Review Open Access
Copyright ©The Author(s) 2004. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Feb 15, 2004; 10(4): 463-470
Published online Feb 15, 2004. doi: 10.3748/wjg.v10.i4.463
Roles of main pro- and anti-angiogenic factors in tumor angiogenesis
Zhi Huang, School of Life Sciences, Xiamen University, Xiamen 361005, Fujian Province, China
Shi-Deng Bao, the Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen 361005, Fujian Province, China
Author contributions: All authors contributed equally to the work.
Supported by the National Natural Science Foundation of China, No.30170463
Correspondence to: Zhi Huang, Cell Biology Research Lab, School of Life Sciences, Xiamen University, Xiamen 361005, Fujian Province, China. subhiv@hotmail.com
Telephone: +86-592-2186091 Fax: +86-592-2186091
Received: June 21, 2003
Revised: July 8, 2003
Accepted: July 24, 2003
Published online: February 15, 2004

Abstract

Tumor growth without size restriction depends on vascular supply. The ability of tumor to induce new blood-vessel formation has been a major focus of cancer research over the past decade. It is now known that members of the vascular endothelial growth factor and angiopoietin families, mainly secreted by tumor cells, induce tumor angiogenesis, whereas other endogenous angiogenic inhibitors, including thrombospondin-1 and angiostatin, keep tumor in dormancy. Experimental and clinical evidence has suggested that the process of tumor metastasis depends on angiogenesis or lymphangiogenesis. This article summarizes the recent research progress for some basic pro- or anti-angiogenic factors in tumor angiogenesis.




INTRODUCTION

In the early time of the 20th century, Lewis observed that the vascular architecture depended on the tumor type, and proposed that the tumor environment determined the growth and morphological characteristics of tumor vessels. Hitherto it is the cognition that tumor vascularization is a vital process for the progression of all solid tumors from a small, localized focus to a large tumor with the capability to metastasize[1,2]. This progression begins with the sprouting or intussusception from pre-existing host vessels. Circulating endothelial precursors, shed from the vessel wall or mobilized from the bone marrow, also contribute to tumor angiogenesis[3,4].

Contrarily, adult normal tissue vasculature is quiescent because of the balance between angiogenic promoters and inhibitors. The early development of tumor is dormant because of the same reason. Break of the balance results in physiological angiogenesis such as wound healing and pathological angiogenesis such as tumor angiogenesis.

Tumor-induced angiogenesis is thought to depend on the production of pro-angiogenic growth factors by the tumor cells, which affect the existing vessels[5]. This turns on the tumor “angiogenic switch”, and then promotes tumor growth and metastases.

ANGIOGENIC FACTORS
Vascular endothelial growth factors and vascular endothelial growth factor receptors

In 1983, Senger and his colleagues discovered a protein that induced vascular leakage and named it as tumor vascular permeability factor (VPF)[6]. Subsequently Ferrara and Connolly showed that VPF and vascular endothelial growth factor (VEGF) were the same molecule[7,8]. In 1992, Breier and Risau found that the expression of VEGF mRNA temporally and spatially correlated with blood-vessel growth in the developing mouse embryo. This supported the idea that VEGF was an angiogenic factor in vivo[9]. Later Veikkola and Alitalo also demonstrated that inactivation of one of the two VEGF alleles in the mouse embryos led to severe defects in vascular formation, ultimately resulted in embryonic lethality, whereas increased expression of VEGF seemed to be essential for tumor growth beyond size restriction[10]. In-situ hybridization studies have identified that the expression of VEGF mRNA is increased in renal[11], ovarian[12], gastric carcinomas[13], and particularly the highly vascular and aggressive glioblastoma[14]. VEGF is considered as a prognostic molecular marker in hepatocellular carcinoma[15]. VEGF gene is localized on the short arm of chromosome 6[16], with eight exons and seven introns[17]. Predominant isoforms of VEGF are VEGF189, VEGF165, VEGF121, and VEGF205. They are generated by alternative mRNA splicing and proteolytic processing. VEGF121 differs from all the other higher molecular weight species by the absence of the heparin-binding domain encoded by exon 7 that can bind to neuropilin-1 (NP-1)[18,19]. Therefore, VEGF121–diphtheria toxin (DT385) conjugation treatment completely prevents tumor angiogenesis in vivo without toxicity to NP-1[20]. VEGF is a major inducer of angiogenesis and structurally resembles (placenta growth factor), VEGF-B, VEGF-C, VEGF-D and the Orf virus derived VEGF (also called VEGF-E)[21-26].

Interestingly, VEGF is highly expressed in most ischemic areas of the tumor, indicating that VEGF expression might be induced by hypoxia. In hypoxic tissue the hypoxia inducible factor-1 (HIF-1) has been proven to play a central role in inducing the transcription of genes that are involved in glycolysis and angiogenesis, including VEGF[27]. Subsequent studies have shown that the mechanisms of hypoxic regulation of VEGF and erythropoietin are similar[28]. VEGF gene contains two hypoxia-regulated element (HRE) in the 5’ and 3’ regions (Figure 1). HIF-1 recognizes and binds HRE[29,30]. Hypoxia also induces an RNA-binding protein, HuR, which can bind to the 3’ untranslated region (UTR) of VEGF mRNA and increase the mRNA stability[31]. Oncogenes including members of the ras and erbB families also up-regulate the expression of VEGF[32,33]. The products of ras gene activate the HIF-1α and promote the stability of the HIF-1αvia Raf/MEK/MAPK and PI3K/PDK-1/AKT pathways[34]. The activated AKT increases the phosphorylation of GSK3β on Ser9, and then enhances HIF-1α stabilization, since unphosphorylated GSK3β inhibits HIF-1α accumulation[35] (Figure 1). Recent studies have indicated that the chronic myelogenous leukemia (CML)-associated oncogene BCR/ABL induces VEGF expression in growth factor-dependent Ba/F3 cells by promoting the expression of HIF-1[36]. However, the product of the von Hippel-Lindau tumor suppressor gene (vHL) inhibits the expression of VEGF[37,38]. The ubiquitin ligase E3 complex containing the vHL tumor suppressor protein increases the HIF-1α degradation[39]. In addition to vHL, other tumor suppressor proteins such as p53 also inhibit the transcriptional activity of HIF-1 by binding to the HIF-1/P300 complex and then promoting Mdm2-mediated ubiquitination and proteasomal degradation of the HIF-1α, which ultimately results in the decrease of VEGF expression[40,41]. These findings indicate that HIF-1 is critical for the regulation of VEGF expression. VEGF expression is also regulated by estradiol in human breast cancer cells[42]. Similarly, 17 beta-estradiol also enhances VEGF expression via the regulation of adenylate cyclase in differentiated THP-1 cells[43].

Figure 1
Figure 1 The regulation of the VEGF expression. HIF1 plays central roles in this regulation.

