1
|
VEGF-A promotes the motility of human melanoma cells through the VEGFR1-PI3K/Akt signaling pathway. In Vitro Cell Dev Biol Anim 2022; 58:758-770. [PMID: 35997849 PMCID: PMC9550759 DOI: 10.1007/s11626-022-00717-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022]
Abstract
Vascular endothelial growth factor A (VEGF-A) and its receptors (VEGFR1 and R2) play important roles in the progression of malignant melanoma through tumor angiogenesis. However, it is not clear whether the VEGF-A/VEGFR1 signaling pathway is involved in the proliferation and migration of melanoma cells. Thus, the effect of VEGF-A on cell migration was investigated in human melanoma cell lines. Of several splicing variants of VEGF-A, VEGF165 is the most abundant and responsible for VEGF-A biological potency. VEGF165 facilitated the migration of melanoma cells in both a chemotactic and chemokinetic manner, but cell proliferation was not affected by VEGF165. VEGF165 also induced the phosphorylation of Akt. In addition, VEGF165-induced cell migration was inhibited significantly by VEGFR1/2 or a VEGFR1-neutralizing antibody. Furthermore, the downregulation of VEGFR1 via the transfection of VEGFR1-targeting antisense oligonucleotides suppressed VEGF165-induced cell migration. Moreover, wortmannin, an inhibitor of phosphatidylinositol-3 kinase (PI3K) in the PI3K/Akt pathway, suppressed VEGF165-induced Akt phosphorylation and VEGF165-induced cell migration. These findings suggest that the motility of melanoma cells is regulated by signals mediated through the PI3K/Akt kinase pathway with the activation of VEGFR1 tyrosine kinase by VEGF165. Thus, the downregulation of signaling via VEGF-A/VEGFR1 might be an effective therapeutic approach that could prevent the progression of malignant melanoma.
Collapse
|
2
|
Compounds for radionuclide imaging and therapy of malignant foci characterized by the increased angiogenesis. Russ Chem Bull 2016. [DOI: 10.1007/s11172-016-1309-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
3
|
Cheung CY. Vascular Endothelial Growth Factor: Possible Role in Fetal Development and Placental Function. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155769700400401] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Cecilia Y. Cheung
- Division of Perinatal Medicine, Department of Reproductive Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
4
|
Lim EK, Kim T, Paik S, Haam S, Huh YM, Lee K. Nanomaterials for Theranostics: Recent Advances and Future Challenges. Chem Rev 2014; 115:327-94. [DOI: 10.1021/cr300213b] [Citation(s) in RCA: 916] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Eun-Kyung Lim
- Department
of Radiology, Yonsei University, Seoul 120-752, Korea
- BioNanotechnology
Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Korea
| | - Taekhoon Kim
- Department
of Chemistry, Korea University, Seoul 136-701, Korea
- Electronic
Materials Laboratory, Samsung Advanced Institute of Technology, Mt. 14-1,
Nongseo-Ri, Giheung-Eup, Yongin-Si, Gyeonggi-Do 449-712, Korea
| | - Soonmyung Paik
- Severance
Biomedical Research Institute, Yonsei University College of Medicine, Seoul 120-749, Korea
- Division
of Pathology, NSABP Foundation, Pittsburgh, Pennsylvania 15212, United States
| | - Seungjoo Haam
- Department
of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea
| | - Yong-Min Huh
- Department
of Radiology, Yonsei University, Seoul 120-752, Korea
| | - Kwangyeol Lee
- Department
of Chemistry, Korea University, Seoul 136-701, Korea
| |
Collapse
|
5
|
Pourrajab F, Babaei Zarch M, Baghi Yazdi M, Rahimi Zarchi A, Vakili Zarch A. Application of stem cell/growth factor system, as a multimodal therapy approach in regenerative medicine to improve cell therapy yields. Int J Cardiol 2014; 173:12-9. [PMID: 24612559 DOI: 10.1016/j.ijcard.2014.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/26/2013] [Accepted: 02/08/2014] [Indexed: 12/13/2022]
Abstract
Stem cells hold a great promise for regenerative medicine, especially for replacing cells in infarcted organ that hardly have any intrinsic renewal capacity, including heart and brain. Signaling pathways that regulate pluripotency or lineage-specific gene and protein expression have been the major focus of stem cell research. Between them, there are some well known signaling pathways such as GF/GFR systems, SDF-1α/CXC4 ligand receptor interaction and PI3K/Akt signaling, and cytokines may regulate cell fate decisions, and can be utilized to positively influence cell therapy outcomes or accentuate synergistic compliance. For example, contributing factors in the progression of heart failure are both the loss of cardiomyocytes after myocardial infarction, and the absence of an adequate endogenous repair signaling. Combining cell engraftment with therapeutic signaling factor delivery is more exciting in terms of host progenitor/donor stem cell survival and proliferation. Thus stem cell-based therapy, besides triggering signaling pathways through GF/GFR systems can become a realistic option in regenerative processes for replacing lost cells and reconstituting the damaged organ, as before.
Collapse
Affiliation(s)
- Fatemeh Pourrajab
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Clinical Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | | | | | - Abolfazl Rahimi Zarchi
- School of Nursing, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Abbas Vakili Zarch
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
6
|
Jing M, Bowser MT. Tracking the emergence of high affinity aptamers for rhVEGF165 during capillary electrophoresis-systematic evolution of ligands by exponential enrichment using high throughput sequencing. Anal Chem 2013; 85:10761-70. [PMID: 24125636 PMCID: PMC3892959 DOI: 10.1021/ac401875h] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Capillary electrophoresis-systematic evolution of ligands by exponential enrichment (CE-SELEX) is a powerful technique for isolating aptamers for various targets, from large proteins to small peptides with molecular weights of several kilodaltons. One of the unique characteristics of CE-SELEX is the relatively high heterogeneity of the ssDNA pools that remains even after multiple rounds of selection. Enriched sequences or highly abundant oligonucleotide motifs are rarely reported in CE-SELEX studies. In this work, we employed 454 pyrosequencing to profile the evolution of an oligonucleotide pool through multiple rounds of CE-SELEX selection against the target recombinant human vascular endothelial growth factor 165 (rhVEGF165). High throughput sequencing allowed up to 3 × 10(4) sequences to be obtained from each selected pool and compared to the unselected library. Remarkably, the highest abundance contiguous sequence (contig) was only present in 0.8% of sequences even after four rounds of selection. Closer analyses of the most abundant contigs, the top 1000 oligonucleotide fragments, and even the eight original FASTA files showed no evidence of prevailing motifs in the selected pools. The sequencing results also provided insight into why many CE-SELEX selections obtain pools with reduced affinities after many rounds of selection (typically >4). Preferential amplification of a particular short polymerase chain reaction (PCR) product allowed this nonbinding sequence to overtake the pool in later rounds of selection suggesting that further refinement of primer design or amplification optimization is necessary. High affinity aptamers with 10(-8) M dissociation constants for rhVEGF165 were identified. The affinities of the higher abundance contigs were compared with aptamers randomly chosen from the final selection pool using affinity capillary electrophoresis (ACE) and fluorescence polarization (FP). No statistical difference in affinity between the higher abundance contigs and the randomly chosen aptamers was observed, supporting the premise that CE-SELEX selects a uniquely heterogeneous pool of high affinity aptamers.
Collapse
Affiliation(s)
- Meng Jing
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota, 55455, United States
| | - Michael T. Bowser
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota, 55455, United States
| |
Collapse
|
7
|
Coiled-coil-mediated grafting of bioactive vascular endothelial growth factor. Acta Biomater 2013; 9:6806-13. [PMID: 23485856 DOI: 10.1016/j.actbio.2013.02.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 02/01/2013] [Accepted: 02/18/2013] [Indexed: 11/20/2022]
Abstract
Chimeric growth factors may represent a powerful alternative to their natural counterparts for the functionalization of tissue-engineered scaffolds and applications in regenerative medicine. Their rational design should provide a simple, readily scalable production strategy while improving retention at the site of action. In that endeavor, we here report the synthesis of a chimeric protein corresponding to human vascular endothelial growth factor 165 being N-terminally fused to an E5 peptide tag (E5-VEGF). E5-VEGF was successfully expressed as a homodimer in mammalian cells. Following affinity purification, in vitro surface plasmon resonance biosensing and cell survival assays confirmed diffusible E5-VEGF ability to bind to its receptor ectodomains, while observed morphological phenotypes confirmed its anti-apoptotic features. Additional surface plasmon resonance assays highlighted that E5-VEGF could be specifically captured with high stability when interacting with covalently immobilized K5 peptide (a synthetic peptide designed to bind to the E5 moiety of chimeric hVEGF). This immobilization strategy was applied to glass substrates and chimeric hVEGF was shown to be maintained in a functionally active state following capture. Altogether, our data demonstrated that stable hVEGF capture can be performed via coiled-coil interactions without impacting hVEGF bioactivity, thus opening up the way to future applications in the field of tissue engineering and regenerative medicine.
Collapse
|
8
|
Lim EK, Kim B, Choi Y, Ro Y, Cho EJ, Lee JH, Ryu SH, Suh JS, Haam S, Huh YM. Aptamer-conjugated magnetic nanoparticles enable efficient targeted detection of integrin αvβ3 via magnetic resonance imaging. J Biomed Mater Res A 2013; 102:49-59. [PMID: 23568770 DOI: 10.1002/jbm.a.34678] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 01/30/2013] [Accepted: 02/05/2013] [Indexed: 12/17/2022]
Abstract
An understanding of neovascularization and/or angiogenesis in cancer is acutely required for effective cancer therapy due to concerns about tumor growth and metastasis. In particular, integrin αvβ3 is closely associated with cell migration and invasion during angiogenesis. Hence, we developed aptamer(αvβ3)-conjugated magnetic nanoparticles (Apt(αvβ3)-MNPs) to enable precise detection of integrin-expressing cancer cells using magnetic resonance imaging. Apt(αvβ3)-MNPs exhibited not only cytocompatibility, but also an efficient targeting ability with high magnetic sensitivity through in vitro/in vivo studies. The results of this study demonstrate that Apt(αvβ3)-MNPs have the potential to be used for accurate tumor diagnosis and therapy.
