9
Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs Jeong Heon Lee c , Taehoon Chun d , Sang-Yoon Park b , Seung Bae Rho a, a Research Institute, National Cancer Center, 809, Madu 1-dong, Ilsan-gu, Goyang-si, Gyeonggi-do 411-769, Republic of Korea b Department of Obstetrics and Gynecology, Research Institute, National Cancer Center, Goyang, Gyeonggi 411-769, Republic of Korea c Department of Obstetrics and Gynecology, Chonbuk National University Medical School, Jeonju 561-712, Republic of Korea d Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea ABSTRACT ARTICLE INFO Article history: Received 11 February 2008 Received in revised form 31 March 2008 Accepted 7 April 2008 Available online 22 April 2008 Keywords: Interferon regulatory factor Angiogenesis Alternative splicing Endothelial cell Vessel sprouting CAM Interferon regulatory factor-1 (IRF-1) is a tumor suppressor and transcriptional modulator that can regulate gene expression involved in cell growth control, induction of apoptosis, and post-translation modication. In this study, we found that IRF-1 inhibits endothelial cell angiogenesis using human umbilical vein endothelial cell (HUVECs) culture system. In addition, IRF-1 directly inhibited the tube formation of endothelial cells on Matrigel and reduced the expression of p-Akt, and p-eNOS, which play a signicant role in angiogenesis when stimulated by VEGF. We also demonstrate that C-terminal region including transactivation domain (TA) of IRF-1 functions as a signal for its angiostatic activity, and is spliced in human tumor tissues. These ndings indicate that splicing variant involving exons 7 of IRF-1 could potentially modulate anti-angiogenic effect of IRF-1. In overall, this study provides the rst evidence for anti-angiogenic role of IRF-1, which may have therapeutic values for cancer and angiogenesis-associated diseases. © 2008 Elsevier B.V. All rights reserved. 1. Introduction Interferon regulatory factor-1 (IRF-1), the founding member of the IRF family, is a transcription factor involved in cell growth regulation, induction of apoptosis, immune responses, post-transcription mod- ication, and cell transformation by oncogenes [15]. The best characterized activity of IRF-1 is as a sequence specic DNA-binding domain and transcriptional activator of certain IFN-stimulated genes [6]. Accumulating evidence supports the theory that IRF-1 functions as a tumor suppressor [710] and revert the transformed phenotype [9,1113]. In human tumors, IRF-1 is inactivated to prevent cell cycle arrest and apoptosis by genetic alteration, such as gene and exon deletion [10,1416]. IRF-1 also has been shown to play roles in regulating both ubiquitination and SUMOylation in cancer [5,17,18]. Angiogenesis, the formation of new blood vessels from pre- existing vasculature, is a crucial step in tumor growth [19,20] and progression because it enables the supply of oxygen and nutrients to the growing tumor [21]. One of the key mediators of blood vessel formation during development is vascular endothelial growth factor (VEGF), which can stimulate the proliferation and migration in endothelial cells [22]. In tumors, the activity of endothelial cells plays an essential role for the regulation of various vascular biological function and diseases. IRF family members share signicant homology with the N-terminal 120 amino acids, which comprise a DNA-binding domain (DBD) characterized by ve tryptophan repeats [2,23]. The DBD also revealed as a region required for homodimerization and nuclear localization [24]. The C-terminus of IRF-1 contains a transactivation domain (TA) between amino acids 185 and 256, but this amino acid region is not present in the transcriptional repressor IRF-2. A functional antagonist of IRF-1 is IRF-2 [2], which functions as a tumor suppressor and as an oncoprotein [11,25]. In the present work, we evaluated the role of IRF-1 during angio- genesis using a human umbilical vein endothelial cell (HUVECs) culture system with puried protein of IRF-1 in vitro. We found that IRF-1 inhibited HUVECs migration and tube formation on Matrigel. Further, we dened the functional domains of IRF-1 causing angiogenesis. This study may provide evidence for the potential application of the IRF-1 as an anti-angiogenic therapy for the angiogenesis-associated disease such as cancer. 2. Materials and methods 2.1. Cell culture and antibiotics Human umbilical vein endothelial cells (HUVECs) were obtained from Clonetics (Walkersville, MD), and maintained on 0.3% gelatin (Sigma, St. Louis, MO) coated dishes using the EGM-2 BulletKit medium (Clonetics) in an atmosphere of 5% CO 2 in air at 37 °C. The EGM-2 BulletKit medium consists of a base medium containing 20μM HEPES (Sigma), 5% heat inactivated fetal calf serum, and 1% PenicillinStreptomycin (Gibco, New York, NY). HUVECs were used for experiments at passages 35. The following antibodies were used in this study: anti-IRF-1 (Santa Cruz Biotechnology, Santa Cruz, Biochimica et Biophysica Acta 1783 (2008) 16541662 Corresponding author. Tel.: +82 31 920 2383; fax: +82 31 920 2337. E-mail address: [email protected] (S.B. Rho). 0167-4889/$ see front matter © 2008 Elsevier B.V. All rights reserved. doi:10.1016/j.bbamcr.2008.04.006 Contents lists available at ScienceDirect Biochimica et Biophysica Acta journal homepage: www.elsevier.com/locate/bbamcr

Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

Embed Size (px)

Citation preview

Page 1: Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

Biochimica et Biophysica Acta 1783 (2008) 1654–1662

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

j ourna l homepage: www.e lsev ie r.com/ locate /bbamcr

Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesisin HUVECs

Jeong Heon Lee c, Taehoon Chun d, Sang-Yoon Park b, Seung Bae Rho a,⁎a Research Institute, National Cancer Center, 809, Madu 1-dong, Ilsan-gu, Goyang-si, Gyeonggi-do 411-769, Republic of Koreab Department of Obstetrics and Gynecology, Research Institute, National Cancer Center, Goyang, Gyeonggi 411-769, Republic of Koreac Department of Obstetrics and Gynecology, Chonbuk National University Medical School, Jeonju 561-712, Republic of Koread Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea

⁎ Corresponding author. Tel.: +82 31 920 2383; fax: +E-mail address: [email protected] (S.B. Rho).

0167-4889/$ – see front matter © 2008 Elsevier B.V. Aldoi:10.1016/j.bbamcr.2008.04.006

A B S T R A C T

A R T I C L E I N F O

Article history:

Interferon regulatory factor- Received 11 February 2008Received in revised form 31 March 2008Accepted 7 April 2008Available online 22 April 2008

Keywords:Interferon regulatory factorAngiogenesisAlternative splicingEndothelial cellVessel sproutingCAM

1 (IRF-1) is a tumor suppressor and transcriptional modulator that can regulategene expression involved in cell growth control, induction of apoptosis, and post-translation modification. Inthis study, we found that IRF-1 inhibits endothelial cell angiogenesis using human umbilical vein endothelialcell (HUVECs) culture system. In addition, IRF-1 directly inhibited the tube formation of endothelial cells onMatrigel and reduced the expression of p-Akt, and p-eNOS, which play a significant role in angiogenesiswhen stimulated by VEGF. We also demonstrate that C-terminal region including transactivation domain (TA)of IRF-1 functions as a signal for its angiostatic activity, and is spliced in human tumor tissues. These findingsindicate that splicing variant involving exons 7 of IRF-1 could potentially modulate anti-angiogenic effect ofIRF-1. In overall, this study provides the first evidence for anti-angiogenic role of IRF-1, which may havetherapeutic values for cancer and angiogenesis-associated diseases.