VEGFs induce angiogenesis through the binding to their relevant receptors that are mainly expressed in endothelial cells. The first VEGF receptor was identified to be Flt1 (FMS-like-tyrosine kinase), also known as VEGFR1[44]. Subsequently, the other highly specific VEGF-binding receptors (KDR, Flk1, or VEGFR2) were discovered[45,46]. Scientists investigated the function of VEGFR1 and VEGFR2 in endothelial cells via Flt1 or Flk1 mutant endothelial cells or an embryonic model. They counted the number of mitotic endothelial cells in embryo to confirm the effect of proliferation of endothelial cells. In VEGFR1 knockout mice, the reason causing early embryonic lethality was not the absence of vascularisation but the disorganized embryonic and extra-embryonic vasculature[47]. This suggests that Flt1 signal pathways mainly regulate normal endothelial cell-cell or cell-matrix interactions during the process of vascular development. The ability of VEGFR1 to bind to VEGF is 10 times stronger than VEGFR2, but the activity of tyrosine kinase of VEGFR1 is weaker than that of VEGFR2. A study demonstrated that Flt1 null mutation resulted in early embryonic death, but Flt1 lacking tyrosine kinase domain homozygous mice developed normal vessels and survive[48]. These observations indicate that Flt1 without tyrosine kinase domain is sufficient to promote embryonic normal angiogenesis. The effects of VEGFR2 in endothelial cells, such as cell survival and proliferation, are not induced by treatment with VEGFR1 specific ligands or in the VEGFR1 overexpressing cells lacking VEGFR2[49,50]. These data suggest that VEGFR2 plays the central role in VEGF signal transduction in endothelial cells. VEGFR1 is likely to be the antagonist of VEGFR2, acting as a negative regulator of angiogenesis via sequestering VEGF and thus blocking the VEGF signal transduction mediated by VEGFR2. Recent studies have confirmed this conclusion. The Flt1 gene encodes not only the full-length Flt-1, but also a soluble short form of Ftl-1, designated sFlt1. In placenta, sFlt-1 is abundant in the trophoblast layer during pregnancy, suggesting that it is a negative regulator to excess angiogenesis at the feto-maternal border in mammals[51]. Subsequently Kearney confirmed that Flt-1 normally modulated vascular growth by controlling the rate of endothelial cell division in vitro and in vivo[52]. The downstream signaling events of VEGF signal transduction pathway via VEGFR2 includes receptor dimerization, transphosphorylation of intracellular VEGFR2 tyrosine residues (Tyr951/996 and Tyr1054/1059)[53], the activation of protein kinase C, and then the activation of the MEK/MAPK pathway to promote endothelial cell proliferation (Figure 2). However, Meyer’s data showed that phosphorylation of tyrosine 1212 of VEGFR2 was also necessary for VEGFR2-mediated angiogenesis[54]. In addition to VEGF, scientists have found that there are other molecules that activate VEGFR2 or enhance the expression of VEGFR2. Recent studies have indicated that tumor necrosis factor (TNF) has anti-tumor activity by down-regulating Flk-1 expression in tumor endothelial cells[55]. Moreover, in cardiac capillary endothelial cells, Bradykinin (BK) also stimulates angiogenesis via the activation of the B2-type receptor, which leads to the tyrosine phosphorylation and dimerization of Flk-1 as does VEGF itself[56]. This implies a novel mechanism by which a G protein-coupled receptor activates a receptor tyrosine kinase to generate biological response. In addition to the stimulation of endothelial cell migration and proliferation, VEGF also elevates the survival capacity of endothelial cells in vitro. The underlying mechanism is that VEGF activates VEGFR2, which further activates phosphatidylinositol 3-kinase (PI3-K) and Akt[57]. VEGF also prolongs the survival of human dermal microvascular endothelial cells (HDMECs) in vitro by inducing expression of the anti-apoptotic protein Bcl-2[58].

Figure 2
Figure 2 The intracellular downstream signaling of VEGF signal transduction via VEGFR2.

Interestingly, new data have revealed that NP-1, originally found to control the neuronal axon guidance, also enhances the mitogenic effects of VEGFR2 signaling in endothelial cells[59]. So far, the potential role of NP-1 in pathologic angiogenesis remains unknown. Some experiments have indicated that VEGF up-regulates NP-1 via VEGFR2-dependent pathway. Endothelial proliferation stimulated by VEGF165 is inhibited significantly by antibody neutralization of NP-1. In a murine model of VEGF-dependent angioproliferative retinopathy, intense NP1 mRNA expression was observed in the newly formed vessels[60]. In breast carcinoma cells neuropilin expression also enhances VEGF-inducing cells survival signal[61]. Furthermore, in rat uterus the neuropilins (NP-1 and NP-2) might participate in hormonally regulated changes occurring throughout the female reproductive cycle[62].

The other factor VEGF-C and its receptor (VEGFR3) are essential to the lymphangiogenesis and play a direct role in the metastatic dissemination of tumor cells. It is known that VEGFR3 is mainly expressed in lymphatic endothelium during development[63,64]. Later in embryonic development, VEGFR3 is expressed in lymphatic cells, and further induces lymphangiogenesis[63,64]. However, in early embryonic development without lymphatic vessels, the embryos lacking VEGFR3 gene usually die due to cardiovascular failure[65]. These data indicate that VEGFR3 is necessary in the formation of the primary cardiovascular network. In addition, VEGFR3 is also expressed in the tumor endothelial cells, which helps tumor neovascularization[66].

Angiopoietins and Tie receptors

The second family of growth factors that specifically act on the vascular endothelium has been identified, namely the angiopoietins[67,68]. To date, there are four members of this family, mainly including Ang-1 and Ang-2, which both only bind to the Tie-2 receptor and control the blood vessel stabilization signals[67,68]. The results of early experiments showed that the mouse embryonic development lacking Ang-1 or Tie-2 died in the later stage of angiogenesis because the remodeling and stabilization of the primitive vasculature induced by VEGF was severely perturbed[69-71]. Similarly, the increased Ang-1 expression in human cervical cancer HeLa cells by transgene promotes the growth of human cervical cancers in mice via promoting tumor angiogenesis[72]. In addition to in the active angiogenesis tissue, Ang-1 seems to be widely expressed in the normal adult tissues[73], indicating that Ang-1 may act on the stabilization of existing vessels. The underlying mechanism is thought to modulate the interaction between endothelial cells and surrounding support cells, such as smooth muscle cells[71,74]. Ang-2 was cloned in 1997. It is thought to be a natural antagonist for the Ang-1/Tie-2 interaction now. The transgenic overexpression of Ang-2 in embryo results in early embryonic lethality, similar to the phenomenon in embryonic development without Ang-1 or Tie-2[68].