Collapse
Affiliation(s)
- Eun-Kyung Lim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Ectomesenchymal chondromyxoid tumor of the tongue: insights on histogenesis. Oral Surg Oral Med Oral Pathol Oral Radiol 2013; 115:233-40. [PMID: 23312916 DOI: 10.1016/j.oooo.2012.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 11/06/2012] [Accepted: 11/08/2012] [Indexed: 12/17/2022]
|
10
|
Briane D, Slimani H, Tagounits A, Naejus R, Haddad O, Coudert R, Charnaux N, Cao A. Inhibition of VEGF expression in A431 and MDA-MB-231 tumour cells by cationic lipid-mediated siRNA delivery. J Drug Target 2012; 20:347-54. [PMID: 22475204 DOI: 10.3109/1061186x.2012.656645] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In order to promote siRNA transfer in tumour cells, we used an original cationic lipid, synthesized in our laboratory, dimethyl-hydroxyethyl-aminopropane-carbamoyl-cholesterol (DMHAPC-Chol). Liposomes were prepared from this lipid and dioleoylphosphatidylethanolamine (DOPE) in equimolar proportion. Its transfecting capacity was evaluated using ELISA, cell cytometry, and RT-PCR in estimating the silencing effect of VEGF siRNA. This liposome efficiently delivered VEGF siRNA in two human cancer cell lines abundantly secreting VEGF, A431 and MDA-MB-231. Results showed that 50 nM of VEGF siRNA carried by DMHAPC-Chol/DOPE liposomes already silenced more than 90% of VEGF in these cells. A comparative study with two commercial carriers indicated that the inhibition induced by VEGF siRNA transported by cationic DMHAPC-Chol/DOPE liposomes was comparable to that induced by INTERFERin and better than lipofectamine 2000. Moreover, a transfection by a GFP plasmid followed by a GFP siRNA showed that DMHAPC-Chol/DOPE liposomes compared to lipofectamine were less efficient for plasmid but better for siRNA transport. Following one of our previous works concerning cell delivery of plasmid ( Percot et al., 2004 ), the main interest of results presented here resides in the double potential of DMHAPC-Chol/DOPE liposomes to deliver little-sized siRNA as well as large nucleic acids in cells.
Collapse
Affiliation(s)
- Dominique Briane
- Groupe Vectorisation, UFR de Médecine, Université Paris 13, 74 rue Marcel Cachin, F93017 Bobigny Cedex, France.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Imaging of angiogenesis has become increasingly important with the rising use of targeted antiangiogenic therapies like bevacizumab (Avastin). Non-invasive assessment of angiogenic activity is in this respect interesting, e.g. for response assessment of such targeted antiangiogenic therapies. One promising approach of angiogenesis imaging is imaging of specific molecular markers of the angiogenic cascade like the integrin α(v)β(3). For molecular imaging of integrin expression, the use of radiolabelled peptides is still the only approach that has been successfully translated into the clinic. In this review we will summarize the current data on imaging of α(v)β(3) expression using radiolabelled RGD peptides with a focus on tracers already in clinical use. A perspective will be presented on the future clinical use of radiolabelled RGD peptides including an outlook on potential applications for radionuclide therapy.
Collapse
|
12
|
Bellou S, Karali E, Bagli E, Al-Maharik N, Morbidelli L, Ziche M, Adlercreutz H, Murphy C, Fotsis T. The isoflavone metabolite 6-methoxyequol inhibits angiogenesis and suppresses tumor growth. Mol Cancer 2012; 11:35. [PMID: 22583931 PMCID: PMC3406996 DOI: 10.1186/1476-4598-11-35] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 05/14/2012] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Increased consumption of plant-based diets has been linked to the presence of certain phytochemicals, including polyphenols such as flavonoids. Several of these compounds exert their protective effect via inhibition of tumor angiogenesis. Identification of additional phytochemicals with potential antiangiogenic activity is important not only for understanding the mechanism of the preventive effect, but also for developing novel therapeutic interventions. RESULTS In an attempt to identify phytochemicals contributing to the well-documented preventive effect of plant-based diets on cancer incidence and mortality, we have screened a set of hitherto untested phytoestrogen metabolites concerning their anti-angiogenic effect, using endothelial cell proliferation as an end point. Here, we show that a novel phytoestrogen, 6-methoxyequol (6-ME), inhibited VEGF-induced proliferation of human umbilical vein endothelial cells (HUVE) cells, whereas VEGF-induced migration and survival of HUVE cells remained unaffected. In addition, 6-ME inhibited FGF-2-induced proliferation of bovine brain capillary endothelial (BBCE) cells. In line with its role in cell proliferation, 6-ME inhibited VEGF-induced phosphorylation of ERK1/2 MAPK, the key cascade responsible for VEGF-induced proliferation of endothelial cells. In this context, 6-ME inhibited in a dose dependent manner the phosphorylation of MEK1/2, the only known upstream activator of ERK1/2. 6-ME did not alter VEGF-induced phosphorylation of p38 MAPK or AKT, compatible with the lack of effect on VEGF-induced migration and survival of endothelial cells. Peri-tumor injection of 6-ME in A-431 xenograft tumors resulted in reduced tumor growth with suppressed neovasularization compared to vehicle controls (P < 0.01). CONCLUSIONS 6-ME inhibits VEGF- and FGF2-induced proliferation of ECs by targeting the phosphorylation of MEK1/2 and it downstream substrate ERK1/2, both key components of the mitogenic MAPK pathway. Injection of 6-ME in mouse A-431 xenograft tumors results to tumors with decreased neovascularization and reduced tumor volume suggesting that 6-ME may be developed to a novel anti-angiogenic agent in cancer treatment.
Collapse
Affiliation(s)
- Sofia Bellou
- Department of Biomedical Research, Foundation of Research and Technology-Hellas, Institute of Molecular Biology & Biotechnology, University Campus, 45110, Ioannina, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Shah NJ, Macdonald ML, Beben YM, Padera RF, Samuel RE, Hammond PT. Tunable dual growth factor delivery from polyelectrolyte multilayer films. Biomaterials 2011; 32:6183-93. [PMID: 21645919 DOI: 10.1016/j.biomaterials.2011.04.036] [Citation(s) in RCA: 185] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/18/2011] [Indexed: 02/07/2023]
Abstract
A promising strategy to accelerate joint implant integration and reduce recovery time and failure rates is to deliver a combination of certain growth factors to the integration site. There is a need to control the quantity of growth factors delivered at different times during the healing process to maximize efficacy. Polyelectrolyte multilayer (PEM) films, built using the layer-by-layer (LbL) technique, are attractive for releasing controlled amounts of potent growth factors over a sustained period. Here, we present PEM films that sequester physiological amounts of osteogenic rhBMP-2 (recombinant human bone morphogenetic protein-2) and angiogenic rhVEGF₁₆₅ (recombinant human vascular endothelial growth factor) in different ratios in a degradable [poly(β-amino ester)/polyanion/growth factor/polyanion] LbL tetralayer repeat architecture where the biologic load scaled linearly with the number of tetralayers. No burst release of either growth factor was observed as the films degraded. The release of rhBMP-2 was sustained over a period of 2 weeks, while rhVEGF₁₆₅ eluted from the film over the first 8 days. Both growth factors retained their efficacy, as quantified with relevant in vitro assays. rhBMP-2 initiated a dose dependent differentiation cascade in MC3T3-E1S4 pre-osteoblasts while rhVEGF₁₆₅ upregulated HUVEC proliferation, and accelerated closure of a scratch in HUVEC cell cultures in a dose dependent manner. In vivo, the mineral density of ectopic bone formed de novo by rhBMP-2/rhVEGF₁₆₅ PEM films was approximately 33% higher than when only rhBMP-2 was introduced, with a higher trabecular thickness, which would indicate a decrease in the risk of osteoporotic fracture. Bone formed throughout the scaffold when both growth factors were released, which suggests more complete remodeling due to an increased local vascular network. This study demonstrates a promising approach to delivering precise doses of multiple growth factors for a variety of implant applications where control over spatial and temporal release profile of the biologic is desired.
Collapse
Affiliation(s)
- Nisarg J Shah
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | |
Collapse
|
14
|
Yang QR, Zwijsen A, Slegers H, Berghe DV. Purification and Characterization of VEGFNPF Secreted by Human Retinal Pigment Epithelial Cells. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/10623329409024636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
15
|
Wu FTH, Stefanini MO, Mac Gabhann F, Popel AS. A compartment model of VEGF distribution in humans in the presence of soluble VEGF receptor-1 acting as a ligand trap. PLoS One 2009; 4:e5108. [PMID: 19352513 PMCID: PMC2663039 DOI: 10.1371/journal.pone.0005108] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 03/10/2009] [Indexed: 12/25/2022] Open
Abstract
Vascular endothelial growth factor (VEGF), through its activation of cell surface receptor tyrosine kinases including VEGFR1 and VEGFR2, is a vital regulator of stimulatory and inhibitory processes that keep angiogenesis--new capillary growth from existing microvasculature--at a dynamic balance in normal physiology. Soluble VEGF receptor-1 (sVEGFR1)--a naturally-occurring truncated version of VEGFR1 lacking the transmembrane and intracellular signaling domains--has been postulated to exert inhibitory effects on angiogenic signaling via two mechanisms: direct sequestration of angiogenic ligands such as VEGF; or dominant-negative heterodimerization with surface VEGFRs. In pre-clinical studies, sVEGFR1 gene and protein therapy have demonstrated efficacy in inhibiting tumor angiogenesis; while in clinical studies, sVEGFR1 has shown utility as a diagnostic or prognostic marker in a widening array of angiogenesis-dependent diseases. Here we developed a novel computational multi-tissue model for recapitulating the dynamic systemic distributions of VEGF and sVEGFR1. Model features included: physiologically-based multi-scale compartmentalization of the human body; inter-compartmental macromolecular biotransport processes (vascular permeability, lymphatic drainage); and molecularly-detailed binding interactions between the ligand isoforms VEGF(121) and VEGF(165), signaling receptors VEGFR1 and VEGFR2, non-signaling co-receptor neuropilin-1 (NRP1), as well as sVEGFR1. The model was parameterized to represent a healthy human subject, whereupon we investigated the effects of sVEGFR1 on the distribution and activation of VEGF ligands and receptors. We assessed the healthy baseline stability of circulating VEGF and sVEGFR1 levels in plasma, as well as their reliability in indicating tissue-level angiogenic signaling potential. Unexpectedly, simulated results showed that sVEGFR1 - acting as a diffusible VEGF sink alone, i.e., without sVEGFR1-VEGFR heterodimerization--did not significantly lower interstitial VEGF, nor inhibit signaling potential in tissues. Additionally, the sensitivity of plasma VEGF and sVEGFR1 to physiological fluctuations in transport rates may partially account for the heterogeneity in clinical measurements of these circulating angiogenic markers, potentially hindering their diagnostic reliability for diseases.
