10
Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin Olsson, A ˚ sa Thulin, Chunsik Lee, Irja Johansson, and Lena Claesson-Welsh Rudbeck Laboratory, Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden Abstract Histidine-rich glycoprotein (HRGP) is an abundant heparin- binding plasma protein that efficiently arrests growth and vascularization of mouse tumor models. We have shown that the antiangiogenic effect of HRGP is dependent on its histidine/proline–rich domain, which needs to be released from the mother protein to exert its effects. Here we identify a 35-amino-acid peptide, HRGP330, derived from the histidine/ proline–rich domain as endowed with antiangiogenic proper- ties in vitro and in vivo . The mechanism of action of HRGP330 involves subversion of focal adhesion function by disruption of integrin-linked kinase (ILK) and focal adhesion kinase (FAK) functions, inhibition of vascular endothelial growth factor (VEGF)–induced tyrosine phosphorylation of the FAK sub- strate A-actinin, and, as a consequence, an arrest in endo- thelial cell motility. The disturbed focal adhesion function is reflected in the ability of HRGP as well as of HRGP330 to prevent endothelial cell adhesion to vitronectin in a manner involving A v B 3 integrin. In conclusion, HRGP330, which we define as the minimal antiangiogenic domain of HRGP, exerts its effects through signal transduction targeting focal adhe- sions, thereby interrupting VEGF-induced endothelial cell motility. (Cancer Res 2006; 66(4): 2089-97) Introduction Angiogenesis, formation of new blood vessels from preexisting ones, is a physiologic process that occurs during development, as a result of exercise, in wound healing and during the menstrual cycle. Angiogenesis is tightly regulated by a number of proangiogenic and antiangiogenic factors acting in concert to adjust the distribution of oxygen and nutrients to meet the demands of the tissue. A number of pathologic conditions including rheumatoid arthritis, diabetic retinopathy, tumor growth, atherosclerosis, and diabetic loss of peripheral circulation are characterized by deregulated angiogenesis (1, 2). The consequences range from inadequate tissue growth to ischemia. Vascular endothelial growth factor (VEGF; first identified as vascular permeability factor; refs. 3, 4) acts as a proangiogenic factor by promoting endothelial migration, proliferation, differen- tiation, and tissue degradation. These effects are primarily mediated by the receptor tyrosine kinase VEGF receptor 2 (VEGFR-2; also known as KDR in the human and Flk-1 in the mouse). Most, if not all, tumors express VEGF, which induces angiogenesis in neighboring VEGFR-2-expressing vessels and which promotes endothelial cell permeability, leading to extravasation of provisional matrix compo- nents such as fibrin, fibronectin, and vitronectin. Binding of endothelial cell integrins to these matrix components further modulates the angiogenic response by inducing downstream signaling pathways converging with those induced by activation of VEGFR-2. Targeting the vasculature by neutralizing the action of VEGF with antibodies (Avastin/bevacizumab) prolonged the lives of patients with metastatic renal or colorectal cancer in phase III clinical trials (5, 6). This provides evidence that targeting the vasculature is a valid approach in cancer treatment. A number of endogenous antiangiogenic factors, derived from naturally occur- ring proteins, have been identified and shown to inhibit growth of mouse tumors (see ref. 7 for a review). A common theme for inhibitors such as endostatin, tumstatin, and kininostatin, fragments of collagen XVIII, collagen IV, and kininogen, respectively, is their interference with integrin function either by using integrins as receptors (8, 9) or by specifically binding to different matrix components (10). Endostatin is reported to act via a5h1 integrins and tumstatin via a v h 3 . Binding of endostatin to endothelial cells also involves heparan sulfates (11, 12) and the leukocyte adhesion antigen E-selectin (13), indicating that the receptor mechanism may be complex and dependent on several components. We and others have identified histidine-rich glycoprotein (HRGP; also known as HRG/HPRG, for review see ref. 14) as a potent inhibitor of angiogenesis, an effect mediated via the histidine/ proline–rich (His/Pro–rich) domain of the protein (15, 16). We have shown that HRGP reduces endothelial cell migration and tumor vascularization and growth in mice. HRGP is a member of the cystatin superfamily together with a2-HS glycoprotein/fetuin, fetuin-B, cystatin, and kininogen (17, 18). The central domain of HRGP is rich in histidine and proline residues and contains multiple more or less conserved tandem repeats of the pentapeptide GHHPH. We have shown that this domain can be proteolytically released from the full-length protein, which is required for the antiangiogenic activity (16). HRGP preparations always contain proteolytic frag- ments of distinct masses (16, 19) and one of these fragments corresponds to the His/Pro–rich domain (16). In the present study, we further define the minimal active domain within the His/Pro– rich fragment of HRGP and describe its mechanism of action. Materials and Methods Antibodies. The antibodies used were neutralizing anti-a v h 3 (MAB 1976z, Chemicon, Temecula, CA); anti–focal adhesion kinase (FAK; Santa Cruz Biotechnology, Santa Cruz, CA); anti-paxillin (Transduction Lab, Franklin Lakes, NJ); anti-a-actinin; horseradish peroxidase (HRP) anti-goat Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). J. Dixelius and A-K. Olsson contributed equally to this work. J. Dixelius is presently at Department of Medical Biochemistry and Biophysics, Karolinska Institute, Scheeles va ¨g 2, SE-17177 Stockholm, Sweden. Requests for reprints: Lena Claesson-Welsh, Rudbeck Laboratory, Department of Genetics and Pathology, Uppsala University, Dag Hammarskjo ¨lds va ¨g 20, SE-751 85 Uppsala, Sweden. E-mail: [email protected]. I2006 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-05-2217 www.aacrjournals.org 2089 Cancer Res 2006; 66: (4). February 15, 2006 Research Article Research. on April 24, 2020. © 2006 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

Minimal Active Domain and Mechanism of Action of the

Angiogenesis Inhibitor Histidine-Rich Glycoprotein

Johan Dixelius, Anna-Karin Olsson, Asa Thulin, Chunsik Lee, Irja Johansson,and Lena Claesson-Welsh

Rudbeck Laboratory, Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden

Abstract

Histidine-rich glycoprotein (HRGP) is an abundant heparin-binding plasma protein that efficiently arrests growth andvascularization of mouse tumor models. We have shownthat the antiangiogenic effect of HRGP is dependent on itshistidine/proline–rich domain, which needs to be releasedfrom the mother protein to exert its effects. Here we identify a35-amino-acid peptide, HRGP330, derived from the histidine/proline–rich domain as endowed with antiangiogenic proper-ties in vitro and in vivo . The mechanism of action of HRGP330involves subversion of focal adhesion function by disruption ofintegrin-linked kinase (ILK) and focal adhesion kinase (FAK)functions, inhibition of vascular endothelial growth factor(VEGF)–induced tyrosine phosphorylation of the FAK sub-strate A-actinin, and, as a consequence, an arrest in endo-thelial cell motility. The disturbed focal adhesion function isreflected in the ability of HRGP as well as of HRGP330 toprevent endothelial cell adhesion to vitronectin in a mannerinvolving AvB3 integrin. In conclusion, HRGP330, which wedefine as the minimal antiangiogenic domain of HRGP, exertsits effects through signal transduction targeting focal adhe-sions, thereby interrupting VEGF-induced endothelial cellmotility. (Cancer Res 2006; 66(4): 2089-97)