The relation of VEGF and angiopoietins in angiogenesis has become a research hotspot (Figure 3). In the onset of cerebral ischemia, the increase of VEGF mRNA goes with the decrease of Ang-1 mRNA. Later increased expression of VEGF/VEGFRs and Ang-1/Tie2 is detected[75]. This data show that Ang-1 works at the later stage of angiogenesis. Other experiments directly demonstrate the above mentioned role of VEGF and Ang-1. The transgenic overexpression of VEGF in the skin of mice induces the formation of numerous leaky blood vessels. In contrast, overexpression of Ang-1 leads to an enlargement of dermal vessels with less leaking, instead of an increase in the number of vessels. If both VEGF and Ang-1 are overexpressed, the size and number of skin vessels are both increased. Furthermore, these vessels are not leaky. Remarkably, it is found that Ang-1 also reduces VEGF-stimulated inflammation by suppressing expression of adhesion molecules, including intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1)[76] and E-selectin[77]. The expression of Ang-2 is not broader than that of Ang-1. Holash et al[78] demonstrated that endothelial cells apoptosis occurred prior to the wide spread loss of tumor cells in the necrotic tumor core. This implies that there is a mechanism to induce this process. Ang-2 promotes endothelial cell death and vessel regression if the activity of VEGF is inhibited. By contrast, Ang-2 promotes a rapid increase in capillary diameter and stimulates sprouting of new blood vessels in the presence of endogenous VEGF in vivo[79]. These results indicate that the pre-effect of existing blood vessels by Ang-2 is in favor of the step of VEGF-inducing angiogenesis. Thereby, in the regulation of angiogenesis, VEGF can convert the role of Ang-2 from an anti-angiogenic factor to a pro-angiogenic factor. Other data further confirm that Ang-2 expression precedes that of VEGF in the early stage of tumor angiogenesis[80].

Figure 3
Figure 3 The interaction relation between VEGF and angiopoietins in the development of tumor angiogenesis.

Altogether, VEGF/VEGFRs and angiopoietins/Ties co-regulate the tumor vessels regression, growth and maturation. Expression of Ang-2 without VEGF induces vessels regression in core of a necrotic tumor. Then, hypoxia induces tumor cells to express VEGF. The angiogenic properties of VEGF may be easier to act on the vessels, which are destabilized by Ang-2. Newly formed tumor vessels are abnormal, including highly disorganization, poor differentiation, tortuousness and excessive branch, because continuous expression of Ang-2 in newly tumor vessels inhibits the maturation and stabilization signals via Ang-1. In the later development of tumor angiogenesis, the increasing expression of Ang-1 promotes the maturation of neovascularization. In fact, hyper-vascular hepatomas with aberrant vasculature show high levels of Ang-2 expression in the endothelium.

Other angiogenic factors

The mechanism of tumor angiogenesis is complicated. It involves many factors and some different signal pathways. Therefore, if only one signal molecule (for example VEGF) is blocked, tumor may switch to another molecule (for example bFGF) for the induction of angiogenesis. The basic fibroblast growth factor (bFGF, also known as FGF-2) isolated from a chondrosarcoma was identified to be the first pro-angiogenic factor[81]. bFGF is the main member of FGF family which is a structurally homologous protein family, including at least 20 members. FGF has a high affinity to heparin sulfate protepglycans (HSPGs), which locates on the surface of most cells and within the extracellular matrix[82].

Four receptor tyrosine kinases (known as FGFR-1,-2,-3,-4) can interact with FGF. The FGF/FGFRs signal pathway may play a crucial role in angiogenesis. The FGFR-1 mutant mouse embryos exhibited abnormal embryonic and extra-embryonic vascularization, suggesting that FGF/FGFR-1 plays a role in the development and maintenance of a matured vascular network in the embryo[83]. The mechanisms for VEGF- and FGF-mediated angiogenesis are not the same. In astrocytoma, bFGF and VEGF expression levels positively correlated with tumor growth and angiogenesis. However, bFGF and VEGF expression levels were significantly different in various grades of astrocytoma[84]. In the culture of embryonic stem cell-derived vascular precursors in vitro, bFGF and VEGF both exhibited effects on the survival of angioblasts. However, VEGF induced the formation of primitive endothelial tubes, whereas bFGF did not[85]. This finding suggests that VEGF but not bFGF plays a major role in angiogenesis. The mitogen-activated protein kinase kinase (MEK) inhibitor PD98059 suppresses FGF-induced angiogenesis, indicating that the Ras-MEK-MAPK pathway is important for the biological effect of FGF[86]. Subsequently, Cross et al[87] found that the Shb adaptor protein binding to Tyr766 in the FGFR-1 promoted FRS2 phosphorylation, and further regulated the Ras-MEK-MAPK pathway. FGFR-1-mediated intracellular signal cascades include the Ras pathway, Src family tyrosine kinases, phosphoinositide 3-kinase (PI3K) and the PLC pathway[88]. Some members of the FGF family (such as FGF-3) promote oncogenesis, and are identified as proto-oncogenes[89].

ANTI-ANGIOGENIC FACTORS

In humans or vertebrates, normal vascularization is quiescent, suggesting that in vivo humans or vertebrates should have anti-angiogenic factors against angiogenic factors. Recently, experiments in vitro or in vivo confirm the presence of endogenous anti-angiogenic factors.

The primary anti-angiogenic factors include thrombospondins (TSP), angiostatin and endostatin. Now the TSP family includes five members, known as TSP-1, -2, -3, -4 and TSP-5/COMP. They play multiple functions via binding to matrix proteins, plasma proteins and cytokines[90-93]. Some reports have shown that TSP-1 and TSP-2 have anti-angiogenic properties. TSP-1 inhibits angiogenesis via a direct way that suppresses migration and induces apoptosis of endothelial cells or via an indirect way that inhibits the mobilization of pro-angiogenic factors and blocks their access to co-receptors on the endothelial cell surface[94]. TSP-2 has the similar functions[95]. The signal pathway of TSP includes TSP/CD36 receptor, cytoplasmic tyrosine kinase p59fyn, caspase-3-like proteases and p38 mitogen-activated protein kinase (MAPK), and results in apoptosis of endothelial cells[94,96,97]. It has been proven that TSP is an anti-angiogenic factor or acts as prognostic predictor in pancreatic ductal carcinoma[98], bladder cancer[99], non-small cell lung cancer[100,101], breast cancer[102], cervical cancer[103] and colon cancer[104]. TSP expression is regulated by several factors. Hypoxia increases TSP-1 expression in endothelial cells by promoting the post-transcriptional stabilization of the TSP-1 mRNA[105]. This property does not match with its anti-angiogenic property. Contrarily, in vitro hypoxia suppresses TSP-1 expression and induces VEGF expression in rodent cells, regardless of the p53 genotype[106]. In cultured fibroblasts, loss of the wild-type allele of the p53 results in reduced expression of TSP-1. Transfection assays have revealed that p53 stimulates the expression of the endogenous TSP-1 gene and positively regulate TSP-1 gene promoter sequences[107,108]. A novel p53-inducible gene, named BAI1, was found in 1997[109]. BAI1 contains five TSP-type 1 repeats, and significantly suppresses angiogenesis. Some oncogenes have been also found to regulate TSP1 expression, including c-jun[110] and H-ras[106]. These data indicate that oncogenes or loss of tumor suppressor genes also regulate TSP expression to turn on the angiogenic switch and promote tumor growth.