Collapse
Affiliation(s)
- Florence T H Wu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America.
| | | | | | | |
Collapse
|
16
|
Kim SJ, Lee JS, Im KC, Kim SY, Park SA, Lee SJ, Oh SJ, Lee DS, Moon DH. Kinetic Modeling of 3′-Deoxy-3′-18F-Fluorothymidine for Quantitative Cell Proliferation Imaging in Subcutaneous Tumor Models in Mice. J Nucl Med 2008; 49:2057-66. [DOI: 10.2967/jnumed.108.053215] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
17
|
Hadj-Bouazza A, Teste K, Colombeau L, Chaleix V, Zerrouki R, Kraemer M, Sainte Catherine O. Synthesis and biological activity of mustard derivatives of thymine. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2008; 27:439-48. [PMID: 18569783 DOI: 10.1080/15257770802086872] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The synthesis and biological activity of a novel DNA cross-linking antitumor agent is presented. The new alkylating agent significantly inhibited cell proliferation, migration and invasion as tested in vitro on the A431 vulvar epidermal carcinoma cell line.
Collapse
Affiliation(s)
- Amel Hadj-Bouazza
- Laboratoire de Chimie des Substances Naturelles, Faculte des Sciences et Techniques Limoges, Universite de Limoges, France
| | | | | | | | | | | | | |
Collapse
|
18
|
Barbat A, Gloaguen V, Moine C, Sainte-Catherine O, Kraemer M, Rogniaux H, Ropartz D, Krausz P. Structural characterization and cytotoxic properties of a 4-O-methylglucuronoxylan from castanea sativa. 2. Evidence of a structure-activity relationship. JOURNAL OF NATURAL PRODUCTS 2008; 71:1404-1409. [PMID: 18646856 DOI: 10.1021/np800207g] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Xylans were purified from delignified holocellulose alkaline extracts of Castanea sativa (Spanish chestnut) and Argania spinosa (Argan tree) and their structures analyzed by means of GC of their per-trimethylsilylated methylglycoside derivatives and (1)H NMR spectroscopy. The structures deduced were characteristic of a 4-O-methylglucuronoxylan (MGX) and a homoxylan (HX), respectively, with degrees of polymerization ranging from 182 to 360. In the case of MGX, the regular or random distribution of 4-O-methylglucuronic acid along the xylosyl backbone--determined by MALDI mass spectrometry after autohydrolysis of the polysaccharide--varied and depended both on the botanical source from which they were extracted and on the xylan extraction procedure. The MGX also inhibited in different ways the proliferation as well as the migration and invasion capability of A431 human epidermoid carcinoma cells. These biological properties could be correlated with structural features including values of the degree of polymerization, 4-O-MeGlcA to xylose ratios, and distribution of 4-O-MeGlcA along the xylosyl backbone, giving evidence of a defined structure-activity relationship.
Collapse
Affiliation(s)
- Aline Barbat
- Laboratoire de Chimie des Substances Naturelles, EA 1069, Université de Limoges, F-87060, France
| | | | | | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Tramentozzi E, Pagetta A, Frasson M, Brunati AM, Montopoli M, Finotti P. Angiogenic transforming capacity of IgG purified from plasma of type 1 diabetic patients. J Cell Mol Med 2008; 13:1336-47. [PMID: 18429934 PMCID: PMC4496147 DOI: 10.1111/j.1582-4934.2008.00354.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
We previously demonstrated that plasma of type 1 diabetic patients contains antibodies complexed irreversibly with Grp94 that also display proteolytic activity. In this work, we wanted to test whether antibodies obtained from diabetic plasma may convey an inflammatory risk on vascular cells. To this aim, IgG were purified on the Protein-G column from individual plasma of eight type 1 diabetic patients, and then tested on HUVECs to measure effects on cell growth and morphologic changes at different incubation times. The purified fractions of IgG contained a significant amount of Fab/(Fab)(2), both free and in big aggregates, and anti-Grp94 antibodies, mostly irreversibly linked with, but also free of Grp94. The purified fractions of both Fab/(Fab)(2) and whole IgG stimulated the proliferation and sustained the angiogenic differentiation of human umbilical vein endothelial cells (HUVECs) at sub-nanomolar concentrations. IgG from normal plasma neither stimulated the cell growth nor induced any differentiation of HUVECs. The maximum cell growth stimulation occurred at 6-9 hrs and associated with the strong activation of the ERK1/2 pathway, whereas angiogenic transformation was completed later when the ERK1/2 activation was silenced and cell growth stimulation significantly reduced. Neither proteolytic activity of MMP-9 nor VEGF were apparently involved in mediating the angiogenic differentiation of HUVECs that mostly correlated with an increased expression of HSP70 closely coupled with cell membrane-bound inactive species of MMP-9. Results indicate that effects displayed on HUVECs by antibodies purified from diabetic plasma are likely sustained by immune complexes with Grp94 that may thus predict an increased risk of angiogenic transformation in vivo.
Collapse
Affiliation(s)
- Elisa Tramentozzi
- Department of Pharmacology and Anesthesiology, University of Padova, Padova, Italy
| | | | | | | | | | | |
Collapse
|
21
|
Colombeau L, Teste K, Hadj-Bouazza A, Chaleix V, Zerrouki R, Kraemer M, Catherine OS. Synthesis and biological activity of chloroethyl pyrimidine nucleosides. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2008; 27:110-20. [PMID: 18205066 DOI: 10.1080/15257770701795813] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The synthesis and biological activity of chloroethyl pyrimidine nucleosides is presented. One of these new nucleosides analogues significantly inhibited cell proliferation, migration and invasion as tested in vitro on the A431 vulvar epidermal carcinoma cell line.
Collapse
Affiliation(s)
- Ludovic Colombeau
- Université de Limoges, Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences et Techniques, Limoges, France
| | | | | | | | | | | | | |
Collapse
|
22
|
Haubner R. Alphavbeta3-integrin imaging: a new approach to characterise angiogenesis? Eur J Nucl Med Mol Imaging 2007; 33 Suppl 1:54-63. [PMID: 16791598 DOI: 10.1007/s00259-006-0136-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OVERVIEW The field of angiogenesis research is one of the most rapidly growing biomedical disciplines. Great efforts are being made to develop anti-angiogenesis drugs for treatment of cancer as well as non-oncological diseases. Thus, imaging techniques allowing non-invasive monitoring of corresponding molecular processes will be of great interest. One target structure involved in the angiogenic process is the integrin alphavbeta3, which mediates the migration of activated endothelial cells during vessel formation. MATERIALS AND METHODS A variety of radiolabelled RGD peptides have been introduced for monitoring of alphavbeta3 expression using nuclear medicine tracer techniques. OBJECTIVES This review discusses tracer development and highlights some strategies for tracer optimisation. It summarises the preclinical and clinical data and discusses the potential of this class of tracer to characterise angiogenesis.
Collapse
Affiliation(s)
- Roland Haubner
- Universitätsklinik für Nuklearmedizin, Medizinische Universität Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
23
|
Mac Gabhann F, Popel AS. Dimerization of VEGF receptors and implications for signal transduction: a computational study. Biophys Chem 2007; 128:125-39. [PMID: 17442480 PMCID: PMC2711879 DOI: 10.1016/j.bpc.2007.03.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2006] [Revised: 03/12/2007] [Accepted: 03/12/2007] [Indexed: 01/13/2023]
Abstract
Vascular endothelial growth factor (VEGF) is a potent cytokine involved in the induction of neovascularization. Secreted as a cysteine-linked dimer, it has two binding sites at opposite poles through which it may bind VEGF receptors (VEGFRs), receptor tyrosine kinases found on the surface of endothelial and other cells. The binding of a VEGF molecule to two VEGFR molecules induces transphosphorylation of the intracellular domains of the receptors, leading to signal transduction. The dominant mechanism of receptor dimerization is not clear: the receptors may be present in an inactive pre-dimerized form, VEGF binding first to one of the receptors, the second receptor then ideally located for dimerization; or VEGF may bind receptor monomers on the cell surface, which then diffuse and bind to available unligated receptor monomers to complete the activation. Both processes take place and one or other may dominate on different cell types. We demonstrate the impact of dimerization mechanism on the binding of VEGF to the cell surface and on the formation of active signaling receptor complexes. We describe two methods to determine which process dominates, based on binding and phosphorylation assays. The presence of two VEGF receptor populations, VEGFR1 and VEGFR2, can result in receptor heterodimer formation. Our simulations predict that heterodimers will comprise 10-50% of the active, signaling VEGF receptor complexes, and that heterodimers will form at the expense of homodimers of VEGFR1 when VEGFR2 populations are larger. These results have significant implications for VEGF signal transduction and interpretation of experimental studies. These results may be applicable to other ligand-receptor pairs, in particular PDGF.
Collapse
Affiliation(s)
- Feilim Mac Gabhann
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
24
|
Abstract
Current focus on cancer metastasis has centered on the intrinsic factors regulating the cell autonomous homing of the tumor cells to the metastatic site. Specific up-regulation of fibronectin and clustering of bone marrow-derived cellular infiltrates coexpressing matrix metalloproteinases in distant tissue sites before tumor cell arrival are proving to be indispensable for the initial stages of metastasis. These bone marrow-derived hematopoietic progenitors that express vascular endothelial growth factor receptor 1 mobilize in response to the unique array of growth factors produced by the primary tumor. Their arrival in distant sites represents early changes in the local microenvironment, termed the "premetastatic niche," which dictate the pattern of metastatic spread. Focus on the early cellular and molecular events in cancer dissemination and selectivity will likely lead to new approaches to detect and prevent metastasis at its earliest inception.
Collapse
Affiliation(s)
- Rosandra N. Kaplan
- Department of Pediatrics, Weill College of Medicine at Cornell University, New York, New York
- Department of Cell and Developmental Biology, Weill College of Medicine at Cornell University, New York, New York
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Shahin Rafii
- Department of Cell and Developmental Biology, Weill College of Medicine at Cornell University, New York, New York
- Department of Genetic Medicine, Weill College of Medicine at Cornell University, New York, New York
| | - David Lyden
- Department of Pediatrics, Weill College of Medicine at Cornell University, New York, New York
- Department of Cell and Developmental Biology, Weill College of Medicine at Cornell University, New York, New York
- Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
25
|
Moine C, Krausz P, Chaleix V, Sainte-Catherine O, Kraemer M, Gloaguen V. Structural characterization and cytotoxic properties of a 4-O-methylglucuronoxylan from Castanea sativa. JOURNAL OF NATURAL PRODUCTS 2007; 70:60-6. [PMID: 17253851 DOI: 10.1021/np060354p] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
A glucuronoxylan was purified from a delignified holocellulose alkaline extract of Castanea sativa (Spanish chestnut) and its structure analyzed by means of FT-IR, GC of the per-trimethylsilylated methylglycoside derivatives, and 1H and 13C NMR spectroscopy. The results supported a structure based on a linear polymer of xylopyranose units linked with beta(1-->4) bonds in which, on average, one out of every six units is substituted at C-2 by a 4-O-methylglucuronic acid unit; this structure is typical of a hardwood acidic 4-O-methylglucuronoxylan (MGX) with an estimated degree of polymerization of 200. The MGX from C. sativa inhibited the proliferation of A431 human epidermoid carcinoma cells with an IC50 value of 50 microM. In addition, this xylan inhibited A431 cell migration and invasion. Preliminary experiments showing that secretion of metalloproteinases MMP2 and MMP9 by A431 tumor cells was inhibited by the purified C. sativa MGX strongly suggest that this mechanism of action may play a role in its antimigration and anti-invasive properties.
