1101
|
Voelkel NF, Cool C, Lee SD, Wright L, Geraci MW, Tuder RM. Primary pulmonary hypertension between inflammation and cancer. Chest 1998; 114:225S-230S. [PMID: 9741573 DOI: 10.1378/chest.114.3_supplement.225s] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
We believe that the monoclonal cell expansion in primary pulmonary hypertension is the result of autonomous growth of stem cell-like endothelial cells, whereas the polyclonal proliferation in secondary pulmonary hypertension occurs as a response of endothelial cells to exogenous stimuli (like viral infection or high shear stress). In this context, we propose that different transcriptional and translational events govern the growth and expansion of monoclonal when compared with polyclonal pulmonary endothelial cells. The availability of antibodies directed against specific tyrosine kinase proteins involved in vasculogenesis/angiogenesis now permits the identification and localization of the components of such a misguided angiogenesis cell proliferation program in the pulmonary hypertensive vascular lesions.
Collapse
Affiliation(s)
- N F Voelkel
- Pulmonary Hypertension Center, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | | | | | | | |
Collapse
|
1102
|
Migdal M, Huppertz B, Tessler S, Comforti A, Shibuya M, Reich R, Baumann H, Neufeld G. Neuropilin-1 is a placenta growth factor-2 receptor. J Biol Chem 1998; 273:22272-8. [PMID: 9712843 DOI: 10.1074/jbc.273.35.22272] [Citation(s) in RCA: 235] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Placenta growth factor (PlGF) belongs to the family of vascular endothelial growth factors (VEGFs). It binds to the flt-1 VEGF receptor but not to the KDR/flk-1 receptor which is thought to mediate most of the angiogenic and proliferative effects of VEGF. Three PlGF isoforms are produced by alternative splicing. PlGF-1 and PlGF-3 differ from PlGF-2 since they lack the exon 6 encoded peptide which bestows upon PlGF-2 its heparin binding properties. Cross-linking experiments revealed that 125I-PlGF-2 binds to two endothelial cell surface receptors in a heparin dependent fashion. The binding of 125I-PlGF-2 to these receptors was inhibited by an excess of PlGF-2 and by the 165-amino acid form of VEGF (VEGF165), but not at all by VEGF121 and very marginally if at all by PlGF-1. The apparent molecular weight and the binding characteristics of these receptors correspond to those of the recently identified VEGF165 specific receptor neuropilin-1, and we therefore conclude that neuropilin-1 is a receptor for PlGF-2. The binding of 125I-PlGF-2 as well as the binding of 125I-VEGF165 to these receptors was inhibited by a synthetic peptide derived from exon 6 of PlGF. Furthermore, the binding of 125I-PlGF-2, but not that of 125I-VEGF165, was also inhibited by a synthetic peptide derived from exon 7 of PlGF. These observations indicate that the peptides encoded by these exons probably participate in the formation of the domain which mediates the binding of PlGF-2 to these receptors. We have also determined, using chemically modified heparin species, that the presence of sulfate moieties on the glucosamine-O-6 and on the iduronic acid-O-2 groups of heparin was required for the potentiation of 125I-PlGF-2 binding to these receptors. To determine if PlGF-2 is able to induce biological responses that are not induced by PlGF-1, we compared the effects of PlGF-1 and PlGF-2 on the migration and proliferation of endothelial cells. Both PlGF forms induced migration of endothelial cells. However, there was no quantitative difference between the response to PlGF-2 and the response to PlGF-1. Furthermore, neither PlGF-1 nor PlGF-2 had any effect upon the proliferation of the endothelial cells.
