scispace - formally typeset
Search or ask a question

Showing papers by "Kari Alitalo published in 2003"


Journal ArticleDOI
TL;DR: It is shown here that VEGF-A controls angiogenic sprouting in the early postnatal retina by guiding filopodial extension from specialized endothelial cells situated at the tips of the vascular sprouts.
Abstract: Vascular endothelial growth factor (VEGF-A) is a major regulator of blood vessel formation and function. It controls several processes in endothelial cells, such as proliferation, survival, and migration, but it is not known how these are coordinately regulated to result in more complex morphogenetic events, such as tubular sprouting, fusion, and network formation. We show here that VEGF-A controls angiogenic sprouting in the early postnatal retina by guiding filopodial extension from specialized endothelial cells situated at the tips of the vascular sprouts. The tip cells respond to VEGF-A only by guided migration; the proliferative response to VEGF-A occurs in the sprout stalks. These two cellular responses are both mediated by agonistic activity of VEGF-A on VEGF receptor 2. Whereas tip cell migration depends on a gradient of VEGF-A, proliferation is regulated by its concentration. Thus, vessel patterning during retinal angiogenesis depends on the balance between two different qualities of the extracellular VEGF-A distribution, which regulate distinct cellular responses in defined populations of endothelial cells.

2,737 citations


Journal ArticleDOI
TL;DR: Therapeutic induction of vascular growth may provide a treatment option for those patients with myocardial or peripheral ischemia who are not suited to conventional revascularization therapies, and some lymphatic vascular disorders may also be amenable to this therapy.
Abstract: Therapeutic induction of vascular growth may provide a treatment option for those patients with myocardial or peripheral ischemia who are not suited to conventional revascularization therapies Some lymphatic vascular disorders may also be amenable to this therapy However, clear evidence of efficacy must be obtained from phase 2 and 3 clinical trials before these new treatments can be entered into clinical practice Apart from the clinical applications, gene transfer aimed at stimulating or blocking vascular growth with various growth factors, cytokines, transcription factors and receptors or their antagonists is useful for analyzing the effects of those molecules on the vasculature, especially when gene targeting results in lethality or when large animal models are required

429 citations


Journal ArticleDOI
TL;DR: This study compares, for the first time, angiogenesis and lymphangiogenesis induced by gene transfer of different human VEGFs, and shows that VEGF-D is the most potent member when delivered via an adenoviral vector into skeletal muscle.
Abstract: Optimal angiogenic and lymphangiogenic gene therapy requires knowledge of the best growth factors for each purpose. We studied the therapeutic potential of human vascular endothelial growth factor (VEGF) family members VEGF-A, VEGF-B, VEGF-C, and VEGF-D as well as a VEGFR-3–specific mutant (VEGF-C 156S ) using adenoviral gene transfer in rabbit hindlimb skeletal muscle. The significance of proteolytic processing of VEGF-D was explored using adenoviruses encoding either full-length or mature (ΔNΔC) VEGF-D. Adenoviruses expressing potent VEGFR-2 ligands, VEGF-A and VEGF-D ΔNΔC , induced the strongest angiogenesis and vascular permeability effects as assessed by capillary vessel and perfusion measurements, modified Miles assay, and MRI. The most significant feature of angiogenesis induced by both VEGF-A and VEGF-D ΔNΔC was a remarkable enlargement of microvessels with efficient recruitment of pericytes suggesting formation of arterioles or venules. VEGF-A also moderately increased capillary density and created glomeruloid bodies, clusters of tortuous vessels, whereas VEGF-D ΔNΔC –induced angiogenesis was more diffuse. Vascular smooth muscle cell proliferation occurred in regions with increased plasma protein extravasation, indicating that arteriogenesis may be promoted by VEGF-A and VEGF-D ΔNΔC . Full-length VEGF-C and VEGF-D induced predominantly and the selective VEGFR-3 ligand VEGF-C 156S exclusively lymphangiogenesis. Unlike angiogenesis, lymphangiogenesis was not dependent on nitric oxide. The VEGFR-1 ligand VEGF-B did not promote either angiogenesis or lymphangiogenesis. Finally, we found a positive correlation between capillary size and vascular permeability. This study compares, for the first time, angiogenesis and lymphangiogenesis induced by gene transfer of different human VEGFs, and shows that VEGF-D is the most potent member when delivered via an adenoviral vector into skeletal muscle.

