antibody against junctional adhesion molecule-c inhibits ... · adhesion molecules with...

9
Antibody against Junctional Adhesion Molecule-C Inhibits Angiogenesis and Tumor Growth Chrystelle Lamagna, 1 Kairbaan M. Hodivala-Dilke, 2 Beat A. Imhof, 1 and Michel Aurrand-Lions 1 1 Department of Pathology and Immunology, Centre Me´dical Universitaire, Geneva, Switzerland and 2 Cell Adhesion and Disease Laboratory/Department of Tumour Biology, Bart’s and The London Queen Mary’s School of Medicine and Dentistry, John Vane Science Centre, London, United Kingdom Abstract The junctional adhesion molecule-C (JAM-C) was recently described as an adhesion molecule localized at interendothe- lial contacts and involved in leukocyte transendothelial mi- gration. The protein JAM-C interacts with polarity complex molecules and regulates the activity of the small GTPase Cdc42. The angiogenesis process involves rearrangement of endothelial junctions and implicates modulation of cell polarity. We tested whether JAM-C plays a role in angiogenesis using tumor grafts and hypoxia-induced retinal neovasculari- zation. Treatment with a monoclonal antibody directed against JAM-C reduces tumor growth and infiltration of macrophages into tumors. The antibody decreases angiogen- esis in the model of hypoxia-induced retinal neovasculariza- tion in vivo and vessel outgrowth from aortic rings in vitro . Importantly, the antibody does not induce pathologic side effects in vivo . These findings show for the first time a role for JAM-C in angiogenesis and define JAM-C as a valuable target for antitumor therapies. (Cancer Res 2005; 65(13): 5703-10) Introduction Angiogenesis is the formation of new blood vessels from the preexisting vasculature (1) and occurs in a multistep process involving migration, proliferation, and differentiation of endothelial cells leading to the formation of vascular loops (2). Angiogenesis occurs during embryonic development and adulthood during a variety of physiologic processes, including wound healing and the menstrual cycle of the endometrium (3, 4). Angiogenesis is also a key event in pathologic processes, such as tumor development (1) and diabetic retinopathy (5). The formation of immature endothelial sprouts is promoted by angiogenic factors, including vascular endothelial growth factor (VEGF) and angiopoietins. Maturation into functional vessels is then accomplished by the establishment of new interendothelial junctions, the organization of a new basement membrane, and the recruitment of pericytes (2). Altogether, these mechanisms result in the stabilization of a novel vascular network. Macrophages play an important role in regulating blood vessel formation by secreting angiogenic factors during tumor develop- ment as well as during physiologic angiogenesis (6, 7). Indeed, monocytes extravasate and migrate toward hypoxic or inflamma- tory regions created by a growing tumor (8). Although angiogen- esis is induced by angiogenic factors, additional molecules contribute to proliferation and remodeling of the vessel wall. As an example, VEGF leads to loosening of the pericyte-endothelial contacts, allowing proliferation and interaction of endothelial adhesion molecules with extracellular matrix (9). The integrins a v h 3 and a v h 5 participate in blood vessel development via a signaling cross-talk with receptors of angiogenic factors (10, 11). Other adhesion molecules implicated in the organization of interendothelial junctions are essential in maintaining integrity of the endothelium. For example, the targeted disruption in mice of the adherens junction molecule vascular endothelial-cadherin (VE-cadherin) leads to embryonic lethality due to impaired remodeling and maturation of vascular plexus (12, 13). More recently, it has been shown that the targeted disruption of the tight junction molecule, endothelial cell–selective adhesion mole- cule, inhibits angiogenesis in vitro and in vivo (14). In addition, in vitro experiments have shown that signaling through junc- tional adhesion molecule (JAM)-A and a v h 3 integrin is required for the angiogenic action of basic fibroblast growth factor (bFGF; refs. 15, 16). We recently described JAM-C and found it expressed in vascular cell-cell contacts (17, 18). When JAM-C is transfected into epithelial cells, it localizes in tight junctions, whereas it has been found in desmosomes of enterocytes (18, 19). We and others have shown that JAM-C is involved in leukocyte transendothelial migration (17, 18, 20, 21). Furthermore, JAM-C coimmunoprecipitates with polarity complex molecules, such as PAR-3, PAR-6, or PATJ, and regulates the activity of the small GTPase Cdc42 (22, 23). These results show that JAM-C plays a role in the formation and main- tenance of intercellular contacts and suggest that it may contribute to the remodeling of endothelial junctions. We therefore investigated whether JAM-C participates in angiogenesis, a mechanism involving rearrangement of endothelial junctions. Here, we show that a monoclonal antibody directed against JAM-C totally abolishes outgrowth of microvessels in ex vivo aortic ring assays. When injected in vivo , the antibody reduces hypoxia-induced angiogenesis in the retina and the growth of experimental tumors. These results show a role for JAM-C in angiogenesis and underline the importance of endothelial cell adhesion molecules in the formation of new blood vessels. Materials and Methods Antibodies. Rat monoclonal antibodies (CRAM) against human and mouse JAM-C (H33 for functional assays and H36 for immunocytochemistry) and rat monoclonal antibodies against mouse platelet/endothelial cell adhesion molecule-1 (PECAM-1)/CD31 (GC51) were described previously (18, 24). Anti-human CD44 (Hermes, 9B5) used as irrelevant antibody control rat IgG2a was kindly provided by Dr. B. Engelhardt (Theodor-Kocher- Institute, Bern, Switzerland) (25, 26). Monoclonal rat anti-mouse intercel- lular adhesion molecule-2 (ICAM-2; 3C4) and monoclonal rat anti-mouse Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Requests for reprints: Beat A. Imhof, Department of Pathology and Immunology, Centre Me ´dical Universitaire, 1 rue Michel-Servet, 1204 Geneva, Switzerland. Phone: 41-22-379-57-47; Fax: 41-22-379-57-46; E-mail: [email protected]. I2005 American Association for Cancer Research. www.aacrjournals.org 5703 Cancer Res 2005; 65: (13). July 1, 2005 Research Article Research. on December 22, 2020. © 2005 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Upload: others

Post on 31-Aug-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Antibody against Junctional Adhesion Molecule-C Inhibits ... · adhesion molecules with extracellular matrix (9). The integrins a vh 3 and a vh 5 participate in blood vessel development

Antibody against Junctional Adhesion Molecule-C Inhibits

Angiogenesis and Tumor Growth

Chrystelle Lamagna,1Kairbaan M. Hodivala-Dilke,

2Beat A. Imhof,

1and Michel Aurrand-Lions

1

1Department of Pathology and Immunology, Centre Medical Universitaire, Geneva, Switzerland and 2Cell Adhesion and DiseaseLaboratory/Department of Tumour Biology, Bart’s and The London Queen Mary’s School of Medicine and Dentistry,John Vane Science Centre, London, United Kingdom