Collapse
Affiliation(s)
- M Migdal
- Department of Biology, Technion, Israel Institute of Technology, Haifa, 32000, Israel
| | | | | | | | | | | | | | | |
Collapse
|
1103
|
Giraudo E, Primo L, Audero E, Gerber HP, Koolwijk P, Soker S, Klagsbrun M, Ferrara N, Bussolino F. Tumor necrosis factor-alpha regulates expression of vascular endothelial growth factor receptor-2 and of its co-receptor neuropilin-1 in human vascular endothelial cells. J Biol Chem 1998; 273:22128-35. [PMID: 9705358 DOI: 10.1074/jbc.273.34.22128] [Citation(s) in RCA: 201] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-alpha) modulates gene expression in endothelial cells and is angiogenic in vivo. TNF-alpha does not activate in vitro migration and proliferation of endothelium, and its angiogenic activity is elicited by synthesis of direct angiogenic inducers or of proteases. Here, we show that TNF-alpha up-regulates in a dose- and time-dependent manner the expression and the function of vascular endothelial growth factor receptor-2 (VEGFR-2) as well as the expression of its co-receptor neuropilin-1 in human endothelium. As inferred by nuclear run-on assay and transient expression of VEGFR-2 promoter-based reporter gene construct, the cytokine increased the transcription of the VEGFR-2 gene. Mithramycin, an inhibitor of binding of nuclear transcription factor Sp1 to the promoter consensus sequence, blocked activation of VEGFR-2, suggesting that the up-regulation of the receptor required Sp1 binding sites. TNF-alpha increased the cellular amounts of VEGFR-2 protein and tripled the high affinity 125I-VEGF-A165 capacity without affecting the Kd of ligand-receptor interaction. As a consequence, TNF-alpha enhanced the migration and the wound healing triggered by VEGF-A165. Since VEGFR-2 mediates angiogenic signals in endothelium, our data indicate that its up-regulation is another mechanism by which TNF-alpha is angiogenic and may provide insight into the mechanism of neovascularization as occurs in TNF-alpha-mediated pathological settings.
Collapse
Affiliation(s)
- E Giraudo
- Vascular Biology Laboratory, Department of Genetics, Biology and Biochemistry, Medical School, University of Torino, Torino, 10126 Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
1104
|
Moriyama T, Kataoka H, Hamasuna R, Yokogami K, Uehara H, Kawano H, Goya T, Tsubouchi H, Koono M, Wakisaka S. Up-regulation of vascular endothelial growth factor induced by hepatocyte growth factor/scatter factor stimulation in human glioma cells. Biochem Biophys Res Commun 1998; 249:73-7. [PMID: 9705834 DOI: 10.1006/bbrc.1998.9078] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Recent findings suggest that hepatocyte growth factor/scatter factor (HGF/SF) contributes to the malignant progression of human gliomas. We investigated the effect of HGF/SF on vascular endothelial growth factor (VEGF) expression of c-Met/HGF receptor-positive human glioma cell lines. Treatment of the glioma cells with various concentrations of HGF/SF resulted in an enhanced secretion of VEGF proteins accompanying increased transcription of VEGF mRNA in a dose-dependent fashion. Since malignant gliomas frequently co-express HGF/SF and its receptor, these results suggest that HGF/SF could act as an indirect angiogenic factor through autocrine induction of VEGF expression and secretion in malignant gliomas.
Collapse
Affiliation(s)
- T Moriyama
- Department of Neurosurgery, Miyazaki Medical College, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1105
|
Maru Y, Yamaguchi S, Takahashi T, Ueno H, Shibuya M. Virally activated Ras cooperates with integrin to induce tubulogenesis in sinusoidal endothelial cell lines. J Cell Physiol 1998; 176:223-34. [PMID: 9648910 DOI: 10.1002/(sici)1097-4652(199808)176:2<223::aid-jcp1>3.0.co;2-q] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Four cell lines, named nonparenchymal 11 (NP11), NP26, NP31, and NP32, were established from sinusoidal endothelial cells (SECs) of rat liver. They still retained expression of receptors for vascular endothelial growth factor (VEGF), Fit-1, and kinase domain-containing receptor (KDR). NP31 and NP32 turned out to be incapable of tubulogenesis in basement membrane matrix (Matrigel), which belongs to endothelial properties, as shown by SECs in primary culture. Expression of temperature-sensitive, virally activated Ras (ts-v-Ras) restored tubulogenic behaviors back to NP31 only at permissive temperature. Matrigel induced long-lasting tyrosine phosphorylation of Shc, with recruitment of Grb-2 and microtubule-associated protein kinase (MAPK) activation in both parental NP31 and NP31 transformed by ts-v-Ras, which was blocked by anti-beta1 integrin antibody. Tubulogenesis was inhibited by adenovirus-mediated expression of dominant-negative Ras in human umbilical vein endothelial cells (HUVECs). PD 098059, a selective inhibitor of MAPK kinase (MEK), nearly perfectly blocked tubulogenesis by ts-v-Ras-expressing NP31 cells at permissive temperature. Furthermore, the botulinum C3 toxin, an inhibitor for Rho, caused fragmentation of branching cords in networks formed by NP31 that expressed ts-v-Ras at permissive temperature. These data suggest that the integrin-mediated Ras signals may be necessary but are not sufficient for tubulogenesis and that an artificial expression of v-Ras might substitute for the second signal required in this system.