Collapse
Affiliation(s)
- Charlotte Moine
- Laboratoire de Chimie des Substances Naturelles, EA 1069, Faculté des Sciences et Techniques, Université de Limoges, F-87060, France
| | | | | | | | | | | |
Collapse
|
26
|
Banerjee S, Sengupta K, Dhar K, Mehta S, D'Amore PA, Dhar G, Banerjee SK. Breast cancer cells secreted platelet-derived growth factor-induced motility of vascular smooth muscle cells is mediated through neuropilin-1. Mol Carcinog 2006; 45:871-80. [PMID: 16847823 DOI: 10.1002/mc.20248] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Motility of vascular smooth muscle cells (SMCs) is an essential step for both normal and pathologic angiogenesis. We report here that breast tumor cells, such as MCF-7 and MDA-MB-231, can modulate this SMC migration. We present evidence that the tumor cell-derived platelet-derived growth factor (PDGF) is the key regulator of vascular SMCs motility induced by breast cancer cells. PDGF significantly upregulates neuropilin-1 (NRP-1) mRNA expression and protein production in aortic smooth muscle cells (AOSMCs) and depletion of NRP-1 production by AOSMCs with specific short hairpin RNA (shRNA) prevents the PDGF-dependent migration of vascular SMCs. Moreover, we demonstrate that PDGF physically interacts with NRP-1. We propose that tumor-derived PDGF and NRP-1 of AOSMCs function as a relay system that promotes motility of vascular SMCs.
Collapse
Affiliation(s)
- Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Vascular endothelial growth factor (VEGF) is an essential peptide in new vessel growth in physiology (endometrial growth, embryonic development); pathological conditions (diabetic retinopathy, rheumatoid arthritis); as well as in tumor cell growth, particularly distant metastases. This study focused on VEGF structure, receptors, and angiogensis in tumors, especially their roles in thyroid cancer. The VEGF mRNA undergoes alternative splicing events that generate four homodimeric isoforms, including VEGF121, VEGF165, VEGF189, or VEGF206. Using VEGF purified from a culture medium conditioned by A-431 human epidermoid carcinoma cells, VEGF-binding site complexes of 230, 170, and 125 kDa were detected on human umbilical vein endothelial cells. The VEGF specifically induced the tyrosine phosphorylation of a 190-kDa polypeptide, which had similar mass to the largest binding site detected through affinity cross-linking. A transmembrane receptor belongs to the tyrosine kinase family, fms-like tyrosine kinase (FLT). These receptor tyrosine kinases encoded by the FLT gene family have distinct functions in regulating blood vessel growth and differentiation. Regulation of VEGF is a complex, multistep mechanism in various kinds of cells and tissues. Hypoxia-dependent and -independent mechanisms are illustrated in different cancer tissues. Hypoxic tumor cells may switch to a proangiogenic phenotype, which increases VEGF transcription. Clinical applications of VEGF in cancer have included diagnosis, prediction of prognosis, and treatment in different solid tumors, including thyroid tumors. Studies involving thyroid cancer cell lines, serum level determination, immunohistocytochemical staining, molecular biological studies, and gene therapy to the in vivo clinical trials, have shown that antiangiogensis therapy can provide another treatment modality for thyroid cancer. Future studies focused on recombinant human anti-VEGF research involving patients with advanced thyroid cancer, and investigation of the protection of high-risk patients by using novel antiangiogenic vaccines, are warranted.
Collapse
Affiliation(s)
- Jen-Der Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung University, Taiwan, Republic of China.
| | | |
Collapse
|
28
|
Mac Gabhann F, Yang MT, Popel AS. Monte Carlo simulations of VEGF binding to cell surface receptors in vitro. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1746:95-107. [PMID: 16257459 DOI: 10.1016/j.bbamcr.2005.09.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Revised: 09/01/2005] [Accepted: 09/19/2005] [Indexed: 11/26/2022]
Abstract
The vascular endothelial growth factor (VEGF) family binds multiple endothelial cell surface receptors. Our goal is to build comprehensive models of these interactions for the purpose of simulating angiogenesis. In view of low concentrations of growth factors in vivo and in vitro, stochastic modeling of molecular interactions may be necessary. Here, we compare Monte Carlo simulations of the stochastic binding of VEGF and two of its major receptors on cells in vitro to equivalent deterministic simulations. In the range of typical VEGF concentrations, the stochastic and deterministic models are in agreement. However, we observe significant variability in receptor binding, which may be linked to biological stochastic events, e.g., blood vessel sprout initiation. We study patches of cell surface of varying sizes to investigate spatial integration of the signal by the cell, which impacts directly the variability of binding, and find significant variability up to the single-cell level. Dimerization of VEGF receptors does not significantly alter the variability in ligand binding. A 'sliding window' approach demonstrated no reduction in the variability of binding by temporal integration. The variability is expected to be more prominent in in vivo situations where the number of ligand molecules available for binding is less.
Collapse
Affiliation(s)
- Feilim Mac Gabhann
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Ave., #613 Traylor, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
29
|
Mac Gabhann F, Popel AS. Differential binding of VEGF isoforms to VEGF receptor 2 in the presence of neuropilin-1: a computational model. Am J Physiol Heart Circ Physiol 2005; 288:H2851-60. [PMID: 15708957 DOI: 10.1152/ajpheart.01218.2004] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
A comprehensive, biophysically accurate, computational model of vascular endothelial growth factor (VEGF) family member interactions with endothelial cell surface receptors was developed to study angiogenesis. Neuropilin-1 (NRP1) and the signaling VEGF receptor, VEGFR2, do not interact directly but are bridged by one VEGF isoform, VEGF165. Using the model and published experimental data, we estimated the kinetic rate of this VEGFR2-NRP1 coupling in vitro. With the use of this rate, our model gives predictions in good quantitative agreement with several independent in vitro experiments involving VEGF121and VEGF165isoforms, confirming that VEGFR2-NRP1 coupling through VEGF165can fully explain the observed differences in receptor binding and phosphorylation in response to these isoforms. Model predictions also determine the mechanism of action of a commonly used NRP1 antibody and predict the results of potential future experiments. This is the first model to include VEGF isoforms or NRPs, and it is a necessary step toward a quantitative molecular level description of VEGF that can be extended to in vivo situations. The model has applications for both proangiogenic and antiangiogenic therapies, such as for heart disease and cancer, as well as in tissue engineering.
Collapse
Affiliation(s)
- Feilim Mac Gabhann
- Dept. of Biomedical Engineering, Johns Hopkins Univ. School of Medicine, 720 Rutland Ave., #611 Traylor, Baltimore, MD 21205, USA.
| | | |
Collapse
|
30
|
Haubner R, Weber WA, Beer AJ, Vabuliene E, Reim D, Sarbia M, Becker KF, Goebel M, Hein R, Wester HJ, Kessler H, Schwaiger M. Noninvasive visualization of the activated alphavbeta3 integrin in cancer patients by positron emission tomography and [18F]Galacto-RGD. PLoS Med 2005; 2:e70. [PMID: 15783258 PMCID: PMC1069665 DOI: 10.1371/journal.pmed.0020070] [Citation(s) in RCA: 376] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2004] [Accepted: 01/28/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The integrin alphavbeta3 plays an important role in angiogenesis and tumor cell metastasis, and is currently being evaluated as a target for new therapeutic approaches. Several techniques are being studied to enable noninvasive determination of alphavbeta3 expression. We developed [(18)F]Galacto-RGD, a (18)F-labeled glycosylated alphavbeta3 antagonist, allowing monitoring of alphavbeta3 expression with positron emission tomography (PET). METHODS AND FINDINGS Here we show by quantitative analysis of images resulting from a small-animal PET scanner that uptake of [(18)F]Galacto-RGD in the tumor correlates with alphavbeta3 expression subsequently determined by Western blot analyses. Moreover, using the A431 human squamous cell carcinoma model we demonstrate that this approach is sensitive enough to visualize alphavbeta3 expression resulting exclusively from the tumor vasculature. Most important, this study shows, that [(18)F]Galacto-RGD with PET enables noninvasive quantitative assessment of the alphavbeta3 expression pattern on tumor and endothelial cells in patients with malignant tumors. CONCLUSIONS Molecular imaging with [(18)F]Galacto-RGD and PET can provide important information for planning and monitoring anti-angiogenic therapies targeting the alphavbeta3 integrins and can reveal the involvement and role of this integrin in metastatic and angiogenic processes in various diseases.
Collapse
Affiliation(s)
- Roland Haubner
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Strawn LM, Shawver LK. Tyrosine kinases in disease: overview of kinase inhibitors as therapeutic agents and current drugs in clinical trials. Expert Opin Investig Drugs 2005; 7:553-73. [PMID: 15991993 DOI: 10.1517/13543784.7.4.553] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tyrosine kinases, first described as oncogenes, have been shown to play a role in normal cellular processes. Aberrations in tyrosine kinase activity lead to disease states. For fifteen years it has been postulated that the inhibition of tyrosine kinases may have therapeutic utility and the design and testing of inhibitors have been major focuses of research and development in both academic institutions and pharmaceutical companies. While early research focused on developing chemical entities that mimic phosphotyrosine, later research has focused on developing competitive adenosine triphosphate (ATP) inhibitors with various levels of selectivity on kinase targets. This review focuses on a discussion of tyrosine kinases thought to be important in disease, including platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), vascular endothelial cell growth factor (VEGF), epidermal growth factor (EGF) receptors, HER-2 and Src. In addition, the classes of inhibitors designed to affect these targets and that have overcome research and development challenges and entered clinical trials are discussed. These include isoxazole, quinazoline, substituted pyrimidines and indolinone compounds, all of which are in clinical trials or near clinical development by SUGEN, Zeneca, Novartis, Pfizer and Parke-Davis. A summary of the chemistry and activity of these agents is provided.