Abstract

The junctional adhesion molecule-C (JAM-C) was recentlydescribed as an adhesion molecule localized at interendothe-lial contacts and involved in leukocyte transendothelial mi-gration. The protein JAM-C interacts with polarity complexmolecules and regulates the activity of the small GTPaseCdc42. The angiogenesis process involves rearrangement ofendothelial junctions and implicates modulation of cellpolarity. We tested whether JAM-C plays a role in angiogenesisusing tumor grafts and hypoxia-induced retinal neovasculari-zation. Treatment with a monoclonal antibody directedagainst JAM-C reduces tumor growth and infiltration ofmacrophages into tumors. The antibody decreases angiogen-esis in the model of hypoxia-induced retinal neovasculariza-tion in vivo and vessel outgrowth from aortic rings in vitro.Importantly, the antibody does not induce pathologic sideeffects in vivo . These findings show for the first time a role forJAM-C in angiogenesis and define JAM-C as a valuable targetfor antitumor therapies. (Cancer Res 2005; 65(13): 5703-10)

Introduction

Angiogenesis is the formation of new blood vessels from thepreexisting vasculature (1) and occurs in a multistep processinvolving migration, proliferation, and differentiation of endothelialcells leading to the formation of vascular loops (2). Angiogenesisoccurs during embryonic development and adulthood during avariety of physiologic processes, including wound healing and themenstrual cycle of the endometrium (3, 4). Angiogenesis is also akey event in pathologic processes, such as tumor development (1)and diabetic retinopathy (5). The formation of immatureendothelial sprouts is promoted by angiogenic factors, includingvascular endothelial growth factor (VEGF) and angiopoietins.Maturation into functional vessels is then accomplished by theestablishment of new interendothelial junctions, the organizationof a new basement membrane, and the recruitment of pericytes (2).Altogether, these mechanisms result in the stabilization of a novelvascular network.

Macrophages play an important role in regulating blood vesselformation by secreting angiogenic factors during tumor develop-ment as well as during physiologic angiogenesis (6, 7). Indeed,monocytes extravasate and migrate toward hypoxic or inflamma-tory regions created by a growing tumor (8). Although angiogen-

esis is induced by angiogenic factors, additional moleculescontribute to proliferation and remodeling of the vessel wall. Asan example, VEGF leads to loosening of the pericyte-endothelialcontacts, allowing proliferation and interaction of endothelialadhesion molecules with extracellular matrix (9). The integrinsavh3 and avh5 participate in blood vessel development via asignaling cross-talk with receptors of angiogenic factors (10, 11).

Other adhesion molecules implicated in the organization ofinterendothelial junctions are essential in maintaining integrity ofthe endothelium. For example, the targeted disruption in miceof the adherens junction molecule vascular endothelial-cadherin(VE-cadherin) leads to embryonic lethality due to impairedremodeling and maturation of vascular plexus (12, 13). Morerecently, it has been shown that the targeted disruption of thetight junction molecule, endothelial cell–selective adhesion mole-cule, inhibits angiogenesis in vitro and in vivo (14). In addition,in vitro experiments have shown that signaling through junc-tional adhesion molecule (JAM)-A and avh3 integrin is requiredfor the angiogenic action of basic fibroblast growth factor (bFGF;refs. 15, 16).

We recently described JAM-C and found it expressed in vascularcell-cell contacts (17, 18). When JAM-C is transfected into epithelialcells, it localizes in tight junctions, whereas it has been found indesmosomes of enterocytes (18, 19). We and others have shownthat JAM-C is involved in leukocyte transendothelial migration(17, 18, 20, 21). Furthermore, JAM-C coimmunoprecipitates withpolarity complex molecules, such as PAR-3, PAR-6, or PATJ, andregulates the activity of the small GTPase Cdc42 (22, 23). Theseresults show that JAM-C plays a role in the formation and main-tenance of intercellular contacts and suggest that it may contributeto the remodeling of endothelial junctions.

We therefore investigated whether JAM-C participates inangiogenesis, a mechanism involving rearrangement of endothelialjunctions. Here, we show that a monoclonal antibody directedagainst JAM-C totally abolishes outgrowth of microvessels inex vivo aortic ring assays. When injected in vivo , the antibodyreduces hypoxia-induced angiogenesis in the retina and the growthof experimental tumors. These results show a role for JAM-C inangiogenesis and underline the importance of endothelial celladhesion molecules in the formation of new blood vessels.

Materials and Methods

Antibodies. Rat monoclonal antibodies (CRAM) against human and

mouse JAM-C (H33 for functional assays and H36 for immunocytochemistry)and rat monoclonal antibodies against mouse platelet/endothelial cell

adhesion molecule-1 (PECAM-1)/CD31 (GC51) were described previously

(18, 24). Anti-human CD44 (Hermes, 9B5) used as irrelevant antibody controlrat IgG2a was kindly provided by Dr. B. Engelhardt (Theodor-Kocher-

Institute, Bern, Switzerland) (25, 26). Monoclonal rat anti-mouse intercel-

lular adhesion molecule-2 (ICAM-2; 3C4) and monoclonal rat anti-mouse

Note: Supplementary data for this article are available at Cancer Research Online(http://cancerres.aacrjournals.org/).

Requests for reprints: Beat A. Imhof, Department of Pathology and Immunology,Centre Medical Universitaire, 1 rue Michel-Servet, 1204 Geneva, Switzerland. Phone:41-22-379-57-47; Fax: 41-22-379-57-46; E-mail: [email protected].

I2005 American Association for Cancer Research.

www.aacrjournals.org 5703 Cancer Res 2005; 65: (13). July 1, 2005

Research Article

Research. on December 22, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Antibody against Junctional Adhesion Molecule-C Inhibits ... · adhesion molecules with extracellular matrix (9). The integrins a vh 3 and a vh 5 participate in blood vessel development

Fcg II/III receptor (2.4G2) were from BD PharMingen (Franklin Lakes, NJ).Polyclonal rabbit antibody against PECAM-1/CD31 was described previously

(27). A polyclonal rabbit serum against murine JAM-C was generated using

recombinant soluble molecule as immunogen (Covalab, Lyon, France). The

specificity of polyclonal antibody was confirmed by immunohistochemistryon tissue sections, Western blot, and flow cytometry using transfected cells.

Anti-JAM-C antibody H33 and isotype-matched control antibody 9B5

used in functional assays were tested for endotoxin using the Pyrogent Plus

Limulus Amebocyte Lysate kit (BioWhittaker, Walkersville, MD).Isolation and culture of primary endothelial cells. Human umbilical

vein endothelial cells (HUVEC) were isolated and cultured as described

previously in ref. 28. Cells were used between passages 3 and 5.

Lung murine endothelial cells were isolated from wild-type C56BL6/J

mice (Charles River Laboratories, L’Arbresle, France) as described in ref. 29.