Introduction

Angiogenesis, formation of new blood vessels from preexistingones, is a physiologic process that occurs during development, as aresult of exercise, in wound healing and during the menstrual cycle.Angiogenesis is tightly regulated by a number of proangiogenic andantiangiogenic factors acting in concert to adjust the distributionof oxygen and nutrients to meet the demands of the tissue. Anumber of pathologic conditions including rheumatoid arthritis,diabetic retinopathy, tumor growth, atherosclerosis, and diabeticloss of peripheral circulation are characterized by deregulatedangiogenesis (1, 2). The consequences range from inadequate tissuegrowth to ischemia.Vascular endothelial growth factor (VEGF; first identified as

vascular permeability factor; refs. 3, 4) acts as a proangiogenicfactor by promoting endothelial migration, proliferation, differen-tiation, and tissue degradation. These effects are primarily mediated

by the receptor tyrosine kinase VEGF receptor 2 (VEGFR-2; alsoknown as KDR in the human and Flk-1 in the mouse). Most, if not all,tumors express VEGF, which induces angiogenesis in neighboringVEGFR-2-expressing vessels and which promotes endothelial cellpermeability, leading to extravasation of provisional matrix compo-nents such as fibrin, fibronectin, and vitronectin. Binding ofendothelial cell integrins to these matrix components furthermodulates the angiogenic response by inducing downstreamsignaling pathways converging with those induced by activationof VEGFR-2. Targeting the vasculature by neutralizing the action ofVEGF with antibodies (Avastin/bevacizumab) prolonged the lives ofpatients with metastatic renal or colorectal cancer in phase IIIclinical trials (5, 6). This provides evidence that targeting thevasculature is a valid approach in cancer treatment. A number ofendogenous antiangiogenic factors, derived from naturally occur-ring proteins, have been identified and shown to inhibit growth ofmouse tumors (see ref. 7 for a review).A common theme for inhibitors such as endostatin, tumstatin,

and kininostatin, fragments of collagen XVIII, collagen IV, andkininogen, respectively, is their interference with integrin functioneither by using integrins as receptors (8, 9) or by specifically bindingto differentmatrix components (10). Endostatin is reported to act viaa5h1 integrins and tumstatin via avh3. Binding of endostatin toendothelial cells also involves heparan sulfates (11, 12) and theleukocyte adhesion antigen E-selectin (13), indicating that thereceptor mechanism may be complex and dependent on severalcomponents.We and others have identified histidine-rich glycoprotein (HRGP;

also known as HRG/HPRG, for review see ref. 14) as a potentinhibitor of angiogenesis, an effect mediated via the histidine/proline–rich (His/Pro–rich) domain of the protein (15, 16). We haveshown that HRGP reduces endothelial cell migration and tumorvascularization and growth in mice. HRGP is a member of thecystatin superfamily together with a2-HS glycoprotein/fetuin,fetuin-B, cystatin, and kininogen (17, 18). The central domain ofHRGP is rich in histidine and proline residues and contains multiplemore or less conserved tandem repeats of the pentapeptide GHHPH.We have shown that this domain can be proteolytically released fromthe full-length protein, which is required for the antiangiogenicactivity (16). HRGP preparations always contain proteolytic frag-ments of distinct masses (16, 19) and one of these fragmentscorresponds to the His/Pro–rich domain (16). In the present study,we further define the minimal active domain within the His/Pro–rich fragment of HRGP and describe its mechanism of action.

Materials and Methods

Antibodies. The antibodies used were neutralizing anti-avh3 (MAB

1976z, Chemicon, Temecula, CA); anti–focal adhesion kinase (FAK; SantaCruz Biotechnology, Santa Cruz, CA); anti-paxillin (Transduction Lab,

Franklin Lakes, NJ); anti-a-actinin; horseradish peroxidase (HRP) anti-goat

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

J. Dixelius and A-K. Olsson contributed equally to this work. J. Dixelius is presentlyat Department of Medical Biochemistry and Biophysics, Karolinska Institute, Scheelesvag 2, SE-17177 Stockholm, Sweden.

Requests for reprints: Lena Claesson-Welsh, Rudbeck Laboratory, Department ofGenetics and Pathology, Uppsala University, Dag Hammarskjolds vag 20, SE-751 85Uppsala, Sweden. E-mail: [email protected].

I2006 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-05-2217

www.aacrjournals.org 2089 Cancer Res 2006; 66: (4). February 15, 2006

Research Article

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

(Sigma-Aldrich, Stockholm, Sweden); anti–integrin-linked kinase (ILK; CellSignaling, Danvers, MA); anti-phosphotyrosine (4G10, Upstate Biotechnol-

ogy, Charlottesville, VA); Alexa 488 antimouse; Alexa 555 antimouse; Alexa

488 antirabbit and Alexa 568 antirabbit (Molecular Probes, Eugene, OR);

HRP antirabbit and antimouse (Amersham Biosciences, Uppsala, Sweden);antimouse CD31 (Becton Dickinson, Franklin Lakes, NJ); and biotinylated

antirat antibody (Vector Laboratories, Burlingame, CA).

HRGP and HRGP-derived synthetic peptides. Recombinant His-tagged

HRGP was produced and purified as previously described (16). In

preparations of purified HRGP, there are always proteolytic fragments of

distinct masses present (16, 19). One of these fragments corresponds to the

His/Pro–rich domain of HRGP (16). The designation ‘‘HRGP’’ in this study

refers to the purified preparation consisting of the mix of full-length HRGP

and proteolytic fragments thereof. HRGP-derived synthetic peptides

HRGP330, HRGP365, and HRGP398 were purchased from Innovagen AB

(Lund, Sweden). The peptides were analyzed by reverse-phase high-

performance liquid chromatography and mass spectral analysis and were

dissolved in TBS [20mmol/LTris-HCl (pH 7.5), 150mmol/L NaCl] containing

1 mmol/L ZnCl2. The peptides have the following sequences: HRGP330

(amino acids 330-364), DLHPHKHHSHEQHPHGHHPHAHHPHEHDTHRQ-

HPH-COOH; HRGP365 (amino acids 365-397), GHHPHGHHPHGHHPH-

GHHPHGHHPHCHDFQDYG-COOH; and HRGP398 (amino acid 398-439),

PCDPPPHNQGHCCHGHGPPPGHLRRRGPGKGPRPFHCRQIGS-COOH.

Cell culture. Bovine capillary endothelial cells, a kind gift from Dr. R.Christofferson (Department of Medical Cell Biology, Uppsala University),

were cultured on gelatin-coated dishes in DMEM (Life Technologies, Inc.,

Rockville, MD), 10% newborn calf serum, and 2 ng fibroblast growth factor

2 (FGF-2)/mL (complete medium). Telomerase immortalized endothelialcells, derived from human dermal microvascular endothelial cells, were a

kind gift from Dr. Martin McMahon (Cancer Research Institute, University

of California San Francisco, San Francisco, CA; ref. 20). Telomerase

immortalized endothelial cells were cultured in complete endothelial cellbasal medium (EBM MV2, PromoCell, Heidelberg, Germany) without

antibiotics on gelatin-coated cell culture plastic.