A number of experimental studies have demonstrated that a primary tumor inhibits a secondary distant tumor[111,112], indicating that there are some inhibitors from the primary tumor to inhibit the growth of metastases. Endostatin is a circulatory inhibitor, and identified to be an anti-angiogenic factor[113]. It is a 20 kD C-terminal fragment of collagen XVIII. Treatment with endostatin by transgene inhibits angiogenesis and liver tumor growth[114]. In other similar diseases such as rheumatoid arthritis, treatment with endostatin showed beneficial effects[115]. Interestingly, endostatin immunostaining was stronger in area accompanied with blood vessel maturation. Endostatin expression was significantly prominent in vessels of tumor marginal zone where angiogenesis was highly active. These observations suggest that endostatin inhibits angiogenesis by stabilization of newly formed endothelial tubes in angiogenic active region in tumors[116]. Intracellular signaling mechanisms by endostatin remain poorly understood. Jiang et al[117] found that endostatin increases intracellular Ca2+ concentration approximately three-fold, indicating that endostatin induces intracellular Ca2+ signal. Endostatin also modulates the Wnt signaling by regulating β-catenin stability via a novel glycogen synthase kinase 3(GSK3)-independent mechanism[118].

Angiostatin is another circulatory anti-angiogenic factor. It was isolated from a lung carcinoma cell line and is particularly effective in suppressing metastatic growth when the primary tumor is removed[119]. Angiostatin is a 38 kD fragment of plasminogen. Treatment with angiostatin inhibits angiogenesis in a murine breast cancer model[120]. Now it is found that angiostatin plays a crucial role in inflammation via directly inhibiting neutrophil migration and neutrophil-mediated angiogenesis[121]. Recent studies have shown that angiostatin has structural similarities to hepatocyte growth factor (HGF), which induces proliferation and migration of both endothelial cells and smooth muscle cells via its cell surface receptor, c-met. Therefore, the disruption of HGF/c-met signaling is a novel mechanism for the anti-angiogenic effect of angiostatin[122]. Experiments by Mauceri et al[123] indicated that TNF-α enhances the production of angiostatin in tumor cells by inducing the activity of the plasminogen activator and the release of MMP-9 by tumor cells.

CONCLUSIONS

Tumor angiogenesis plays crucial roles in tumor growth and metastases. Tumor angiogenesis is a complex process involving interactions of many molecules and several different or similar signal pathways. Despite the rapid research progress in the field, there are many mysteries regarding the molecular mechanism of tumor angiogenesis. Once we fully understand the precise functions of these pro- and anti-angiogenic factors in tumor angiogenesis, clinical application of those novel research results for tumor therapy would become practical.