424 citations


Journal ArticleDOI
01 Jan 2003-Blood
TL;DR: It is demonstrated that human CD34(+)CD133(+) cells expressing VEGFR-3 constitute a phenotypically and functionally distinct population of endothelial stem and precursor cells that may play a role in postnatal lymphangiogenesis and/or angiogenesis.

405 citations


Journal ArticleDOI
TL;DR: The data suggest that the carboxyl-terminal tail of VEGFR-3 provides important regulatory tyrosine phosphorylation sites with potential signal transduction capacity and that these sites are differentially used in ligand-induced homo- and heterodimeric receptor complexes.

280 citations


Journal ArticleDOI
TL;DR: It is reported that the serine protease plasmin cleaved both propeptides from the V EGF homology domain of human VEGF-D and thereby generated a mature form exhibiting greatly enhanced binding and cross-linking of VEGFR-2 and V EGFR-3 in comparison to full-length material.
Abstract: Vascular endothelial growth factor (VEGF) C and VEGF-D stimulate lymphangiogenesis and angiogenesis in tissues and tumors by activating the endothelial cell surface receptor tyrosine kinases VEGF receptor (VEGFR) 2 and VEGFR-3. These growth factors are secreted as full-length inactive forms consisting of NH2- and COOH-terminal propeptides and a central VEGF homology domain (VHD) containing receptor binding sites. Proteolytic cleavage removes the propeptides to generate mature forms, consisting of dimers of the VEGF homology domain, that bind receptors with much greater affinity than the full-length forms. Therefore, proteolytic processing activates VEGF-C and VEGF-D, although the proteases involved were unknown. Here, we report that the serine protease plasmin cleaved both propeptides from the VEGF homology domain of human VEGF-D and thereby generated a mature form exhibiting greatly enhanced binding and cross-linking of VEGFR-2 and VEGFR-3 in comparison to full-length material. Plasmin also activated VEGF-C. As lymphangiogenic growth factors promote the metastatic spread of cancer via the lymphatics, the proteolytic activation of these molecules represents a potential target for antimetastatic agents. Identification of an enzyme that activates the lymphangiogenic growth factors will facilitate development of inhibitors of metastasis.

202 citations


Journal ArticleDOI
TL;DR: It is suggested that Ang2 plays a critical role in inducing tumor cell infiltration, and that this invasive phenotype is caused by activation of MMP-2.
Abstract: A hallmark of highly malignant human gliomas is their infiltration of the brain. We analyzed a large number of primary human glioma biopsies and found high levels of expression of an angiogenic regulator, angiopoietin-2 (Ang2), in the invasive areas, but not in the central regions, of those tumors. In the invasive regions where Ang2 was overexpressed, increased levels of matrix metalloprotease-2 (MMP-2) were also apparent. Consonant with these features, intracranial xenografts of glioma cells engineered to express Ang2 were highly invasive into adjacent brain parenchyma compared with isogenic control tumors. In regions of the Ang2-expressing tumors that were actively invading the brain, high levels of expression of MMP-2 and increased angiogenesis were also evident. A link between these two features was apparent, because stable expression of Ang2 by U87MG cells or treatment of several glioma cell lines with recombinant Ang2 in vitro caused activation of MMP-2 and acquisition of increased invasiveness. Conversely, MMP inhibitors suppressed Ang2-stimulated activation of MMP-2 and Ang2-induced cell invasion. These results suggest that Ang2 plays a critical role in inducing tumor cell infiltration, and that this invasive phenotype is caused by activation of MMP-2.

158 citations


Journal ArticleDOI
TL;DR: It is demonstrated that TNF induces transactivation between Etk and V EGFR2, and Etk directly activates PI3K-Akt angiogenic signaling independent of VEGF-induced VEGFR2-PI3K -Akt signaling pathway.

145 citations


Journal ArticleDOI
TL;DR: Observations that endostatin inhibits endothelial cell migration and induces disassembly of the actin cytoskeleton provide putative cellular mechanisms for this effect, which plausibly explains the anti-angiogenic mechanisms ofendostatin in vivo.