Conditions for microscopy and immunoblotting. For all microscopy

and immunoblotting experiments, cells were seeded on vitronectin-coated

(V8379, Sigma-Aldrich) Petri dishes or cell culture slides in complete

medium, incubated for 24 hours, washed, and incubated in DMEM/1%

newborn calf serum (starvation medium) for 20 to 24 hours. To prevent

contribution from serum-derived HRGP, the medium was replaced with

fresh EBM/0.1% bovine serum albumin (BSA), 20 mmol/L HEPES (pH 7.2),

and incubated for 90 minutes. The cells were then stimulated as indicated.Microphotography. The slides were put on ice, washed in ice-cold TBS

[20 mmol/L Tris-HCl (pH 7.5), 150 mmol/L NaCl], and fixed in Zn-fix [0.05%

Ca-acetate, 0.5% Zn-acetate, 0.5% ZnCl2 in TBS (pH 6.6), and 0.2% TritonX-100] for 15 minutes at room temperature. The preparations were washed

in TBS, blocked in TBS/10% FCS, and stained using standard protocols. The

60�/NA 1.4 or 10�/NA 0.45 objectives of a Nikon E1000 microscope and a

Nikon DXM1200 camera were used.Immunoblotting. Cells were put on ice and quickly washed once in

ice-cold TBS/100 Amol/L Na3VO4, then lysed in NP40 lysis buffer [1% NP40,

20 mmol/L HEPES (pH 7.5), 150 mmol/L NaCl, 10% glycerol, 2.5 mmol/L

EDTA, 10 kallikrein inhibitory units (KIE) aprotinin/mL, 1 mmol/L phenyl-methylsulfonyl fluoride, and 100 Amol/L Na3VO4]. Lysates were clarified

by centrifugation. Immunoprecipitation, SDS-PAGE, and immunoblotting

were done using standard procedures. Immunoreactive sites on the filterswere detected by enhanced chemiluminescence.

Time lapse. Telomerase immortalized endothelial cells were seeded on

vitronectin-coated coverslips at a subconfluent density in complete EBM

medium and incubated for 24 hours, washed, and starved in EBM/1% FCSfor 20 hours. The coverslips were mounted in observation dishes in a 1:1

mixture of conditioned EBM/1% FCS and EBM/20 mmol/L HEPES (pH 7.2)

in a temperature-controlled chamber set to 37jC on a Zeiss LSM 510 Meta

confocal microscope. The cells were allowed to stabilize for 60 minutes;after which, one differential interference contrast image per minute was

collected for 2 hours using the 40�/NA 1.3 objective. The image in Fig. 4A

was processed in Adobe Photoshop software (San Jose, CA) using the ‘‘find

edges’’ function. Coloring was done by manually outlining the cells using the‘‘pen tool.’’

Adhesion assay. Ninety-six-well cell culture plates were incubated

overnight with vitronectin (1 Ag/mL in PBS) or BSA (2% BSA in PBS),

blocked for 2 hours with BSA in all wells, and washed thrice with PBS. Cellsplated the previous day in complete medium were washed once and

incubated for 60 minutes in medium [0.2% BSA, 20 mmol/L HEPES (pH 7.2),

2 ng FGF-2/mL; assay medium]. Cells were dissociated using nonenzymatic

‘‘Cell dissociation solution’’ (Sigma), washed once, and resuspended in assaymedium. Reagents were added to the cell suspension at the following

concentrations: HRGP, 100 ng/mL; HRGP330, 100 ng/mL; LM609, 10 Ag/mL,

and incubated for 15 or 5 minutes before seeding to allow complete or only

partial inhibition of adhesion, respectively. After 45 minutes in 37jC, cellswere washed thrice in TBS and fixed in Zn-fix for 10 minutes. Finally,

cells were stained with Hoechst 33342 (1 Ag/mL) washed once in TBS,

photographed (2� objective), and cell number was quantified using the EasyImage software (Tekno Optik AB, Skarholmen, Sweden). The experiments

were repeated at least thrice and the mean value of at least four wells per

condition was used. Error bars represent 1 SD. Statistical significance was

calculated using Students t test. Differences with P < 0.05 were consideredsignificant.

Chemotaxis assay. The chemotaxis assay was done using a modified

Boyden chamber, as described earlier (16), with 8-Am micropore polycar-

bonate filters (PFB8-50; Neuro Probe, Inc., Gaithersburg, MD) coated

with type 1 collagen solution at 100 Ag/mL (Vitrogen 100; Collagen Corp.,

Palo Alto, CA). Bovine capillary endothelial cells that had been starved

overnight in 0.5% NCS were trypsinized and resuspended at 7.5 � 105/mL

in DMEM, 0.25% BSA, and trasylol (aprotinin) at 1,000 KIE/mL. The cell

suspension was added in the upper chamber and VEGF-A (Peprotech, Rocky

Hill, NJ) at 10 ng/mL in the lower chamber. HRGP peptides (100 ng/mL)

and avh3 neutralizing antibody (LM609; 10 Ag/mL) were added both in the

upper and lower chambers. After 5 hours at 37jC, cells that had migrated

through the filter were stained with Giemsa and counted using the Easy

Image Analysis software (Tekno Optik). All samples were analyzed in at least

six wells for each treatment and at several separate occasions. Error bars

represent 1 SD. Statistical significance was calculated using Students t test.

Differences with P < 0.05 were considered significant.

Animal studies. Animal work was approved by the Uppsala University

board of animal experimentation and thus was done according to the UnitedKingdom Coordinating Committee on Cancer Research guidelines for

the welfare of animals in experimental neoplasia (21). The mice were

anesthetized with isoflurane (Forene; Abbott, Abbott Park, IL) during all

manipulations. Six-week-old female Fox Chase severe combined immunode-ficient mice were injected s.c. with 2.5 � 106 BxPC3 pancreatic carcinoma

cells on their left flank. When the tumors had reached a size of f200 mm3,

the animals were randomized into two groups (n = 7 per group) and

treatment was initiated with daily s.c injections on the right flank witheither 5 mg/kg/d HRGP330 (dissolved in 1 mmol/L ZnCl2, 68 mmol/L NaCl,

1.4 mmol/L KCl, 2.1 mmol/L Na2HPO4�7H2O, 0.7 mmol/L KH2PO4) or vehicle.

The tumors were measured with a caliper, in a blind procedure, and volumeswere calculated by the formula p / 6� width2� length. Tumor material was

snap-frozen in dry ice/isopentane and sectioned at 3 Am.

CD31 staining and quantification of vascular variables. Frozen

sections of the tumor tissue were fixed in acetone and blocked in 2% rabbit

serum before incubation with a rat anti-mouse CD31 antibody (Becton

Dickinson), diluted 1:500, and incubated at 4jC overnight. Detection of

the primary antibody binding sites was done using a biotinylated antirat

antibody (BA-4001; Vector Laboratories), diluted 1:300, and streptavidin-

HRP (SA-5004; Vector Laboratories) diluted 1:200. The enzymatic signal was

visualized using AEC peroxidase substrate kit (SK-4200; Vector Laborato-

ries). Hematoxylin staining was done to visualize all cells. Stereological

quantification of vascular variables was done as described earlier (16) on at

least three different tumors from each treatment group. This method of

quantifying tumor angiogenesis relates the length, volume, and surface area

of the vessels to tumor volume (22). Error bars represent 1 SD. Statistical

significance was calculated using Students t test. Differences with P < 0.05

were considered significant.