Briefly, lungs were harvested from two mice, carefully dissected, and washed

twice in 20 mL Ham’s F-12 medium supplemented with antibiotics. Then,

lungs were finely minced with scissors and digested in 10 mL collagenase

(180-200 units/mL) at 37jC for 1 hour. The digested tissue was then

mechanically dissociated by triturating, filtered through a 70 Am disposable

cell strainer (Becton Dickinson Labware, Bedford, MA), and centrifuged at

1,200 � g for 5 minutes. The cell pellet was resuspended in 50:50 mix of

Ham’s F-12 medium/DMEM (Life Technologies/Invitrogen Corporation,

Basel, Switzerland) supplemented with 20% FCS, 20 Ag/mL endothelial cell

mitogen (Biogenesis, Poole, United Kingdom), 1 Ag/mL heparin (Sigma-

Aldrich Corp., St. Louis, MO), and antibiotics (complete medium). The cell

suspension was plated on flasks precoated with 0.1% gelatin, 10 Ag/mL

fibronectin (Sigma-Aldrich), and 30 Ag/mL Vitrogen (Nutacon B.V.,

Leimuiden, the Netherlands) in PBS. Endothelial cells were purified by

magnetic immunosorting with a single negative sort for Fcg III/II receptor-

positive macrophages and at least two positive sorts for ICAM-2-positive

endothelial cells. Immunosorting was done using sheep anti-rat IgG Dynal

beads (Dynal Biotech, Oslo, Norway). Cells were cultured routinely in the

complete medium described above.Vascular endothelial growth factor stimulation and immunocyto-

chemistry. HUVECs (1 � 105) were plated on 22 mm2 glass slides coated

with growth factor–reduced Matrigel (Becton Dickinson, Bedford, MA) and

after 24 hours starved for endothelial cell growth factors. Twenty-four hourslater, cells were incubated with 100 ng/mL recombinant human VEGF-165

(PeproTech House, London, United Kingdom) for 15 minutes.

For immunocytochemistry, cells were fixed with 4% paraformaldehyde

in PBS for 15 minutes before permeabilization with 0.01% Triton X-100in PBS for 10 minutes. Cells were washed with PBS/0.2% bovine serum

albumin (BSA), incubated with primary antibody H36 for 1 hour, and

washed before further incubation with secondary antibody coupled toFITC (Jackson Immunoresearch Laboratories, Inc., West Grove, PA).

Pictures were acquired using a Zeiss LSM510 confocal microscope (Zeiss,

Oberkochen, Germany).

Flow cytometry. HUVEC and LLC1 were incubated on ice with H36and H33 anti-JAM-C monoclonal antibodies, respectively. After washing with

PBS/0.2% BSA, binding of antibody was detected using a phycoerythrin-

coupled anti-rat antibody ( Jackson Immunoresearch Laboratories).

As control, the primary antibody was omitted. The surface content ofproteins was analyzed using FACSCalibur and CellQuest software (Becton

Dickinson, Mountain View, CA).

Histology and quantification of vascular volume fraction, apoptoticcells, and macrophage contents into the tumors. For immunohisto-

chemistry on tumor cryosections with monoclonal antibody anti-PECAM-1

(GC51), sections were fixed with acetone/methanol (1:1) for 5 minutes at

�20jC, dried, and hydrated in PBS/0.2% gelatin/0.05% Tween 20. Sectionswere incubated with antibody for 1 hour at room temperature and after

three washes in PBS incubated with a secondary antibody coupled to

peroxidase (Jackson Immunoresearch Laboratories).

For immunohistochemistry on paraformaldehyde-fixed and paraffin-embedded eye sections with polyclonal antibodies against PECAM-1 and

JAM-C, sections were dewaxed following the classic procedure. Tissue

sections were then treated with H2O2 0.3% in methanol for 10 minutes,

washed in PBS, and blocked with PBS/3% BSA/0.1% Tween 20 for

30 minutes. Sections were incubated with polyclonal antibodies for 1 hourat room temperature and after washes in PBS incubated with EnVision

system for 30 minutes (DakoCytomation AG, Baar, Switzerland). Peroxidase

activity was detected using 3-amino-9-ethylcarbazol (Sigma-Aldrich) as

substrate and sections were counterstained for 1 minute with hemalumbefore mounting in Aquatex (Merck, Darmstadt, Germany).

Acid phosphatase activity was detected on tumor cryosections using

the method described previously in ref. 30. Detection of apoptotic cells on

tumor cryosections was based on labeling of DNA strand breaks [terminaldeoxynucleotidyl transferase–mediated dUTP nick end labeling (TUNEL)-

fluorescence method] and done using terminal transferase and biotin-16-

dUTP according to the manufacturer’s instructions (Roche Diagnostics

AG, Rotkreuz, Switzerland). Bound biotin-16-dUTP was detected withstreptavidin coupled to Texas red dye ( Jackson Immunoresearch

Laboratories).

Pictures were acquired using a Zeiss LSM510 confocal microscope or aZeiss Axiophot 1 microscope equipped with an Axiocam color CCD camera.

Images were recorded and treated using the AxioVision software (Zeiss).

To quantify vascular volume fraction, the number of macrophages, and

the number of apoptotic cells into tumors, pictures of the entire cryosection

( four cryosections per tumor) were analyzed using Zeiss KS400 or Openlab

software. The vascular volume fraction was quantified by determining the

total area of PECAM-1-positive staining across whole sections of tumors.

Results are expressed as percentage of PECAM-1 staining by applying the

formula: % PECAM-1 staining = [Total area of PECAM-1 staining (mm2)] /

[Total area of tumor section (mm2)] � 100. The number of acid

phosphatase–positive cells and the number of TUNEL-positive cells present

across the entire area of each tumor section were counted and divided by

the area of the whole section. This determined the number of macrophages

per square millimeter and the number of apoptotic cells per square

millimeter, respectively.

Ex vivo aortic ring assay. Mouse aortic ring assay was adapted from

ref. 31. Briefly, 1 mm thoracic aortic rings were placed between two layers of50 AL growth factor–reduced Matrigel and overlaid with 100 AL DMEM

supplemented with 20% FCS, 20 units/mL heparin, and endothelial cell

growth supplement (Upstate Biotechnology, Lake Placid, NY) in the

presence or absence of 50 Ag/mL 9B5 or H33 antibodies. Microvesseloutgrowth was visualized by phase microscopy using a Zeiss Axioskop

microscope.

Tumor graft. Eight- to 10-week-old female C56BL6/J mice were

inoculated s.c. with 1 � 106 murine Lewis lung carcinoma cells (LLC1;obtained from the European Collection of Cell Cultures, Salisbury, United

Kingdom). Mice were then injected i.p. every second day with 150 Agmonoclonal antibody H33, isotype-matched control antibody 9B5, or PBS.When the control tumors (PBS-injected mice) had reached 1 cm, animals

were sacrificed and tumors were excised and analyzed. Tumor volume

was measured by using a caliper, applying the following formula for

approximating the volume of an ellipsoid: Volume (mm3) = 4/3p �(Length / 2) � (Width / 2) � (Height / 2).

Hypoxia-induced retinal angiogenesis. Postnatal day 7 (P7) mice

were placed in 75% oxygen in air for 5 days causing central avascularization

of retinas followed by housing the mice for 5 additional days (until P17)

under normoxic conditions. Mice were injected i.p. with 50 Ag monoclonal

antibodies at P12, P14, and P16. After anesthesia with 150 mg/kg ketamin

plus 12.5 mg/kg xylazine, 17-day-old mice were perfused with a

nondiffusible fluorescein-dextran solution (Sigma-Aldrich). Neovasculariza-

tion was visualized and quantified on flat-mounted retinas by counting the

number of vascular glomeruli under the microscope. Pictures were acquired

using a Zeiss LSM510 confocal microscope.Proliferation assays. Primary mouse lung endothelial cells or LLC1

tumor cells were plated at a density of 2,500 cells/cm2 in 24- or 6-well plates,

respectively. Twenty-four hours later, the medium was replaced by medium

supplemented with control or H33 antibodies at 50 Ag/mL. Cells weretrypsinized from wells daily for 4 days and counted using a Casy-1 Coulter

counter (Scharfe System GmbH, Reutlingen, Germany).