Collapse
Affiliation(s)
- L M Strawn
- SUGEN, INC., 351 Galveston Drive, Redwood City, CA 94063, USA
| | | |
Collapse
|
32
|
Bagli E, Stefaniotou M, Morbidelli L, Ziche M, Psillas K, Murphy C, Fotsis T. Luteolin inhibits vascular endothelial growth factor-induced angiogenesis; inhibition of endothelial cell survival and proliferation by targeting phosphatidylinositol 3'-kinase activity. Cancer Res 2004; 64:7936-46. [PMID: 15520200 DOI: 10.1158/0008-5472.can-03-3104] [Citation(s) in RCA: 144] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In an attempt to identify phytochemicals contributing to the well-documented preventive effect of plant-based diets on cancer incidence and mortality, we have previously shown that certain flavonoids inhibit in vitro angiogenesis. Here, we show that the flavonoid luteolin inhibited tumor growth and angiogenesis in a murine xenograft model. Furthermore, luteolin inhibited vascular endothelial growth factor (VEGF)-induced in vivo angiogenesis in the rabbit corneal assay. In agreement, luteolin inhibited both VEGF-induced survival and proliferation of human umbilical vein endothelial cells (HUVECs) with an IC(50) of about 5 mumol/L. Luteolin inhibited VEGF-induced phosphatidylinositol 3'-kinase (PI3K) activity in HUVECs, and this inhibition was critical for both the antisurvival and antimitotic affects of the compound. Indeed, luteolin abolished VEGF-induced activation of Akt, a downstream target of PI3K conveying both survival and mitotic downstream signals. Because overexpression of a constitutively active form of Akt rescued HUVECs only from the antisurvival effects of luteolin, the result indicated that luteolin targeted mainly the survival signals of the PI3K/Akt pathway. With regard to its antimitotic activity, luteolin inhibited VEGF-induced phosphorylation of p70 S6 kinase (S6K), a downstream effector of PI3K responsible for G(1) progression. Indeed, VEGF-induced proliferation of HUVECs was sensitive to rapamycin, an inhibitor of p70 S6K activation. Surprisingly, luteolin did not affect VEGF-induced phosphorylation of extracellular signal-regulated kinase 1/2 mitogen-activated protein kinases, a pathway that is considered important for the mitotic effects of VEGF. Thus, blockade of PI3K by luteolin was responsible for the inhibitory effects of the compound on VEGF-induced survival and proliferation of HUVECs. The antisurvival effects of luteolin were mediated via blockage of PI3K/Akt-dependent pathways, whereas inhibition of the PI3K/p70 S6K pathway mediated the antimitotic effects of the compound.
Collapse
Affiliation(s)
- Eleni Bagli
- Laboratory of Biological Chemistry and Department of Ophthalmology, Medical School, University of Ioannina, Ioannina, Greece
| | | | | | | | | | | | | |
Collapse
|
33
|
Mac Gabhann F, Popel AS. Model of competitive binding of vascular endothelial growth factor and placental growth factor to VEGF receptors on endothelial cells. Am J Physiol Heart Circ Physiol 2004; 286:H153-64. [PMID: 12714333 DOI: 10.1152/ajpheart.00254.2003] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Placental growth factor (PlGF) competes with vascular endothelial growth factor (VEGF) for binding to VEGF receptor (VEGFR)-1 but does not bind VEGFR2. Experiments show that PlGF can augment the response to VEGF in pathological angiogenesis and in models of endothelial cell survival, migration, and proliferation. This synergy has been hypothesized to be due to a combination of the following: signaling by PlGF through VEGFR1 and displacement of VEGF from VEGFR1 to VEGFR2 by PlGF, causing increased signaling through VEGFR2. In this study, the relative contribution of PlGF-induced VEGF displacement to the synergy is quantified using a mathematical model of ligand-receptor binding to examine the effect on ligand-receptor complex formation of VEGF and PlGF acting together. Parameters specific to the VEGF-PlGF system are used based on existing data. The model is used to simulate in silico a specific in vitro experiment in which VEGF-PlGF synergy is observed. We show that, whereas a significant change in the formation of endothelial surface growth factor-VEGFR1 complexes is predicted in the presence of PlGF, the increase in the number of VEGFR2-containing signaling complexes is less significant; these results were shown to be robust to significant variation in the kinetic parameters of the model. Synergistic effects observed in that experiment thus appear unlikely to be due to VEGF displacement but to a shift from VEGF-VEGFR1 to PlGF-VEGFR1 complexes and an increase in total VEGFR1 complexes. These results suggest that VEGFR1 signaling can be functional in adult-derived endothelial cells.
Collapse
Affiliation(s)
- Feilim Mac Gabhann
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Traylor 613, 720 Rutland Ave., Baltimore, MD 21205, USA.
| | | |
Collapse
|
34
|
Hamma-Kourbali Y, Starzec A, Vassy R, Martin A, Kraemer M, Perret G, Crépin M. Carboxymethyl benzylamide dextran inhibits angiogenesis and growth of VEGF-overexpressing human epidermoid carcinoma xenograft in nude mice. Br J Cancer 2003; 89:215-21. [PMID: 12838326 PMCID: PMC2394212 DOI: 10.1038/sj.bjc.6601029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) expression is elevated in a wide variety of solid tumours. Inhibition of VEGF activities is able to reduce angiogenesis and tumour growth. We have recently shown in vitro that carboxymethyl dextran benzylamide (CMDB7) prevents the binding of VEGF(165) to its cell surface receptors and thus inhibits VEGF activities on endothelial cells. In the present study, we explored the effects of CMDB7 on highly aggressive human epidermoid carcinoma A431 cells known to overexpress epidermal growth factor receptors (EGFRs) and produce a high amount of VEGF and a minor quantity of bFGF. In vitro, CMDB7 blocked the mitogenic activity of A431-conditioned medium on endothelial cells. Concerning A431 cells, CMDB7 inhibited their proliferation and the VEGF(165) binding to them. In vivo, administration of CMDB7 (10 mg kg(-1)) three times per week for 2 weeks inhibited the growth of A431 xenografts in nude mice by 73% as compared to the control group. Immunostaining of endothelial cells with mouse-specific GSL-1 lectin in tumour sections revealed that CMDB7 also inhibited the density of intratumour endothelial cells by 66%. These findings demonstrate that CMDB7 has an efficient antiangiogenic and antitumour action in vivo even when tumour cells produce a high level of VEGF and EGFRs.
Collapse
Affiliation(s)
- Y Hamma-Kourbali
- Laboratoire de Ciblage Fonctionnel des Tumeurs Solides, UPRES 2360, Faculté de Médecine, Université Paris 13, 74 rue Marcel Cachin, 93017 Bobigny cedex, France
| | - A Starzec
- Laboratoire de Ciblage Fonctionnel des Tumeurs Solides, UPRES 2360, Faculté de Médecine, Université Paris 13, 74 rue Marcel Cachin, 93017 Bobigny cedex, France
| | - R Vassy
- Laboratoire de Ciblage Fonctionnel des Tumeurs Solides, UPRES 2360, Faculté de Médecine, Université Paris 13, 74 rue Marcel Cachin, 93017 Bobigny cedex, France
| | - A Martin
- Service d'Anatomie Pathologie, Hôpital Avicenne, 125, route de Stalingrad, F-93017 Bobigny cedex, France
| | - M Kraemer
- Laboratoire de Ciblage Fonctionnel des Tumeurs Solides, UPRES 2360, Faculté de Médecine, Université Paris 13, 74 rue Marcel Cachin, 93017 Bobigny cedex, France
| | - G Perret
- Laboratoire de Ciblage Fonctionnel des Tumeurs Solides, UPRES 2360, Faculté de Médecine, Université Paris 13, 74 rue Marcel Cachin, 93017 Bobigny cedex, France
| | - M Crépin
- Laboratoire de Ciblage Fonctionnel des Tumeurs Solides, UPRES 2360, Faculté de Médecine, Université Paris 13, 74 rue Marcel Cachin, 93017 Bobigny cedex, France
- Service d'Anatomie Pathologie, Hôpital Avicenne, 125, route de Stalingrad, F-93017 Bobigny cedex, France
- Laboratoire d'Hémostase, Endothélium et Angiogénèse, Unité INSERM 553, Hôpital Saint-Louis, 75010 Paris, France
- Laboratoire d'Hémostase, Endothélium et Angiogénèse, Unité INSERM 553, Hôpital Saint Louis,1 Av. Claude Vellefaux, 75010 Paris, France. E-mail:
| |
Collapse
|
35
|
Di Benedetto M, Starzec A, Vassy R, Perret GY, Crépin M, Kraemer M. Inhibition of epidermoid carcinoma A431 cell growth and angiogenesis in nude mice by early and late treatment with a novel dextran derivative. Br J Cancer 2003; 88:1987-94. [PMID: 12799647 PMCID: PMC2741107 DOI: 10.1038/sj.bjc.6600985] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We investigated the effect of a new dextran derivative, phenylacetate carboxymethyl benzylamide dextran (NaPaC), on epidermoid carcinoma A431 cells secreting a large quantity of angiogenic factor, vascular endothelial growth factor (VEGF). In vitro, NaPaC inhibited the proliferation of A431 cells (IC(50)=5 micro M). Also, NaPaC decreased the binding of radiolabelled VEGF(165) to endothelial cells (IC(50)=0.2 micro M). In vivo, we explored the effects of NaPaC (15 mg kg(-1)) on A431 xenograft growth starting the drug administration at the time of tumour cell inoculation (early treatment) and 1 week later, when tumours were well established (late treatment). Early treatment was more efficient on tumour inhibition (70% vs control) than late treatment (50% vs control). Early and late NaPaC-treatment increased the aponecrosis in tumour by 70 and 30%, respectively. Whatever treatment, NaPaC inhibited the intratumour endothelial cell density in the same manner. In contrast, vessel area was decreased only when NaPaC was injected early (35%). These results show that NaPaC has a potent inhibitory effect, dependent on treatment outset, on epidermoid carcinoma growth associated with an intratumour microvascular network diminution and an aponecrosis increase. As this drug is nontoxic at efficient dose, it offers interesting perspectives for the therapy of malignant lesions.
Collapse
Affiliation(s)
- M Di Benedetto
- Laboratoire d'Oncologie Cellulaire et Moléculaire, UPRES 2360, Université Paris 13, 74 rue Marcel Cachin, 93017 Bobigny cedex, France.
| | | | | | | | | | | |
Collapse
|
36
|
Sawaji Y, Sato T, Takeuchi A, Hirata M, Ito A. Anti-angiogenic action of hyperthermia by suppressing gene expression and production of tumour-derived vascular endothelial growth factor in vivo and in vitro. Br J Cancer 2002; 86:1597-603. [PMID: 12085210 PMCID: PMC2746582 DOI: 10.1038/sj.bjc.6600268] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2001] [Revised: 02/19/2002] [Accepted: 03/01/2002] [Indexed: 01/22/2023] Open
Abstract
Vascular endothelial growth factor is an important angiogenic factor for tumour progression because it increases endothelial-cell proliferation and remodels extracellular matrix in blood vessels. We demonstrated that hyperthermia at 42 degrees C, termed heat shock, suppressed the gene expression and production of vascular endothelial growth factor in human fibrosarcoma HT-1080 cells and inhibited its in vitro angiogenic action on human umbilical vein endothelial cells. The gene expression of alternative splicing variants for vascular endothelial growth factor, VEGF121, VEGF165 and VEGF189, was constitutively detected in HT-1080 cells, but the VEGF189 transcript was less abundant than VEGF121 and VEGF165. When HT-1080 cells were treated with heat shock at 42 degrees C for 4 h and then maintained at 37 degrees C for another 24 h, the gene expression of all vascular endothelial growth factor variants was suppressed. In addition, HT-1080 cells were found to produce abundant VEGF165, but much less VEGF121, both of which were inhibited by heat shock. Furthermore, the level of vascular endothelial growth factor in sera from six cancer patients was significantly diminished 2-3 weeks after completion of whole-body hyperthermia at 42 degrees C (49.9+/-36.5 pg x ml(-1), P<0.01) as compared with that prior to the treatment (177.0+/-77.5 pg x ml(-1)). On the other hand, HT-1080 cell-conditioned medium showed vascular endothelial growth factor-dependent cell proliferative activity and the augmentation of pro-matrix metalloproteinase-1 production in human umbilical vein endothelial cells. The augmentation of endothelial-cell proliferation and pro-matrix metalloproteinase-1 production was poor when human umbilical vein endothelial cells were treated with conditioned medium from heat-shocked HT-1080 cells. These results suggest that hyperthermia acts as an anti-angiogenic strategy by suppressing the expression of tumour-derived vascular endothelial growth factor production and thereby inhibiting endothelial-cell proliferation and extracellular matrix remodelling in blood vessels.
Collapse
MESH Headings
- Cell Division/drug effects
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Collagenases/biosynthesis
- Collagenases/genetics
- Culture Media, Conditioned/pharmacology
- Endothelial Growth Factors/biosynthesis
- Endothelial Growth Factors/blood
- Endothelial Growth Factors/genetics
- Endothelial Growth Factors/physiology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Enzyme Induction/drug effects
- Enzyme Precursors/biosynthesis
- Enzyme Precursors/genetics
- Extracellular Matrix/metabolism
- Female
- Fibrosarcoma/pathology
- Gene Expression Regulation, Neoplastic/drug effects
- Hot Temperature
- Humans
- Hyperthermia, Induced
- Lymphokines/biosynthesis
- Lymphokines/blood
- Lymphokines/genetics
- Lymphokines/physiology
- Male
- Matrix Metalloproteinase 1
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/blood
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Neoplasms/blood
- Neoplasms/blood supply
- Neoplasms/therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/therapy
- Protein Isoforms/biosynthesis
- Protein Isoforms/genetics
- RNA, Messenger/biosynthesis
- RNA, Neoplasm/biosynthesis
- Transcription, Genetic
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/pathology
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- Y Sawaji
- Department of Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | | | | | | | |
Collapse
|
37
|
Lal BK, Varma S, Pappas PJ, Hobson RW, Durán WN. VEGF increases permeability of the endothelial cell monolayer by activation of PKB/akt, endothelial nitric-oxide synthase, and MAP kinase pathways. Microvasc Res 2001; 62:252-62. [PMID: 11678628 DOI: 10.1006/mvre.2001.2338] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
VEGF is a key regulator of vascular permeability. However, its signaling pathways are incompletely understood. We tested the hypothesis that VEGF regulates endothelial cell (EC) permeability by activating PKB/akt, NOS, and MAP kinase dependent pathways using human umbilical vein EC (HUVEC). Permeability was measured from FITC-dextran 70-kDa flux across the EC monolayer at baseline and after VEGF at 0.034, 0.068, 1, 10, and 100 nM. VEGF increased HUVEC permeability to FITC-dextran in a dose-dependent manner. VEGF (1 nM) increased permeability from 3.9 x 10(-6) +/- 0.7 x 10(-6) to 14.0 x 10(-6) +/- 1.7 x 10(-6) cm/s (mean +/- SEM; P < 0.001). Permeability changes were also assessed after treatment with 1, 10, and 100 nM wortmannin (PI 3-kinase inhibitor); 0.01, 0.1, and 1.0 nM LY294002 (PI 3-kinase inhibitor); 200 microM l-NMMA (NOS inhibitor); 2.7 microM AG126 (p42/44(MAPK) inhibitor); and 0.006, 0.06, and 0.6 microM SB203580 (p38(MAPK) inhibitor). All inhibitors blocked VEGF-induced permeability changes. Our data demonstrate that (1) VEGF increases permeability of EC monolayers in a dose-dependent fashion, and (2) VEGF-induced permeability is mediated through PI-3 kinase-PKB, NOS, and MAP-kinase signaling cascades. These observations suggest that microvascular hyperpermeability associated with inflammation and vascular disease is mediated by activation of these EC signaling pathways.
Collapse
Affiliation(s)
- B K Lal
- Program in Vascular Biology, Department of Pharmacology & Physiology, UMDNJ-New Jersey Medical School, Newark, New Jersey 07103-2714, USA
| | | | | | | | | |
Collapse
|
38
|
Yu Y, Hulmes JD, Herley MT, Whitney RG, Crabb JW, Sato JD. Direct identification of a major autophosphorylation site on vascular endothelial growth factor receptor Flt-1 that mediates phosphatidylinositol 3'-kinase binding. Biochem J 2001; 358:465-72. [PMID: 11513746 PMCID: PMC1222080 DOI: 10.1042/0264-6021:3580465] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Progress has been made in our understanding of the mechanism by which the binding of vascular endothelial growth factor (VEGF) to cognate receptors induces a range of biological responses, but it is far from complete. Identification of receptor autophosphorylation sites will allow us to determine how activated VEGF receptors are coupled to specific downstream signalling proteins. In the present study, we have expressed human VEGF receptors in insect cells using the baculovirus expression system, identified a major autophosphorylation site on the VEGF receptor fms-like tyrosine kinase-1 (Flt-1) by HPLC-electrospray ionization (ESI)-MS, and characterized in vitro interactions between Flt-1 and phosphatidylinositol 3'-kinase (PI3-kinase). Infection of High 5 insect cells with Flt-1 recombinant virus resulted in the expression of a 170 kDa glycoprotein, which bound VEGF with a K(d) of 2 x 10(-10) M in intact insect cells. The overexpressed recombinant Flt-1 receptors exhibited tyrosine kinase activity and were constitutively phosphorylated. Analysis of Flt-1 tryptic peptides by HPLC-ESI-MS with selective phosphate ion monitoring identified a hexapeptide (YVNAFK; where single-letter amino-acid code has been used) containing a phosphotyrosine (pTyr) residue at position 1213. Using synthetic phosphopeptides, this pTyr residue was found to be directly involved in the binding of PI3-kinase in vitro even though it did not fall within a consensus pYM/VXM PI3-kinase binding motif. These results suggest that phosphorylated Flt-1 associates with PI3-kinase at pTyr(1213) to mediate the activation of this pathway in VEGF signalling.
Collapse
Affiliation(s)
- Y Yu
- Surgical Research Laboratory, Children's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
39
|
Tan DC, Kini RM, Jois SD, Lim DK, Xin L, Ge R. A small peptide derived from Flt-1 (VEGFR-1) functions as an angiogenic inhibitor. FEBS Lett 2001; 494:150-6. [PMID: 11311231 DOI: 10.1016/s0014-5793(01)02314-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic stimulator which functions through two endothelial specific tyrosine kinase receptors, Flt-1 and Flk-1. In this work, we show that an 11-amino acid peptide derived from the second immunoglobulin-like domain of Flt-1 functions as an angiogenic inhibitor in chick chorioallantoic membrane and inhibited VEGF-induced vascular permeability in Miles' assay without binding to VEGF directly. Circular dichroism and nuclear magnetic resonance analyses indicate that this peptide forms a stable extended structure in solution, presumably beta-sheet structure and is most likely existing as a dimer. Our results suggest that this small peptide functions as an angiogenic inhibitor by inhibiting VEGF function through a non-VEGF binding mechanism.
Collapse
Affiliation(s)
- D C Tan
- Department of Biological Sciences, National University of Singapore, Singapore 119260
| | | | | | | | | | | |
Collapse
|
40
|
Gallo O, Franchi A, Magnelli L, Sardi I, Vannacci A, Boddi V, Chiarugi V, Masini E. Cyclooxygenase-2 pathway correlates with VEGF expression in head and neck cancer. Implications for tumor angiogenesis and metastasis. Neoplasia 2001; 3:53-61. [PMID: 11326316 PMCID: PMC1505025 DOI: 10.1038/sj.neo.7900127] [Citation(s) in RCA: 231] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2000] [Accepted: 10/23/2000] [Indexed: 12/21/2022] Open
Abstract
We evaluated the role of COX-2 pathway in 35 head and neck cancers (HNCs) by analyzing COX-2 expression and prostaglandin E2 (PGE2) production in relation to tumor angiogenesis and lymph node metastasis. COX-2 activity was also correlated to vascular endothelial growth factor (VEGF) mRNA and protein expression. COX-2 mRNA and protein expression was higher in tumor samples than in normal mucosa. PGE2 levels were higher in the tumor front zone in comparison with tumor core and normal mucosa (P<.0001). Specimens from patients with lymph node metastasis exhibited higher COX-2 protein expression (P=.0074), PGE2 levels (P=.0011) and microvessel density (P<.0001) than specimens from patients without metastasis. A significant correlation between COX-2 and tumor vascularization (r(s)=0.450, P=.007) as well as between COX-2 and microvessel density with VEGF expression in tumor tissues was found (r(s)=0.450, P=.007; r(s)=0.620, P=.0001, respectively). The induction of COX-2 mRNA and PGE2 synthesis by EGF and Escherichia coli lipopolysaccharide (LPS) in A-431 and SCC-9 cell lines, resulted in an increase in VEGF mRNA and protein production. Indomethacin and celecoxib reversed the EGF- and LPS-dependent COX-2, VEGF, and PGE2 increases. This study suggests a central role of COX-2 pathway in HNC angiogenesis by modulating VEGF production and indicates that COX-2 inhibitors may be useful in HNC treatment.