Cancer Research

Cancer Res 2006; 66: (4). February 15, 2006 2090 www.aacrjournals.org

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

Results

Identification of the minimal active domain of HRGP. Wehave shown that inhibition of endothelial cell chemotaxis by theangiogenesis inhibitor HRGP is mediated by the His/Pro–richdomain (16). HRGP is a 75-kDa multidomain molecule whichengages in interactions with a broad spectrum of molecules (14).To avoid complications in our studies from such interactivepartners (e.g., thrombospondins 1 and 2), we wished to identify theminimal active domain of HRGP. A series of synthetic peptides

(HRGP330, amino acids 330-364; HRGP365, amino acids 365-397;

and HRGP398, amino acids 398-439) derived sequentially from

the His/Pro–rich region were screened for their ability to inhibit

endothelial cell chemotaxis (Fig. 1A-C). Of these, only HRGP330

was able to inhibit VEGF-induced chemotaxis of primary endo-

thelial cells (Fig. 1A). As seen in Fig. 1D , HRGP330 attenuated

chemotaxis in a dose-dependent manner.HRGP330 is antiangiogenic in vivo . We next tested whether

HRGP330 retained the antiangiogenic properties of HRGP in vivo .

Figure 1. Identification of the minimal active domainof HRGP. A to C, chemotaxis of primary endothelialcells induced by 10 ng/mL VEGF was inhibited by100 ng/mL HRGP330 (H330; A) but not by HRGP365 (B)or HRGP398 (C ). D, the 35-amino-acid residue peptideHRGP330 inhibited VEGF-induced endothelial cellchemotaxis in a dose-dependent manner. E, growthcurves of BxPC3 tumors in HRGP330-treated(5 mg/kg/d; w ) and control-treated (5) mice (n = 7 pergroup). F, visualization of the tumor vasculature byimmunohistochemical staining against CD31 revealeda striking reduction in CD31-positive vessels in tumorsfrom HRGP330-treated animals. Bar, 100 Am. G,stereological quantification of vascular parameters intumors from control- or HRGP330-treated mice showeda significant reduction in vessel length, volume, andsurface area. Bars, 1 SD. Significance at the P < 0.01 orP < 0.05 level is indicated.

Angiogenesis Inhibition by HRGP

www.aacrjournals.org 2091 Cancer Res 2006; 66: (4). February 15, 2006

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

Female severe combined immunodeficient mice (n = 7 per group)were injected with BxPC3 human pancreatic carcinoma cells ontheir left flank. The tumors were allowed to become well establishedand reach f200 mm3 before treatment was initiated with dailys.c. injections on their right flank with HRGP330 (5 mg/kg/d in1 mmol/L ZnCl2) or vehicle. The tumor growth curves (Fig. 1E)showed no significant difference in tumor size between the groupsalthough there was a tendency for reduced growth in the treatmentgroup. After 31 days, the study had to be interrupted due to weightloss of animals in both groups. To determine whether HRGP330exerted antiangiogenic effect in vivo , the degree of tumorvascularization was analyzed by immunohistochemical stainingfor expression of the vascular marker CD31. As seen in Fig. 1F, tumorvascularization of HRGP330-treated mice was dramatically reduced.The control mice tumor vasculature was composed of larger vesselsand an even, dense capillary network. In contrast, tumors from theHRGP330-treated mice retained the large vessels whereas the smallcapillaries were essentially absent (Fig. 1F). Quantification of tumorvascularization according to the method by Gunderssen et al. (22)

showed a striking reduction primarily in length and volume butalso in surface area of the blood vessels in tumors from HRGP330-treated mice as compared with control-treated mice (Fig. 1G).These data show that HRGP330 possesses potent antiangiogenicproperties in an in vivo setting.HRGP330 inhibits VEGF-induced complex formation of ILK

and paxillin. Migration requires coordinated signaling fromgrowth factor receptors and integrins. The focal adhesioncomponents ILK and FAK are instrumental integrators of thesesignals. ILK is a serine/threonine kinase; however, its contributionto focal adhesion function seems to be dependent on its role as anadapter for a large number of molecules (e.g., paxillin). Complexformation between ILK and paxillin is needed for subcellularlocalization of ILK to focal adhesions and for cellular migration(23). We have previously shown that HRGP induces rearrangementof focal adhesions in endothelial cells after 10 minutes of treatment.To seek the mechanism of action of HRGP330, we subjectedendothelial cells to treatment with the peptide for 10 minutes inthe presence and absence of VEGF. Immunoprecipitation of either

Figure 2. HRGP330 disrupts ILK-paxillin complexformation. A, endothelial cells on vitronectin weretreated with HRGP330 (100 ng/mL) and VEGF (50 ng/mL),individually or in combination, for 10 minutes. VEGFinduced increased complex formation between paxillinand ILK as shown by immunoprecipitation (IP ) of paxillinand immunoblotting (IB ) for ILK (top; arrow, ILK).Cotreatment with HRGP330 and VEGF disruptedILK-paxillin complex formation. Control immunoblottingfor paxillin on the paxillin immunoprecipitates (middle )and immunoblotting for actin on total cell lysates (bottom ),show equal protein amounts in cell lysates used forimmunoprecipitation. B, the reciprocal experiment(IP:ILK, IB:paxillin ) gave the same result. The broadpaxillin band is indicated. Immunoblotting for ILK andactin showed equal amount of protein in total celllysates. Control immunoblotting for the amount ofimmunoprecipitated ILK was not feasible due tocomigration with the immunoglobulin heavy chain. C,immunocytochemical staining for paxillin (red ) and ILK(green ), using the same conditions as in (A ), revealedcolocalization of paxillin and ILK after VEGF stimulation(Merge; yellow , indicated by arrowheads), which wasdisrupted by cotreatment with VEGF and HRGP330.Bar, 4 Am.

Cancer Research

Cancer Res 2006; 66: (4). February 15, 2006 2092 www.aacrjournals.org

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

paxillin (Fig. 2A) or ILK (Fig. 2B) followed by immunoblotting forthe partner showed that ILK-paxillin complex formation wasenhanced by VEGF whereas cotreatment with HRGP330 blockedthe VEGF-induced increase. The reduced ILK-paxillin complexformation was not due to decreased protein expression levels; incontrast, there was a tendency for increased recovery of paxillin inthe biochemical analysis of cells treated with HRGP330 (Fig. 2A).Because ILK migrates at the same position of the immunoglobulinheavy chain, immunoblotting for ILK was done on total celllysates from stimulated cells (Fig. 2B, middle), which revealedsimilar recovery of ILK independent of treatment. Immunostain-ing of endothelial cells (Fig. 2C), using the same experimentalconditions as above, confirmed increased colocalization of ILKand paxillin in focal adhesions in response to treatment withVEGF. The colocalization was blocked by inclusion of HRGP330together with VEGF. Disruption of VEGF-induced complexformation between ILK and paxillin by HRGP330, visualized byimmunoblotting or immunostaining of intact cells, provides amechanistic explanation for the inhibition of migration by theangiogenesis inhibitor.

HRGP330 inhibits VEGF-induced tyrosine phosphorylationof FAK and A-actinin. The cytoplasmic tyrosine kinase FAK iscrucial for focal adhesion function. Several angiogenesis inhibitorssuch as angiostatin and endostatin have been shown to activateFAK in endothelial cells (24, 25). Both VEGF and HRGP330individually stimulated tyrosine phosphorylation (reflecting acti-vation) of FAK in endothelial cells, with a slight increase after 10minutes and a more pronounced effect after 2 hours of treatment(Fig. 3A). In contrast, in cells cotreated with VEGF and HRGP330,FAK tyrosine phosphorylation was attenuated. One importantsubstrate for the FAK kinase is a-actinin, of which binding to actinis negatively regulated by its tyrosine phosphorylation (26, 27).Tyrosine phosphorylation of a-actinin was studied in cells treatedwith HRGP330 and VEGF for different time periods under the sameconditions as above (Fig. 3B). Immunoblotting analyses showedthat VEGF as well as HRGP330 induced a robust phosphorylationof a-actinin both at 10 minutes and 2 hours. However, cotreatmentof the cells with VEGF and HRGP330 for 2 hours blocked a-actininphosphorylation, indicating persistent binding to actin andreduced cellular motility.