Real-time quantitative PCR. Retinal mRNA from newborn miceundergone hypoxia-induced angiogenesis were extracted by Trizol

Cancer Research

Cancer Res 2005; 65: (13). July 1, 2005 5704 www.aacrjournals.org

Research. on December 22, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Antibody against Junctional Adhesion Molecule-C Inhibits ... · adhesion molecules with extracellular matrix (9). The integrins a vh 3 and a vh 5 participate in blood vessel development

according to the manufacturer’s instructions (Life Technologies/Invitrogen). Reverse transcription was done by using 1 Ag total RNA,

random hexanucleotide primers, and SuperScript II reverse transcriptase

(Invitrogen). 1:25 Dilution of the resulting cDNA was used for real-time

quantitative PCR using the SYBR Green PCR Master Mix kit asrecommended by the provider and an ABI PRISM 7900HT Sequence

Detection System (Applied Biosystems, Foster City, CA). The following

primers were used: JAM-C forward 5V-GCTGGGAGAGCACATGCAA and

reverse 5V-CAGGAGCTCTGGGCTCACA, RPS-9 forward 5V-GACCAGGAGC-TAAAGTTGATTGGA and reverse 5V-TCTTGGCCAGGGTAAACTTGA, andTBP forward 5V-TTGACCTAAAGACCATTGCACTTC and reverse 5V-TTCTC-ATGATGACTGCAGCAAA. JAM-C relative mRNA content was normalized

by geometric averaging of internal control genes RPS-9 and TBP accordingto ref. 32.

Statistical analysis. Each column in graphs represents the mean F SE.

All experiments were evaluated with the Mann-Whitney’s t test usingthe statistical software StatView (Abacus Concepts, Inc., Berkeley, CA).

P < 0.05 was considered as statistically significant.

Animal regulation. All animal experiments were done under the ethical

approval and the recommendations of the Veterinary Office of Geneva stateaccording to the Swiss federal laws.

Results

Vascular endothelial cells express junctional adhesionmolecule-C and vascular endothelial growth factor changesits localization. The adhesion molecule JAM-C is constitutivelyexpressed by endothelial cells and partially localized in cell-cellcontacts (21). We investigated by fluorescence-activated cellsorting analysis whether the level of JAM-C proteins at thesurface of endothelial cells can be regulated by angiogenicstimuli. Treatment with the angiogenic factor VEGF did notchange JAM-C surface expression levels (Fig. 1A). However,analysis of the distribution of the molecule in VEGF-stimulatedendothelial monolayer by immunocytochemistry revealed aredistribution of the existing JAM-C in cell-cell contacts withinminutes (Fig. 1B and C). Other endothelial adhesion molecules,such as JAM-A or PECAM-1, were not redistributed on VEGFstimulation (data not shown). Because VEGF is a promoter ofangiogenesis, this result prompted us to investigate whetherJAM-C may play a role in the reorganization of endothelial cellsjunctions during angiogenesis.

In vitro vessel outgrowth is inhibited by anti–junctionaladhesion molecule-C monoclonal antibody. We first testedwhether anti-JAM-C antibody would interfere with angiogenesisusing ex vivo aortic ring assays. This consisted of embeddingfreshly dissected mouse aortic rings into Matrigel in the presenceof endothelial growth factors. Outgrowths of vascular sprouts fromaortic rings were followed over a period of 12 days of culture(Fig. 2). The anti-JAM-C antibody H33 blocked vessel outgrowth,whereas control antibody had no effect (Fig. 2). These in vitroexperiments suggested a role for JAM-C in angiogenesis andshowed that anti-JAM-C antibodies might be valuable toolspreventing angiogenesis in vivo .Anti–junctional adhesion molecule-C monoclonal antibody

reduces tumor growth and angiogenesis in vivo . Given thefinding that in vitro vessel outgrowth can be blocked with theH33 anti-JAM-C antibody, we investigated whether it affectedtumor angiogenesis and tumor growth. Mice were s.c. injectedwith Lewis lung carcinoma cells (LLC1) and the antibodies weregiven i.p. every second day. Tumor size, volume, and weightwere significantly decreased when mice were treated with H33 anti-JAM-C antibody compared with isotype-matched control antibodyor PBS (Fig. 3A-C). Because LLC1 tumor cells did not expressJAM-C (Fig. 4A), we suggested that the reduction in tumorgrowth was due to an effect of the H33 antibody on endothelialcells.To visualize the tumor vasculature, tumor vessels were labeled

with the endothelial marker PECAM-1 (Fig. 4). The vascularvolume fraction was then quantified by calculating the surface ofPECAM-1 staining across tumor sections. Tumors excised fromH33-treated animals exhibited a reduction in the percentage ofPECAM-1 staining when compared with control animals (Fig. 4B).To deliver a conclusive proof of JAM-C implication in angio-

genesis, we then used the antibody in hypoxia-induced retinopathyin neonatal mice, a model of tumor-independent neovasculariza-tion. Central avascularization of the retina of P7 mice was causedby exposure of the animals to 75% oxygen in air during 5 days. Then,by housing the mice until P17 under normoxia, retinal neo-vascularization was induced. During this period, mice were injectedwith anti-JAM-C or control monoclonal antibodies. Neovasculariza-tion was detected by fluorescence microscopy after perfusion of

Figure 1. JAM-C is redistributed to interendothelial junctions of HUVECs on VEGF stimulation. HUVECs were stimulated with 100 ng/mL VEGF. Surface expressionlevel and subcellular localization of JAM-C were analyzed by flow cytofluorometry (A) and immunofluorescent staining (B and C ), respectively. A, JAM-C surfaceexpression level is not changed on VEGF stimulation. Gray line, negative control, primary antibody omitted; dashed line, nontreated cells labeled with anti-JAM-Cantibody H36; bold line, VEGF-treated cells labeled with anti-JAM-C antibody H36. B, JAM-C is barely detectable at interendothelial contacts in the absence of VEGFstimulation. C, after 15 minutes of VEGF treatment, the JAM-C molecule is redistributed to intercellular contacts of HUVEC. Bar, 20 Am.