139 citations


Journal ArticleDOI
TL;DR: Recent results show that the homeodomain transcription factor Prox-1 is an important lymphatic endothelial cell (LEC) fate-determining factor which can induce LEC-specific gene transcription even in blood vascular endothelial cells (BECs), which suggests that the distinct phenotypes of cells in the adult vascular endothelium are plastic and sensitive to transcriptional reprogramming, which might be useful for future therapeutic applications involving endothelial Cells.
Abstract: The lymphatic vasculature is essential for the maintenance of normal fluid balance and for the immune responses, but it is also involved in a variety of diseases. Hypoplasia or dysfuction of the lymphatic vessels can lead to lymphedema, whereas hyperplasia or abnormal growth of these vessels are associated with lymphangiomas and lymphangiosarcomas. Lymphatic vessels are also involved in lymph node and systemic metastasis of cancer cells. Recent novel findings on the molecular mechanisms involved in lymphatic vessel development and regulation allow the modulation of the lymphangiogenic process and specific targeting of the lymphatic endothelium. Recent results show that the homeodomain transcription factor Prox-1 is an important lymphatic endothelial cell (LEC) fate-determining factor which can induce LEC-specific gene transcription even in blood vascular endothelial cells (BECs). This suggests that the distinct phenotypes of cells in the adult vascular endothelium are plastic and sensitive to transcriptional reprogramming, which might be useful for future therapeutic applications involving endothelial cells Vascular endothelial growth factor-C (VEGF-C) and VEGF-D are peptide growth factors capable of inducing the growth of new lymphatic vessels in vivo in a process called lymphangiogenesis. They belong to the larger family which also includes VEGF, placenta growth factor (PlGF) and VEGF-B. VEGF-C and VEGF-D are ligands for the endothelial cell specific tyrosine kinase receptors VEGFR-2 and VEGFR-3. In adult human as well as mouse tissues VEGFR-3 is expressed predominantly in lymphatic endothelial cells which line the inner surface of lymphatic vessels. While VEGFR-2 is thought to be the main mediator of angiogenesis, VEGFR-3 signaling is crucial for the development of the lymphatic vessels. Heterozygous inactivation of the VEGFR-3 tyrosine kinase leads to primary lymphedema due to defective lymphatic drainage in the limbs. Other factors that seem to be involved in lymphangiogenesis include the Tie/angiopoietin system, neuropilin-2 and integrin α9. VEGF-C induces lymphatic vessel growth, but high levels of VEGF-C also resulted in blood vessel leakiness and growth. The VEGFR-3-specific mutant form of VEGF-C called VEGF-C156S lacks blood vascular side effects but is sufficient for therapeutic lymphangiogenesis in a mouse model of lymphedema. As VEGF-C156S is a specific lymphatic endothelial growth factor in the skin, it provides an attractive molecule for pro-lymphangiogenic therapy. This publication was partially financed by Serono. Part of this paper was originally presented at the 2nd International Workshop on New Therapeutic Targets in Vascular Biology, which took place in Geneva, Switzerland from February 6-9, 2003.

134 citations


Journal ArticleDOI
TL;DR: An important role for HB‐EGF and ErbB receptors in the recruitment of SMCs by ECs is suggested and the mechanism by which angiopoietins exert their vascular effects is elaborate.
Abstract: Recruitment of vascular smooth muscle cells (SMC) by endothelial cells (EC) is essential for angiogenesis. Endothelial-derived heparin binding EGF-like growth factor (HB-EGF) was shown to mediate this process by signaling via ErbB1 and ErbB2 receptors in SMCs. 1) Analysis of ErbB-ligands demonstrated that primary ECs expressed only HB-EGF and neuregulin-1. 2) Primary SMCs expressed ErbB1 and ErbB2, but not ErbB3 or ErbB4. 3) Consistent with their known receptor specificities, recombinant HB-EGF, but not neuregulin-1, stimulated tyrosine phosphorylation of ErbB1 and ErbB2 and migration in SMCs. 4) Neutralization of HB-EGF or inhibition of ErbB1 or ErbB2 blocked 70-90% of the potential of ECs to stimulate SMC migration. Moreover, 5) angiopoietin-1, an EC effector with a role in recruitment of SMC-like cells to vascular structures in vivo, enhanced EC-stimulated SMC migration by a mechanism involving up-regulation of endothelial HB-EGF. Finally, 6) immunohistochemical analysis of developing human tissues demonstrated that HB-EGF was expressed in vivo in ECs associated with SMCs or pericytes but not in ECs of the hyaloid vessels not associated with SMCs. These results suggest an important role for HB-EGF and ErbB receptors in the recruitment of SMCs by ECs and elaborate on the mechanism by which angiopoietins exert their vascular effects.