Figure 3. HRGP330 inhibits VEGF-induced activation of FAK and tyrosine phosphorylation of a-actinin and induces redistribution of a-actinin. Endothelial cellson vitronectin were treated with HRGP330 (100 ng/mL) and VEGF (50 ng/mL in A and B, 10 ng/ml in C ), individually or in combination, for 10 minutes and2 hours. A, both VEGF and HRGP330 added individually induced tyrosine phosphorylation of FAK at 10 minutes and more pronounced after 2 hours, as shown byimmunoprecipitation of FAK and subsequent immunoblotting for phosphotyrosine (PY ). Cotreatment of the cells with VEGF and HRGP330, both at 10 minutes and2 hours, abolished FAK phosphorylation. Bottom, control immunoblotting for FAK. Levels of phosphorylated FAK normalized to total FAK levels are depicted graphicallybelow the blot (PY/FAK). B, VEGF and HRGP330 added individually induced a pronounced increase in tyrosine phosphorylation of a-actinin (a-act), both at 10minutes and 2 hours, as shown by immunoprecipitation for a-actinin, followed by immunoblotting for phosphotyrosine or a-actinin. Cotreatment of cells with VEGF andHRGP330 for 10 minutes allowed a-actinin phosphorylation whereas, at 2 hours, the level of phosphorylation had returned to basal, in contrast to treatment withthe individual factors. Bottom, control immunoblotting for a-actinin. Levels of phosphorylated a-actinin normalized to total a-actinin levels are depicted graphically belowthe blot (PY/a-act). C, immunocytochemical staining for a-actinin (red) was done in endothelial cells on vitronectin treated with VEGF and HRGP330, individuallyor in combination, for 10 minutes and 2 hours (i.e., the same experimental setup as in A and B). FAK staining (green ) was used to visualize focal contacts. No significantchange in a-actinin staining pattern compared with control cells was detected after individual treatment with VEGF or HRGP330 or after the combined treatmentfor 10 minutes. Cotreatment with VEGF and HRGP330 for 2 hours resulted in accumulation of a-actinin along actinlike fiber structures. Bar, 8 Am.

Angiogenesis Inhibition by HRGP

www.aacrjournals.org 2093 Cancer Res 2006; 66: (4). February 15, 2006

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

HRGP330 and VEGF cotreatment induces redistribution ofA-actinin. Migrating cells have a rapid turnover of focal adhesionsand a dynamic actin cytoskeleton whereas nonmigrating cells arecharacterized by localization of a-actinin to their stable focaladhesions (27). Further, a-actinin incorporation into actin fibers islikely to promote a less dynamic cytoskeleton (28). The distributionof a-actinin was investigated by immunofluorescent staining ofendothelial cells treated with VEGF and HRGP330 as indicated(Fig. 3C). Untreated cells or cells treated with VEGF or HRGP330alone, for 10 minutes or 2 hours, showed low a-actinin immuno-reactivity. However, in cells cotreated with VEGF and HRGP330 for2 hours, a-actinin-positive fibers were clearly visible, merging withfocal adhesions visualized by FAK staining. These data reinforce thenotion that HRGP330 acts to inhibit endothelial cell motilityresponses by subverting the function of focal adhesions.HRGP330 interferes with VEGF-induced polarization and

lamellopodia formation but does not inhibit VEGFR-2 activa-tion. Cell migration requires the concerted regulation of growthfactor–induced lamellopodia formation and cell polarization. Endo-thelial cells on coverslips were monitored by time-lapse microscopyto study the effect of HRGP330 on these cellular responses (Fig. 4A ;Supplementary data). Untreated cells were nonpolarized with abasal lamellopodia activity. Treatment with HRGP330 alone(Supplementary data) did not affect the cell morphology whereasinclusion of VEGF resulted in a transient burst of increasedformation of lamellopodia at 30 minutes and, within two hours, ashift towards a polarized phenotype with elongated cells, consistentwith amigratory phenotype. Cotreatment of the cells with VEGFandHRGP330 abrogated the VEGF-induced changes, indicating thatHRGP330 directly or indirectly interferes with signaling eventsinduced by VEGF (Fig. 4A ; Supplementary data).To show that HRGP330 did not simply exert its effects by direct

inhibition of activation of VEGFR-2, the main transducer ofendothelial cell responses to VEGF, cells were treated with VEGF inthe presence and absence of HRGP330 (i.e., similar conditions asthose used above to show HRGP330 activation of FAK). As shownin Fig. 4B , VEGFR-2 tyrosine phosphorylation, indicative of itsactivation, was induced by VEGF irrespective of whether HRGP330was included in the treatment or not.Involvement of AvB3 integrin in HRGP action. We have

previously shown that HRGP interfereswith the ability of endothelialcells to adhere to vitronectin (16). As shown in Fig. 5A , HRGP330also inhibited adhesion of endothelial cells to vitronectin, possiblyimplicating the avh3 integrin, the main vitronectin receptor onendothelial cells, as a target molecule for HRGP330. To addressthe role of avh3 in the adhesion assay, we compared the effect oftreatment with the neutralizing anti-avh3 antibodies and HRGP/HRGP330 on endothelial cell adhesion to vitronectin. As shown inFig. 5A , treatment with HRGP/HRGP330 and anti-avh3 antibodiesinhibited the vitronectin-induced adhesion. Under conditions whereadhesion was only partially inhibited by individual treatment withHRGP or by the neutralizing antibody LM609, we noted that thecombined treatment did not further reduce adhesion of theendothelial cells (Fig. 5B). These data indicate an overlapping targetfor HRGP and the anti-avh3 antibody, suggesting that HRGP/HRGP330 may act via interference with avh3 function.That avh3 integrin function was important for endothelial cell

motility under the conditions used in the chemotaxis assay wasshown by inclusion of a neutralizing anti-avh3 antibody, whichreduced VEGF-induced chemotaxis to basal levels (Fig. 5C). Thechemotaxis experiment was done using collagen I–coated filters

because vitronectin coating of the polycarbonate filters used in thechemotaxis assay did not support endothelial cell adhesion. It isconceivable that avh3 integrins used cryptic sites exposed in thepartially denatured collagen I coat for movement and that theseinteractions were obliterated by the neutralizing antibody, therebyattenuating the migratory response (Fig. 5C). This result shows thatmigration of the endothelial cells in the present experimental setupis dependent on avh3 integrins and supports the notion thatHRGP330 may act by interfering with integrin signaling.

Discussion

HRGP330 exerts its antiangiogenic effect by arresting the actincytoskeleton in an immobile state and preventing growth factor–induced promigratory events, such as lamellopodia formation andcell polarization. The molecular mechanism involves disruption ofVEGF-induced complex formation between paxillin and ILK, as well

Figure 4. HRGP330 reverts VEGF-induced polarization and lamellopodiaformation. A, time-lapse images of endothelial cells on vitronectin treatedwith VEGF alone (10 ng/mL) or in combination with HRGP330 (100 ng/mL).VEGF stimulation resulted in increased lamellopodia formation (arrows ) at 30minutes and a shift towards a polarized phenotype with elongated cells within2 hours, consistent with a migratory phenotype. In the presence of HRGP330,these VEGF-induced morphologic changes were abrogated. B, VEGFR-2tyrosine phosphorylation induced by VEGF (50 ng/mL) was unaffected by thepresence of HRGP330 (100 ng/mL) as shown by immunoprecipitation ofVEGFR-2 and immunoblotting for phosphotyrosine. The expression level ofVEGFR-2 is low in primary endothelial cells and immunoblotting for Erk wastherefore used as a control for protein amount in total cell lysates.