Anti–JAM-C Inhibits Angiogenesis

www.aacrjournals.org 5705 Cancer Res 2005; 65: (13). July 1, 2005

Research. on December 22, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Antibody against Junctional Adhesion Molecule-C Inhibits ... · adhesion molecules with extracellular matrix (9). The integrins a vh 3 and a vh 5 participate in blood vessel development

the entire vasculature with a nondiffusible fluorescein-dextran.Vascular glomeruli, corresponding to highly proliferating clustersof vessels, were counted as a measure of neovascularization (10).As shown in Fig. 5A and B , the number of glomeruli wassignificantly reduced when mice were treated with the H33 anti-JAM-C antibody. It is important to note that de novo formedvessels and vascular glomeruli expressed JAM-C, as do retinalvessels of control mice that have not been exposed to hypoxia(Fig. 5C). Taken together, these in vivo experiments showed thatanti-JAM-C antibody reduced pathologic angiogenesis.Junctional adhesion molecule-C expression is not regulated

during retinal angiogenesis. In Fig. 1, we described that JAM-Csurface expression level was not modified on VEGF stimulationof endothelial cells in vitro . We explored whether this absence ofregulation would also be found in vivo . To this end, retinal mRNAextracts from mice that have undergone hypoxia-induced retinalangiogenesis were isolated at different time points of the exper-iment and subjected to real-time quantitative PCR. As a control,

retinal mRNA from nonexposed newborn and adult mice wereprocessed in parallel. In agreement with in vitro observations, JAM-C mRNA contents were not regulated during the time course ofthe angiogenic process (Fig. 5D). These results support the hypo-thesis that the redistribution of JAM-C might play an integralpart in the angiogenic function as already found in vitro (Fig. 1Band C).H33 anti–junctional adhesion molecule-C antibody has no

effect on endothelial cell proliferation or apoptosis. Angio-genesis is a complex process orchestrated by the proliferation andarchitectural reorganization of endothelial cells on angiogenicstimuli. We first tested whether the H33 antibody blocksangiogenesis by inhibiting proliferation of lung primary endothe-lial cells in vitro and found no effect (Fig. 6A). To avoid any directconsequence of H33 treatment on tumor cell proliferation, wealso did the experiment with tumor cells in vitro and no effectwas detectable (Fig. 6B). These results indicated that thereduction of angiogenesis observed after H33 administration

Figure 2. Anti-JAM-C monoclonal antibody abolishes angiogenesis in vitro . Aortic rings from C57BL6/J mice were embedded in Matrigel layers in the presenceor absence of antibodies (50 Ag/mL). Neovascularization was followed over a period of 12 days. Micrographs show outgrowth of microvessels from aortic rings.Only H33 anti-JAM-C antibody blocked angiogenic sprouting. Nontreated (n = 11), isotype-matched control antibody (n = 6), and anti-JAM-C antibody H33 (n = 11).Bar, 500 Am.

Figure 3. Anti-JAM-C antibody H33treatment reduces tumor growth. C57BL6/Jmice were injected s.c. with Lewis lungcarcinoma cells (LLC1) into the flank. Micereceived i.p. injections of either PBS,isotype-matched control antibody, oranti-JAM-C antibody H33 every secondday (150 Ag). When control tumors(PBS-injected mice) reached the maximumallowed size, all tumors were excised andanalyzed. A, macroscopic aspects of12-day-old tumors grown in mice treatedwith PBS, control antibody, or H33anti-JAM-C antibody. Mice treated withH33 antibody showed reduced tumorgrowth as indicated by measurement oftumor volume (B ) and tumor weight (C ).PBS (n = 6), control antibody (n = 6), andH33 (n = 9). Columns, mean; bars, SE.Representative experiment of four.*, P < 0.05; **, P < 0.01. Bar, 0.5 cm.

Cancer Research

Cancer Res 2005; 65: (13). July 1, 2005 5706 www.aacrjournals.org

Research. on December 22, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Antibody against Junctional Adhesion Molecule-C Inhibits ... · adhesion molecules with extracellular matrix (9). The integrins a vh 3 and a vh 5 participate in blood vessel development

was not caused by prevention of vascular or tumor cell growth.Stimulation of apoptosis would be another explanation for thereduction of angiogenesis induced by H33 treatment. We testedthis hypothesis on tumor sections by identifying endothelial cellswith PECAM-1 staining and apoptosis by a standard labelingprotocol of apoptotic cells (TUNEL). Results revealed that H33antibody had no consequence on endothelial cell apoptosis in vivoand in vitro (Fig. 6C ; data not shown). However, tumor cellsshowed increased TUNEL labeling in vivo , suggesting thatapoptosis occurred as a consequence of antibody-mediatedreduced vascularization (Fig. 6C).H33 anti–junctional adhesion molecule-C antibody has no

pathologic side effect in vivo . It has been shown that antibodiescan be toxic when injected in vivo by increasing vascularpermeability or inducing organ failure (33, 34). We injected animalswith the H33 anti-JAM-C antibody and tested whether it inducedpathologic side effects in vivo . Because JAM-C is expressed byendothelium in the kidney, we first analyzed whether the antibodywould affect the normal function of the kidney. Using histochem-istry and periodic acid-Schiff staining, we did not observe accu-mulation of proteins in tubules and the morphology of glomeruliappeared normal (Supplementary Fig. S1A). Because JAM-C hasbeen involved in controlling paracellular permeability of mono-layers (18), we tested whether the antibody would increasepermeability of blood vessels in vivo . No change in permeabilitywas detectable in the organs analyzed, such as heart, kidney, andbrain (Supplementary Fig. S1B).H33 anti–junctional adhesion molecule-C antibody reduces

the recruitment of macrophages into the tumors. Tumorangiogenesis is often accompanied by inflammation and macro-phages represent prominent tumor-associated inflammatory cells(6). Indeed, macrophages participate in angiogenesis by secretingangiogenic factors, such as VEGF, mostly under hypoxicconditions (8). JAM-C is implicated in leukocyte adhesion andtransmigration through endothelial and epithelial cells (19, 20, 28,35). We thus investigated whether H33 antibody might affectrecruitment of macrophages into tumors. As shown in Fig. 6D ,

mice treated with H33 antibody showed reduced macrophagecontent in tumors compared with control mice. This indicatesthat the H33 effect on angiogenesis is mediated in part via itsaction on recruitment of macrophages.

Discussion

In the present study, we show that the administration of amonoclonal antibody directed against JAM-C reduces angiogenesisin two different in vivo models: hypoxia-induced neovasculariza-tion of the retina and angiogenic vascularization of tumors. Weshow for the first time an important role for a member of the JAMfamily in angiogenesis in vivo . More importantly, JAM-C is nowvalidated as a potential target for antiangiogenic treatment.We reported previously that the molecule JAM-C is present on

resting endothelium in different organs (18). Thus, the angiogenesis-blocking antibody H33 does not exclusively bind to angiogeniczones of the vasculature. However, the H33 antibody has no effecton in vivo vascular permeability, suggesting that the antibodydoes not interfere with established interendothelial junctions. Inaddition, the antibody does not induce other detrimental effectson the resting endothelium, such as kidney endothelial cells,which express large amount of JAM-C. These observations suggestthat H33 antibody neither interferes with the integrity ofestablished interendothelial junctions nor creates vascular immu-nopathologies but rather interferes with a specific function ofangiogenic endothelium.In the exploration of anticancer treatments, antibody-based ther-

apies have been shown to be good strategies to reducevascularization of tumors and to limit their growth (36). Severalgrowth factor receptors and adhesion molecules, such as VEGFreceptor-2 (VEGFR-2), VE-cadherin, or avh3 integrin, have beensuccessfully exploited as targets for antibody-based therapies(33, 37, 38). Although the mechanisms of action and the doses ofantibodies required to get antiangiogenic effect differ from onetarget to the other, these strategies always consist in perturbing themechanisms involved in migration, proliferation, or apoptosis ofendothelial cells (33, 37, 38).