© 2008 Elsevier B.V. All rights reserved.

1. Introduction

Interferon regulatory factor-1 (IRF-1), the founding member of theIRF family, is a transcription factor involved in cell growth regulation,induction of apoptosis, immune responses, post-transcription mod-ification, and cell transformation by oncogenes [1–5]. The bestcharacterized activity of IRF-1 is as a sequence specific DNA-bindingdomain and transcriptional activator of certain IFN-stimulated genes[6]. Accumulating evidence supports the theory that IRF-1 functions asa tumor suppressor [7–10] and revert the transformed phenotype[9,11–13]. In human tumors, IRF-1 is inactivated to prevent cell cyclearrest and apoptosis by genetic alteration, such as gene and exondeletion [10,14–16]. IRF-1 also has been shown to play roles inregulating both ubiquitination and SUMOylation in cancer [5,17,18].

Angiogenesis, the formation of new blood vessels from pre-existing vasculature, is a crucial step in tumor growth [19,20] andprogression because it enables the supply of oxygen and nutrients tothe growing tumor [21]. One of the key mediators of blood vesselformation during development is vascular endothelial growth factor(VEGF), which can stimulate the proliferation and migration inendothelial cells [22]. In tumors, the activity of endothelial cellsplays an essential role for the regulation of various vascular biologicalfunction and diseases.

82 31 920 2337.

l rights reserved.

IRF familymembers share significant homologywith the N-terminal120 amino acids, which comprise a DNA-binding domain (DBD)characterized by five tryptophan repeats [2,23]. The DBD also revealedas a region required for homodimerization and nuclear localization [24].The C-terminus of IRF-1 contains a transactivation domain (TA) betweenamino acids 185 and 256, but this amino acid region is not present in thetranscriptional repressor IRF-2. A functional antagonist of IRF-1 is IRF-2[2], which functions as a tumor suppressor and as an oncoprotein[11,25].

In the present work, we evaluated the role of IRF-1 during angio-genesis using a human umbilical vein endothelial cell (HUVECs)culture system with purified protein of IRF-1 in vitro. We found thatIRF-1 inhibited HUVECs migration and tube formation on Matrigel.Further, we defined the functional domains of IRF-1 causingangiogenesis. This study may provide evidence for the potentialapplication of the IRF-1 as an anti-angiogenic therapy for theangiogenesis-associated disease such as cancer.

2. Materials and methods

2.1. Cell culture and antibiotics

Human umbilical vein endothelial cells (HUVECs) were obtained from Clonetics(Walkersville, MD), and maintained on 0.3% gelatin (Sigma, St. Louis, MO) coated dishesusing the EGM-2 BulletKit medium (Clonetics) in an atmosphere of 5% CO2 in air at37 °C. The EGM-2 BulletKit medium consists of a base medium containing 20μMHEPES(Sigma), 5% heat inactivated fetal calf serum, and 1% Penicillin–Streptomycin (Gibco,New York, NY). HUVECs were used for experiments at passages 3–5. The followingantibodies were used in this study: anti-IRF-1 (Santa Cruz Biotechnology, Santa Cruz,

Page 2: Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

Fig. 1. The effect of human IRF-1 on endothelial cell migration. (A) Variousconcentrations of IRF-1 were used for the assays. Serum-starved HUVECs were platedonto the Transwell plate with different doses of IRF-1 (25–200 ng/ml). After 24 hincubation, HUVECs migrated to the 96-well chamber were calculated by a fluorescencestaining and quantitation. The data are expressed as the means ± SD of threeindependent experiments which were performed in duplicate. (B) In the time course,serum-starved HUVECs were plated onto the Transwell platewith or without 100 ng/mlIRF-1. HUVECs migration was investigated at 4, 6, 12, and 24 h. Each bar represents themeans ± SD. ⁎, P b 0.05 and ⁎⁎, P b 0.01 as compared to untreated cells.

1655J.H. Lee et al. / Biochimica et Biophysica Acta 1783 (2008) 1654–1662

CA), anti-Akt (Santa Cruz), anti-phospho-specific Akt (Santa Cruz), anti-eNOS (SantaCruz), anti-phospho-specific eNOS (Santa Cruz), anti-PI3K (Santa Cruz), anti-VEGF165(Ab-1, Oncogene), anti-VEGF121 (Santa Cruz), anti-β-actin (Sigma), and anti-GFP (SantaCruz).

2.2. Small interfering RNA (siRNA) construction

The siRNA oligonucleotide sequence targeting IRF-1 (CTACTCCTTCCCTTTAATG)corresponded to nucleotides 879 to 897 in the human sequence. siRNAwas synthesizedby using a siRNA Construction kit (Ambion, Austin, TX) and transfected by using theRNAi shuttle (Orbigen, San Diego, CA) according to the manufacturer's protocols.HUVECs were then transfected with GFP-IRF-1 or GFP-IRF-1 with siRNA transfection.GFP imageswere captured using a fluorescencemicroscope (Zeiss, Oberoken, Germany).Total RNA was isolated using a TRIZOL Reagent (Life Technologies, Gaithersburg, MD)and reverse transcription (RT) PCR was then performed. The transfection efficiency wasexamined with respect to the expression level of co-transfected green fluorescentprotein (GFP), which was confirmed by immunoblotting with an anti-IRF-1 antibody.

2.3. Protein expression, purification and biochemical analysis

DNA encoding the three splicing variants of IRF-1 and full-length IRF-1 wereisolated by polymerase chain reaction and subcloned into pET28a (Novagen) usingEcoRI and XhoI. Then each construct was expressed in Escherichia coli strain BL21 (DE3)grown in Luria Bertani medium supplemented with kanamycin (75mg/ml). Culture wasgrown at 37 °C to an A600 = 0.4 ~ 0.5, transferred at 30 °C, and were induced by theaddition of 0.4 mM isopropyl-1-thio-β-D-galactopyranoside (IPTG) for 4h. The inducedcells were then harvested and resuspended in 20 mM Tris–HCl (pH 8.0) containing 10%glycerol, 50 mM NaCl, 0.1 mM EDTA, and 1 mM dithiothreitol. Cells were lysed byultrasonication, and were centrifuged for 15 min to remove cell debris. The supernatantwas filtered and loaded onto a nickel affinity column matrix (Invitrogen) and incubatedat 4 °C for 2h. The slurry was pelleted by centrifugation and washed with washingbuffer (20 mMNa2HPO4, pH 6.0, 500 mMNaCl) three times. The pellet of the gel matrixwas resuspended in elution buffer (20 mM Na2HPO4, pH 6.0, 500 mM NaCl containing400mMimidazole) and incubated at 4 °C for 20min to elute the boundHis fusionproteins.The His-tag was cleaved off with thrombin, and the each protein was further purifiedusing high performance liquid chromatography (Waters 600). Protein concentrationwas determined by the Bradford assay with bovine serum albumin (Sigma) as a standard(Bio-Rad).