Cancer Research

Cancer Res 2006; 66: (4). February 15, 2006 2094 www.aacrjournals.org

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

as prevention of VEGF-induced phosphorylation of FAK and itsdownstream substrate a-actinin. VEGFR-2 activation is not affectedby the presence of HRGP330. Instead, our data implicate avh3, themain vitronectin receptor on endothelial cells, in the mechanism ofaction of HRGP330. However, we do not exclude that the receptormechanism involves one or more additional cell-surface compo-nents. A summary and possible model for the mechanism of actionof HRGP330, based on the above findings, is depicted in Fig. 6.HRGP interacts with a number of plasma proteins (14). Complex

formation with thrombospondins 1 and 2, involving a COOH-terminal CLESH-1 domain of HRGP (29, 30), has received attentionas it apparently leads to neutralization of the antiangiogenic effect ofthrombospondin. Therefore, the identification of a 35-amino-acidresidue peptide, HRGP330, which lacks the thrombospondin bindingdomain but retains the antiangiogenic properties, is important infurther development of HRGP as an angiogenesis inhibitor (15, 16,31). Although it is likely that the stability of HRGP330 is low in vivo ,we achieved a significant reduction in BxPC3 vascularization witha virtual disappearance of small vessels in tumors in the treatedanimals. Donate et al. (31) recently reported on a multimerizedpentapeptide based on the repetitive sequence GHHPH fromthe His/Pro–rich domain of HRGP, which was used to inhibit

angiogenesis in s.c. Matrigel plugs and tumor growth in mice at80 mg/kg/d. Five repeats of the sequence GHHPH are containedwithin the peptide HRGP365 that did not show any antichemotacticeffect on endothelial cells in our hands (Fig. 1B) whereas HRGP330contains one GHHPH repeat. Another difference between HRGP330and HRGP365 and HRGP398 is that HRGP330 binds to heparin/heparan sulfate, showing that HRGP330 is dependent on sequencesdistinct from GHHPH for its biological properties.1

How does HRGP330 interact with the endothelial cell surfaceand transduce its effects? Considering the effect of HRGP/HRGP330 on endothelial cell adhesion, migration, and focaladhesions, integrins are obvious receptor candidates. The majorvitronectin receptor on endothelial cells is avh3 integrin, which is

Figure 6. Mechanism of action of HRGP330. The antiangiogenic effect ofHRGP330 is exerted through an arrest of the endothelial cell actin cytoskeletonand a consequent inhibition of VEGF-induced effects such as lamellopodiaformation and polarization, events needed for migration, and an angiogenicresponse. HRGP330 disrupts VEGF-induced complex formation between paxillinand ILK in focal adhesions, as well as VEGF-induced phosphorylation/activationof FAK and its downstream substrate a-actinin. VEGFR-2 activation is notaffected by the presence of HRGP330. The main vitronectin receptor onendothelial cells, avh3 integrin, is implicated in the mechanism of action ofHRGP330. However, it is likely that the receptor mechanism involves one orseveral cell-surface molecules, such as heparan sulfates, or other thus farunidentified components (?). Vn, vitronectin; HSPG, heparan sulfateproteoglycan; pax, paxillin; a-act, a-actinin.

Figure 5. Involvement of avh3 integrin in HRGP action. A, adhesion ofendothelial cells to vitronectin was significantly reduced in the presence ofHRGP, HRGP330, or neutralizing avh3 antibody (LM609). B, under conditionswhere adhesion was only partially decreased (compare extent of HRGP andHRGP330 block in relation to BSA between A and B), no further reductionwas seen as a result of combined treatment with HRGP and LM609. C,chemotaxis of endothelial cells towards VEGF in a modified Boyden chamberwas reduced to similar extents by inclusion of HRGP, HRGP330, or neutralizingavh3 antibody (LM609). Bars, 1 SD. All changes induced by HRGP, HRGP330,or LM609, compared with control in (A and B ), or by the combination ofVEGF and HRGP, HRGP330, or LM609, compared with VEGF in (C ), weresignificant (P < 0.01; P < 0.05).

1 M. Vanwildemeersch, A.K. Olsson, E. Gottfridsson, L. Claesson-Welsh, U. Lindahl,D. Spillmann. Angiogenesis inhibition by HRGP involves Zn2+-dependent binding ofheparan sulfate to the His/Pro–rich domain. 2005, submitted for publication.

Angiogenesis Inhibition by HRGP

www.aacrjournals.org 2095 Cancer Res 2006; 66: (4). February 15, 2006

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

expressed on angiogenic endothelial cells in conjunction withvessel remodeling (32). We show that endothelial cell chemotaxis isdependent on avh3; moreover, neutralizing anti-avh3 antibodiesblock endothelial cell adhesion to vitronectin to the same extent asHRGP330. These data are compatible with the notion that themechanism of action of HRGP330 involves interference with avh3

function. Moreover, ongoing studies implicate heparan sulfate inthe mechanism of action of HRGP330.1 It is of interest that avh3

has been shown to occur in complex with VEGFR-2 and to berequired for VEGFR-2 responsiveness (33). In this context, it isimportant to note that HRGP330 does not simply interfere withVEGFR-2 activation by binding either to heparin/heparan sulfate orto avh3 (see Fig. 4B). Thus, we suggest that there may be more thanone interactive partner for HRGP330 on the endothelial cellsurface. The His/Pro–rich region of HRGP is able to bind Zn2+,which is also required for HRGP330 to exert its antiangiogenicfunction.1 Spectra obtained using far-UV technique indicate thatHRGP belongs to the category of intrinsically unstructured proteins(34). A probable effect of Zn2+ binding is stabilization of theconformation of the His/Pro–rich domain, which may be requiredfor its interaction with a receptor.Other potential mechanisms of action of HRGP330 include the

possibility of interaction with circulating vitronectin, which maypromote homing of HRGP330 to sites of active angiogenesis. Thistype of mechanism has recently been proposed for the antiangio-genic molecules anginex and anastellin (10). Moreover, two recentreports from the same group (31, 35) suggest membrane-boundtropomyosin as a receptor for HRGP. Tropomyosins constitute alarge family of widely expressed intracellular actin-binding mole-cules lacking transmembrane domain (36). It is noteworthy thattropomyosin has been suggested to also serve as a receptor forkininogen (37) as well as for endostatin (38). The current consensusis that endostatin requires a5h1 integrins and heparan sulfate toexert its effects on endothelial cells.Our in vitro data show that HRGP330 potently inhibits chemo-