Figure 4. Anti-JAM-C antibody H33reduces tumor vascularization. Todetermine whether LLC1 express JAM-C,tumor cells were incubated with H33anti-JAM-C monoclonal antibody (A, bold )and the protein content was analyzed byfluorescence-activated cell sorting. Ascontrol, the primary antibody was omitted(A, gray). LLC1 tumor cells do not expressJAM-C. Tumor cryosections fromPBS-, control antibody–, or H33-treatedmice were thus stained for the endothelialmarker PECAM-1 and the vascular volumefraction was quantified by counting thepercentage of PECAM-1 staining persection as described in Materials andMethods (B). Representative pictures areshown in (C ). Tumors from H33-treatedanimals show reduced vascularizationcompared with control animals. Columns,mean obtained from four sections of n miceper group (PBS, n = 6; control antibody,n = 6; H33, n = 9); bars, SE. Bar, 160 Am.

Anti–JAM-C Inhibits Angiogenesis

www.aacrjournals.org 5707 Cancer Res 2005; 65: (13). July 1, 2005

Research. on December 22, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Antibody against Junctional Adhesion Molecule-C Inhibits ... · adhesion molecules with extracellular matrix (9). The integrins a vh 3 and a vh 5 participate in blood vessel development

Based on the comparison of results obtained with antibodiesand knockout mice on angiogenesis, it is possible to classify thetargets into two categories: (a) targets antagonized in a fashionmimicking their absence and (b) targets used to transduce a spe-cific signal in angiogenic endothelial cells. The endothelial mol-ecules VE-cadherin and VEGFR-2 belong to the first category.Indeed, the targeted inactivation of these genes lead to embryoniclethality due to vasculogenesis defects (12, 39). In contrast, micelacking av or h3 integrin subunits survive and maintaindevelopmental blood vessel formation as well as angiogenesis inadult mice (10, 40). The integrin avh3 is known to mediate celladhesion to extracellular matrix. This receptor-ligand interactionleads to outside-in signaling, which delivers intracellular survivalsignals. The contradiction between the antiangiogenic effect ofavh3 integrin inhibitors and enhanced angiogenesis in avh3-deficient animals has been explained as follows: when antagonistsblock engagement of avh3 with its ligands, the survival signal isswitched into a death signal. In contrast, when the avh3 integrin isnot expressed, the induction of apoptotic signals is absent (9).Likewise to avh3 integrin, we show that antibody against JAM-Creduces angiogenesis, although preliminary data suggest that the

genetic inactivation of JAM-C may not lead to apparent vasculardefects (23). This indicates that the antibody H33 targets a specificfunction of JAM-C on angiogenic endothelial cells. However, we canexclude an effect on apoptosis or proliferation of endothelial cells,indicating that the antibody acts by a different mechanism.Our findings indicate that the effect of H33 antibody on tumor

growth may be due to impaired recruitment of monocytes intoneovascularized areas. It is well described that local hypoxiainduces secretion of chemokines by tumor cells responsible formonocyte recruitment (8, 41). Hypoxia also stimulates thesecretion of angiogenic factors by monocytes/macrophages (42).Hence, tumor-associated angiogenesis is enhanced by thepresence of macrophages. Recently, we showed that antibodyH33 affects the adhesion of monocytes to the endothelium bymodulating the availability of endothelial JAM-C for themonocyte integrin aMh2.

3 We propose that JAM-C is alsoimplicated in the recruitment of macrophages into tumors.Nevertheless, we cannot distinguish whether the reduction of

Figure 5. H33 antibody reduces hypoxia-induced retinal angiogenesis. Newborn mice (P7) were exposed for 5 days to 75% oxygen in air, producing a centralavascularization of the retina. Mice were then housed from P12 to P17 under normoxia leading to a hypoxia-induced situation and to neovascularization of the retina.Mice at P12, P14, and P16 were i.p. injected with PBS (n = 9), isotype-matched control antibody (n = 9), or H33 antibody (n = 13; 50 Ag/injection). Mice at P17 wereperfused with a nondiffusible FITC-dextran solution to visualize vasculature and were sacrificed, and retinal angiogenesis was analyzed. A, flat mounts of retinas frommice treated with control or H33 antibody. H33-treated retinas showed a reduced number of vascular glomeruli, which corresponded to highly proliferative clusters ofvessels produced in response to hypoxia (arrowheads ) compared with control retinas. Bar , 200 Am. B, number of vascular glomeruli was counted under themicroscope. Columns, mean; bars, SE. ***, P < 0.001. C, serial sections of retina from P17 mice nonexposed (top ) or exposed to hypoxia (bottom ) wereimmunostained with polyclonal antibodies raised against JAM-C or PECAM-1. JAM-C is exclusively detected on blood vessels (arrow ) and glomeruli (arrowhead ).Bar, 20 Am. D, retinal JAM-C mRNA contents were quantified by real-time quantitative PCR. JAM-C expression level is not regulated by hypoxia compared withcontrol animals.

3 Submitted for publication.

Cancer Research

Cancer Res 2005; 65: (13). July 1, 2005 5708 www.aacrjournals.org

Research. on December 22, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Antibody against Junctional Adhesion Molecule-C Inhibits ... · adhesion molecules with extracellular matrix (9). The integrins a vh 3 and a vh 5 participate in blood vessel development

tumor-associated macrophages is cause or consequence ofreduced tumor vascularization in H33-treated mice. Indeed, theantibody abolishes vessel outgrowth in ex vivo aortic ring assaysdone in the presence of angiogenic growth factors and in theabsence of possible recruitment of macrophages in culturedishes. This indicates that H33 antibody has also a direct,monocyte-independent effect on angiogenesis.Although we can exclude an effect of the antibody H33

on endothelial cell proliferation or apoptosis, an alternateexplanation for a direct effect of H33 antibody on endothelialcells could have been the reduction of endothelial cell migration.Indeed, angiogenesis depends in part on migration of endothelialcells, mainly supported by the coordination of signals fromgrowth factors and the extracellular matrix (10, 43, 44). Moreover,regulation of cell-cell adhesion is also critical for migration. Forexample, VE-cadherin supports a cross-talk with VEGFR-2signaling, which allows sprouting and endothelial cell migrationduring angiogenesis (45). A cross-talk was also suggested between

JAM-A and bFGF to induce endothelial cell migration (15, 16).Nevertheless, at this time, no results have confirmed a role forJAM-A in angiogenesis in vivo . We found that H33 antibody doesnot affect migration of primary lung endothelial cells in responseto VEGF in vitro (Supplementary Fig. S2). However, we cannotexclude that JAM-C plays a role in migration of endothelial cellsin vivo , indicating that the antiangiogenic effect of anti-JAM-Cantibody is indeed more complex. Whether JAM-C interacts withvascular growth factor receptors or extracellular matrix receptors,such as integrins, awaits further investigations.Interestingly, we observe that JAM-C is relocalized in cell-cell

contacts on VEGF or tumor necrosis factor-a stimulation (Fig. 1;data not shown). Such stimuli have been involved in activation ofsmall Rho GTPases in human endothelial cells (46–48). Inaddition, the small GTPases Cdc42 and Rac have been implicatedin capillary lumen formation in three-dimensional extracellularmatrixes in vitro (49). We showed recently that JAM-C modulatesthe localization and activity of Cdc42, which is essential for the