2.4. Transient transfection and in vitro specific activity assay

Transient transfections of 70–80% confluent HUVECs were performed using theFugene 6 Reagent (Roche, Mannheim, Germany) according to the manufacturer'sinstructions. Luciferase reporter assay was performed as previously described [16]. TheHUVECs plated on 60-mm dishes and triply transfected using Fugene 6 Reagent withfull-length IRF-1 or its splicing variants (200 ng each), pGL-3 containing the interferon-stimulated response element (150 ng), and pRL containing the Renilla luciferase cDNA(50 ng). Twenty-four hours after transfection, the cells were harvested and dissolved in40ml of 1× passive lysis buffer (Promega, Madison,WI). After lysis, the cell extracts wereincubatedwith luciferase substrate for 30min at room temperature. Lysateswere clearedby centrifugation for 15 min at 14,000 rpm, and then a 10 μl aliquot of each sample wastransferred to a 96-well assay luminometer plate containing 50 μl/well of the providedLuciferase Assay Reagent II. The provided Stop and Glo Reagent (50 μl/well) was thenadded to initiate Renilla luciferase activity, and the ratio of firefly luciferase activity toRenilla luciferase activity was calculated.

2.5. [3H]thymidine uptake assay

To measure cell proliferation [26], HUVECs were seeded at a density of 9 × 103 cellsper well in DMEM containing 5% heat inactivated fetal calf serum, and 1% Penicillin–Streptomycin in the gelatinized plates on day 0. After 18h, cells were incubated for 6 h inM199 containing 1% FBS and then stimulated with VEGF (10 ng/ml, R&D Systems,Minneapolis, MN) for 24 h in M199 containing 1% FBS. [3H]thymidine (0.5 μCi/ml,Amersharm, Arlington, IL) was then added 4h prior to the assay. High molecular mass[3H]-radioactivity was precipitated using 5% trichloroacetic acid at 4 °C for 1 h. Cellswere solubilized in 0.2N NaOH and 0.1% SDS and [3H]thymidine uptake was evaluatedwith a liquid scintillation counter (Beckman Instrument). Three independent experi-ments were conducted in triplicate; values shown represent means ± SD of total cpm.

2.6. Migration and invasion assay

Migration and invasion were assayed using Transwells (Costar, 8 μm pore size) aspreviously described [26]. For migration assays, the lower surface of a filter was coatedwith 10 μg of gelatin. M199 containing 1% FBS with VEGF (25 ng/ml) was placed in thelower wells. Cells were fixed and stained with H&E. Non-migrating cells on the upperfilter surface were removed by wiping with a cotton swab. The numbers of cells thatmigrated to the lower side of the filter were counted under a light microscope and themean values of eight fields were determined. For the invasion assay, the lower andupper surfaces of a filter were coated with 10 μg of gelatin and 10 μg of Matrigel (BDbiosciences), respectively. The fixation and quantification methods used were the same

as described for the migration assay. Three independent experiments were conductedin triplicate; and the values shown represent means ± SD.

2.7. Tube formation assay

Growth factor-reduced Matrigel (200 μl of 10 mg/ml) was added to a 24-well plateand polymerized for 30min at 37 °C. Untransfected, pEGFP, pEGFP-IRF-1, or siIRF-1-transfected HUVECs (1×105 cells) were seeded on the surface of theMatrigel. Cells werethen incubated for 48 h with or without 10 ng/ml of VEGF in M199 containing 1% FBS.Morphological changes were photographed at ×40 magnification. HUVEC tube lengthswere determined using an inverted microscope equipped with a digital CCD camera(Zeiss) and quantified using ImageLab imaging software (MCM Design).

2.8. Ex vivo angiogenesis assay

A novel ex vivo angiogenesis assay using an explant culture of skeletal muscle onMatrigel was done with some modifications, according to Jang et al. [27]. Six-week-oldBALB/c mice were anesthetized and the legs were shaved. The tibialis anterior musclewas extracted and the cross-sections taken from the muscle were washed thrice withPBS. The washed muscle was placed in a 24-well plate containing 200 μl of growthfactor-reduced Matrigel and polymerized for 30min at 37 °C. M199 containing 1% FBSwith or without 10 ng/ml of VEGF was added. After 6 days, outgrowths of capillary-likestructures were observed. The observed structures were then treated with Mock, IRF-1,or IRF-1 siRNA for 5 days. The mean area of the microvessels was measured byemploying an optical imaging technique and the measurements were then quantifiedusing ImageLab imaging software. Independent experiments were repeated thrice andeach value represents the mean ± SD of triplicate samples.

2.9. Chick chorioallantoic membrane (CAM) assay

The CAM assay was performed with minor modifications [28]. Fertilized chickembryos were pre-incubated at 37 °C with 70% humidity in order to conduct thechorioallantoic membrane assay. After 3 days, a square window was opened after theremoval of 2–3 ml of albumin to detach the developing CAM from the shell. A1.5×1.5 cm window in the shell was made to expose the CAM. Clear tape was used toseal the windows that were formed and the eggs were incubated for 60 h. On day 8,CAMs were implanted, under sterile conditions within a laminar flow hood, withsterilized Thermanox discs. Thermanox discs were loaded with 100 ng Mock, IRF-1, or

Page 3: Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

1656 J.H. Lee et al. / Biochimica et Biophysica Acta 1783 (2008) 1654–1662

siRNA-treated recombinant protein as negative and positive controls, respectively. TheCAM was examined daily until day 12 and photographed in ovo with an Axioskope2plus microscope (Zeiss) equipped with color CCD camera (ProgResC14, Jenoptik,Germany). Recombinant human VEGF (100 ng), incorporated into Thermanox discs,induced branching of blood vessels. At a dose of 100 ng/disk, IRF-1 inhibited theseresponses in 90% (n = 9/10) of CAMs. At similar dose, both Mock or siRNA-treated groupdid not show angiogenesis inhibition (0%, n = 0/10). Two independent, blindedinvestigators performed the count of blood vessels for each group.

2.10. Western blot analysis of PI3K, Akt, and eNOS phosphorylation

A Western blot analysis of PI3K, Akt, and eNOS was conducted. VEGF was used tostimulate the HUVECs for 20 min. The cells were then lysed, and equal amounts ofprotein were separated using SDS-PAGE. The separated protein was then transferred toa nitrocellulose membrane. After incubation in blocking solution, membranes wereincubated with anti-PI3K, anti-p-Akt, and anti-p-eNOS antibody for 1.5 h at roomtemperature. An ECL system (Amersham) was used for detection.