Journal ArticleDOI
TL;DR: The findings suggest that the antiinflammatory properties of Ang1 may offer an entirely new therapeutic approach to prevent cardiac allograft arteriosclerosis.
Abstract: Background— Angiopoietin (Ang)–1 is an angiogenic growth factor that counteracts the permeability and proinflammatory effects of vascular endothelial growth factor and other proinflammatory cytokines. Recently, we demonstrated that vascular endothelial growth factor enhances cardiac allograft arteriosclerosis. Here, we studied the roles of Ang1, its natural antagonist Ang2, and their receptor Tie2 in rat cardiac allograft arteriosclerosis. Methods and Results— Heterotopic cardiac allografts and syngrafts were transplanted from Dark Agouti (DA) to Wistar-Furth rats and from DA to DA rats, respectively. Immunohistochemistry disclosed that only a few mesenchymal cells expressed Ang1 in normal hearts and syngrafts, whereas no immunoreactivity was found in cardiac allografts undergoing chronic rejection. Ang2 and Tie2 immunoreactivity was induced mainly in capillaries and postcapillary venules in chronic allografts when compared with syngeneic controls, but no immunoreactivity was found in arterial endothelium...

Journal ArticleDOI
TL;DR: An overview of the structure, function, and development of the lymphatics is given, with special emphasis on the recently discovered lymphangiogenic growth factors.
Abstract: The lymphatic system is generally regarded as supplementary to the blood vascular system, in that it transports interstitial fluid, macromolecules, and immune cells back into the blood. However, in insects, the open hemolymphatic (or lymphohematic) system ensures the circulation of immune cells and interstitial fluid through the body. The Drosophila homolog of the mammalian vascular endothelial growth factor receptor (VEGFR) gene family is expressed in hemocytes, suggesting a close relationship to the endothelium that develops later in phylogeny. Lymph hearts are typical organs for the propulsion of lymph in lower vertebrates and are still transiently present in birds. The lymphatic endothelial marker VEGFR-3 is transiently expressed in embryonic blood vessels and is crucial for their development. We therefore regard the question of whether the blood vascular system or the lymphatic system is primary or secondary as open. Future molecular comparisons should be performed without any bias based on the current prevalence of the blood vascular system over the lymphatic system. Here, we give an overview of the structure, function, and development of the lymphatics, with special emphasis on the recently discovered lymphangiogenic growth factors.

Journal ArticleDOI
TL;DR: The results suggest that in large arteries VEGF-D is mainly expressed in smooth muscle cells and that it may have a role in the maintenance of vascular homeostasis and in complicated lesions it was also expressed in macrophages and may contribute to plaque neovascularization.
Abstract: Objective: Vascular endothelial growth factor-D (VEGF-D) is a recently characterized member of the VEGF family, but its expression in atherosclerotic lesions remains unknown. We studied the expression of VEGF-D and its receptors (VEGFR-2 and VEGFR-3) in normal and atherosclerotic human arteries, and compared that to the expression pattern of VEGF-A. Methods: Human arterial samples ( n =39) obtained from amputation operations and fast autopsies were classified according to the stage of atherosclerosis and studied by immunohistochemistry. The results were confirmed by in situ hybridization and RT-PCR. Results: We found that while VEGF-A expression increased during atherogenesis, VEGF-D expression remained relatively stable only decreasing in complicated lesions. In normal arteries and in early lesions VEGF-D was mainly expressed in smooth muscle cells, whereas in complicated atherosclerotic lesions the expression was most prominent in macrophages and also colocalized with plaque neovascularization. By comparing the staining profiles of different antibodies, we found that proteolytic processing of VEGF-D was efficient in the vessel wall. VEGFR-2, but not VEGFR-3, was expressed in the vessel wall at every stage of atherosclerosis. Conclusions: Our results suggest that in large arteries VEGF-D is mainly expressed in smooth muscle cells and that it may have a role in the maintenance of vascular homeostasis. However, in complicated lesions it was also expressed in macrophages and may contribute to plaque neovascularization. The constitutive expression of VEGFR-2 in arteries suggests that it may be one of the principal mediators of the VEGF-D effects in large arteries.