Collapse
MESH Headings
- Aged
- Blotting, Northern
- Blotting, Western
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/enzymology
- Carcinoma, Squamous Cell/pathology
- Celecoxib
- Cyclooxygenase 2
- Dinoprostone/metabolism
- Endothelial Growth Factors/genetics
- Endothelial Growth Factors/metabolism
- Epidermal Growth Factor/pharmacology
- Escherichia coli
- Female
- Gene Expression
- Head and Neck Neoplasms/blood supply
- Head and Neck Neoplasms/enzymology
- Head and Neck Neoplasms/pathology
- Humans
- Immunoenzyme Techniques
- Indomethacin/pharmacology
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Lipopolysaccharides/pharmacology
- Lymph Nodes/enzymology
- Lymphatic Metastasis
- Lymphokines/genetics
- Lymphokines/metabolism
- Male
- Membrane Proteins
- Middle Aged
- Neovascularization, Pathologic/enzymology
- Neovascularization, Pathologic/pathology
- Prostaglandin-Endoperoxide Synthases/genetics
- Prostaglandin-Endoperoxide Synthases/metabolism
- Pyrazoles
- RNA, Messenger/metabolism
- Signal Transduction
- Sulfonamides/pharmacology
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/metabolism
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- O Gallo
- Department of Oto-Neuro-Ophthalmologic Surgery, University of Florence, 50139 Florence, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lee CG, Yoon HJ, Zhu Z, Link H, Wang Z, Gwaltney JM, Landry M, Elias JA. Respiratory syncytial virus stimulation of vascular endothelial cell growth Factor/Vascular permeability factor. Am J Respir Cell Mol Biol 2000; 23:662-9. [PMID: 11062145 DOI: 10.1165/ajrcmb.23.5.4188] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We hypothesized that respiratory syncytial virus (RSV)-induced pathologies could be mediated, in part, by vascular active cytokines elaborated during virus infection. To address this hypothesis, we determined whether RSV stimulated vascular endothelial cell growth factor (VEGF)/vascular permeability factor (VPF) elaboration in vitro. Supernatants from unstimulated A549 cells and normal human bronchial epithelial cells contained modest levels of VEGF. In contrast, supernatants from RSV-infected cells contained elevated levels of VEGF/VPF. This stimulation was seen after as little as 2 h, was still prominent after 48 h, and, by immunoblot, was specific for the 165- and 121-amino acid isoforms of VEGF/VPF. It was not associated with significant cell cytotoxicity or alterations in VEGF messenger RNA. It did, however, require new protein biosynthesis. In accordance with these findings, the 165- and 121-amino acid isoforms of VEGF/VPF were also found in the nasal washings from patients with RSV infections. These studies demonstrate that RSV is a potent stimulator of VEGF/VPF elaboration and that, in vitro, this stimulation is mediated via a noncytotoxic translational and/or post-translational biosynthetic mechanism. VEGF/VPF may play an important role in the pathogenesis of RSV-induced disorders.
Collapse
Affiliation(s)
- C G Lee
- Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8057, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Hiltunen MO, Laitinen M, Turunen MP, Jeltsch M, Hartikainen J, Rissanen TT, Laukkanen J, Niemi M, Kossila M, Häkkinen TP, Kivelä A, Enholm B, Mansukoski H, Turunen AM, Alitalo K, Ylä-Herttuala S. Intravascular adenovirus-mediated VEGF-C gene transfer reduces neointima formation in balloon-denuded rabbit aorta. Circulation 2000; 102:2262-8. [PMID: 11056103 DOI: 10.1161/01.cir.102.18.2262] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Gene transfer to the vessel wall may provide new possibilities for the treatment of vascular disorders, such as postangioplasty restenosis. In this study, we analyzed the effects of adenovirus-mediated vascular endothelial growth factor (VEGF)-C gene transfer on neointima formation after endothelial denudation in rabbits. For comparison, a second group was treated with VEGF-A adenovirus and a third group with lacZ adenovirus. Clinical-grade adenoviruses were used for the study. METHODS AND RESULTS Aortas of cholesterol-fed New Zealand White rabbits were balloon-denuded, and gene transfer was performed 3 days later. Animals were euthanized 2 and 4 weeks after the gene transfer, and intima/media ratio (I/M), histology, and cell proliferation were analyzed. Two weeks after the gene transfer, I/M in the lacZ-transfected control group was 0. 57+/-0.04. VEGF-C gene transfer reduced I/M to 0.38+/-0.02 (P:<0.05 versus lacZ group). I/M in VEGF-A-treated animals was 0.49+/-0.17 (P:=NS). The tendency that both VEGF groups had smaller I/M persisted at the 4-week time point, when the lacZ group had an I/M of 0.73+/-0.16, the VEGF-C group 0.44+/-0.14, and the VEGF-A group 0. 63+/-0.21 (P:=NS). Expression of VEGF receptors 1, 2, and 3 was detected in the vessel wall by immunocytochemistry and in situ hybridization. As an additional control, the effect of adenovirus on cell proliferation was analyzed by performing gene transfer to intact aorta without endothelial denudation. No differences were seen in smooth muscle cell proliferation or I/M between lacZ adenovirus and 0.9% saline-treated animals. CONCLUSIONS Adenovirus-mediated VEGF-C gene transfer may be useful for the treatment of postangioplasty restenosis and vessel wall thickening after vascular manipulations.
Collapse
MESH Headings
- Adenoviridae/genetics
- Adenoviridae/metabolism
- Angioplasty, Balloon/adverse effects
- Animals
- Aortic Valve Stenosis/etiology
- Aortic Valve Stenosis/metabolism
- Aortic Valve Stenosis/prevention & control
- Cell Division/drug effects
- Cells, Cultured
- Endothelial Growth Factors/biosynthesis
- Endothelial Growth Factors/genetics
- Endothelial Growth Factors/pharmacology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Gene Transfer Techniques
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Proto-Oncogene Proteins/biosynthesis
- Rabbits
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptors, Cell Surface/biosynthesis
- Receptors, Growth Factor/biosynthesis
- Receptors, Vascular Endothelial Growth Factor
- Transfection
- Tunica Intima/drug effects
- Tunica Intima/metabolism
- Tunica Intima/pathology
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factor C
- Vascular Endothelial Growth Factor Receptor-1
- Vascular Endothelial Growth Factor Receptor-3
Collapse
Affiliation(s)
- M O Hiltunen
- A.I. Virtanen Institute, University of Kuopio, Kuopio, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bae DG, Gho YS, Yoon WH, Chae CB. Arginine-rich anti-vascular endothelial growth factor peptides inhibit tumor growth and metastasis by blocking angiogenesis. J Biol Chem 2000; 275:13588-96. [PMID: 10788475 DOI: 10.1074/jbc.275.18.13588] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor angiogenesis is a critical step for the growth and metastasis of solid tumors. Vascular endothelial growth factor (VEGF) is a specific and potent angiogenic factor and contributes to the development of solid tumors by promoting tumor angiogenesis. Therefore, it is a prime therapeutic target for the development of antagonists for treatment of cancer. We identified from peptide libraries arginine-rich hexapeptides that inhibit the interaction of VEGF(165) with VEGF receptor (IC(50) = 2-4 micrometer). They have no effect on binding of basic fibroblast growth factor to cellular receptor. The hexapeptides inhibit the proliferation of human umbilical vein endothelial cells induced by VEGF(165) without toxicity. The peptides bind to VEGF and inhibit binding of both VEGF(165) and VEGF(121), suggesting that the peptides interact with the main body of VEGF but not the heparin-binding domain that is absent in VEGF(121). The identified peptides block the angiogenesis induced by VEGF(165) in vivo in the chick chorioallantoic membrane and the rabbit cornea. Furthermore, one of the hexapeptides, RRKRRR, blocks the growth and metastasis of VEGF-secreting HM7 human colon carcinoma cells in nude mice. Based on our results, the arginine-rich hexapeptides may be effective for the treatment of various human tumors and other angiogenesis-dependent diseases that are related to the action of VEGF and could also serve as leads for development of more effective drugs.
Collapse
Affiliation(s)
- D G Bae
- Division of Molecular and Life Science, Pohang University of Science and Technology, Pohang, 790-784, Korea
| | | | | | | |
Collapse
|
44
|
Binétruy-Tournaire R, Demangel C, Malavaud B, Vassy R, Rouyre S, Kraemer M, Plouët J, Derbin C, Perret G, Mazié JC. Identification of a peptide blocking vascular endothelial growth factor (VEGF)-mediated angiogenesis. EMBO J 2000; 19:1525-33. [PMID: 10747021 PMCID: PMC310222 DOI: 10.1093/emboj/19.7.1525] [Citation(s) in RCA: 227] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) binding to the kinase domain receptor (KDR/FLK1 or VEGFR-2) mediates vascularization and tumor-induced angiogenesis. Since there is evidence that KDR plays an important role in tumor angiogenesis, we sought to identify peptides able to block the VEGF-KDR interaction. A phage epitope library was screened by affinity for membrane-expressed KDR or for an anti-VEGF neutralizing monoclonal antibody. Both strategies led to the isolation of peptides binding KDR specifically, but those isolated by KDR binding tended to display lower reactivities. Of the synthetic peptides corresponding to selected clones tested to determine their inhibitory activity, ATWLPPR completely abolished VEGF binding to cell-displayed KDR. In vitro, this effect led to the inhibition of the VEGF-mediated proliferation of human vascular endothelial cells, in a dose-dependent and endothelial cell type-specific manner. Moreover, in vivo, ATWLPPR totally abolished VEGF-induced angiogenesis in a rabbit corneal model. Taken together, these data demonstrate that ATWLPPR is an effective antagonist of VEGF binding, and suggest that this peptide may be a potent inhibitor of tumor angiogenesis and metastasis.