Fig. 2. Activity of human IRF-1 domain on HUVECs. (A) Ablation of GFP-IRF-1 protein andvisualized under fluorescence microscope, and analyzed the mRNA or protein expression levhuman IRF-1. Human IRF-1 (100 ng) was assayed on HUVECs in the presence of VEGF165 in ascintillation counter. (C) and (D) To investigate whether overexpressed IRF-1 modulates the ethe migration assays (C) or on Matrigel-coated Transwell for the invasion assays (D) followinvaded cells were counted under a light microscope and mean values were determined. ⁎,

2.11. Total RNA extraction and RT-PCR

Total cellular RNAwas isolated using TRIZOL Reagent according to themanufacturer'sinstructions, and 2 μg of total RNA was reverse transcribed in a reaction volume of 20 μlusing the Superscript II enzyme (Invitrogen). The resulting cDNAwas used as the templatefor PCRamplificationof the full-length IRF-1gene (forward5′-GCGCTCCGCGCGCCCTCACT-3′ and reverse 5′-GCCAGGTCCTGCTTGCCTAG-3′), or two fragments of the IRF-1 encodinggene (N-terminal half forward 5′-GCGCTCCGCGCGCCCTCACT-3′ and reverse 5′-TAGCTGCTGTGGTCATCA-3′, comprising exons 2, 3, 4, and 5; C-terminal half forward 5′-AAGTCATGTGGGGATTCCAG-3′ and reverse 5′-GCCAGGTCCTGCTTGCCTAG-3′ comprisingexons6, 7, 8, 9, and10). The three splicingvariantswere amplifiedusing theC-terminalhalfforward primer and specific reverse primers (Δ7, 5′-TGTAGCTTCAGAGGTGGA-3′; Δ8, 5′-CTCCAAGAGCTGGAGTCA-3′; Δ9, 5′-CCCAATATCCCCCCCTGG-3′). The PCR conditionsconsisted of the indicated number of cycles at 95 °C for 30s, 61 °C for 30s (but 58 °C forΔ9), and 72 °C for 30s. The resulting fragments were resolved by 1% agarose gel electro-phoresis and confirmed by automatic sequencing (ABI 373, PerkinElmer Life Sciences).

mRNA expression by siRNA in HUVECs. Bar, 50 μm. After transfection, the cells wereels by RT-PCRs and immunoblots. (B) Inhibition of DNA synthesis in endothelial cells by72h proliferation experiment. cpm value of [3H]thymidine was determined with a liquidffects of VEGF on endothelial cell migration and invasion, we tested using Transwells fored by stimulation with or without VEGF (25 ng/ml) for 48 h. Numbers of migrated orPb0.05; ⁎⁎, Pb0.01 compared with VEGF alone.

Page 4: Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

1657J.H. Lee et al. / Biochimica et Biophysica Acta 1783 (2008) 1654–1662

2.12. Yeast two-hybrid analysis and coimmunoprecipitation

The cDNAs encoding human VEGFR1 or VEGFR2 were constructed into pGildaexpression vector, respectively. The resulting plasmid pGilda-VEGFR1 or VEGFR2 wereintroduced into yeast strain EGY48 [MATa, his3, trp1, ura3-52, leu2: :pLeu2-LexAop6/pSH18-34 (LexAop-lacZ reporter)] by a modified lithium acetate method. The cDNAsencoding B42-IRF-1 fusion protein were introduced into the competent yeast cells thatalready contained pGilda-VEGFR1 or VEGFR2 and the tryptophan prototrophy (plasmidmarker) transformants were selected on a synthetic medium (Ura−, His−, Trp−) containing2% glucose. The interaction between the VEGFR1 or VEGFR2 and IRF-1 was compared bymeasuring the expression level of the two reporter genes. Theβ-galactosidase activitywasdetermined according to the method as described [29]. Yeast cells containing each of theconstructions were cultured in SD media until they reached a mid-log phase. The culturebroth (0.4 ml) was taken and mixed with Z buffer (1.4 ml) containing 2-mercaptoethanol.Chloroform (100 μl) and 0.1% SDS (100 μl) were added to the mixture, and the cellswere vortex mixed for 45s. The reaction substrate O-nitrophenyl β-D-galactopyranoside(ONPG) (0.32 ml) was added and the reactionwas carried out at 30 °C until a yellow colorappeared, and was then quenched by adding 1M Na2CO3. Samples were then centrifugedbriefly to remove cell debris, and the absorbance of the supernatant was measured at420nm.

For co-immunoprecipitation, HUVECs were co-transfected with cDNA constructs ofpEGFPC1-IRF-1 and pcDNA4/HisMax-VEGGR1 or VEGFR2 using Fugene 6 Reagent. As anegative control, pEGFPC1-IRF-1 and empty vector pcDNA4/HisMax were also co-transfected. Two days after transfection, cells were harvested by trypsinization andcentrifugation. Cell pellets were washed in PBS, resuspended in cell lysis solution(50mM Tris, pH 7.2,150mMNaCl,1% Triton X-100,1 μg/ml leupeptin, 1 μg/ml pepstatin,2 μg/ml aprotinin, 200 μg/ml PMSF). Lysates were incubated with anti-His antibody(Santa Cruz) and precipitated with protein A-agarose beads. Immunoprecipitates wereresolved by SDS-PAGE, and immunoblottedwith anti-GFP antibody or anti-His antibody(Santa Cruz). An ECL system (Amersham) was used for detection.

2.13. s.c. tumor models and immunohistochemistry

Specific pathogen-free BALB/c and nu/nu mice were purchased from Biogenomics(Seoul, Korea) and Charles River Labs (Wilmington, MA), respectively. To establishtumors in mice, 1×106 of 2774 tumor cells were injected s.c. in the mid-dorsal region.Tumors were allowed to grow for 11 days. Then, an intratumoral injection of IRF-1 wasdone thrice, once every 3 days. Tumor growthwas determined by calipermeasurementsevery 3 days. Tumor volume was calculated by the following formula: tumor volume(mm3) = (a×b2) / 2, where a = length in mm and b = width in mm. Tumor volume and

Fig. 3. IRF-1 inhibits tube formation in vitro and vessel sprouting ex vivo. (A) HUVECs were eithreducedMatrigel and then treatedwith orwithout VEGF (10 ng/ml) for 48 h. The formation ofand are expressed as themeans±SD. ⁎, Pb0.05 comparedwith VEGFalone. (B) Cross-sections oor without VEGF for 6 days and treated with Mock, IRF-1, or IRF-1 siRNA for 5 days. Outgrowt×12.5 and ×40, top and bottom, respectively). Bar, 500 µm. Mean area of vascular sprouting w

body weight was measured every other day. Mice were sacrificed on day 24 after finalinjection. Tumors were then excised and prepared for immunohistochemistry.

2.14. Data and statistical analysis

All datavalues are presented as themean± SDormeans ± SEM. Statistical comparisonswere carried out using the Student's t-test. P b 0.05 were considered relevant.

3. Results

3.1. IRF-1 decreases the VEGF-induced migration of HUVECs in adose-dependent manner

In order to confirm the effect of IRF-1 on HUVECs migration, cellswere treated with several concentrations of IRF-1 (25–200 ng/ml). Weobserved that IRF-1 decreased the VEGF-induced migration ofHUVECs in a concentration-time-dependent manner and maximumeffect was at 100 ng/ml (Fig. 1A). To confirm the above finding, weinvestigated a time course at 4, 6, 12, and 24 h and observed aninhibitory effect of IRF-1 (100 ng/ml) on cell migration (Fig. 1B). After24 h of treatment with increasing concentrations (25–200 ng/ml) ofIRF-1, no cell migration change was observed in the treated sample(data not shown). The results indicate that VEGF-induced endothelialcell migration is specifically inhibited by IRF-1, whereas was notcaused by its cytotoxic effect.

3.2. IRF-1 inhibits VEGF-induced proliferation, migration, and invasion ofHUVECs

To further confirm the effect of IRF-1 on endothelial proliferation,weused RNA interference. As shown in Fig. 2A, siRNA markedly inhibitedthe expression of GFP-IRF-1 protein and mRNA in HUVECs (lane 2).However, siRNA did not affect the expression of an irrelevant gene(GAPDH). HUVECs were either untreated or treated with Mock, IRF-1 or

er untreated or treated withMock, IRF-1, or IRF-1 siRNA, were seeded on growth factor-tubular structureswas detected by an invertedmicroscope. Tube lengthswere quantifiedfmouse tibialis anteriormusclewere embedded in growth factor-reducedMatrigelwithh of capillary-like structures was observed with an invertedmicroscope (magnifications,as quantified and expressed as the means±SD. ⁎, Pb0.05 compared with VEGF alone.

Page 5: Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

Fig. 5. IRF-1 interacts with VEGFR2 in vivo and in vitro. (A) Testing the interactionbetween IRF-1 and VEGFR2 in the yeast two-hybrid system. Positive interactions wererevealed by observing cell growth over 3 days at 30 °C on medium lacking leucine, andwith the formation of blue colonies on medium containing X-gal (left). β-galactosidaseactivity was then measured adding O-nitrophenyl β-D-galactopyranoside (ONPG)(right). (B) Co-immunoprecipitation of IRF-1 with VEGFR2 and VEGFR1. Lane 1; lysatefrom pEGFPC1-IRF-1 and pcDNA4/HisMax (vector only) co-transfectant, 2; lysate frompEGFPC1-IRF-1 and pcDNA4/HisMax-VEGFR2 co-transfectant (left), Lane 1; lysate frompEGFPC1-IRF-1 and pcDNA4/HisMax (vector only) co-transfectant, 2; lysate frompEGFPC1-IRF-1 and pcDNA4/HisMax-VEGFR1 co-transfectant (right). IP means ‘immu-noprecipitation’ and WB means ‘immunoblotting’ with the indicated antibodies.

Fig. 4. Angiostatic activity of IRF-1 in the chicken chorioallantoic membrane (CAM)assay. (A) Digital images of in vivo angiogenesis assay in CAM. The open box indicatesthe point at which the Mock, IRF-1, or siIRF-1-treated coverslips were loaded onto theCAM of the developing eggs. (B) A quantitative evaluation of the angiostatic activity. Thedata are presented as the mean number of blood vessel branches±standard deviationsfrom 6 to 9 embryos in each sample. The VEGF-treated vector (Mock) was only used as apositive control. ⁎, Pb0.05 compared with VEGF alone.

1658 J.H. Lee et al. / Biochimica et Biophysica Acta 1783 (2008) 1654–1662

IRF-1 with siRNA (IRF-1-targeted short interfering RNA), then DNAsynthesis was assayed using [3H]thymidine incorporation (Fig. 2B). TheVEGF-induced DNA synthesis was observed in both untreated HUVECsand empty Mock-treated HUVECs when compared with those ofunstimulated cells [30]. Overexpression of IRF-1 markedly inhibitedVEGF-induced DNA synthesis. This inhibitory effect was not due to thecytotoxicity of IRF-1 in endothelial cells, since IRF-1 had no effect on theviability of HUVECs in the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenylte-trazolium bromide assay (data not shown). In addition, IRF-1 did notexhibit cytotoxic effects on other normal cell types tested, such asnormal fibroblasts, MRC-5 and IMR-90 (data not shown). The inhibitoryeffect of IRF-1 on VEGF-induced endothelial cell proliferation wascompletely recovered by siRNA infection. The results indicate that IRF-1specifically inhibits VEGF-induced endothelial cell proliferation.

To investigate whether overexpressed IRF-1 modulates the effectsof VEGF on endothelial cell migration and invasion, we conductedTranswell migration and invasion assays. VEGF enhanced the migra-tion (Fig. 2C) and invasion (Fig. 2D) of untreated HUVECs and of emptyMock-treatedHUVECswhen comparedwith that of unstimulated cells.However, overexpression of IRF-1 significantly reduced VEGF-inducedmigration and invasion ofHUVECs. The ablation of overexpressed IRF-1by siRNAmaintained the stimulatory effects of VEGF onmigration andinvasion of HUVECs. Therefore, the overexpression of IRF-1 potentlyinhibited key events of the angiogenic process induced by VEGF, suchas proliferation, migration, and invasion of endothelial cells in vitro.

3.3. IRF-1 inhibits tube formation in vitro and vessel sprouting ex vivo

To confirm IRF-1′s direct anti-angiogenic effects, we investigatedwhether overexpression of IRF-1 could alter endothelial tube formation.Untreated or empty Mock-treated cells incubated with VEGF formed anorganized network of endothelial cells on Matrigel (Fig. 3A). In contrasthowever, overexpression of IRF-1 markedly inhibited VEGF-induced

tube formation. The inhibitory effect of IRF-1 on VEGF-induced tubeformation was completely recovered by siRNA transfection.

To evaluate whether IRF-1 inhibits vessel sprouting, an ex vivoexplant assay [27] was done (Fig. 3B). A vast amount of vessel sproutingwas detected in empty Mock-treated explants in the presence of VEGF.Overexpression of IRF-1 showed significantly reduced VEGF-inducedvessel sprouting, and was almost recovered by siRNA. These observa-tions suggest that IRF-1 effectively suppressed capillary formation invitro and ex vivo.

3.4. Angiostatic activity of IRF-1 in the chicken chorioallantoic membrane(CAM) assay

Chick CAM assays were performed to test for angiostatic activity ofIRF-1 on in vivo angiogenesis. As shown in Fig. 4, VEGF-inducedangiogenesis in CAMs were clearly inhibited by about 80%. Again,activity was completely recovered by IRF-1-siRNA treatment. Theseobservations suggest that IRF-1 effectively suppressed the formationof blood vessels in vitro and in vivo.

3.5. IRF-1 interacts with VEGFR2

We next tried to determine the mechanism involved in theinhibition of angiogenesis by identifying proteins which bind to IRF-1.

Page 6: Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

1659J.H. Lee et al. / Biochimica et Biophysica Acta 1783 (2008) 1654–1662

Thus, we investigated for IRF-1 binding proteins using the yeast two-hybrid system. One prominent gene identified was VEGFR2. Theinteractions between IRF-1 and VEGFR2 were examined bymeasuringthe relative expression levels of β-galactosidase and by co-immuno-precipitation. As shown in Fig. 5A, β-galactosidase activity indicatedinteractions between IRF-1 and VEGFR2, which were fully observed,whereas little β-galactosidase activity was observed from the interac-tions between IRF-1 and the empty vector (vector only). VEGFR1 wasused to conclude the binding specificity of IRF-1 to VEGFR2. IRF-1 wasshown to bind VEGFR2 but not VEGFR1 as illustrated in Fig. 5A.

For co-immunoprecipitation, cDNA constructs of IRF-1 (pEGFPC1-IRF-1) and VEGFR1, 2 (pcDNA4/HisMax-VEGFR1, 2), along withpEGFPC1-IRF-1 and vector only (pcDNA4/HisMax) were co-trans-fected into HUVECs. Likewise, as above, IRF-1 was used to confirm thespecificity of VEGFR2′s binding to IRF-1. Subsequently, immunopre-cipitationwas also performed using an anti-GFP antibody with lysatesfrom both transfected cells. After immunoprecipitation, precipitatedproteins were immunoblotted using anti-VEGFR1, 2 and anti-IRF-1antibodies. As shown in Fig. 5B, pcDNA4/HisMax-VEGFR2 was co-immunoprecipitated with pEGFPC1-IRF-1 (lane 2, upper left panel),whereas no interaction was observed between pcDNA4/HisMax(vector only) and pEGFPC1-IRF-1 (lane 1, upper left panel). Immuno-blotting using anti-IRF-1 antibody confirmed that an equal amount ofIRF-1 was precipitated in both samples (middle left panel). Whole celllysates from both samples contained equivalent amounts of protein asdetermined through immunoblotting using anti-β-actin antibody(lower left panel). Also shown in Fig. 5B (upper right panel), is thecomparison made with VEGFR1. The panel clearly shows the absenceof binding between VEGFR1 and IRF-1. Taken together, our resultsstrongly suggest that the interaction between IRF-1 and VEGFR2 ishighly specific.

Fig. 6. IRF-1 interfereswith VEGF-mediated PI3K-Akt signaling in HUVECs. (A) HUVECswere trafor 20min. PI3K (p85) were detected byWestern blot analysis. Detection of the tubulin proteinPhosphorylated Akt and eNOSwere visualized by immunoblotting with antibodies that were spprotein served as a loading control (Akt, eNOS). Each band indexwas calculated by computer-asrepresentative of three separate experiments. Data is shown as means±SEM. ⁎, Pb0.05; ⁎⁎, Pb

3.6. IRF-1 inhibits PI3K/Akt signaling pathway

Based on the findings presented, we first investigated whether thehuman IRF-1 inhibits Akt upstream phosphatidylinotidol 3-kinase(PI3K). PI3K pathway is implicated in upregulating VEGF [31]. Asshown in Fig. 6A, VEGF-stimulated PI3K activation was markedlyreduced by overexpressed IRF-1. Akt is one of the important down-stream targets of PI3K. We also determined whether IRF-1 inhibits thephosphorylation of Akt (Ser-473) and eNOS (Ser-1172). For example,eNOS, endothelial NO synthase, inhibitors block endothelial cellmigration, proliferation, and tube formation that are induced by VEGFin vitro as well as in vivo. As a result, the conclusion that VEGF inducedphosphorylation of Akt and eNOS plays a key role in VEGF-stimulatedangiogenesis can bemade. As presented in Fig. 6B, VEGF-stimulated Aktand eNOS phosphorylation were dramatically reduced by the IRF-1.Therefore, it seems that IRF-1 inhibits the PI3K/Akt signaling pathway inHUVECs.

3.7. The exon 7 is essential for angiogenesis in vitro and reduces VEGFexpression in endothelial cells

We next used RT-PCR to examine splicing variants in 17 humanuterine cervical tissue samples (5 normal and 12 cancerous samples).Previously, our results revealed that all tested cervical cancer tissuesamples showed a trio of lower bands in addition to the major wild-type band [16]. In contrast, the uterine myometrial tissue samples didnot yield any IRF-1 splicing variants. Thus, this alternative splicingmay be tissue specific. To facilitate characterization of the IRF-1splicing variants in cervical cancer, we subcloned the Δ7, Δ8, and Δ9variants and the wild-type into pEGFP expression vectors, which werethen transfected into HUVECs. To confirm IRF-1′s direct anti-

nsfectedwithMock, IRF-1, or siIRF-1 for 48 h followed by treatmentwith VEGF (50 ng/ml)served as a loading control. (B) HUVECs were stimulated for 20minwith VEGF (50 ng/ml).ecific for the phosphorylated protein (p-Akt, p-eNOS). Detection of the unphosphorylatedsisted densitometric signal intensities of PI3K, p-Akt, and p-eNOS (lower panel). Results are0.01 compared with VEGF alone.

Page 7: Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

1660 J.H. Lee et al. / Biochimica et Biophysica Acta 1783 (2008) 1654–1662

angiogenic effects, we investigated whether overexpression of IRF-1splicing variants could alter endothelial tube formation. Empty Mock-treated cells incubated with VEGF formed an organized network ofendothelial cells on Matrigel (Fig. 7A). In contrast however, over-expression of IRF-1 and IRF-1(Δ7) markedly inhibited VEGF-inducedtube formation. The inhibitory effects of IRF-1 and IRF-1(Δ7) on VEGF-induced tube formation were completely recovered by IRF-1(Δ8) andIRF-1(Δ9) transfection. In order to find the key exons in the IRF-1, all ofthe IRF-1 splicing variants revealed the protein expression in HUVECs.The protein encoded by Δ7, Δ8, and Δ9 could be visualized using ananti-IRF-1 antibody (Fig. 7B). Consistent with these findings, over-expression of IRF-1(wt) and IRF-1(Δ7) is shown to completely inhibit

Fig. 7. Anti-angiogenic effects of alternative splicing variants on the VEGF-induced tubeformation in HUVECs. (A) The HUVECs were plated onto 48-well and pre-coated withMatrigel. Next, HUVECs were incubated with Mock, IRF-1(wt), or each alternativesplicing variant (Δ7, Δ8, Δ9- 100 ng) in the presence or without VEGF (10 ng/ml) for48 h. The formation of tubular structures was detected by an inverted phase contrastmicroscope image (top panel). The bar graph indicates the total length of the tubesformed by the HUVECs (bottom panel) and expressed as the means±SD. ⁎, Pb0.05compared with VEGF alone. (B) The total cell lysate of HUVECs in A was used for VEGFdetection.

Fig. 8. IRF-1 suppresses tumor growth and angiogenesis in vivo. Intratumoral injectionof IRF-1 inhibits s.c. 2774 human ovarian carcinoma growth as xenografts in nude mice(n=8). Mean tumor volume was determined by caliper measurements (upper panel).Frozen sections of the tumors were stained for endothelial cells using an anti-CD31antibody (lower panel). ⁎, Pb0.05 compared with Mock.

VEGF expression in HUVECs, the IRF-1(Δ8) and IRF-1(Δ9) splicingvariants did not (Fig. 7B). These indicate that IRF-1(wt) and IRF-1(Δ7)inhibits theautocrineeffectof VEGF in endothelial cells and thus, a directanti-angiogenic effect. These results strongly suggest that the exon 7within the C-terminal region of IRF-1 is involved in the activation ofangiogenic signaling in vitro and in vivo.

3.8. IRF-1 suppresses tumor growth and associated angiogenesis

To explore whether IRF-1 has direct antitumor activity, we testedthe effects of overexpressed IRF-1 on tumor cell growth in vitro. Wefound that IRF-1 markedly decreased 2774 ovarian cancer cell growthwhen compared with controls (data not shown).

The anti-angiogenic activity of IRF-1 was then evaluated in vivo.2774 cells were implanted s.c. in nudemice. We allowed the tumors togrow until they reached a mean volume of 100 mm3. On day 11, anintratumoral injection of IRF-1was done and repeated every 3 days for9 days. Tumors from IRF-1-treated mice were excised on day 24 afterthe final injection. The volume of IRF-1-treated tumors was 80%smaller than those from Mock mice (Fig. 8). Moreover, immunohis-tologic staining of endothelial cells in the IRF-1-treated mice showedan 85% decrease in the number of blood vessels stained with anti-CD31. Taken together, these results show that overexpressed IRF-1potently inhibits tumor growth and angiogenesis in vivo.

4. Discussion

IRF-1was thefirstmember of the IRF family, and originally identifiedas a regulatorof IFNα/β and its homolog IRF-2 [3]. Functional antagonistof IRF-1 as a tumor suppressor is IRF-2. IRF family diverse expressionscould be due to their multifunctional properties with respect to cell

Page 8: Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

1661J.H. Lee et al. / Biochimica et Biophysica Acta 1783 (2008) 1654–1662

growth control, induction of apoptosis, and susceptibility to transforma-tion by oncogenes [32,33]. Recently, it was reported that SUMOylatedIRF-1 interferes with IRF-1-mediated apoptosis in tumor cells. TheSUMOylated protein represses IRF-1-mediated transcriptional activa-tion and apoptosis [5]. Thus, IRF-1 of tumor suppressor can enhance thetumor cell evasion of immune system. It is known that IRF-1 isfrequently inactivated to prevent apoptosis by genetic alteration such asexon deletion in human cancer populations [10,14–16].

In this study, IRF-1 significantly inhibited endothelial cell migra-tion in dose–time-dependent manner. In addition, IRF-1 directlyinhibited the tube formation of endothelial cells on Matrigel andreduced the expression of VEGF, p-Akt, and p-eNOS in HUVECs. Aktand eNOS play a significant role in angiogenesis when stimulated byVEGF. VEGF is known to stimulate Akt dependent phosphorylation ofeNOS, therefore activating eNOS. The observations from our studyshow that IRF-1 had little to no effect on unphosphorylated Akt andeNOS. Once phosphorylated though, p-Akt produced a faint expres-sion of protein, and it was observed that protein expression becamefainter when p-Akt was associated with IRF-1. Phosphorylation ofeNOS also showed protein expression like p-Akt, and the expressiondecreased as it was associated with IRF-1. These data indicate that IRF-1 have a directly inhibitive effect on angiogenesis in vitro. We alsoobserved that exogenous IRF-1 has a direct inhibitive effect onendothelial cell migration and tube formation, and using a CAMmodel. Endothelial cell proliferation and migration are initial stepscritical to the angiogenesis process.

To further examine the biological significance of the splicingvariants of IRF-1 in cancer, we performed two types of methodsrelevant to its physiological functions of tube formation and expres-sion level of VEGF via Western blot. Our results revealed that thevariants differed in their abilities to angiogenic effect (Fig. 5A). Inaddition, VEGF protein expression followed an induction patternsimilar to the angiostatic activity of IRF-1 and its splicing variants(Fig. 5B). Here we also demonstrate that C-terminal region includingtransactivation domain (TA) of IRF-1 functions as a signal for itsangiostatic activity is spliced in human tumor tissues. These findingssuggest that splicing variant involving exon 7 could potentiallymodulate IRF-1 anti-angiogenic effect.

Recently, many molecules that inhibit tumor angiogenesis havebeen identified and characterized. These include antagonists ofangiogenic growth factors, receptors, integrin-adhesion molecules,and matrix proteinases, some of which are currently subjects ofclinical trials [34,35]. However, one of themajor challenges in terms ofdesigning such therapies is that there are numerous direct andindirect mechanisms by which tumors can induce new blood vesselgrowth. It is therefore reasonable to assume that successful anti-angiogenic protocols must address each of the possible mechanismsby which tumors can induce new blood vessel growth. These newtherapeutic agents could prospectively be added to chemotherapy orradiotherapy regimens, or could also be used in combination withimmunotherapy or vaccine therapy.

In summary, the results of the present study show that exogenoushuman IRF-1 exerts a direct anti-angiogenic effect on endothelial cellsby inhibiting cell proliferation, migration, tube formation in vitro, andvessel sprouting ex vivo, and CAM in vivo. We also have shown thatalternative splicing of IRF-1 in the region of exon 7 could potentiallyregulate anti-angiogenic effect. This study provides further evidencefor anti-angiogenic role of IRF-1, which may have therapeutic valuesfor cancer and angiogenesis-associated diseases.

Acknowledgements

We thank Drs. S. A. Martinis (Department of Biochemistry,University of Illinois at Urbana-Champaign, USA), and Richard Yoo(University of Washington) for critically reading the manuscript. Thiswork was supported by the National Cancer Center (NCC-0810410-1).

References

[1] M. Miyamoto, T. Fujita, Y. Kimura, M. Maruyama, H. Harada, Y. Sudo, T. Miyata,T. Taniguchi, Regulated expression of a gene encoding a nuclear factor, IRF-1,that specifically binds to IFN-beta gene regulatory elements, Cell 54 (1988)903–913.

[2] T. Fujita, Y. Kimura, M. Miyamoto, E.L. Barsoumian, T. Taniguchi, Induction ofendogenous IFN-alpha and IFN-beta genes by a regulatory transcription factor, IRF-1,Nature 337 (1989) 270–272.

[3] H. Harada, T. Fujita, M. Miyamoto, Y. Kimura, M. Maruyama, A. Furia, T. Miyata,T. Taniguchi, Structurally similar but functionally distinct factors, IRF-1 andIRF-2, bind to the same regulatory elements of IFN and IFN-inducible genes,Cell 58 (1989) 729–739.

[4] T. Taniguchi, K. Ogasawara, A. Takaoka, N. Tanaka, IRF family of transcriptionfactors as regulators of host defense, Annu. Rev. Immunol. 19 (2001) 623–655.

[5] J. Park, K.K. Kim, E.J. Lee, Y.J. Seo, S.N. Lim, K. Park, S.B. Rho, S.H. Lee, J.H. Lee,Elevated level of SUMOylated IRF-1 in tumor cells interferes with IRF-1-mediatedapoptosis, Proc. Natl. Acad. Sci. U. S. A. 104 (2007) 17028–17033.

[6] A. Kroge, M. Koster, K. Schroeder, H. Hauser, P.P. Mueller, Activities of IRF-1,J. Interferon Cytokine Res. 22 (2002) 5–14.

[7] P. Lengyel, Tumor-suppressor genes: news about the interferon connection, Proc.Natl. Acad. Sci. U. S. A. 90 (1993) 5893–5895.

[8] N. Tanaka, M. Ishihara, M. Kitagawa, H. Harada, T. Kimura, T. Matsuyama, M.S.Lamphier, S. Aizawa, T.W. Mak, T. Taniguchi, Cellular commitment to oncogene-induced transformation or apoptosis is dependent on the transcription factor IRF-1,Cell 77 (1994) 829–839.

[9] T. Taniguchi, M.S. Lamphier, N. Tanaka, IRF-1: the transcription factor linkingthe interferon response and oncogenesis, Biochim. Biophys. Acta 1333 (1997)M9–M17.

[10] Y. Moriyama, S. Nishiguchi, A. Tamori, N. Koh, Y. Yano, S. Kubo, K. Hirohashi, S.Otani, Tumor-suppressor effect of interferon regulatory factor-1 in humanhepatocellular carcinoma, Clin. Cancer Res. 7 (2001) 1293–1298.

[11] H. Harada, M. Kitagawa, N. Tanaka, H. Yamamoto, K. Harada, M. Ishihara, T.Taniguchi, Anti-oncogenic and oncogenic potential of interferon regulatoryfactors-1 and -2, Science 259 (1993) 971–974.

[12] N. Tanaka, M. Ishihara, T. Taniguchi, Suppression of c-myc or fosB-induced celltransformation by the transcription factor IRF-1, Cancer Lett. 83 (1994) 191–196.

[13] E. Pizzoferrato, Y. Liu, A. Gambotto, M.J. Armstrong, M.T. Stang, W.E. Gooding, S.M.Alber, S.H. Shand, S.C. Watkins, W.J. Storkus, J.H. Yim, Ectopic expression ofinterferon regulatory factor-1 promotes human breast cancer cell death andresults in reduced expression of surviving, Cancer Res. 64 (2004) 8381–8388.

[14] C.L. Willman, C.E. Sever, M.G. Pallavicini, H. Harada, N. Tanaka, M.L. Slovak, H.Yamamoto, K. Harada, T.C. Meeker, A.F. List, T. Taniguchi, Deletion of IRF-1,mapping to chromosome 5q31.1, in human leukemia and preleukemic myelodys-plasia, Science 259 (1993) 968–971.

[15] H. Harada, T. Kondo, S. Ogawa, T. Tamura, M. Kitagawa, N. Tanaka, M.S. Lamphier, H.Hirai, T. Taniguchi, Accelerated exon skipping of IRF-1 mRNA in humanmyelodysplasia/leukemia; a possible mechanism of tumor suppressor inactiva-tion, Oncogene 9 (1994) 3313–3320.

[16] E.J. Lee, M. Jo, J. Park, W. Zhang, J.H. Lee, Alternative splicing variants of IRF-1lacking exons 7, 8, and 9 in cervical cancer, Biochem. Biophys. Res. Commun.347 (2006) 882–888.

[17] K. Nakagawa, H. Yokosawa, Degradation of transcription factor IRF-1 by theubiquitin-proteasome pathway. The C-terminal region governs the proteinstability, Eur. J. Biochem. 267 (2000) 1680–1686.

[18] K. Nakagawa, H. Yokosawa, PIAS3 induces SUMO-1 modification and transcrip-tional repression of IRF-1, FEBS Lett. 530 (2002) 204–208.

[19] J. Folkman, Y. Shing, Angiogenesis, J. Biol. Chem. 267 (1992) 10931–10934.[20] W. Risau, Mechanisms of angiogenesis, Nature 386 (1997) 671–674.[21] G.D. Yancopoulos, S. Davis, N.W. Gale, J.S. Rudge, S.J. Wiegand, J. Holash,

Vascular-specific growth factors and blood vessel formation, Nature 407 (2000)242–248.

[22] N. Ferrara, VEGF and the quest for tumour angiogenesis factors, Nat. Rev. Cancer2 (2002) 795–803.

[23] C.R. Escalante, J. Yie, D. Thanos, A.K. Aggarwal, Structure of IRF-1 with bound DNAreveals determinants of interferon regulation, Nature 391 (1998) 103–106.

[24] F. Schaper, S. Kirchhoff, G. Posern, M. Koster, A. Oumard, R. Sharf, B.Z. Levi, H.Hauser, Functional domains of interferon regulatory factor I (IRF-1), Biochem. J.335 (1998) 147–157.

[25] H. Harada, K. Willison, J. Sakakibara, M. Miyamoto, T. Fujita, T. Taniguchi, Absenceof the type I IFN system in EC cells: transcriptional activator (IRF-1) and repressor(IRF-2) genes are developmentally regulated, Cell 63 (1990) 303–312.

[26] O.H. Lee, Y.M. Kim, Y.M. Lee, E.J. Moon, D.J. Lee, J.H. Kim, K.W. Kim, Y.G. Kwon,Sphingosine 1-phosphate induces angiogenesis: its angiogenic action andsignaling mechanism in human umbilical vein endothelial cells, Biochem. Biophys.Res. Commun. 264 (1999) 743–750.

[27] H.S. Jang, H.J. Kim, J.M. Kim, Y.S. Lee, K.L. Kim, J.A. Kim, J.Y. Lee, W. Suh, J.H. Choi,E.S. Jeon, J. Byun, D.K. Kim, A novel ex vivo angiogenesis assay based onelectroporation-mediated delivery of naked plasmid DNA to skeletal muscle,Mol. Ther. 9 (2004) 464–474.

[28] M. Nguyen, Y. Shing, J. Folkman, Quantitation of angiogenesis and anti-angiogenesis in the chick embryo chorioallantoic membrane, Microvasc. Res.47 (1994) 31–40.

[29] S.B. Rho, K.H. Lee, J.W. Kim, K. Shiba, Y.J. Jo, S. Kim, Interaction between humantRNA synthetases involves repeated sequence elements, Proc. Natl. Acad. Sci. U. S. A.93 (1996) 10128–10133.

Page 9: Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs

1662 J.H. Lee et al. / Biochimica et Biophysica Acta 1783 (2008) 1654–1662

[30] K.H.Plate,G.Breier,H.A.Weich,W.Risau,Vascularendothelial growth factor is apotentialtumour angiogenesis factor in human gliomas in vivo, Nature 29 (1992) 845–848.

[31] J. Fang, M. Ding, L. Yang, L.Z. Liu, B.H. Jiang, PI3K/PTEN/AKT signaling regulatesprostate tumor angiogenesis, Cell. Signal. 19 (2007) 2487–2497.

[32] N. Tanaka, T. Kawakami, T. Taniguchi, Recognition DNA sequences of interferonregulatory factor (IRF-1) and IRF-2, regulators of cell growth and the interferonsystem, Mol. Cell. Biol. 13 (1993) 4531–4538.

[33] M. Sato, T. Taniguchi, N. Tanaka, The interferon system and interferon regulatoryfactor transcription factors-studies from gene knockout mice, Cytokine GrowthFactor Rev. 12 (2001) 133–142.

[34] V. Brower, Tumor angiogenesis-new drugs on the block, Nat. Biotechnol. 17 (1999)963–968.

[35] R. Kerbel, J. Folkman, Clinical translation of angiogenesis inhibitors, Nat. Rev.Cancer 2 (2002) 727–739.