taxis of endothelial cells in the nanogram range. Cell motility is acomplex process, which requires cells to polarize and concentratelamellopodia and filopodia activity to the leading edge, where smallfocal adhesions are formed de novo . Actin stress fibers, microtubuli,and RhoA activity are confined to and necessary for the trailing endwhere focal adhesions are larger and need to be remodeled ordisrupted. The chemoattractant signal relayed by receptor tyrosinekinases, such as VEGFR-2, converge with signals from integrinreceptors to orchestrate the motility response. This integration maybe achieved by direct interaction (e.g., between VEGFR-2 and avh3)serving to amplify reciprocal signals (see ref. 39 for review).Furthermore, downstream signaling pathways integrate to transmita permissive migratory signal. Such pathways include the cytoplas-mic kinases FAK and ILK, both of which interact directly or indirectlywith integrins as well as growth factor receptors. FAK has beenascribed a critical role in focal adhesion turnover (40) and FAK-dependent tyrosine phosphorylation of a-actinin blocks its associ-ation with actin, allowing increased actin dynamics (26). In FAK�/�

cells, a-actinin fails to become tyrosine phosphorylated, indicating akey role for FAK in this signaling pathway (26). ILK has a structural

role in linking integrins to the actin cytoskeleton (41). Endothelialcell–specific ablation of the serine/threonine kinase ILK leads tosevere vascular defects and embryonic death at E11.5 (42) andinhibition of ILK kinase activity inhibits tumor angiogenesis inmice (43). The importance of ILK serine/threonine kinase activity,however, has been questioned as genetic data from C. elegans and D.melanogaster suggest that the kinase activity is dispensable in vivo(41, 44). Of particular relevance for interpretation of data in thisstudy, ovexpression of a mutant form of ILK that lacks the ability toengage in complex formation with paxillin leads to severely reducedmotility (23). Moreover, cells overexpressing wild-type ILK fail topolarize and display a poorly spread phenotype similar to thatof cells overexpressing RhoA. Forced expression of FAK, whichnegatively regulates RhoA (45), rescues this phenotype in wild-typeILK–transfected cells, suggesting that a balanced activation of FAKand ILK is required for polarization andmigration. Our results on theeffect of HRGP330 on FAK and ILK are in excellent agreement withthe well-established and central role of these cytoplasmic kinases incell migration.Cancer is a major cause of premature death. Proof of the concept

that angiogiogenesis inhibition has clinical benefits in tumortreatment is given by the VEGF-blocking antibody Avastin(bevacizumab). Recent reports indicate that VEGFR-2 expressionand function is not restricted to angiogenic endothelium and effectson neuronal survival cannot be excluded in long-term anti-VEGFtreatments (46). We have, at this point, no indications of adverseeffects on normal vasculature by systemic HRGP/HRGP330 treat-ment but these studies will continue. It seems clear that members ofthe cystatin superfamily, to which HRGP belongs, present a numberof interesting features that support our focus on HRGP as anangiogenesis inhibitor. Thus, fetuin-B has recently been implicatedas a tumor suppressor (47) and kininogen possesses antiangiogenicproperties, also mediated via a histidine-rich domain (37). Deletionof the proximal region of mouse chromosome 16, containing fetuin,fetuin-B, HRGP, and kininogen of the cystatin superfamily, as well asother genes, has been associated with the ‘‘angiogenic switch’’ in atransgenic model of multistage tumorigenesis (48). A recent reporton HRGP gene inactivation implicates HRGP in regulation of bloodcoagulation (49). The consequence of loss of HRGP expressionfor pathologic angiogenesis awaits further characterization. Inconclusion, antiangiogenic drugs have a large potential, perhaps inparticular when combined with cytotoxic drugs or surgery, and theirdevelopment will be aided by mechanistic insights.

Acknowledgments

Received 6/23/2005; revised 11/27/2005; accepted 12/16/2005.Grant support: Swedish Cancer Society (project no. 3820-B04-09XAC), Innoventus

Project AB, and the Sixth European Union Framework Programme (Integrated Project‘‘Angiotargeting’’; contract no. 504743) in the area of Life Sciences, Genomics, andBiotechnology for Health (L. Claesson-Welsh); Magnus Bergvalls Foundation and Erik,Karin, and Gosta Selanders Foundation (A-K. Olsson); and Swedish Society of MedicalResearch (J. Dixelius).

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 Dr. Martin McMahon for the kind gift of telomerase immortalizedendothelial cells.

References

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

2. Carmeliet P, Jain RK. Angiogenesis in cancer and otherdiseases. Nature 2000;407:249–57.

3. Senger DR, Perruzzi CA, Feder J, Dvorak HF. A highlyconserved vascular permeability factor secreted by a

variety of human and rodent tumor cell lines. CancerRes 1986;46:5629–32.

4. Ferrara N, Henzel WJ. Pituitary follicular cells secrete anovel heparin-binding growth factor specific for vascular

Cancer Research

Cancer Res 2006; 66: (4). February 15, 2006 2096 www.aacrjournals.org

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

Angiogenesis Inhibition by HRGP

www.aacrjournals.org 2097 Cancer Res 2006; 66: (4). February 15, 2006

endothelial cells. Biochem Biophys Res Commun 1989;161:851–8.

5. Yang JC, Haworth L, Sherry RM, et al. A randomizedtrial of bevacizumab, an anti-vascular endothelialgrowth factor antibody, for metastatic renal cancer.N Engl J Med 2003;349:427–34.

6. Hurwitz H, Fehrenbacher L, Novotny W, et al.Bevacizumab plus irinotecan, fluorouracil, and leuco-vorin for metastatic colorectal cancer. N Engl J Med2004;350:2335–42.

7. Folkman J. Endogenous angiogenesis inhibitors.APMIS 2004;112:496–507.

8. Colman RW, Jameson BA, Lin Y, Johnson D, MousaSA. Domain 5 of high molecular weight kininogen(kininostatin) down-regulates endothelial cell prolifer-ation and migration and inhibits angiogenesis. Blood2000;95:543–50.

9. Sudhakar A, Sugimoto H, Yang C, Lively J, Zeisberg M,Kalluri R. Human tumstatin and human endostatinexhibit distinct antiangiogenic activities mediated byavh3 and a5h1 integrins. Proc Natl Acad Sci U S A 2003;100:4766–71.

10. Akerman ME, Pilch J, Peters D, Ruoslahti E.Angiostatic peptides use plasma fibronectin to hometo angiogenic vasculature. Proc Natl Acad Sci U S A2005;102:2040–5.

11. Karumanchi SA, Jha V, Ramchandran R, et al. Cellsurface glypicans are low-affinity endostatin receptors.Mol Cell 2001;7:811–22.

12. Olsson AK, Johansson I, Akerud H, et al. The minimalactive domain of endostatin is a heparin-binding motifthat mediates inhibition of tumor vascularization.Cancer Res 2004;64:9012–7.

13. Yu Y, Moulton KS, Khan MK, et al. S. E-selectin isrequired for the antiangiogenic activity of endostatin.Proc Natl Acad Sci U S A 2004;101:8005–10.

14. Jones AL, Hulett MD, Parish CR. Histidine-richglycoprotein: A novel adaptor protein in plasma thatmodulates the immune, vascular and coagulationsystems. Immunol Cell Biol 2005;83:106–18.

15. Juarez JC, Guan X, Shipulina NV, et al. Histidine-proline-rich glycoprotein has potent antiangiogenicactivity mediated through the histidine-proline-richdomain. Cancer Res 2002;62:5344–50.

16. Olsson AK, Larsson H, Dixelius J, et al. A fragment ofhistidine-rich glycoprotein is a potent inhibitor of tumorvascularization. Cancer Res 2004;64:599–605.

17. Brown WM, Dziegielewska KM. Friends and relationsof the cystatin superfamily—new members and theirevolution. Protein Sci 1997;6:5–12.

