdV-IκBα) or for<br />

overexpression <strong>of</strong> a mutant dominant negative IκB kinase 2 (AdV-dnIKK2), respectively, as<br />

described previously. 11, 12 Infection was performed in M199 supplemented with 20% FCS,<br />

2mM L-glutamine, 100 U/mL penicillin, 100 µg/mL streptomycin, 5 U/mL heparin, <strong>and</strong> 25<br />

µg/mL endothelial cell growth supplement (Promocell, Heidelberg, Germany) for 4-6 h with<br />

the AdV-IκBα, the AdV-dnIKK2 or control adenovirus (AdV-green fluorescent protein<br />

(GFP)) 13 at a multiplicity <strong>of</strong> infection <strong>of</strong> 100. 48 h post infection cells were stimulated with<br />

IL-<strong>33</strong> (100 ng/mL) for 6 h.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


References<br />

1. Zhang WJ, Hufnagl P, Binder BR, Wojta J. Antiinflammatory activity <strong>of</strong> astragaloside<br />

IV is mediated by inhibition <strong>of</strong> NF-kappaB activation <strong>and</strong> adhesion molecule<br />

expression. Thromb Haemost. 2003;90:904-914.<br />

2. Choi EY, Chavakis E, Czabanka MA, Langer HF, Fraemohs L, Economopoulou M,<br />

Kundu RK, Orl<strong>and</strong>i A, Zheng YY, Prieto DA, Ballantyne CM, Constant SL, Aird WC,<br />

Papayannopoulou T, Gahmberg CG, Udey MC, Vajkoczy P, Quertermous T,<br />

Dimmeler S, Weber C, Chavakis T. Del-1, an endogenous leukocyte-endothelial<br />

adhesion inhibitor, limits inflammatory cell recruitment. Science. 2008;322:1101-<br />

1104.<br />

3. Philipose S, Konya V, Sreckovic I, Marsche G, Lippe IT, Peskar BA, Heinemann A,<br />

Schuligoi R. The prostagl<strong>and</strong>in E2 receptor EP4 is expressed by human platelets <strong>and</strong><br />

potently inhibits platelet aggregation <strong>and</strong> thrombus formation. Arterioscler Thromb<br />

Vasc Biol. 2010;30:2416-2423.<br />

4. Konya V, Philipose S, Balint Z, Olschewski A, Marsche G, Sturm EM, Schicho R,<br />

Peskar BA, Schuligoi R, Heinemann A. Interaction <strong>of</strong> eosinophils with endothelial<br />

cells is modulated by prostagl<strong>and</strong>in EP4 receptors. Eur J Immunol. 2011.<br />

5. Niessner A, Sato K, Chaik<strong>of</strong> EL, Colmegna I, Goronzy JJ, Wey<strong>and</strong> CM. Pathogen-<br />

sensing plasmacytoid dendritic cells stimulate cytotoxic T-cell function in the<br />

atherosclerotic plaque through interferon-alpha. Circulation. 2006;114:2482-2489.<br />

6. Demyanets S, Pfaffenberger S, Kaun C, Rega G, Speidl WS, Kastl SP, Weiss TW,<br />

Hohensinner PJ, Dietrich W, Tschugguel W, Bochkov VN, Awad EM, Maurer G,<br />

Huber K, Wojta J. The estrogen metabolite 17beta-dihydroequilenin counteracts<br />

interleukin-1alpha induced expression <strong>of</strong> inflammatory mediators in human<br />

endothelial cells in vitro via NF-kappaB pathway. Thromb Haemost. 2006;95:107-<br />

116.<br />

7. Carriere V, Roussel L, Ortega N, Lacorre DA, Americh L, Aguilar L, Bouche G,<br />

Girard JP. IL-<strong>33</strong>, the IL-1-like cytokine lig<strong>and</strong> for ST2 receptor, is a chromatin-<br />

associated nuclear factor in vivo. Proc Natl Acad Sci U S A. 2007;104:282-287.<br />

8. Baekkevold ES, Roussigne M, Yamanaka T, Johansen FE, Jahnsen FL, Amalric F,<br />

Br<strong>and</strong>tzaeg P, Erard M, Haraldsen G, Girard JP. Molecular characterization <strong>of</strong> NF-<br />

HEV, a nuclear factor preferentially expressed in human high endothelial venules. Am<br />

J Pathol. 2003;163:69-79.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


9. Sponheim J, Pollheimer J, Olsen T, Balogh J, Hammarstrom C, Loos T, Kasprzycka<br />

M, Sorensen DR, Nilsen HR, Kuchler AM, Vatn MH, Haraldsen G. Inflammatory<br />

bowel disease-associated interleukin-<strong>33</strong> is preferentially expressed in ulceration-<br />

associated my<strong>of</strong>ibroblasts. Am J Pathol. 2010;177:2804-2815.<br />

10. Beltran CJ, Nunez LE, Diaz-Jimenez D, Farfan N, C<strong>and</strong>ia E, Heine C, Lopez F,<br />

Gonzalez MJ, Quera R, Hermoso MA. Characterization <strong>of</strong> the novel ST2/IL-<strong>33</strong><br />

system in patients with inflammatory bowel disease. Inflamm Bowel Dis.<br />

2010;16:1097-1107.<br />

11. Oitzinger W, H<strong>of</strong>er-Warbinek R, Schmid JA, Koshelnick Y, Binder BR, de Martin R.<br />

Adenovirus-mediated expression <strong>of</strong> a mutant IkappaB kinase 2 inhibits the response <strong>of</strong><br />

endothelial cells to inflammatory stimuli. Blood. 2001;97:1611-1617.<br />

12. Wrighton CJ, H<strong>of</strong>er-Warbinek R, Moll T, Eytner R, Bach FH, de Martin R. Inhibition<br />

<strong>of</strong> endothelial cell activation by adenovirus-mediated expression <strong>of</strong> I kappa B alpha,<br />

an inhibitor <strong>of</strong> the transcription factor NF-kappa B. J Exp Med. 1996;183:1013-1022.<br />

13. de Martin R, Raidl M, H<strong>of</strong>er E, Binder BR. Adenovirus-mediated expression <strong>of</strong> green<br />

fluorescent protein. Gene Ther. 1997;4:493-495.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


Supplemental Table I. IL-1 mRNA is up-regulated by IL-<strong>33</strong> in HUVEC <strong>and</strong> HCAEC.<br />

1 h 3 h 6 h 9 h 24 h<br />

HUVEC 30.3±7.7* 20.8±0.6* 68.2±15.3* 19.8±0.4* 5.3±0.6*<br />

HCAEC 3.5±0.7* 7.5±2.2* 6.9±2.6* 5.5±2.9* 3.8±0.8*<br />

Confluent monolayers <strong>of</strong> HUVEC or HCAEC were incubated for 1, 3, 6, 9 or 24 h in the<br />

absence or presence <strong>of</strong> IL-<strong>33</strong> (100 ng/mL). mRNA was prepared <strong>and</strong> RealTime-PCR with<br />

primers specific for IL-1 was performed as described in “Methods”. IL-1 mRNA levels were<br />

normalized according to the respective GAPDH mRNA level. Each experiment was<br />

performed in triplicates. Values are given as x-fold <strong>of</strong> control, which was set as 1 <strong>and</strong><br />

represent mean ± SD <strong>of</strong> 2 different experiments.*p≤0.05 as compared to respective<br />

unstimulated control.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


Supplemental Table II. IL-1 receptor antagonist does not counteract IL-<strong>33</strong>-induced<br />

ICAM-1, VCAM-1, E-selectin <strong>and</strong> MCP-1 mRNA expressions in HUVEC.<br />

IL-<strong>33</strong> IL-<strong>33</strong> + IL-1Ra IL-1β IL-1β+ IL-1Ra<br />

ICAM-1 55.8±8.7 54.1±5.7 20.3±1.0 0.5±0.1<br />

VCAM-1 72.6±2.2 76.8±3.1 <strong>33</strong>.2±5.7 0.3±0.1<br />

E-selectin 1032.1±99.2 1021.8±17.1 343.3±5.7 0.4±0.1<br />

MCP-1 73.3±0.7 65.2±9.2 20.4±2.2 1.2±0.1<br />

Confluent monolayers <strong>of</strong> HUVEC were incubated for 6 h in the absence or presence <strong>of</strong> IL-<strong>33</strong><br />

(100 ng/mL) or IL-1β (200 U/mL) with or without IL-1Ra (10 µg/mL). mRNA was prepared<br />

<strong>and</strong> RealTime-PCR with primers specific for ICAM-1, VCAM-1, E-selectin <strong>and</strong> MCP-1 was<br />

performed as described in “Methods”. <strong>Adhesion</strong> molecules <strong>and</strong> MCP-1 mRNA levels were<br />

normalized according to the respective GAPDH mRNA level. Each experiment was<br />

performed in triplicates. Values are given as x-fold <strong>of</strong> control, which was set as 1 <strong>and</strong><br />

represent mean ± SD <strong>of</strong> 2 different experiments. The overall ANOVA for each tested<br />

molecule remained significant after correction for 4 comparisons.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


Supplemental Table III. Mitogen-activated protein/extracellular signal regulated kinase<br />

(MEK) inhibitor U0126 or phosphoinoside-3-kinase (PI3K) inhibitor LY-294002 do not<br />

abrogate IL-<strong>33</strong> induced ICAM-1, VCAM-1, E-selectin or MCP-1 protein in HUVEC.<br />

Control IL-<strong>33</strong> U+IL-<strong>33</strong> LY+ IL-<strong>33</strong> U LY<br />

ICAM 297±50 5<strong>33</strong>8±799 4886±959 5354±983 304±61 254±44<br />

VCAM 177±63 547±81 591±121 318±80 170±56 182±49<br />

E-<br />

selectin<br />

151±<strong>33</strong> 4214±434 3666±754 4447±857 156±42 143±40<br />

MCP-1 2565±44 41945±6670 46288±7007 36147±8406 2496±65 2201±78<br />

Confluent monolayers <strong>of</strong> HUVEC were pre-incubated for 1 h with the MEK inhibitor U0126<br />

(U) at 1 µM or the PI3K inhibitor LY-294002 (LY) at 5µM. Thereafter, the cells were treated<br />

with or without IL-<strong>33</strong> at a concentration <strong>of</strong> 100 ng/mL for 24 h (for ICAM-1, VCAM-1,<br />

MCP-1 measurement) or 4 h (for E-selectin determination) or left untreated (control). Cells<br />

were gently detached by detachment buffer <strong>and</strong> ICAM-1, VCAM-1 <strong>and</strong> E-selectin<br />

expressions at the cell surface were measured by means <strong>of</strong> flow cytometry as described in<br />

“Methods” <strong>and</strong> MCP-1 protein was determined in conditioned media by ELISA as described<br />

in “Methods”. Each experiment was performed in triplicates. Values are given in mean<br />

fluorescence intensity (MFI) for ICAM-1, VCAM-1, E-selectin or in pg/10 4 cells for MCP-1<br />

<strong>and</strong> represent mean values ± SD <strong>of</strong> 3 different experiments. Post hoc analyses indicated<br />

significant differences between IL-<strong>33</strong> stimulations <strong>and</strong> controls for each molecule but no<br />

significant differences between IL-<strong>33</strong> stimulations in the presence <strong>and</strong> absence <strong>of</strong> the MEK<br />

inhibitor U0126 or the PI3K inhibitor LY-294002. The overall ANOVA was not affected by<br />

correction for 4 comparisons.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


Supplemental Figure I<br />

A<br />

C<br />

ICAM-1 (MFI)<br />

E-selectin (MFI)<br />

2000<br />

1600<br />

1200<br />

800<br />

400<br />

0<br />

800<br />

600<br />

400<br />

200<br />

0<br />

Control<br />

Control<br />

IL-<strong>33</strong><br />

IL-<strong>33</strong><br />

IL-<strong>33</strong> + ST2-Fc<br />

IL-<strong>33</strong> + ST2-Fc<br />

Supplemental Figure I. Soluble ST2 fusion protein (sST2-Fc) abolished IL-<strong>33</strong> effects in<br />

human endothelial cells. IL-<strong>33</strong> (1 ng/mL) was incubated with rh sST2-Fc (5 µg/mL) or with<br />

IgG1-Fc (5 µg/mL) for 15 min at 37°C before addition to confluent monolayers <strong>of</strong> HUVEC<br />

for 24 h (for ICAM-1, VCAM-1, MCP-1 determination) or 4 h (for E-selectin measurement).<br />

Cells were gently detached by detachment buffer <strong>and</strong> (A) ICAM-1, (B) VCAM-1 <strong>and</strong> (C) E-<br />

selectin expressions at the cell surface were measured by means <strong>of</strong> flow cytometry as<br />

described in “Methods” or (D) MCP-1 protein was determined in conditioned media by<br />

ELISA as described in “Methods”. Each experiment was performed in triplicates. (A, B, C)<br />

Values are given in mean fluorescence intensity (MFI) or (D) in pg/10 4 cells <strong>and</strong> represent<br />

mean values ± SD <strong>of</strong> 3 different experiments. The overall ANOVA for each tested molecule<br />

remained significant after correction for 4 comparisons.<br />

IL-<strong>33</strong> + IgG<br />

IL-<strong>33</strong> + IgG<br />

ST2-Fc<br />

ST2-Fc<br />

IgG<br />

IgG<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011<br />

B<br />

D<br />

VCAM-1 (MFI)<br />

MCP-1 (pg/10 4 cells)<br />

400<br />

300<br />

200<br />

100<br />

0<br />

4000<br />

3000<br />

2000<br />

1000<br />

0<br />

Control<br />

Control<br />

IL-<strong>33</strong><br />

IL-<strong>33</strong><br />

IL-<strong>33</strong> + ST2-Fc<br />

IL-<strong>33</strong> + ST2-Fc<br />

IL-<strong>33</strong> + IgG<br />

IL-<strong>33</strong> + IgG<br />

ST2-Fc<br />

ST2-Fc<br />

IgG<br />

IgG


Supplemental Figure II<br />

A<br />

C<br />

ICAM-1 (x-fold control)<br />

E-selectin (x-fold control)<br />

200<br />

160<br />

120<br />

80<br />

40<br />

0<br />

1800<br />

1600<br />

1400<br />

1200<br />

1000<br />

800<br />

600<br />

400<br />

200<br />

0<br />

1h 3h 6h 9h 24h<br />

IL-<strong>33</strong> (100 ng/mL)<br />

1h 3h 6h 9h 24h<br />

IL-<strong>33</strong> (100 ng/mL)<br />

Supplemental Figure II. Effects <strong>of</strong> IL-<strong>33</strong> on ICAM-1, VCAM-1, E-selectin <strong>and</strong> MCP-1<br />

mRNA expression by HUVEC. Confluent monolayers <strong>of</strong> HUVEC were incubated for 1, 3,<br />

6, 9 or 24 h in the absence or presence <strong>of</strong> IL-<strong>33</strong> (100 ng/mL). mRNA was prepared <strong>and</strong><br />

RealTime-PCR with primers specific for (A) ICAM-1, (B) VCAM-1, (C) E-selectin or (D)<br />

MCP-1 was performed as described in “Methods”. <strong>Adhesion</strong> molecules <strong>and</strong> MCP-1 mRNA<br />

levels were normalized according to the respective GAPDH mRNA level. Each experiment<br />

was performed in triplicates. Values are given as x-fold <strong>of</strong> control, which was set as 1 <strong>and</strong><br />

represent mean ± SD <strong>of</strong> 2 different experiments. The overall ANOVA for each tested<br />

molecule remained significant after correction for 4 comparisons.<br />

B<br />

D<br />

VCAM-1 (x-fold control)<br />

MCP-1 (x-fold control)<br />

500<br />

400<br />

300<br />

200<br />

100<br />

250<br />

200<br />

150<br />

100<br />

0<br />

50<br />

0<br />

1h 3h 6h 9h 24h<br />

IL-<strong>33</strong> (100 ng/mL)<br />

1h 3h 6h 9h 24h<br />

IL-<strong>33</strong> (100 ng/mL)<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011


Supplemental Figure III<br />

A<br />

C<br />

IL-4 (pg/mL)<br />

IL-10 (pg/mL)<br />

16<br />

12<br />

8<br />

4<br />

0<br />

16<br />

12<br />

8<br />

4<br />

0<br />

Control 4h<br />

Control 4h<br />

IL-<strong>33</strong> 4h<br />

IL-<strong>33</strong> 4h<br />

Control 16h<br />

Control 16h<br />

Supplemental Figure III. IL-<strong>33</strong> did not modulate Th2 <strong>and</strong> T regulatory cytokines in<br />

HCAEC. Confluent monolayers <strong>of</strong> HCAEC were incubated for 4, 16, or 24 h in the absence<br />

or presence <strong>of</strong> IL-<strong>33</strong> (100 ng/mL). Supernatants were processed per manufacturer`s<br />

instruction as described in "Methods". (A) IL-4, (B) IL-5 <strong>and</strong> (C) IL-10 were measured by<br />

means <strong>of</strong> flow cytometry. Each experiment was performed in triplicates. Values are given in<br />

pg/mL <strong>and</strong> represent mean values ± SD <strong>of</strong> 3 different experiments. The overall ANOVA for<br />

each tested molecule was ≥0.05.<br />

IL-<strong>33</strong> 16h<br />

IL-<strong>33</strong> 16h<br />

Control 24h<br />

Control 24h<br />

IL-<strong>33</strong> 24h<br />

IL-<strong>33</strong> 24h<br />

Downloaded from<br />

http://atvb.ahajournals.org/ at Bibliothek der MedUniWien (IX0000096057) on September 2, 2011<br />

B<br />

IL-5 (pg/mL)<br />

6<br />

4<br />

2<br />

0<br />

Control 4h<br />

IL-<strong>33</strong> 4h<br />

Control 16h<br />

IL-<strong>33</strong> 16h<br />

Control 24h<br />

IL-<strong>33</strong> 24h

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!