Footnotes

Edited by Xia HHX Proofread by Zhu LH

References
1.  Gupta MK, Qin RY. Mechanism and its regulation of tumor-induced angiogenesis. World J Gastroenterol. 2003;9:1144-1155.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Folkman J. Anti-angiogenesis: new concept for therapy of solid tumors. Ann Surg. 1972;175:409-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 825]  [Cited by in F6Publishing: 795]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
3.  Asahara T, Kalka C, Isner JM. Stem cell therapy and gene transfer for regeneration. Gene Ther. 2000;7:451-457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 111]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
4.  Rafii S. Circulating endothelial precursors: mystery, reality, and promise. J Clin Invest. 2000;105:17-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 319]  [Cited by in F6Publishing: 324]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
5.  Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86:353-364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4848]  [Cited by in F6Publishing: 4703]  [Article Influence: 168.0]  [Reference Citation Analysis (0)]
6.  Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 1983;219:983-985.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2734]  [Cited by in F6Publishing: 2616]  [Article Influence: 63.8]  [Reference Citation Analysis (0)]
7.  Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science. 1989;246:1306-1309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3536]  [Cited by in F6Publishing: 3491]  [Article Influence: 99.7]  [Reference Citation Analysis (0)]
8.  Keck PJ, Hauser SD, Krivi G, Sanzo K, Warren T, Feder J, Connolly DT. Vascular permeability factor, an endothelial cell mitogen related to PDGF. Science. 1989;246:1309-1312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1412]  [Cited by in F6Publishing: 1371]  [Article Influence: 39.2]  [Reference Citation Analysis (0)]
9.  Breier G, Albrecht U, Sterrer S, Risau W. Expression of vascular endothelial growth factor during embryonic angiogenesis and endothelial cell differentiation. Development. 1992;114:521-532.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Veikkola T, Alitalo K. VEGFs, receptors and angiogenesis. Semin Cancer Biol. 1999;9:211-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 358]  [Cited by in F6Publishing: 379]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
11.  Takahashi A, Sasaki H, Kim SJ, Tobisu K, Kakizoe T, Tsukamoto T, Kumamoto Y, Sugimura T, Terada M. Markedly increased amounts of messenger RNAs for vascular endothelial growth factor and placenta growth factor in renal cell carcinoma associated with angiogenesis. Cancer Res. 1994;54:4233-4237.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Olson TA, Mohanraj D, Carson LF, Ramakrishnan S. Vascular permeability factor gene expression in normal and neoplastic human ovaries. Cancer Res. 1994;54:276-280.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Du JR, Jiang Y, Zhang YM, Fu H. Vascular endothelial growth factor and microvascular density in esophageal and gastric carcinomas. World J Gastroenterol. 2003;9:1604-1606.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Plate KH, Breier G, Weich HA, Risau W. Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature. 1992;359:845-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1572]  [Cited by in F6Publishing: 1568]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
15.  Qin LX, Tang ZY. The prognostic molecular markers in hepatocellular carcinoma. World J Gastroenterol. 2002;8:385-392.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Vincenti V, Cassano C, Rocchi M, Persico G. Assignment of the vascular endothelial growth factor gene to human chromosome 6p21.3. Circulation. 1996;93:1493-1495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 346]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
17.  Tischer E, Mitchell R, Hartman T, Silva M, Gospodarowicz D, Fiddes JC, Abraham JA. The human gene for vascular endothelial growth factor. Multiple protein forms are encoded through alternative exon splicing. J Biol Chem. 1991;266:11947-11954.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Soker S, Fidder H, Neufeld G, Klagsbrun M. Characterization of novel vascular endothelial growth factor (VEGF) receptors on tumor cells that bind VEGF165 via its exon 7-encoded domain. J Biol Chem. 1996;271:5761-5767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 256]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
19.  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;272:31582-31588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 148]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
20.  Wild R, Dhanabal M, Olson TA, Ramakrishnan S. Inhibition of angiogenesis and tumour growth by VEGF121-toxin conjugate: differential effect on proliferating endothelial cells. Br J Cancer. 2000;83:1077-1083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
21.  Ferrara N, Davis-Smyth T. The biology of vascular endothelial growth factor. Endocr Rev. 1997;18:4-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2348]  [Cited by in F6Publishing: 2450]  [Article Influence: 90.7]  [Reference Citation Analysis (0)]
22.  Olofsson B, Pajusola K, Kaipainen A, von Euler G, Joukov V, Saksela O, Orpana A, Pettersson RF, Alitalo K, Eriksson U. Vascular endothelial growth factor B, a novel growth factor for endothelial cells. Proc Natl Acad Sci USA. 1996;93:2576-2581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 527]  [Cited by in F6Publishing: 493]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
23.  Hanrahan V, Currie MJ, Gunningham SP, Morrin HR, Scott PA, Robinson BA, Fox SB. The angiogenic switch for vascular endothelial growth factor (VEGF)-A, VEGF-B, VEGF-C, and VEGF-D in the adenoma-carcinoma sequence during colorectal cancer progression. J Pathol. 2003;200:183-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 160]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
24.  Achen MG, Jeltsch M, Kukk E, Mäkinen T, Vitali A, Wilks AF, Alitalo K, Stacker SA. Vascular endothelial growth factor D (VEGF-D) is a ligand for the tyrosine kinases VEGF receptor 2 (Flk1) and VEGF receptor 3 (Flt4). Proc Natl Acad Sci USA. 1998;95:548-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 880]  [Cited by in F6Publishing: 839]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
25.  Ogawa S, Oku A, Sawano A, Yamaguchi S, Yazaki Y, Shibuya M. A novel type of vascular endothelial growth factor, VEGF-E (NZ-7 VEGF), preferentially utilizes KDR/Flk-1 receptor and carries a potent mitotic activity without heparin-binding domain. J Biol Chem. 1998;273:31273-31282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 291]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
26.  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;18:363-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 342]  [Cited by in F6Publishing: 356]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
27.  Gleadle JM, Ratcliffe PJ. Hypoxia and the regulation of gene expression. Mol Med Today. 1998;4:122-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 101]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
28.  Goldberg MA, Schneider TJ. Similarities between the oxygen-sensing mechanisms regulating the expression of vascular endothelial growth factor and erythropoietin. J Biol Chem. 1994;269:4355-4359.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Minchenko A, Salceda S, Bauer T, Caro J. Hypoxia regulatory elements of the human vascular endothelial growth factor gene. Cell Mol Biol Res. 1994;40:35-39.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Ladoux A, Frelin C. Hypoxia is a strong inducer of vascular endothelial growth factor mRNA expression in the heart. Biochem Biophys Res Commun. 1993;195:1005-1010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 238]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
31.  Levy NS, Chung S, Furneaux H, Levy AP. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. J Biol Chem. 1998;273:6417-6423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 489]  [Cited by in F6Publishing: 515]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
32.  Okada F, Rak JW, Croix BS, Lieubeau B, Kaya M, Roncari L, Shirasawa S, Sasazuki T, Kerbel RS. Impact of oncogenes in tumor angiogenesis: mutant K-ras up-regulation of vascular endothelial growth factor/vascular permeability factor is necessary, but not sufficient for tumorigenicity of human colorectal carcinoma cells. Proc Natl Acad Sci USA. 1998;95:3609-3614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 175]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