Figure 6. H33 antibody affects neither proliferation nor apoptosis of endothelial cells. Primary endothelial cells isolated from murine lungs (A) or LLC1 tumor cells (B )were seeded at a density of 2,500 cells/cm2. After 24 hours, the medium was exchanged for that containing 50 Ag/mL control or H33 antibodies (d0). Cells weretrypsinized and counted every subsequent day for 4 days. Points, mean per square centimeter from four counts; bars, SE. H33 antibody does not influence theproliferation of endothelial and tumor cells. C, LLC1 tumor cryosections from isotype-matched control- or H33-treated mice were immunostained for PECAM-1 (green )and for apoptotic cells with TUNEL staining (red, arrowheads ). Endothelial cells do not exhibit an increase in apoptosis when mice were treated with H33 antibodycompared with controls. In contrast, tumor cells show increased apoptosis as shown by the quantification of apoptotic cells within the tumor. D, LLC1 tumor cryosectionsfrom PBS-, control antibody-, or H33-treated mice were stained for acid phosphatase to detect macrophages (arrowheads ), and the number of labeled macrophages persquare millimeter within the tumor was determined. Tumors from H33-treated animals show reduced infiltration of macrophages compared with control animals.Columns, mean obtained from four sections of n mice per group (PBS, n = 6; control antibody, n = 6; H33, n = 9); bars, SE. **, P < 0.01. Bar , 20 Am.

Anti–JAM-C Inhibits Angiogenesis

www.aacrjournals.org 5709 Cancer Res 2005; 65: (13). July 1, 2005

Research. on December 22, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Antibody against Junctional Adhesion Molecule-C Inhibits ... · adhesion molecules with extracellular matrix (9). The integrins a vh 3 and a vh 5 participate in blood vessel development

polarization of round spermatids in vivo (23). In addition, in vitro

studies have revealed that JAM-C interacts with polarity complex

molecules, such as PAR-3 and PAR-6, suggesting a role for JAM-C in

polarity establishment (22). Disassembly of molecular complexes

essential to maintain endothelial polarity and the establishment of

new interendothelial junctions occur during angiogenesis. We

propose that the antibody H33 interferes with one or both steps.

Current experiments aim to determine whether JAM-C is able to

interact with polarity complex proteins in endothelial cells and

participates to the polarization of the endothelium.

Acknowledgments

Received 11/30/2004; revised 3/31/2005; accepted 4/12/2005.Grant support: Swiss National Science Foundation grants 3100.067896.02

(M. Aurrand-Lions) and 3100A0-100697 (B.A. Imhof) and Oncosuisse grant OCS-01335-02-2003.

The costs of publication of this article were defrayed in part by the payment of pagecharges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Dominique Ducrest-Gay, Claude Magnin, and Patricia Ropraz for theirtechnical expertise; Dr. Paul Frederick Bradfield for critical reading of the article; andDr. Louise Reynolds for helpful advice. The real-time quantitative PCR experimentswere done at the Genomics Platform of the National Center of Competence inResearch Frontiers in Genetics with the help of Dr. Mylene Docquier.

Cancer Research

Cancer Res 2005; 65: (13). July 1, 2005 5710 www.aacrjournals.org

References1. Folkman J. Angiogenesis in cancer, vascular, rheuma-toid and other disease. Nat Med 1995;1:27–31.

2. Risau W. Mechanisms of angiogenesis. Nature 1997;386:671–4.

3. Trabucchi E, Radaelli E, Marazzi M, et al. The role ofmast cells in wound healing. Int J Tissue React 1988;10:367–72.

4. Smith SK. Angiogenesis, vascular endothelial growthfactor and the endometrium. Hum Reprod Update 1998;4:509–19.

5. Alon T, Hemo I, Itin A, Pe’er J, Stone J, Keshet E.Vascular endothelial growth factor acts as a survivalfactor for newly formed retinal vessels and hasimplications for retinopathy of prematurity. Nat Med1995;1:1024–8.

6. Crowther M, Brown NJ, Bishop ET, Lewis CE. Micro-environmental influence on macrophage regulation ofangiogenesis in wounds and malignant tumors. J LeukocBiol 2001;70:478–90.

7. Leek RD, Harris AL, Lewis CE. Cytokine networks insolid human tumors: regulation of angiogenesis. J LeukocBiol 1994;56:423–35.

8. Murdoch C, Giannoudis A, Lewis CE. Mechanismsregulating the recruitment of macrophages into hypoxicareas of tumors and other ischemic tissues. Blood 2004;104:2224–34.

9. Hood JD, Cheresh DA. Role of integrins in cell invasionand migration. Nat Rev Cancer 2002;2:91–100.

10. Reynolds LE, Wyder L, Lively JC, et al. Enhancedpathological angiogenesis in mice lacking h3 integrin orh3 and h5 integrins. Nat Med 2002;8:27–34.

11. Hodivala-Dilke KM, Reynolds AR, Reynolds LE.Integrins in angiogenesis: multitalented molecules in abalancing act. Cell Tissue Res 2003;314:131–44.

12. Carmeliet P, Lampugnani MG, Moons L, et al.Targeted deficiency or cytosolic truncation of the VE-cadherin gene in mice impairs VEGF-mediated endo-thelial survival and angiogenesis. Cell 1999;98:147–57.

13. Dejana E, Spagnuolo R, Bazzoni G. Interendothelialjunctions and their role in the control of angiogenesis,vascular permeability and leukocyte transmigration.Thromb Haemost 2001;86:308–15.

14. Ishida T, Kundu RK, Yang E, Hirata K, Ho YD,Quertermous T. Targeted disruption of endothelial cell-selective adhesion molecule inhibits angiogenic proces-ses in vitro and in vivo . J Biol Chem 2003;278:34598–604.

15. Naik MU, Mousa SA, Parkos CA, Naik UP. Signalingthrough JAM-1 and avh3 is required for the angiogenicaction of bFGF: dissociation of the JAM-1 and avh3

complex. Blood 2003;102:2108–14.16. Naik MU, Vuppalanchi D, Naik UP. Essential role ofjunctional adhesion molecule-1 in basic fibroblastgrowth factor-induced endothelial cell migration. Arte-rioscler Thromb Vasc Biol 2003;23:2165–71.

17. Aurrand-Lions MA, Duncan L, Du Pasquier L, ImhofBA. Cloning of JAM-2 and JAM-3: an emerging junctionaladhesion molecular family? Curr Top Microbiol Immu-nol 2000;251:91–8.

18. Aurrand-Lions M, Duncan L, Ballestrem C, Imhof BA.JAM-2, a novel immunoglobulin superfamily molecule,expressed by endothelial and lymphatic cells. J BiolChem 2001;276:2733–41.

19. Zen K, Babbin BA, Liu Y, Whelan JB, Nusrat A,Parkos CA. JAM-C is a component of desmosomesand a ligand for CD11b/CD18-mediated neutrophiltransepithelial migration. Mol Biol Cell 2004;15:3926–37.

20. Chavakis T, Keiper T, Matz-Westphal R, et al. Thejunctional adhesion molecule-C promotes neutrophiltransendothelial migration in vitro and in vivo . J BiolChem 2004;279:55602–8.