18. Olivier E, Soury E, Ruminy P, et al. Fetuin-B, a secondmember of the fetuin family in mammals. BiochemJ 2000;350 Pt 2:589–97.

19. Kluszynski BA, Kim C, Faulk WP. Zinc as a cofactorfor heparin neutralization by histidine-rich glycoprotein.J Biol Chem 1997;272:13541–7.

20. Venetsanakos E, Mirza A, Fanton C, Romanov SR,

Tlsty T, McMahon M. Induction of tubulogenesis intelomerase-immortalized human microvascular endo-thelial cells by glioblastoma cells. Exp Cell Res 2002;273:21–33.

21. Workman P, Balmain A, Hickman JA, et al. UKCCCRguidelines for the welfare of animals in experimentalneoplasia. Lab Anim 1988;22:195–201.

22. Gundersen HJ, Bendtsen TF, Korbo L, et al. Somenew, simple and efficient stereological methods andtheir use in pathological research and diagnosis. APMIS1988;96:379–94.

23. Khyrul WA, LaLonde DP, Brown MC, Levinson H,Turner CE. The integrin-linked kinase regulates cellmorphology and motility in a rho-associated kinase-dependent manner. J Biol Chem 2004;279:54131–9.

24. Claesson-Welsh L, Welsh M, Ito N, et al. Angiostatininduces endothelial cell apoptosis and activation offocal adhesion kinase independently of the integrin-binding motif RGD. Proc Natl Acad Sci U S A 1998;95:5579–83.

25. Dixelius J, Cross M, Matsumoto T, Sasaki T, Timpl R,Claesson-Welsh L. Endostatin regulates endothelial celladhesion and cytoskeletal organization. Cancer Res2002;62:1944–7.

26. Izaguirre G, Aguirre L, Hu YP, et al. The cytoskeletal/non-muscle isoform of a-actinin is phosphorylated onits actin-binding domain by the focal adhesion kinase.J Biol Chem 2001;276:28676–85.

27. von Wichert G, Haimovich B, Feng GS, Sheetz MP.Force-dependent integrin-cytoskeleton linkage forma-tion requires down-regulation of focal complex dynam-ics by Shp2. EMBO J 2003;22:5023–35.

28. Lin SY, Raval S, Zhang Z, et al. The protein-tyrosinephosphatase SHP-1 regulates the phosphorylation ofa-actinin. J Biol Chem 2004;279:25755–64.

29. Simantov R, Febbraio M, Crombie R, Asch AS,Nachman RL, Silverstein RL. Histidine-rich glycoproteininhibits the antiangiogenic effect of thrombospondin-1.J Clin Invest 2001;107:45–52.

30. Simantov R, Febbraio M, Silverstein RL. The anti-angiogenic effect of thrombospondin-2 is mediated byCD36 and modulated by histidine-rich glycoprotein.Matrix Biol 2005;24:27–34.

31. Donate F, Juarez JC, Guan X, et al. Peptides derivedfrom the histidine-proline domain of the histidine-proline-rich glycoprotein bind to tropomyosin and haveantiangiogenic and antitumor activities. Cancer Res2004;64:5812–7.

32. Eliceiri BP, Cheresh DA. Role of av integrins duringangiogenesis. Cancer J 2000;6 Suppl 3:S245–9.

33. 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. EMBO J1999;18:882–92.

34. Tompa P. Intrinsically unstructured proteins. TrendsBiochem Sci 2002;27:527–33.

35. Guan X, Juarez JC, Qi X, et al. Histidine-proline rich

glycoprotein (HPRG) binds and transduces anti-angio-genic signals through cell surface tropomyosin onendothelial cells. Thromb Haemost 2004;92:403–12.

36. Perry SV. Vertebrate tropomyosin: distribution,properties and function. J Muscle Res Cell Motil 2001;22:5–49.

37. Zhang JC, Donate F, Qi X, et al. The antiangiogenicactivity of cleaved high molecular weight kininogen ismediated through binding to endothelial cell tropomy-osin. Proc Natl Acad Sci U S A 2002;99:12224–9.

38. MacDonald NJ, Shivers WY, Narum DL, et al.Endostatin binds tropomyosin. A potential modulatorof the antitumor activity of endostatin. J Biol Chem2001;276:25190–6.

39. Comoglio PM, Boccaccio C, Trusolino L. Interactionsbetween growth factor receptors and adhesion mole-cules: breaking the rules. Curr Opin Cell Biol 2003;15:565–71.

40. Ilic D, Furuta Y, Kanazawa S, et al. Reduced cellmotility and enhanced focal adhesion contact forma-tion in cells from FAK-deficient mice. Nature 1995;377:539–44.

41. Zervas CG, Gregory SL, Brown NH. Drosophilaintegrin-linked kinase is required at sites of integrinadhesion to link the cytoskeleton to the plasmamembrane. J Cell Biol 2001;152:1007–18.

42. Friedrich EB, Liu E, Sinha S, et al. Integrin-linkedkinase regulates endothelial cell survival and vasculardevelopment. Mol Cell Biol 2004;24:8134–44.

43. Tan C, Cruet-Hennequart S, Troussard A, et al.Regulation of tumor angiogenesis by integrin-linkedkinase (ILK). Cancer Cell 2004;5:79–90.

44. Mackinnon AC, Qadota H, Norman KR, MoermanDG, Williams BD. C. elegans PAT-4/ILK functions as anadaptor protein within integrin adhesion complexes.Curr Biol 2002;12:787–97.

45. Chen BH, Tzen JT, Bresnick AR, Chen HC. Roles ofRho-associated kinase and myosin light chain kinase inmorphological and migratory defects of focal adhesionkinase-null cells. J Biol Chem 2002;277:33857–63.

46. Storkebaum E, Lambrechts D, Dewerchin M, et al.Treatment of motoneuron degeneration by intracere-broventricular delivery of VEGF in a rat model of ALS.Nat Neurosci 2005;8:85–92.

47. Hsu SJ, NagaseH, Balmain A. Identification of Fetuin-Bas a member of a cystatin-like gene family on mousechromosome 16 with tumor suppressor activity. Genome2004;47:931–46.

48. Parangi S, Dietrich W, Christofori G, Lander ES,Hanahan D. Tumor suppressor loci on mouse chromo-somes 9 and 16 are lost at distinct stages of tumorigenesisin a transgenic model of islet cell carcinoma. Cancer Res1995;55:6071–6.

49. Tsuchida-Straeten N, Ensslen S, Schafer C, et al.Enhanced blood coagulation and fibrinolysis in micelacking histidine-rich glycoprotein (HRG). J ThrombHaemost 2005;3:865–72.

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: Minimal Active Domain and Mechanism of Action of the ...Minimal Active Domain and Mechanism of Action of the Angiogenesis Inhibitor Histidine-Rich Glycoprotein Johan Dixelius, Anna-Karin

2006;66:2089-2097. Cancer Res   Johan Dixelius, Anna-Karin Olsson, Åsa Thulin, et al.   Angiogenesis Inhibitor Histidine-Rich GlycoproteinMinimal Active Domain and Mechanism of Action of the

  Updated version

  http://cancerres.aacrjournals.org/content/66/4/2089

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/66/4/2089.full#ref-list-1

This article cites 49 articles, 24 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/66/4/2089.full#related-urls

This article has been cited by 8 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/66/4/2089To request permission to re-use all or part of this article, use this link

Research. on April 24, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from