33.  Petit AM, Rak J, Hung MC, Rockwell P, Goldstein N, Fendly B, Kerbel RS. Neutralizing antibodies against epidermal growth factor and ErbB-2/neu receptor tyrosine kinases down-regulate vascular endothelial growth factor production by tumor cells in vitro and in vivo: angiogenic implications for signal transduction therapy of solid tumors. Am J Pathol. 1997;151:1523-1530.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Sodhi A, Montaner S, Miyazaki H, Gutkind JS. MAPK and Akt act cooperatively but independently on hypoxia inducible factor-1alpha in rasV12 upregulation of VEGF. Biochem Biophys Res Commun. 2001;287:292-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 143]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
35.  Mottet D, Dumont V, Deccache Y, Demazy C, Ninane N, Raes M, Michiels C. Regulation of hypoxia-inducible factor-1alpha protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta pathway in HepG2 cells. J Biol Chem. 2003;278:31277-31285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 251]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
36.  Mayerhofer M, Valent P, Sperr WR, Griffin JD, Sillaber C. BCR/ABL induces expression of vascular endothelial growth factor and its transcriptional activator, hypoxia inducible factor-1alpha, through a pathway involving phosphoinositide 3-kinase and the mammalian target of rapamycin. Blood. 2002;100:3767-3775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 228]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
37.  Siemeister G, Weindel K, Mohrs K, Barleon B, Martiny-Baron G, Marmé D. Reversion of deregulated expression of vascular endothelial growth factor in human renal carcinoma cells by von Hippel-Lindau tumor suppressor protein. Cancer Res. 1996;56:2299-2301.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Gnarra JR, Zhou S, Merrill MJ, Wagner JR, Krumm A, Papavassiliou E, Oldfield EH, Klausner RD, Linehan WM. Post-transcriptional regulation of vascular endothelial growth factor mRNA by the product of the VHL tumor suppressor gene. Proc Natl Acad Sci USA. 1996;93:10589-10594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 381]  [Cited by in F6Publishing: 407]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
39.  Maxwell PH, Ratcliffe PJ. Oxygen sensors and angiogenesis. Semin Cell Dev Biol. 2002;13:29-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 252]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
40.  Blagosklonny MV, An WG, Romanova LY, Trepel J, Fojo T, Neckers L. p53 inhibits hypoxia-inducible factor-stimulated transcription. J Biol Chem. 1998;273:11995-11998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 215]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
41.  Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, Zeng Q, Dillehay LE, Madan A, Semenza GL, Bedi A. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1alpha. Genes Dev. 2000;14:34-44.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Hyder SM, Nawaz Z, Chiappetta C, Stancel GM. Identification of functional estrogen response elements in the gene coding for the potent angiogenic factor vascular endothelial growth factor. Cancer Res. 2000;60:3183-3190.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Kanda N, Watanabe S. 17beta-estradiol enhances vascular endothelial growth factor production and dihydrotestosterone antagonizes the enhancement via the regulation of adenylate cyclase in differentiated THP-1 cells. J Invest Dermatol. 2002;118:519-529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
44.  de Vries C, Escobedo JA, Ueno H, Houck K, Ferrara N, Williams LT. The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. Science. 1992;255:989-991.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1469]  [Cited by in F6Publishing: 1447]  [Article Influence: 45.2]  [Reference Citation Analysis (0)]
45.  Terman BI, Dougher-Vermazen M, Carrion ME, Dimitrov D, Armellino DC, Gospodarowicz D, Böhlen P. Identification of the KDR tyrosine kinase as a receptor for vascular endothelial cell growth factor. Biochem Biophys Res Commun. 1992;187:1579-1586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1048]  [Cited by in F6Publishing: 1042]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
46.  Millauer B, Wizigmann-Voos S, Schnürch H, Martinez R, Møller NP, Risau W, Ullrich A. High affinity VEGF binding and developmental expression suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis. Cell. 1993;72:835-846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1447]  [Cited by in F6Publishing: 1379]  [Article Influence: 44.5]  [Reference Citation Analysis (0)]
47.  Fong GH, Rossant J, Gertsenstein M, Breitman ML. Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature. 1995;376:66-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1894]  [Cited by in F6Publishing: 1768]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
48.  Hiratsuka S, Minowa O, Kuno J, Noda T, Shibuya M. Flt-1 lacking the tyrosine kinase domain is sufficient for normal development and angiogenesis in mice. Proc Natl Acad Sci USA. 1998;95:9349-9354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 767]  [Cited by in F6Publishing: 742]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
49.  Seetharam L, Gotoh N, Maru Y, Neufeld G, Yamaguchi S, Shibuya M. A unique signal transduction from FLT tyrosine kinase, a receptor for vascular endothelial growth factor VEGF. Oncogene. 1995;10:135-147.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Kroll J, Waltenberger J. The vascular endothelial growth factor receptor KDR activates multiple signal transduction pathways in porcine aortic endothelial cells. J Biol Chem. 1997;272:32521-32527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 319]  [Cited by in F6Publishing: 319]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
51.  Yamaguchi S, Iwata K, Shibuya M. Soluble Flt-1 (soluble VEGFR-1), a potent natural antiangiogenic molecule in mammals, is phylogenetically conserved in avians. Biochem Biophys Res Commun. 2002;291:554-559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
52.  Kearney JB, Ambler CA, Monaco KA, Johnson N, Rapoport RG, Bautch VL. Vascular endothelial growth factor receptor Flt-1 negatively regulates developmental blood vessel formation by modulating endothelial cell division. Blood. 2002;99:2397-2407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 115]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
53.  Dougher-Vermazen M, Hulmes JD, Böhlen P, Terman BI. Biological activity and phosphorylation sites of the bacterially expressed cytosolic domain of the KDR VEGF-receptor. Biochem Biophys Res Commun. 1994;205:728-738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 94]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
54.  Meyer RD, Dayanir V, Majnoun F, Rahimi N. The presence of a single tyrosine residue at the carboxyl domain of vascular endothelial growth factor receptor-2/FLK-1 regulates its autophosphorylation and activation of signaling molecules. J Biol Chem. 2002;277:27081-27087.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
55.  Menon C, Iyer M, Prabakaran I, Canter RJ, Lehr SC, Fraker DL. TNF-alpha downregulates vascular endothelial Flk-1 expression in human melanoma xenograft model. Am J Physiol Heart Circ Physiol. 2003;284:H317-H329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
56.  Thuringer D, Maulon L, Frelin C. Rapid transactivation of the vascular endothelial growth factor receptor KDR/Flk-1 by the bradykinin B2 receptor contributes to endothelial nitric-oxide synthase activation in cardiac capillary endothelial cells. J Biol Chem. 2002;277:2028-2032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 92]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
57.  Gerber HP, McMurtrey A, Kowalski J, Yan M, Keyt BA, Dixit V, Ferrara N. Vascular endothelial growth factor regulates endothelial cell survival through the phosphatidylinositol 3'-kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR activation. J Biol Chem. 1998;273:30336-30343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1473]  [Cited by in F6Publishing: 1552]  [Article Influence: 59.7]  [Reference Citation Analysis (0)]
58.  Nör JE, Christensen J, Mooney DJ, Polverini PJ. Vascular endothelial growth factor (VEGF)-mediated angiogenesis is associated with enhanced endothelial cell survival and induction of Bcl-2 expression. Am J Pathol. 1999;154:375-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 475]  [Cited by in F6Publishing: 467]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
59.  Soker S, Takashima S, Miao HQ, Neufeld G, Klagsbrun M. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell. 1998;92:735-745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1849]  [Cited by in F6Publishing: 1823]  [Article Influence: 70.1]  [Reference Citation Analysis (0)]
60.  Oh H, Takagi H, Otani A, Koyama S, Kemmochi S, Uemura A, Honda Y. Selective induction of neuropilin-1 by vascular endothelial growth factor (VEGF): a mechanism contributing to VEGF-induced angiogenesis. Proc Natl Acad Sci USA. 2002;99:383-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 124]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