21. Aurrand-Lions M, Johnson-Leger C, Wong C, DuPasquier L, Imhof BA. Heterogeneity of endothelialjunctions is reflected by differential expression andspecific subcellular localization of the three JAM familymembers. Blood 2001;98:3699–707.

22. Ebnet K, Aurrand-Lions M, Kuhn A, et al. Thejunctional adhesion molecule (JAM) family membersJAM-2 and JAM-3 associate with the cell polarity proteinPAR-3: a possible role for JAMs in endothelial cellpolarity. J Cell Sci 2003;116:3879–91.

23. Gliki G, Ebnet K, Aurrand-Lions M, Imhof BA,Adams RH. Spermatid differentiation requires theassembly of a cell polarity complex downstreamof junctional adhesion molecule-C. Nature 2004;431:320–4.

24. Piali L, Albelda SM, Baldwin HS, Hammel P, GislerRH, Imhof BA. Murine platelet endothelial cell adhesionmolecule (PECAM-1)/CD31 modulates h2 integrins onlymphokine-activated killer cells. Eur J Immunol 1993;23:2464–71.

25. Laschinger M, Engelhardt B. Interaction of a4-integrin with VCAM-1 is involved in adhesion ofencephalitogenic T cell blasts to brain endotheliumbut not in their transendothelial migration in vitro .J Neuroimmunol 2000;102:32–43.

26. Berg EL, Robinson MK, Warnock RA, Butcher EC.The human peripheral lymph node vascular addressin isa ligand for LECAM-1, the peripheral lymph nodehoming receptor. J Cell Biol 1991;114:343–9.

27. Piali L, Hammel P, Uherek C, et al. CD31/PECAM-1 is a ligand for avh3 integrin involved in adhesionof leukocytes to endothelium. J Cell Biol 1995;130:451–60.

28. Johnson-Leger CA, Aurrand-Lions M, Beltraminelli N,Fasel N, Imhof BA. Junctional adhesion molecule-2(JAM-2) promotes lymphocyte transendothelial migra-tion. Blood 2002;100:2479–86.

29. Reynolds AR, Reynolds LE, Nagel TE, et al.Elevated Flk1 (vascular endothelial growth factorreceptor 2) signaling mediates enhanced angiogenesisin h3-integrin-deficient mice. Cancer Res 2004;64:8643–50.

30. Kindler V, Sappino AP, Grau GE, Piguet PF, Vassalli P.The inducing role of tumor necrosis factor in thedevelopment of bactericidal granulomas during BCGinfection. Cell 1989;56:731–40.

31. Nicosia RF, Ottinetti A. Modulation of microvasculargrowth and morphogenesis by reconstituted basementmembrane gel in three-dimensional cultures of rataorta: a comparative study of angiogenesis in Matrigel,collagen, fibrin, and plasma clot. In Vitro Cell Dev Biol1990;26:119–28.

32. Vandesompele J, De Preter K, Pattyn F, et al.Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple

internal control genes. Genome Biol 2002;3:RE-SEARCH0034.

33. Liao F, Li Y, O’Connor W, et al. Monoclonal antibodyto vascular endothelial-cadherin is a potent inhibitor ofangiogenesis, tumor growth, and metastasis. Cancer Res2000;60:6805–10.

34. Corada M, Mariotti M, Thurston G, et al. Vascularendothelial-cadherin is an important determinant ofmicrovascular integrity in vivo . Proc Natl Acad Sci U S A1999;96:9815–20.

35. Cunningham SA, Arrate MP, Rodriguez JM, et al. Anovel protein with homology to the junctional adhesionmolecule. Characterization of leukocyte interactions.J Biol Chem 2000;275:34750–6.

36. Folkman J. Tumor angiogenesis: therapeutic implica-tions. N Engl J Med 1971;285:1182–6.

37. Prewett M, Huber J, Li Y, et al. Antivascular endo-thelial growth factor receptor ( fetal liver kinase 1)monoclonal antibody inhibits tumor angiogenesis andgrowth of several mouse and human tumors. Cancer Res1999;59:5209–18.

38. Brooks PC, Montgomery AM, Rosenfeld M, et al.Integrin avh3 antagonists promote tumor regression byinducing apoptosis of angiogenic blood vessels. Cell1994;79:1157–64.

39. Shalaby F, Rossant J, Yamaguchi TP, et al. Failure ofblood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 1995;376:62–6.

40. Bader BL, Rayburn H, Crowley D, Hynes RO.Extensive vasculogenesis, angiogenesis, and organogen-esis precede lethality in mice lacking all av integrins.Cell 1998;95:507–19.

41. Strieter RM, Polverini PJ, Kunkel SL, et al.The functional role of the ELR motif in CXCchemokine-mediated angiogenesis. J Biol Chem 1995;270:27348–57.

42. Leek RD, Hunt NC, Landers RJ, Lewis CE, Royds JA,Harris AL. Macrophage infiltration is associated withVEGF and EGFR expression in breast cancer. J Pathol2000;190:430–6.

43. Eliceiri BP, Cheresh DA. Adhesion events in angio-genesis. Curr Opin Cell Biol 2001;13:563–8.

44. Soldi R, Mitola S, Strasly M, Defilippi P, Tarone G,Bussolino F. Role of avh3 integrin in the activation ofvascular endothelial growth factor receptor-2. EMBOJ 1999;18:882–92.

45. Esser S, Lampugnani MG, Corada M, Dejana E, RisauW. Vascular endothelial growth factor induces VE-cadherin tyrosine phosphorylation in endothelial cells.J Cell Sci 1998;111:1853–65.

46. Soga N, Namba N, McAllister S, et al. Rho familyGTPases regulate VEGF-stimulated endothelial cellmotility. Exp Cell Res 2001;269:73–87.

47. Wojciak-Stothard B, Potempa S, Eichholtz T, RidleyAJ. Rho and Rac but not Cdc42 regulate endothelial cellpermeability. J Cell Sci 2001;114:1343–55.

48. Braga VM, Del Maschio A, Machesky L, Dejana E.Regulation of cadherin function by Rho and Rac:modulation by junction maturation and cellular context.Mol Biol Cell 1999;10:9–22.

49. Bayless KJ, Davis GE. The Cdc42 and Rac1 GTPasesare required for capillary lumen formation in three-dimensional extracellular matrices. J Cell Sci 2002;115:1123–36.

Research. on December 22, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Antibody against Junctional Adhesion Molecule-C Inhibits ... · adhesion molecules with extracellular matrix (9). The integrins a vh 3 and a vh 5 participate in blood vessel development

2005;65:5703-5710. Cancer Res   Chrystelle Lamagna, Kairbaan M. Hodivala-Dilke, Beat A. Imhof, et al.   Angiogenesis and Tumor GrowthAntibody against Junctional Adhesion Molecule-C Inhibits

  Updated version

  http://cancerres.aacrjournals.org/content/65/13/5703

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2005/07/01/65.13.5703.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/65/13/5703.full#ref-list-1

This article cites 48 articles, 23 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/65/13/5703.full#related-urls

This article has been cited by 19 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/65/13/5703To request permission to re-use all or part of this article, use this link

Research. on December 22, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from