Collapse
Affiliation(s)
- R Binétruy-Tournaire
- Université Paris XIII, UFR Léonard de Vinci, UPRES 2360, 'Ciblage Fonctionnel des Tumeurs Solides', 74 rue Marcel Cachin, 93017 Bobigny Cedex, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Inoue T, Kibata K, Suzuki M, Nakamura S, Motoda R, Orita K. Identification of a vascular endothelial growth factor (VEGF) antagonist, sFlt-1, from a human hematopoietic cell line NALM-16. FEBS Lett 2000; 469:14-8. [PMID: 10708747 DOI: 10.1016/s0014-5793(00)01246-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
An antagonistic activity against vascular endothelial growth factor (VEGF) was identified in the culture supernatants of certain human hematopoietic cell lines and the antagonistic protein was purified from NALM-16 (B cell) culture supernatant. Amino acid sequencing of the N-terminus and Western blot analysis confirmed that the antagonist was identical to a soluble truncated form of Flt-1 (sFlt-1). Seventeen of 52 leukemia and lymphoma cell lines investigated expressed sFlt-1 mRNA, and 16 of the sFlt-1 expressing cells also expressed VEGF and membrane-bound Flt-1 (mFlt-1). This report is the first showing that sFlt-1 can be produced by malignant hematopoietic cells, suggesting that the production of VEGF antagonist by hematopoietic cells may play some role in the regulation of VEGF activity in normal and malignant hematopoietic cell proliferation.
Collapse
Affiliation(s)
- T Inoue
- Fujisaki Cell Center, Hayashibara Biochemical Laboratories, Inc., 675-1 Fujisaki, Okayama, Japan.
| | | | | | | | | | | |
Collapse
|
46
|
Herley MT, Yu Y, Whitney RG, Sato JD. Characterization of the VEGF binding site on the Flt-1 receptor. Biochem Biophys Res Commun 1999; 262:731-8. [PMID: 10471394 DOI: 10.1006/bbrc.1999.1282] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The angiogenic growth factor VEGF binds to the receptor tyrosine kinases Flt-1 and KDR/Flk-1. Immunoglobulin (Ig)-like loop-2 of Flt-1 is involved in binding VEGF, but the contribution of other Flt-1 Ig-loops to VEGF binding remains unclear. We tested the ability of membrane-bound chimeras between the extracellular domain of Flt-1 and the cell adhesion molecule embigin to bind VEGF. VEGF bound as well to receptors containing Flt-1 loops 1-2 or 2-3 as it did to the entire Flt-1 extracellular domain. Chimeras containing only loop-2 of Flt-1 bound VEGF with 22-fold lower affinity. We conclude that high-affinity VEGF binding requires Ig-like loop-2 plus either loop-1 or loop-3. In addition, Flt-1 amino acid residues Arg-224 and Asp-231 were not essential for high-affinity binding of VEGF to membrane-bound Flt-1.
Collapse
Affiliation(s)
- M T Herley
- Biochemistry Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | |
Collapse
|
47
|
Streit M, Velasco P, Brown LF, Skobe M, Richard L, Riccardi L, Lawler J, Detmar M. Overexpression of thrombospondin-1 decreases angiogenesis and inhibits the growth of human cutaneous squamous cell carcinomas. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 155:441-52. [PMID: 10433937 PMCID: PMC1866855 DOI: 10.1016/s0002-9440(10)65140-1] [Citation(s) in RCA: 220] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The function of the endogenous angiogenesis inhibitor thrombospondin-1 (TSP-1) in epithelial tumor development has remained controversial. We studied the in vitro growth characteristics and the in vivo tumor xenograft growth of the human squamous cell carcinoma cell lines A431 and SCC-13, stably transfected to overexpress human TSP-1. Overexpression of TSP-1 inhibited tumor growth of A431 xenotransplants, and completely abolished tumor formation by SCC-13 cells. TSP-1 overexpressing A431 tumors were characterized by extensive areas of necrosis and by decreased tumor vessel number and size. The effects of TSP-1 on tumor cell growth were indirect since tumor cell proliferation rates in vivo and in vitro, anchorage-dependent and -independent growth in vitro, and susceptibility to induction of apoptosis by serum withdrawal were unchanged in TSP-1 overexpressing tumor cells. However, TSP-1 overexpression up-regulated the TSP-1 receptor CD36, leading to enhanced adhesion of A431 cells to TSP-1. These findings establish TSP-1 as a potent inhibitor of angiogenesis and tumor growth in carcinomas of the skin.
Collapse
Affiliation(s)
- M Streit
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Boston, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Yu Y, Sato JD. MAP kinases, phosphatidylinositol 3-kinase, and p70 S6 kinase mediate the mitogenic response of human endothelial cells to vascular endothelial growth factor. J Cell Physiol 1999; 178:235-46. [PMID: 10048588 DOI: 10.1002/(sici)1097-4652(199902)178:2<235::aid-jcp13>3.0.co;2-s] [Citation(s) in RCA: 176] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although the significance of vascular endothelial growth factor (VEGF) and its receptors in angiogenesis is well established, the signal transduction cascades activated by VEGF and their involvement in mediating the mitogenic response of endothelial cells to VEGF are incompletely characterized. Here we demonstrate that VEGF activates mitogen-activated protein (MAP) kinases, including the extracellular signal-regulated protein kinase (ERK) and p38 MAP kinase, phosphatidylinositol 3-kinase (PI 3-kinase), and p70 S6 kinase in human umbilical vein endothelial cells (HUVEC). The activation of these enzymes was assayed by kinase phosphorylation and by kinase activity towards substrates. Studies with PI 3-kinase inhibitors revealed that activation of p70 S6 kinase was mediated by PI 3-kinase. Selective inhibition of ERK, PI 3-kinase, and p70 S6 kinase with the inhibitors PD098059, LY294002, and rapamycin, respectively, inhibited VEGF-stimulated HUVEC proliferation. In marked contrast, the p38 MAP kinase inhibitor SB203580 not only failed to inhibit but actually enhanced HUVEC proliferation; this effect was associated with the phosphorylation of Rb protein. Rb phosphorylation resulted from a decrease in the level of the cdk inhibitor p27KiP1. These results indicate that the activities of ERK, PI 3-kinase, and p70 S6 kinase are essential for VEGF-induced HUVEC proliferation. p38 MAP kinase suppresses endothelial cell proliferation by regulating cell-cycle progression.
Collapse
Affiliation(s)
- Y Yu
- Adirondack Biomedical Research Institute, Lake Placid, New York 12946, USA
| | | |
Collapse
|
49
|
Meyer M, Clauss M, Lepple-Wienhues A, Waltenberger J, Augustin HG, Ziche M, Lanz C, Büttner M, Rziha HJ, Dehio C. A novel vascular endothelial growth factor encoded by Orf virus, VEGF-E, mediates angiogenesis via signalling through VEGFR-2 (KDR) but not VEGFR-1 (Flt-1) receptor tyrosine kinases. EMBO J 1999; 18:363-74. [PMID: 9889193 PMCID: PMC1171131 DOI: 10.1093/emboj/18.2.363] [Citation(s) in RCA: 333] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The different members of the vascular endothelial growth factor (VEGF) family act as key regulators of endothelial cell function controlling vasculogenesis, angiogenesis, vascular permeability and endothelial cell survival. In this study, we have functionally characterized a novel member of the VEGF family, designated VEGF-E. VEGF-E sequences are encoded by the parapoxvirus Orf virus (OV). They carry the characteristic cysteine knot motif present in all mammalian VEGFs, while forming a microheterogenic group distinct from previously described members of this family. VEGF-E was expressed as the native protein in mammalian cells or as a recombinant protein in Escherichia coli and was shown to act as a heat-stable, secreted dimer. VEGF-E and VEGF-A were found to possess similar bioactivities, i.e. both factors stimulate the release of tissue factor (TF), the proliferation, chemotaxis and sprouting of cultured vascular endothelial cells in vitro and angiogenesis in vivo. Like VEGF-A, VEGF-E was found to bind with high affinity to VEGF receptor-2 (KDR) resulting in receptor autophosphorylation and a biphasic rise in free intracellular Ca2+ concentration, whilst in contrast to VEGF-A, VEGF-E did not bind to VEGF receptor-1 (Flt-1). VEGF-E is thus a potent angiogenic factor selectively binding to VEGF receptor-2. These data strongly indicate that activation of VEGF receptor-2 alone can efficiently stimulate angiogenesis.
Collapse
Affiliation(s)
- M Meyer
- Department of Infection Biology, Max Planck Institute for Biology, Spemannstrasse 34, D-72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Soker S, Gollamudi-Payne S, Fidder H, Charmahelli H, Klagsbrun M. Inhibition of vascular endothelial growth factor (VEGF)-induced endothelial cell proliferation by a peptide corresponding to the exon 7-encoded domain of VEGF165. J Biol Chem 1997; 272:31582-8. [PMID: 9395496 DOI: 10.1074/jbc.272.50.31582] [Citation(s) in RCA: 148] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a potent mitogen for endothelial cells (EC) in vitro and a major regulator of angiogenesis in vivo. VEGF121 and VEGF165 are the most abundant of the five known VEGF isoforms. The structural difference between these two is the presence in VEGF165 of 44 amino acids encoded by exon 7 lacking in VEGF121. It was previously shown that VEGF165 and VEGF121 both bind to KDR/Flk-1 and Flt-1 but that VEGF165 binds in addition to a novel receptor (Soker, S., Fidder, H., Neufeld, G., and Klagsbrun, M. (1996) J. Biol. Chem. 271, 5761-5767). The binding of VEGF165 to this VEGF165-specific receptor (VEGF165R) is mediated by the exon 7-encoded domain. To investigate the biological role of this domain further, a glutathione S-transferase fusion protein corresponding to the VEGF165 exon 7-encoded domain was prepared. The fusion protein inhibited binding of 125I-VEGF165 to VEGF165R on human umbilical vein-derived EC (HUVEC) and MDA-MB-231 tumor cells. The fusion protein also inhibited significantly 125I-VEGF165 binding to KDR/Flk-1 on HUVEC but not on porcine EC which express KDR/Flk-1 alone. VEGF165 had a 2-fold higher mitogenic activity for HUVEC than did VEGF121. The exon 7 fusion protein inhibited VEGF165-induced HUVEC proliferation by 60% to about the level stimulated by VEGF121. Unexpectedly, the fusion protein also inhibited HUVEC proliferation in response to VEGF121. Deletion analysis revealed that a core inhibitory domain exists within the C-terminal 23-amino acid portion of the exon 7-encoded domain and that a cysteine residue at position 22 in exon 7 is critical for inhibition. It was concluded that the exon 7-encoded domain of VEGF165 enhances its mitogenic activity for HUVEC by interacting with VEGF165R and modulating KDR/Flk-1-mediated mitogenicity indirectly and that exon 7-derived peptides may be useful VEGF antagonists in angiogenesis-associated diseases.
Collapse
Affiliation(s)
- S Soker
- Department of Surgery, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|