Collapse
MESH Headings
- ADP Ribose Transferases/pharmacology
- Adaptor Proteins, Signal Transducing
- Adaptor Proteins, Vesicular Transport
- Animals
- Botulinum Toxins
- Cadherins/analysis
- Calcium-Calmodulin-Dependent Protein Kinases/analysis
- Cell Adhesion Molecules/analysis
- Cell Line, Transformed/chemistry
- Cell Line, Transformed/drug effects
- Cell Line, Transformed/physiology
- Collagen/pharmacology
- Cytoskeletal Proteins/analysis
- Desmoplakins
- Drug Combinations
- Endothelium/chemistry
- Endothelium/cytology
- Endothelium/enzymology
- Enzyme Inhibitors/pharmacology
- Extracellular Matrix/chemistry
- Extracellular Matrix/enzymology
- Flavonoids/pharmacology
- Gene Expression Regulation, Viral
- Humans
- Integrins/physiology
- Laminin/pharmacology
- Liver/blood supply
- Liver/cytology
- Liver/physiology
- Neovascularization, Physiologic/physiology
- Proteins/physiology
- Proteoglycans/pharmacology
- Rats
- Receptor Protein-Tyrosine Kinases/genetics
- Receptors, Growth Factor/genetics
- Receptors, Mitogen/genetics
- Receptors, Vascular Endothelial Growth Factor
- Retroviridae/genetics
- Shc Signaling Adaptor Proteins
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Src Homology 2 Domain-Containing, Transforming Protein 1
- Trans-Activators
- Umbilical Veins/cytology
- alpha Catenin
- beta Catenin
- ras Proteins/genetics
- src Homology Domains/physiology
Collapse
Affiliation(s)
- Y Maru
- Department of Genetics, Institute of Medical Science, University of Tokyo, Japan.
| | | | | | | | | |
Collapse
|
1106
|
Ma J, Fei ZL, Klein-Szanto A, Gallo JM. Modulation of angiogenesis by human glioma xenograft models that differentially express vascular endothelial growth factor. Clin Exp Metastasis 1998; 16:559-68. [PMID: 9872603 DOI: 10.1023/a:1006598419541] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
To evaluate the potential actions of vascular endothelial growth factor (VEGF) on capillary permeability and drug transport, tumorigenic human glioma cell lines were developed that expressed different levels of VEGF. Three human glioma cell lines (i.e. U373, SF126, SF188) were screened for VEGF under normoxic and hypoxic (i.e. induced by CoCl2) conditions by Western blot analysis. Subsequent to these results, sense and antisense VEGF164 cDNA transfections were conducted. It was found that parental SF188 (SF188/V-) and SF188/V+ (sense transfected) cells could serve as an appropriate in vivo model based on their divergent levels of VEGF expression. Media derived from SF188/V+ cells stimulated endothelial cell growth by 30-60%, and enhanced endothelial cell clonogenicity by 5-10-fold compared to SF188/V- or empty vector transfected cells. Nude rats implanted with either SF188/V- or SF188/V+ cells subcutaneously and intracerebrally formed tumors, with those derived from SF188/V+ cells growing at a faster rate. Immunohistochemistry analysis indicated that the expression of VEGF and number of capillaries (factor VIII assay) were approximately 3-fold greater in SF188/V+ tumors compared to SF188/V- tumors. Pharmacological assays, such as measurements of cytotoxicity and DNA adducts, in SF188/V- and SF188/V+ cells treated with carmustine or temozolomide were similar. