• Aucun résultat trouvé

Importance of junctional adhesion molecule-C for neointimal hyperplasia and monocyte recruitment in atherosclerosis-prone

N/A
N/A
Protected

Academic year: 2022

Partager "Importance of junctional adhesion molecule-C for neointimal hyperplasia and monocyte recruitment in atherosclerosis-prone"

Copied!
12
0
0

Texte intégral

(1)

Article

Reference

Importance of junctional adhesion molecule-C for neointimal hyperplasia and monocyte recruitment in atherosclerosis-prone

mice-brief report

SHAGDARSUREN, Erdenechimeg, et al.

Abstract

OBJECTIVE: Although junctional adhesion molecule (JAM)-C has been implicated in the control of inflammatory leukocyte recruitment, its role in neointima formation after arterial injury has not been elucidated. METHODS AND RESULTS: In apolipoprotein E-deficient (Apoe(-/-)) mice fed an atherogenic diet, antibody blockade of JAM-C significantly reduced neointimal hyperplasia after wire injury of carotid arteries without altering medial area and decreased neointimal macrophage but not smooth muscle cell (SMC) content. An increased expression of JAM-C was detected in colocalization with luminal SMCs 1 day after injury and neointimal SMCs after 3 weeks. Blocking JAM-C inhibited monocytic cell arrest and leukocyte adhesion to carotid arteries perfused ex vivo and in vivo. Furthermore, monocyte adhesion to activated coronary artery SMCs under flow conditions in vitro was diminished by blocking JAM-C. CONCLUSIONS: Our data provide the first evidence for a crucial role of JAM-C in accelerated lesion formation and leukocyte recruitment in atherosclerosis-prone mice.

SHAGDARSUREN, Erdenechimeg, et al. Importance of junctional adhesion molecule-C for neointimal hyperplasia and monocyte recruitment in atherosclerosis-prone mice-brief report.

Arteriosclerosis, Thrombosis, and Vascular Biology, 2009, vol. 29, no. 8, p. 1161-3

DOI : 10.1161/ATVBAHA.109.187898 PMID : 19520977

Available at:

http://archive-ouverte.unige.ch/unige:5434

Disclaimer: layout of this document may differ from the published version.

(2)

Integrative Physiology/Experimental Medicine

Importance of Junctional Adhesion Molecule-C for Neointimal Hyperplasia and Monocyte Recruitment in

Atherosclerosis-Prone Mice–Brief Report

Erdenechimeg Shagdarsuren, Yassin Djalali-Talab, Michel Aurrand-Lions, Kiril Bidzhekov, Elisa A. Liehn, Beat A. Imhof, Christian Weber, Alma Zernecke

Objective—Although junctional adhesion molecule (JAM)-C has been implicated in the control of inflammatory leukocyte recruitment, its role in neointima formation after arterial injury has not been elucidated.

Methods and Results—In apolipoprotein E– deficient (Apoe⫺/⫺) mice fed an atherogenic diet, antibody blockade of JAM-C significantly reduced neointimal hyperplasia after wire injury of carotid arteries without altering medial area and decreased neointimal macrophage but not smooth muscle cell (SMC) content. An increased expression of JAM-C was detected in colocalization with luminal SMCs 1 day after injury and neointimal SMCs after 3 weeks. Blocking JAM-C inhibited monocytic cell arrest and leukocyte adhesion to carotid arteries perfused ex vivo and in vivo. Furthermore, monocyte adhesion to activated coronary artery SMCs under flow conditions in vitro was diminished by blocking JAM-C.

Conclusions—Our data provide the first evidence for a crucial role of JAM-C in accelerated lesion formation and leukocyte recruitment in atherosclerosis-prone mice. (Arterioscler Thromb Vasc Biol. 2009;29:1161-1163.)

Key Words:leukocyte recruitmentadhesion moleculesremodelinginflammationrestonosisatherosclerosis

T

he recruitment of inflammatory cells to the vessel wall determines neointimal hyperplasia after injury.1,2A key role herein can be ascribed to junctional adhesion molecules (JAMs) of the immunoglobulin superfamily, which mediate firm arrest but also subsequent transmigration of leukocytes across endothelial cells (ECs).2– 4JAM-C has been identified to be localized in tight junctions of ECs,4,5in smooth muscle cells (SMCs),6but also human platelets7 and blocking anti- bodies to JAM-C were shown to inhibit leukocyte accumu- lation in different mouse models of inflammation.5–7 In Apoe/mice, JAM-C was described to be highly expressed in ECs and intimal SMCs.6 Recently, genetic deficiency in JAM-C was demonstrated to result in respiratory, digestive, and immune disorders, unveiling its importance in diverse biological processes.8

We here addressed the role of JAM-C in neointima formation after wire-induced arterial injury in Apoe/ mice using a blocking antibody.

Materials and Methods

For detailed Materials and Methods, please see the supplemental materials (available online at http://atvb.ahajournals.org).

Mouse Model of Carotid Artery Injury

Apoe⫺/⫺mice on high-fat diet received monoclonal H33 or isotype control9,10 3/wk and underwent wire-induced injury of carotid arteries. Movats’ pentachrome and immunofluorescence staining was performed on paraffin-embedded artery sections.

Adhesion Assay, Ex Vivo Perfusion, and Intravital Microscopy

MonoMac6 cell adhesion to human coronary SMCs was analyzed in a parallel-wall flow-chamber. Leukocyte arrest to carotid arteries was analyzed ex vivo or in vivo.

Results and Discussion

Carotid arteries ofApoe/mice fed an atherogenic diet and treated with a specific neutralizing anti–JAM-C (H33)9,10or isotype control antibody were analyzed 3 weeks after wire- induced denudation-injury. Compared with control-treated mice, a significant reduction in neointimal plaque formation but not medial areas was observed in H33-treated mice (Figure 1A and 1B). Quantitative immunostaining revealed a significant decrease in the relative content of neointimal macrophages (Figure 1C and 1D), whereas SMC content was

Received March 11, 2009; revision accepted May 24, 2009.

From the Institute for Molecular Cardiovascular Research (IMCAR) (E.S., Y.D.-T., K.B., E.A.L., C.W., A.Z.), RWTH Aachen University, Germany;

UMR891, Inserm (M.A.-L.), Centre de Recherche en Cance´rologie de Marseille, Institut Paoli-Calmettes, France; the Department of Pathology and Immunology (B.A.I.), University Medical Centre, Geneva, Switzerland; and the Rudolf-Virchow-Center (A.Z.), DFG-Research-Center for Experimental Biomedicine, University of Wu¨rzburg, Germany.

E.S. and Y.D.-T. contributed equally to this study.

Correspondence to Dr Alma Zernecke, MD, Institut fu¨r Molekulare Herz-Kreislaufforschung, Universita¨tsklinikum der RWTH Aachen, Pauwelsstrasse

(3)

not altered (Figure 1E). The inhibition of plaque formation may thus be attributable to an attenuation of monocyte infiltration.

In contrast to healthy vessels, atherosclerotic arteries dis- play increased expression of JAM-C in EC layers and intimal

SMCs, and oxLDL induces the redistribution of JAM-C from inter endothelial contacts supporting leukocyte recruitment.6 Compared to marginal staining in uninjured arteries, an enhanced expression of JAM-C could be observed in injured segments ofApoe⫺/⫺carotid arteries (not shown). One day

Figure 1.Carotid arteries ofApoe⫺/⫺

mice treated with isotype control or H33 (n6 each) 3 weeks after wire-induced injury. Quantification of pentachrome stained neointimal and medial areas;

representative images (A and B). Neoin- timal Mac-3macrophage staining (red) relative to neointimal area; representa- tive images (C and D). Alpha-smooth- muscle actin (SMA)SMC content rela- tive to neointimal area (E). *P0.01.

Figure 2.Immunofluorescence staining for JAM-C,-SMASMCs, CD31ECs, and DAPIcell nuclei inApoe⫺/⫺carotid arteries after injury (A). Ex vivo adhesion of MonoMac 6 cells (arrowheads, B), and intravital microscopy of leukocyte adhesion (C) inApoe⫺/⫺carotid arteries 1 day after injury treated with H33 or isotype control, **P0.0001. Adhesion of MonoMac 6 cells pretreated with/without anti–

Mac-1 antibody or isotype control (hatched bar) to oxLDL-stimulated SMCs pretreated with/without H33 or isotype control (black bar) in vitro, **P0.001 vs untreated (D). n3 to 4, *P0.05.

1162 Arterioscler Thromb Vasc Biol August 2009

(4)

after injury, double-immunofluorescence staining revealed the expression of JAM-C in colocalization with luminal SMCs (Figure 2A), whereas remaining ECs could only rarely be detected (not shown). This is in line with previous findings that SMCs can replace ECs in denuded vessels and display a proadhesive phenotype with a constitutive upregulation of different chemokines11and implies that luminal SMCs can express and present JAM-C to circulating leukocytes. Three weeks after injury the expression of JAM-C was detected in colocalization with luminal ECs but also SMCs and neointi- mal plaque SMCs (Figure 2A).

After injury, the exposed subendothelial matrix can trigger the adhesion of platelets, which support leukocyte recruit- ment.1,12On human platelets, JAM-C serves as a ligand for leukocyte-expressed Mac-1 facilitating platelet–leukocyte in- teractions.7Whereas double-immunofluorescence staining re- vealed the accumulation of P-selectinplatelets in arteries 1 day after injury, no colocalization with JAM-C was seen.

Likewise, JAM-C was not detectable by FACS analysis on resting or thrombin-activated mouse platelets (supplemental Figure III), contrasting findings in human platelets.7

To further assess a contribution of JAM-C in early leuko- cyte recruitment, monocytic cell arrest was assessed in carotid arteries perfused ex vivo 1 day after injury. A marked accumulation of Mono Mac 6 cells was observed in untreated or isotype control preperfused arteries, which was signifi- cantly reduced after preperfusion with H33 (Figure 2B).

Similarly, intravital microscopy revealed a significant reduc- tion in the number of leukocytes adherent to carotid arteries 1 day after injury in vivo in H33-treated mice, as compared to untreated or isotype control-treated mice (Figure 2C). In vitro, Mono Mac 6 cell arrest supported by oxLDL-activated coronary human SMCs was markedly inhibited by pretreat- ment with H33 (Figure 2D). Furthermore, Mono Mac 6 cell adhesion to oxLDL-activated SMCs was markedly reduced in the presence of a blocking Mac-1 antibody, and the combi- nation of both H33 and anti–Mac-1 antibodies resulted in an inhibition that was more pronounced than either treatment alone, whereas respective isotype controls had no effect (Figure 2D). These data imply that in conjunction with other ligands known to be expressed in oxLDL-activated SMCs, eg, ICAM-1,6 leukocyte-expressed Mac-1 might directly interact with JAM-C on SMCs. These data identify a prom- inent role of JAM-C in leukocyte adhesion and suggests that JAM-C on SMCs supports monocyte recruitment in vivo, thus contributing to neointima formation after injury.

The expression and interaction of endothelial JAM-C with JAM-B at intercellular junctions has been demonstrated.5,10 Although JAM-C can function as a counter-receptor for Mac-1 independently of JAM-B,7antibodies to JAM-C can abolish the formation of JAM-B/JAM-C heterodimers10and diminish monocyte extravasation by promoting reverse mi- gration rather than by reducing orthograde transmigration.8 Whereas these mechanisms might contribute to the decrease in neointimal macrophages in H33-treated mice at later time

points during reendothelialization, early adhesive interactions supported by JAM-C on exposed SMCs might mechanisti- cally differ. In keeping with our data, JAM-C on SMCs may also engage in direct interactions with leukocytic Mac-1 to mediate adhesion to sites of inflammation or injury.

In conclusion, our data for the first time demonstrate that blocking JAM-C inhibits neointima formation and monocyte accumulation after arterial injury inApoe⫺/⫺mice. Although potential side effects may preclude a systemic inhibition, JAM-C may constitute a therapeutic target for a local mod- ulation of arterial remodeling in atherosclerosis.

Acknowledgments

We thank P. Hammel for antibody generation.

Sources of Funding

This work was supported by the Deutsche Forschungsgemeinschaft (FOR809-TP3/TP6), Avenir Program Inserm, Ligue Nationale contre le Cancer, Swiss National Science Foundation (310000-120184/1), and Swiss Cancer League (OCS-01653-02-2005).

Disclosures

None.

References

1. Schober A, Weber C. Mechanisms of monocyte recruitment in vascular repair after injury.Antioxid Redox Signal. 2005;7:1249 –1257.

2. Weber C, Fraemohs L, Dejana E. The role of junctional adhesion mol- ecules in vascular inflammation.Nat Rev Immunol. 2007;7:467– 477.

3. Bradfield PF, Scheiermann C, Nourshargh S, Ody C, Luscinskas FW, Rainger GE, Nash GB, Miljkovic-Licina M, Aurrand-Lions M, Imhof BA. JAM-C regulates unidirectional monocyte transendothelial migration in inflammation.Blood. 2007;110:2545–2555.

4. Arrate MP, Rodriguez JM, Tran TM, Brock TA, Cunningham SA. Cloning of human junctional adhesion molecule 3 (JAM3) and its identification as the JAM2 couter-receptor.J Biol Chem. 2001;276:45826 – 45832.

5. Aurrand-Lions M, Lamagna C, Dangerfield JP, Wang S, Herrera P, Nourshargh S, Imhof BA. Junctional adhesion molecule-C regulates the early influx of leukocytes into tissues during inflammation.J Immunol.

2005;174:6406 – 6415.

6. Keiper T, Al-Fakhri N, Chavakis E, Athanasopoulos AN, Isermann B, Herzog S, Saffrich R, Hersemeyer K, Bohle RM, Haendeler J, Preissner KT, Santoso S, Chavakis T. The role of junctional adhesion molecule-C (JAM-C) in oxidized LDL-mediated leukocyte recruitment. Faseb J.

2005;19:2078 –2080.

7. Santoso S, Sachs UJ, Kroll H, Linder M, Ruf A, Preissner KT, Chavakis T. The junctional adhesion molecule 3 (JAM-3) on human platelets is a counterreceptor for the leukocyte integrin Mac-1.J Exp Med. 2002;196:

679 – 691.

8. Imhof BA, Zimmerli C, Gliki G, Ducrest-Gay D, Juillard P, Hammel P, Adams R, Aurrand-Lions M. Pulmonary dysfunction and impaired gran- ulocyte homeostasis result in poor survival of Jam-C-deficient mice.

J Pathol. 2007;212:198 –208.

9. Aurrand-Lions M, Duncan L, Ballestrem C, Imhof BA. JAM-2, a novel immunoglobulin superfamily molecule, expressed by endothelial and lymphatic cells.J Biol Chem. 2001;276:2733–2741.

10. Lamagna C, Meda P, Mandicourt G, Brown J, Gilbert RJ, Jones EY, Kiefer F, Ruga P, Imhof BA. Dual interaction of JAM-C with JAM-B and alpha(M)beta2 integrin: function in junctional complexes and leukocyte adhesion.Mol Biol Cell. 2005;16:4992–5003.

11. Zeiffer U, Schober A, Lietz M, Liehn EA, Erl W, Emans N, Yan ZQ, Weber C. Neointimal smooth muscle cells display a proinflammatory phenotype resulting in increased leukocyte recruitment mediated by P-selectin and chemokines.Circ Res. 2004;94:776 –784.

12. Zernecke A, Shagdarsuren E, Weber C. Chemokines in atherosclerosis:

an update.Arterioscler Thromb Vasc Biol. 2008;28:1897–1908.

Shagdarsuren et al JAM-C in Neointimal Hyperplasia and Monocyte Recruitment 1163

(5)

Shagdarsuren et al.: JAM-C in neointima formation

1

Supplement Material

Material and Methods

H33 antibody and specificity

The anti-JAM-C antibody (clone H33, full name CRAM-13H33) was generated by Aurrand- Lions et al.1. The specificity of H33 binding selectively to JAM-C was tested in the murine squamous cell carcinoma cell line KLN205 mock-transfected or stably transfected with a JAM- CEGFP expressing fusion vector2. The molecular weight of the fusion protein containing JAM-C (30 kDa) and EGFP (40 kDa) is approximately 70 kDa. In conjunction with EGFP, flow cytometry confirmed the detection of a specific signal for JAM-C by H33 in comparison to isotype control on JAM-CEGFP-expressing cells, while an antibody to integrin aV (RMV 7) produced a staining in both mock and JAM-CEGFP-transfected cells serving as a positive control; antibody binding was detected using an Phycoerythrin-conjugated anti-rat secondary antibody (Jackson ImmunoResearch, Ref #112-116-143) (Online Data Supplement Figure 1A).

A specific band at the expected size of the fusion protein containing JAM-C (70 kDa) was in addition revealed by H33 in lysates of JAM-CEGFP but not mock-transfected KLN205 cells by Western Blotting after immunoprecipitation of JAM-C, performed as described1, while JAM-C could not be detected using anti-integrin or isotype control antibodies (Online Data Supplement Figure 1B). Furthermore, FACS analyses revealed a specific staining for JAM-C using H33 on primary lung microvascular endothelial cells (LMECs) isolated from wild type (Jam-C+/+) but not JAM-C-deficient (Jam-C-/-) mice3-5 compared to control antibodies directed against L-selectin (Mel14, rat IgG2a). Staining for CD31 (GC51, rat IgG2b) on both Jam-C+/+

at Bibliotheque Faculte Medecine Geneve on March 16, 2010 atvb.ahajournals.org

Downloaded from

(6)

Shagdarsuren et al.: JAM-C in neointima formation

2 and Jam-C-/- LMECs mice in comparison to isotype matched control antibodies directed against Mac-1 (M1/70, rat IgG2b) served as a positive control; primary antibody binding was detected using an Phycoerythrin-conjugated secondary anti-rat antibody (Online Data Supplement Figure 1C). Together, these data confirm binding of H33 specifically to JAM-C. In addition, H33 was shown to block JAM-B/JAM-C interactions5, angiogenesis and leukocyte homing to tumors4.

Mouse model of carotid artery injury

Female, 8 week old Apoe-/- mice (C57Bl/6 background) were fed an atherogenic diet containing 21% fat starting 1 weekbefore transluminal carotid artery injury inducedby a 0.014 inch angioplasty guide-wire in anesthetized mice (100 mg/kg ketamine/ 10 mg/ kg xylacine, i.p.)6. Mice received i.p. injections of monoclonal antibody against JAM-C (H33, 50 µ g/mouse, 3/week) or isotype control antibody (rat IgG2a) starting with 2 injections peri-operatively.

After 1 day or 3 weeks, arteries were harvested by in situ perfusion fixation with 4%

paraformaldehyde and embedded in paraffin. Animal experiments were approved by local authoritiesand complied with the German animal protection law.

Morphometry, immunohistochemistry and immunofluoresence stainings

Serial sections (5µm, 250-500µm from the bifurcation)stained with Movats pentachromewere analyzed by planimetry. The relative macrophage and SMC content was determined by antibody staining for Mac-3 (#550292, BD Pharmingen) detected by alkaline-phosphatase enzyme (Vector Laboratories) and counter-staining by Mayers haemalaun and α-smooth muscle actin (1A4; Dako) detected by FITC-conjugated secondary antibody (Jackson ImmunoResearch). For double immunofluorescence stainings, primary affinity purified rabbit

(7)

Shagdarsuren et al.: JAM-C in neointima formation

3 anti-JAM-C antibody (Nr. 526, provided by Prof. Imhof), anti-SM α-actin (α-SMA), anti-P- selectin (clone C-20, Santa-Cruz Biotechnology), anti-CD31 (M-20, Santa-Cruz Biotechnology), or appropriate isotype control antibodies were detected by secondary anti- rabbit Cy-3 and anti-mouse- or anti-goat FITC-antibodies (Jackson ImmunoResearch). Cell nuclei were counterstained with DAPI (Vectashield, Vector Labs). Images were visualized using a LeicaDMLB microscope (Leica Microsystems, Wetzlar) and 10x0.30 numeric aperture or 40x0.75 Leicaobjectives. Images were captured using a JVC digitalcamera KY-F70B 3- CCD (Vitor, Tokyo) and processedusing Diskus software.

Cell culture and flow adhesion assay

Human coronary artery SMCs (Promocell), MonoMac6 cells and human umbilical vein endothelial cells (HUVECs) were maintained as described.6,7 MonoMac6 cell adhesion to SMC was analyzed in parallel wall flow-chambers6. SMCs expressed smooth muscle specific α-actin but not vWF and similarly to vWF+ HUVECs displayed a strong expression of JAM-C, as analyzed by RT-PCR (see Online Data Supplement Figure 2). Confluent SMCs were activated by oxLDL (10 µ g/ml, overnight) and pre-treated with/without H33 or isotype control (10 µg/ml, 30 min). MonoMac6 cells (0.5x106/mL)were pre-treated with/without anti-Mac-1 antibody (CBRM1/29, 20 µ g/ml) or isotype control (mouse IgG1) and perfused over SMC monolayers (1.5 dyn/cm2)in the continuing presence of the antibodies. The number of cells firmly adherent to SMCs was quantifiedby analysis of images recorded with a 3CCD video camera and recorder (JVC, Wayne, NJ).

at Bibliotheque Faculte Medecine Geneve on March 16, 2010 atvb.ahajournals.org

Downloaded from

(8)

Shagdarsuren et al.: JAM-C in neointima formation

4 Reverse-transcriptase and real time PCR

For RNA analysis, total RNA was isolated from HUVECs and SMCs, reverse-transcribed into cDNA using Mo-MLV RT (Invitrogen). RT-PCR was performed using specific primer pairs (hJAM-C forward: GAA GCC AGT GAC CCC TGT CTG TAG AG, reverse: CAT CTG GTT TCC CTG GGT TCT TG; hcalponin forward: GCA GAT GGG CAC CAA CAA AGG AGC, reverse: CCT TGG GGG GAA AAC AGG GAA GG; hSM α-actin forward: GAT GGC TTT GGG CAG CTT GGC AG, reverse: GAG GCA GGC TAA GCG GGA TGG; hvWF forward:

GTT GTG GGA GAT GTT TGC CTA CG, reverse: GAG AAC CTC ATG GTA CAC AAC AGA GC). Products were separated by agarose gel electrophoresis. The expression levels of the target genes were in addition quantified by real time PCR analysis using the QuantiTect SYBR-Green PCR kit (Opticon MJ Research) and specific primer pairs (Jam-C, Hs00230289_m1; 18sRNA, Hs99999901_s1, Applied biosystems).

Platelet isolation and flow cytometric analysis

Platelet-rich-plasma was prepared from 8 week old Apoe-/- mice and platelets were isolated by centrifugation at 1250 g. Platelets were left untreated or incubated with thrombin (1 U/mL, 30 min, 37°C), and reacted with saturating concentrations of affinity purified anti-JAM-C antibody (rabbit anti-mouse, Nr. 526) and detected with FITC-conjugated secondary Ab and stained with anti-CD41-PE (BD Biosciences). Probes were analyzed using a BD FACSAria System (BD Biosciences) and experiments were analyzed via FlowJo Software (Tree Star).

Ex vivo perfusion and intravital microscopy of carotid arteries

Carotid arteries of Apoe-/- mice were isolated for ex vivo perfusion or intravital microscopy8 one day after wire-injury. After transfer onto a microscope stage, arteries were perfused with

(9)

Shagdarsuren et al.: JAM-C in neointima formation

5 MOPS-buffered physiological salt solution (4 µL/min), and after pre-incubation with H33 or isotype control antibodies (10 µg/ml, 30 min), perfused with calcein-AM labeled MonoMac6 cells (106/mL, Molecular Probes) for 8 min. For intravital microscopy, rhodamin-6G (Molecular Probes) was administered i.v. to Apoe-/- mice injected with H33 or isotype control (10 µ g/ml, 30 min before and after wire-injury) to label circulating leukocytes, and injured common carotid arteries were exposed. Leukocyte arrest to arteries was analyzed by epifluorescence microscopy (Zeiss Axiotech, x20 water immersion).

Statistical Analysis

Data represent mean±SEM and were analyzed by 2-tailed Student’s t-test using GraphPad Prism (InStat software; GraphPad).

References

1. Aurrand-Lions M, Duncan L, Ballestrem C, Imhof BA. JAM-2, a novel immunoglobulin superfamily molecule, expressed by endothelial and lymphatic cells. J Biol Chem.

2001;276:2733-2741.

2. Mandicourt G, Iden S, Ebnet K, Aurrand-Lions M, Imhof BA. JAM-C regulates tight junctions and integrin-mediated cell adhesion and migration. J Biol Chem. 2007;282:1830-1837.

3. Imhof BA, Zimmerli C, Gliki G, Ducrest-Gay D, Juillard P, Hammel P, Adams R, Aurrand- Lions M. Pulmonary dysfunction and impaired granulocyte homeostasis result in poor survival of Jam-C-deficient mice. J Pathol. 2007;212:198-208.

4. Lamagna C, Hodivala-Dilke KM, Imhof BA, Aurrand-Lions M. Antibody against junctional adhesion molecule-C inhibits angiogenesis and tumor growth. Cancer Res. 2005;65:5703-5710.

5. Lamagna C, Meda P, Mandicourt G, Brown J, Gilbert, R.J., Jones EY, Kiefer F, Ruga P, Imhof BA. Dual interaction of JAM-C with JAM-B and alpha(M)beta2 integrin: function in junctional complexes and leukocyte adhesion. . Mol Biol Cell. 2005;16:4992-5003.

6. Shagdarsuren E, Bidzhekov K, Djalali-Talab Y, Liehn EA, Hristov M, Matthijsen RA, Buurman WA, Zernecke A, Weber C. C1-esterase inhibitor protects against neointima formation after arterial injury in atherosclerosis-prone mice. Circulation. 2008;117:70-78.

7. Weber C, Aepfelbacher M, Haag H, Ziegler-Heitbrock HW, Weber PC. Tumor necrosis factor induces enhanced responses to platelet-activating factor and differentiation in human monocytic Mono Mac 6 cells. Eur J Immunol. 1993;23:852-859.

8. Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, Dewor M, Georgiev I, Schober A, Leng L, Kooistra T, Fingerle-Rowson G, Ghezzi P, Kleemann R, McColl SR, Bucala R, Hickey MJ, Weber C. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med. 2007;13:587-596.

at Bibliotheque Faculte Medecine Geneve on March 16, 2010 atvb.ahajournals.org

Downloaded from

(10)

Shagdarsuren et al.: JAM-C in neointima formation

6

mock JAM-CEGFP control

RMV 7 H33

control RMV 7

H33

EGFP

isotypecontrol RMV 7 H33

Jam-C+/+

LMECs

Jam-C-/- LMECs

CD31 H33

mock

B

C A

JAM-CEGFP

30 kDa

45 67 97

Jam-C+/+

LMECs

Jam-C-/- LMECs

mock JAM-CEGFP control

RMV 7 H33

control RMV 7

H33

EGFP

isotypecontrol RMV 7 H33

Jam-C+/+

LMECs

Jam-C-/- LMECs

CD31 H33

mock

B

C A

JAM-CEGFP

30 kDa

45 67 97

Jam-C+/+

LMECs

Jam-C-/- LMECs

Online Data Supplement Figure 1

The murine squamous cell carcinoma cell line KLN205 was mock transfected or stably transfected with a JAM-CEGFP expressing vector (A,B). Flow cytometric analysis was performed to detect EGFP in conjunction with JAM-C by H33 or integrin aV by RMV 7 in comparison to isotype matched control antibodies. Representative dot plots are shown (A).

JAM-C was immunoprecipitated from cell lysates and Western Blotting was performed, revealing a specific signal at the expected size of JAM-C (70 kDa) using H33 in JAM-CEGFP but not mock-transfected KLN205, while no signal for JAM-C could be detected using integrin aV or isotype control antibodies. The band at about 75 kDa represents unspecific binding (B).

Flow cytometry of primary lung microvascular endothelial cells (LMECs) revealed the expression of JAM-C by H33 (black line) on Jam-C+/+ LMECs but not Jam-C-/- mice, while CD31 (black line) could be detected on Jam-C+/+ and Jam-C-/- LMECs mice in comparison to the respective isotype matched control antibodies (filled grey profile). Representative histograms are shown (C).

(11)

Shagdarsuren et al.: JAM-C in neointima formation

7 SMαααα-actin

JAM-C calponin vWF

-

hCASMCs

HUVECs

- SMαααα-actin

JAM-C calponin vWF

-

hCASMCs

HUVECs

-

Online Data Supplement Figure 2

HUVECs and SMCs display a robust expression of JAM-C mRNA (459 base pair product), as evaluated by RT-PCR analysis. In addition, hCASMCs show a strong mRNA expression of the smooth muscle cell markers SM α-actin (358 base pairs) and calponin (471 base pairs) but not of the endothelial cell marker von Willebrand factor (vWF, 479 base pair). In contrast, HUVECs show no transcription of SM α-actin or calponin but a strong expression of vWF.

at Bibliotheque Faculte Medecine Geneve on March 16, 2010 atvb.ahajournals.org

Downloaded from

(12)

Shagdarsuren et al.: JAM-C in neointima formation

8 A

P-selectin JAM-C merge + DAPI

P-selectin JAM-C merge + DAPI

B

cellcount

FITC

isotype control JAM-C

control thrombin

cellcount

FITC

isotype control JAM-C

control thrombin

Online Data Supplement Figure 3

One day after injury, double-immunofluorescence staining revealed the accumulation and aggregation of P-selectin+ platelets in some injured segments of Apoe-/- carotid arteries (green), while no co-staining with JAM-C (red) could be detected; cell nuclei were stained by DAPI (blue, A). Flow cytometry of mouse platelets did not reveal any surface expression of JAM-C on unstimulated (control) or thrombin-activated mouse platelets (B).

Références

Documents relatifs

Absence of hyperplasia in Gasp-1 overexpressing mice is dependent on myostatin up-regulation Caroline Brun, Luce Périé, Fabienne Baraige, Barbara Vernus, Anne Bonnieu,

In the right panels, leukocyte infiltration per field was evaluated by semi-quantitative scoring for anti-JAM-C antibody- treated (n = 6, black columns) and isotype-matched

In cells transfected with wt JAM- 2 the few cell contact sites positive for JAM-2 (Fig. 8A, left panel) were also positive for PAR-3 or ZO-1 (data not shown), indicating that the

Analysis of JAM-C subcellular expression by immunoelectron microscopy indicated that in I/R-injured tissues, EC JAM-C was redistributed from cytoplasmic vesicles and EC junctional

GDF‑15 and CXCL10 levels were also significantly elevated at 24 h in irradiated coronary artery and microvascular endothelial (TICAE and TIME) cells in vitro after high dose of

We present two cases involving the late development of a sec- ond primary squamous cell carcinoma (SCC), arising in both the skin of a deltopectoral Xap and that of a free fore- arm

Disrupted polarized neutrophil paracellular TEM responses were relatively selectively observed under conditions of ischemia-reperfusion (I-R), an inflammatory insult that

We identified HNF1α as a human tumor suppressor gene involved in liver tumorigenesis, as we found biallelic inactivating mutations of this gene in ∼35-50% of HCA as well as a