61.  Bachelder RE, Crago A, Chung J, Wendt MA, Shaw LM, Robinson G, Mercurio AM. Vascular endothelial growth factor is an autocrine survival factor for neuropilin-expressing breast carcinoma cells. Cancer Res. 2001;61:5736-5740.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Pavelock K, Braas K, Ouafik L, Osol G, May V. Differential expression and regulation of the vascular endothelial growth factor receptors neuropilin-1 and neuropilin-2 in rat uterus. Endocrinology. 2001;142:613-622.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Kaipainen A, Korhonen J, Mustonen T, van Hinsbergh VW, Fang GH, Dumont D, Breitman M, Alitalo K. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc Natl Acad Sci USA. 1995;92:3566-3570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1026]  [Cited by in F6Publishing: 1013]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
64.  Kukk E, Lymboussaki A, Taira S, Kaipainen A, Jeltsch M, Joukov V, Alitalo K. VEGF-C receptor binding and pattern of expression with VEGFR-3 suggests a role in lymphatic vascular development. Development. 1996;122:3829-3837.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Dumont DJ, Jussila L, Taipale J, Lymboussaki A, Mustonen T, Pajusola K, Breitman M, Alitalo K. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science. 1998;282:946-949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 638]  [Cited by in F6Publishing: 678]  [Article Influence: 26.1]  [Reference Citation Analysis (0)]
66.  Valtola R, Salven P, Heikkilä P, Taipale J, Joensuu H, Rehn M, Pihlajaniemi T, Weich H, deWaal R, Alitalo K. VEGFR-3 and its ligand VEGF-C are associated with angiogenesis in breast cancer. Am J Pathol. 1999;154:1381-1390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 397]  [Cited by in F6Publishing: 411]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
67.  Davis S, Aldrich TH, Jones PF, Acheson A, Compton DL, Jain V, Ryan TE, Bruno J, Radziejewski C, Maisonpierre PC. Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning. Cell. 1996;87:1161-1169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1465]  [Cited by in F6Publishing: 1508]  [Article Influence: 53.9]  [Reference Citation Analysis (0)]
68.  Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C, Compton D, McClain J, Aldrich TH, Papadopoulos N. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science. 1997;277:55-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2548]  [Cited by in F6Publishing: 2440]  [Article Influence: 90.4]  [Reference Citation Analysis (0)]
69.  Dumont DJ, Gradwohl G, Fong GH, Puri MC, Gertsenstein M, Auerbach A, Breitman ML. Dominant-negative and targeted null mutations in the endothelial receptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of the embryo. Genes Dev. 1994;8:1897-1909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 713]  [Cited by in F6Publishing: 677]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
70.  Sato TN, Tozawa Y, Deutsch U, Wolburg-Buchholz K, Fujiwara Y, Gendron-Maguire M, Gridley T, Wolburg H, Risau W, Qin Y. Distinct roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation. Nature. 1995;376:70-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1319]  [Cited by in F6Publishing: 1229]  [Article Influence: 42.4]  [Reference Citation Analysis (0)]
71.  Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, Davis S, Sato TN, Yancopoulos GD. Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell. 1996;87:1171-1180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2120]  [Cited by in F6Publishing: 2010]  [Article Influence: 71.8]  [Reference Citation Analysis (0)]
72.  Shim WS, Teh M, Bapna A, Kim I, Koh GY, Mack PO, Ge R. Angiopoietin 1 promotes tumor angiogenesis and tumor vessel plasticity of human cervical cancer in mice. Exp Cell Res. 2002;279:299-309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 68]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
73.  Wong AL, Haroon ZA, Werner S, Dewhirst MW, Greenberg CS, Peters KG. Tie2 expression and phosphorylation in angiogenic and quiescent adult tissues. Circ Res. 1997;81:567-574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 301]  [Cited by in F6Publishing: 294]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
74.  Folkman J, D'Amore PA. Blood vessel formation: what is its molecular basis. Cell. 1996;87:1153-1155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 934]  [Cited by in F6Publishing: 855]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
75.  Zhang ZG, Zhang L, Tsang W, Soltanian-Zadeh H, Morris D, Zhang R, Goussev A, Powers C, Yeich T, Chopp M. Correlation of VEGF and angiopoietin expression with disruption of blood-brain barrier and angiogenesis after focal cerebral ischemia. J Cereb Blood Flow Metab. 2002;22:379-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 319]  [Cited by in F6Publishing: 340]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
76.  Ding YB, Chen GY, Xia JG, Zang XW, Yang HY, Yang L. Association of VCAM-1 overexpression with oncogenesis, tumor angiogenesis and metastasis of gastric carcinoma. World J Gastroenterol. 2003;9:1409-1414.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Kim I, Moon SO, Park SK, Chae SW, Koh GY. Angiopoietin-1 reduces VEGF-stimulated leukocyte adhesion to endothelial cells by reducing ICAM-1, VCAM-1, and E-selectin expression. Circ Res. 2001;89:477-479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 250]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
78.  Holash J, Maisonpierre PC, Compton D, Boland P, Alexander CR, Zagzag D, Yancopoulos GD, Wiegand SJ. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science. 1999;284:1994-1998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1620]  [Cited by in F6Publishing: 1498]  [Article Influence: 59.9]  [Reference Citation Analysis (0)]
79.  Lobov IB, Brooks PC, Lang RA. Angiopoietin-2 displays VEGF-dependent modulation of capillary structure and endothelial cell survival in vivo. Proc Natl Acad Sci USA. 2002;99:11205-11210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 478]  [Cited by in F6Publishing: 492]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
80.  Péoch M, Farion R, Hiou A, Le Bas JF, Pasquier B, Rémy C. Immunohistochemical study of VEGF, angiopoietin 2 and their receptors in the neovascularization following microinjection of C6 glioma cells into rat brain. Anticancer Res. 2002;22:2147-2151.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Shing Y, Folkman J, Sullivan R, Butterfield C, Murray J, Klagsbrun M. Heparin affinity: purification of a tumor-derived capillary endothelial cell growth factor. Science. 1984;223:1296-1299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 674]  [Cited by in F6Publishing: 720]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
82.  Vlodavsky I, Bar-Shavit R, Ishai-Michaeli R, Bashkin P, Fuks Z. Extracellular sequestration and release of fibroblast growth factor: a regulatory mechanism. Trends Biochem Sci. 1991;16:268-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 212]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
83.  Lee SH, Schloss DJ, Swain JL. Maintenance of vascular integrity in the embryo requires signaling through the fibroblast growth factor receptor. J Biol Chem. 2000;275:33679-33687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 71]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
84.  Bian XW, Du LL, Shi JQ, Cheng YS, Liu FX. Correlation of bFGF, FGFR-1 and VEGF expression with vascularity and malignancy of human astrocytomas. Anal Quant Cytol Histol. 2000;22:267-274.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Kazemi S, Wenzel D, Kolossov E, Lenka N, Raible A, Sasse P, Hescheler J, Addicks K, Fleischmann BK, Bloch W. Differential role of bFGF and VEGF for vasculogenesis. Cell Physiol Biochem. 2002;12:55-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
86.  Fu XB, Yang YH, Sun TZ, Chen W, Li JY, Sheng ZY. Rapid mitogen-activated protein kinase by basic fibroblast growth factor in rat intestine after ischemia/reperfusion injury. World J Gastroenterol. 2003;9:1312-1317.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Cross MJ, Lu L, Magnusson P, Nyqvist D, Holmqvist K, Welsh M, Claesson-Welsh L. The Shb adaptor protein binds to tyrosine 766 in the FGFR-1 and regulates the Ras/MEK/MAPK pathway via FRS2 phosphorylation in endothelial cells. Mol Biol Cell. 2002;13:2881-2893.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 72]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
88.  Boilly B, Vercoutter-Edouart AS, Hondermarck H, Nurcombe V, Le Bourhis X. FGF signals for cell proliferation and migration through different pathways. Cytokine Growth Factor Rev. 2000;11:295-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 202]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
89.  Kim HS, Crow TJ. Human proto-oncogene Int-2/FGF-3. Map position 11q13.3-q13.4. Chromosome Res. 1998;6:579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