Journal ArticleDOI
TL;DR: The results help to define possibilities for local angiogenic therapy around blood vessels and support the concept thatAngiogenic effects may be tissue-specific and depend both on the growth factor ligands and the target tissues.
Abstract: Recent discovery of new members of the vascular endothelial growth factor (VEGF) family has generated much interest as to which members may be best suited for therapeutic angiogenesis in various tissues. In this study we evaluated angiogenic responses of the different members of the VEGF family in vivo using adenoviral gene transfer. Adenoviruses (1 × 109 plaque-forming units [pfu]) encoding for VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-CΔNΔC and VEGF-DΔNΔC (ΔNΔC are proteolytically cleaved forms) were transferred locally to the periadventitial space of the rabbit carotid arteries using a collar technique that allows efficient local transfection of the periadventitial tissue. Expression of the transfected VEGFs was confirmed by immunohistochemistry and reverse transcription-polymerase chain reaction (RT-PCR). Seven days after the gene transfer maximum neovessel formation was observed in VEGF-A-, VEGF-D-, and VEGF-DΔNΔC-transfected arteries. VEGF-CΔNΔC also showed angiogenic activity whereas VEGF-B was not effe...

Journal ArticleDOI
TL;DR: In this article, the responses of lymphatic endothelial cells (LECs) and BECs to vascular endothelial growth factors (VEGFs) were compared to gain further insight into the mechanisms regulating the endothelial cell phenotype.
Abstract: Vascular endothelial cells are characterized by a high degree of functional and phenotypic plasticity, which is controlled both by their pericellular microenvironment and their intracellular gene expression programs. To gain further insight into the mechanisms regulating the endothelial cell phenotype, we have compared the responses of lymphatic endothelial cells (LECs) and blood vascular endothelial cells (BECs) to vascular endothelial growth factors (VEGFs). VEGFR-3-specific signals are sufficient for LEC but not BEC proliferation, as shown by the ability of the specific ligand VEGF-C156S to stimulate cell cycle entry only in LECs. On the other hand, we found that VEGFR-3 stimulation did not induce LEC cell shape changes typical of VEGFR-2-stimulated LECs, indicating receptor-specific differences in the cytoskeletal responses. Genes induced via VEGFR-2 also differed between BECs and LECs: angiopoietin-2 (Ang-2) was induced via VEGFR-2 in BECs and LECs, but the smooth muscle cell (SMC) chemoattractant BMP-2 was induced only in BECs. Both BECs and LECs were able to promote SMC chemotaxis, but contact with SMCs led to down-regulation of VEGFR-3 expression in BECs in a 3-dimensional coculture system. This was consistent with the finding that VEGFR-3 is down-regulated in vivo at sites of endothelial cell-pericyte/smooth muscle cell contacts. Collectively, these data show intrinsic cell-specific differences of BEC and LEC responses to VEGFs and identify a pericellular regulatory mechanism for VEGFR-3 down-regulation in endothelial cells.

Journal ArticleDOI
TL;DR: These studies suggest that Bmx-Cas interaction, phosphorylation of Cas by Bmx, and subsequent Cas·Crk complex formation functionally couple Bmx to the regulation of actin cytoskeleton and cell motility.

Patent
07 Mar 2003
TL;DR: In this paper, the authors provide polynucleotides and genes that are differentially expressed in lymphatic versus blood vascular endothelial cells for treating diseases involving lymphatic vessels, such as lymphedema, various inflammatory diseases, and cancer metastasis via the lymphatic system.
Abstract: The invention provides polynucleotides and genes that are differentially expressed in lymphatic versus blood vascular endothelial cells. These genes are useful for treating diseases involving lymphatic vessels, such as lymphedema, various inflammatory diseases, and cancer metastasis via the lymphatic system.


Journal ArticleDOI
TL;DR: The new findings presented in this issue of the JCI by Bergers and collaborators suggest that the PCs of tumor blood vessels and their signaling mechanisms via the PDGFR-β are functionally important for the maintenance of tumorBlood vessels.
Abstract: As the growth of most cancers is dependent on the growth of tumor blood vessels, inhibition of tumor angiogenesis may provide an efficient strategy to slow down or block tumor growth. The possibility of selectively targeting angiogenic vasculature in a tumor mass depends on molecular, cellular, and structural differences between the tumor vessels and their normal counterparts (Figure ​(Figure1).1). Tumor cells, like normal cells, need to be located close to the blood vessels serving their metabolic demands to the extent that in solid tumors every endothelial cell in a tumor vessel is considered to support several concentric layers of tumor cells (1). A hypoxic tumor generates its own microcirculation mainly via the hypoxia-inducible factor (HIF) complex, which is activated by inhibition of its oxygen-dependent prolyl hydroxylation and proteasomal destruction (2). This leads to increased transcription of several hypoxia-induced genes. One such gene encodes VEGF, which activates endothelial cell (EC) responses during angiogenesis in coordination with, for example, matrix adhesion events. Studies of the signaling pathways of VEGF receptors, integrins, and cadherins have provided new antiangiogenic strategies for inhibition of tumor growth, and inhibition of VEGF-C and VEGF-D signals that stimulate lymphangiogenesis seems to inhibit lymphatic metastasis in mice (3). One of the most striking new developments in antiangiogenesis research concerns the inflammatory cells and pericytes (PCs) associated with the tumor vasculature. The new findings presented in this issue of the JCI by Bergers and collaborators suggest that the PCs of tumor blood vessels and their signaling mechanisms via the PDGFR-β are functionally important for the maintenance of tumor blood vessels (4). These findings add another constituent cell type of tumor stroma to the list of anticancer targets. Figure 1 Growth factor receptor signals involved in tumor angiogenesis and vascular development. A solid tumor is dependent on new blood vessels for growth. (a) Angiogenesis is induced by VEGF produced mainly by hypoxic tumor cells and inflammatory cells (such ...

Journal ArticleDOI
TL;DR: Findings point to the central role of VEGFR-2 in the angiogenic signaling pathways induced by VEGF-C(deltaNdeltaC) and V EGF-A in the plasminogen activator (PA)-plasmin system.

Journal ArticleDOI
TL;DR: The results suggest that c-Myc and pRb can interact in vivo, and may in part control some aspects of cell proliferation and transformation through modulation of odc expression.

Journal ArticleDOI
TL;DR: This issue of Cancer Cell uses gene-targeted mice to show a physiological role for a carboxy-terminal fragment of collagen IV in the regulation of tumor angiogenesis.


Book ChapterDOI
01 Jan 2003
TL;DR: The lymphatic endothelial cell signaling is also of interest in cancer biology because of the metastatic spread of tumor cells via the lymphatic system and tumor lymphangiogenesis.
Abstract: During embryogenesis, blood and lymphatic vasculature develops through the regulated proliferation, migration, and differentiation of endothelial cells. Initially, a primary blood vascular plexus is formed in the process of vasculogenesis, where mesoderm-derived angioblasts differentiate into endothelial cells and assemble to form a network of uniformly sized primitive vessels. The lymphatic vessels arise from the embryonic veins through a process termed lymphangiogenesis. The lymph vessels become organized parallel to the blood vascular system and function to transport interstitial fluid, extravasated plasma proteins, and cells back into the blood circulation. The lymphatic capillaries transport the fluid into progressively larger collecting vessels consisting of endothelial, muscular, and adventitial layers, which ultimately drain into the venous circulation via the thoracic duct. Movement of lymph is brought about by the intrinsic contractility of the smooth muscle cells surrounding the larger vessels, and backflow is prevented by luminal valves. Lymphedema, the accumulation of protein-rich fluid in interstitial tissues, occurs in humans either as hereditary disease or due to lymph vessel damage or removal. Because of the metastatic spread of tumor cells via the lymphatic system and tumor lymphangiogenesis, the lymphatic endothelial cell signaling is also of interest in cancer biology.