90.  Mosher DF. Physiology of thrombospondin. Annu Rev Med. 1990;41:85-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 151]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
91.  Frazier WA. Thrombospondins. Curr Opin Cell Biol. 1991;3:792-799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 164]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
92.  Bornstein P. Diversity of function is inherent in matricellular proteins: an appraisal of thrombospondin 1. J Cell Biol. 1995;130:503-506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 521]  [Cited by in F6Publishing: 520]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
93.  Hogg PJ, Hotchkiss KA, Jiménez BM, Stathakis P, Chesterman CN. Interaction of platelet-derived growth factor with thrombospondin 1. Biochem J. 1997;326:709-716.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Lawler J. Thrombospondin-1 as an endogenous inhibitor of angiogenesis and tumor growth. J Cell Mol Med. 2002;6:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 411]  [Cited by in F6Publishing: 404]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
95.  DiPietro LA. Thrombospondin as a regulator of angiogenesis. EXS. 1997;79:295-314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 16]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
96.  de Fraipont F, Nicholson AC, Feige JJ, Van Meir EG. Thrombospondins and tumor angiogenesis. Trends Mol Med. 2001;7:401-407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 121]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
97.  Bornstein P, Armstrong LC, Hankenson KD, Kyriakides TR, Yang Z. Thrombospondin 2, a matricellular protein with diverse functions. Matrix Biol. 2000;19:557-568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 132]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
98.  Tobita K, Kijima H, Dowaki S, Oida Y, Kashiwagi H, Ishii M, Sugio Y, Sekka T, Ohtani Y, Tanaka M. Thrombospondin-1 expression as a prognostic predictor of pancreatic ductal carcinoma. Int J Oncol. 2002;21:1189-1195.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Goddard JC, Sutton CD, Jones JL, O'Byrne KJ, Kockelbergh RC. Reduced thrombospondin-1 at presentation predicts disease progression in superficial bladder cancer. Eur Urol. 2002;42:464-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 17]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
100.  Mascaux C, Martin B, Paesmans M, Verdebout JM, Verhest A, Vermylen P, Bosschaerts T, Ninane V, Sculier JP. Expression of thrombospondin in non-small cell lung cancer. Anticancer Res. 2002;22:1273-1277.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Yamaguchi M, Sugio K, Ondo K, Yano T, Sugimachi K. Reduced expression of thrombospondin-1 correlates with a poor prognosis in patients with non-small cell lung cancer. Lung Cancer. 2002;36:143-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
102.  Urquidi V, Sloan D, Kawai K, Agarwal D, Woodman AC, Tarin D, Goodison S. Contrasting expression of thrombospondin-1 and osteopontin correlates with absence or presence of metastatic phenotype in an isogenic model of spontaneous human breast cancer metastasis. Clin Cancer Res. 2002;8:61-74.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Kodama J, Hashimoto I, Seki N, Hongo A, Yoshinouchi M, Okuda H, Kudo T. Thrombospondin-1 and -2 messenger RNA expression in invasive cervical cancer: correlation with angiogenesis and prognosis. Clin Cancer Res. 2001;7:2826-2831.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Maeda K, Nishiguchi Y, Kang SM, Yashiro M, Onoda N, Sawada T, Ishikawa T, Hirakawa K. Expression of thrombospondin-1 inversely correlated with tumor vascularity and hematogenous metastasis in colon cancer. Oncol Rep. 2001;8:763-766.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Phelan MW, Forman LW, Perrine SP, Faller DV. Hypoxia increases thrombospondin-1 transcript and protein in cultured endothelial cells. J Lab Clin Med. 1998;132:519-529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 81]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
106.  Laderoute KR, Alarcon RM, Brody MD, Calaoagan JM, Chen EY, Knapp AM, Yun Z, Denko NC, Giaccia AJ. Opposing effects of hypoxia on expression of the angiogenic inhibitor thrombospondin 1 and the angiogenic inducer vascular endothelial growth factor. Clin Cancer Res. 2000;6:2941-2950.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Dameron KM, Volpert OV, Tainsky MA, Bouck N. Control of angiogenesis in fibroblasts by p53 regulation of thrombospondin-1. Science. 1994;265:1582-1584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1008]  [Cited by in F6Publishing: 1073]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
108.  Dameron KM, Volpert OV, Tainsky MA, Bouck N. The p53 tumor suppressor gene inhibits angiogenesis by stimulating the production of thrombospondin. Cold Spring Harb Symp Quant Biol. 1994;59:483-489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 87]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
109.  Nishimori H, Shiratsuchi T, Urano T, Kimura Y, Kiyono K, Tatsumi K, Yoshida S, Ono M, Kuwano M, Nakamura Y. A novel brain-specific p53-target gene, BAI1, containing thrombospondin type 1 repeats inhibits experimental angiogenesis. Oncogene. 1997;15:2145-2150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 227]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
110.  Mettouchi A, Cabon F, Montreau N, Vernier P, Mercier G, Blangy D, Tricoire H, Vigier P, Binétruy B. SPARC and thrombospondin genes are repressed by the c-jun oncogene in rat embryo fibroblasts. EMBO J. 1994;13:5668-5678.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Prehn RT. The inhibition of tumor growth by tumor mass. Cancer Res. 1991;51:2-4.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Prehn RT. Two competing influences that may explain concomitant tumor resistance. Cancer Res. 1993;53:3266-3269.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  O'Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS, Flynn E, Birkhead JR, Olsen BR, Folkman J. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell. 1997;88:277-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3249]  [Cited by in F6Publishing: 3052]  [Article Influence: 113.0]  [Reference Citation Analysis (0)]
114.  Li X, Fu GF, Fan YR, Shi CF, Liu XJ, Xu GX, Wang JJ. Potent inhibition of angiogenesis and liver tumor growth by administration of an aerosol containing a transferrin-liposome-endostatin complex. World J Gastroenterol. 2003;9:262-266.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Matsuno H, Yudoh K, Uzuki M, Nakazawa F, Sawai T, Yamaguchi N, Olsen BR, Kimura T. Treatment with the angiogenesis inhibitor endostatin: a novel therapy in rheumatoid arthritis. J Rheumatol. 2002;29:890-895.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Ergün S, Kilic N, Wurmbach JH, Ebrahimnejad A, Fernando M, Sevinc S, Kilic E, Chalajour F, Fiedler W, Lauke H. Endostatin inhibits angiogenesis by stabilization of newly formed endothelial tubes. Angiogenesis. 2001;4:193-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
117.  Jiang L, Jha V, Dhanabal M, Sukhatme VP, Alper SL. Intracellular Ca(2+) signaling in endothelial cells by the angiogenesis in-hibitors endostatin and angiostatin. Am J Physiol Cell Physiol. 2001;280:C1140-1150121.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Hanai J, Gloy J, Karumanchi SA, Kale S, Tang J, Hu G, Chan B, Ramchandran R, Jha V, Sukhatme VP. Endostatin is a potential inhibitor of Wnt signaling. J Cell Biol. 2002;158:529-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 116]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
119.  O'Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Moses M, Lane WS, Cao Y, Sage EH, Folkman J. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell. 1994;79:315-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2364]  [Cited by in F6Publishing: 2243]  [Article Influence: 74.8]  [Reference Citation Analysis (0)]
120.  Gyorffy S, Palmer K, Podor TJ, Hitt M, Gauldie J. Combined treatment of a murine breast cancer model with type 5 adenovirus vectors expressing murine angiostatin and IL-12: a role for combined anti-angiogenesis and immunotherapy. J Immunol. 2001;166:6212-6217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
121.  Benelli R, Morini M, Carrozzino F, Ferrari N, Minghelli S, Santi L, Cassatella M, Noonan DM, Albini A. Neutrophils as a key cellular target for angiostatin: implications for regulation of angiogenesis and inflammation. FASEB J. 2002;16:267-269.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Wajih N, Sane DC. Angiostatin selectively inhibits signaling by hepatocyte growth factor in endothelial and smooth muscle cells. Blood. 2003;101:1857-1863.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Mauceri HJ, Seetharam S, Beckett MA, Lee JY, Gupta VK, Gately S, Stack MS, Brown CK, Swedberg K, Kufe DW. Tumor production of angiostatin is enhanced after exposure to TNF-alpha. Int J Cancer. 2002;97:410-415.  [PubMed]  [DOI]  [Cited in This Article: ]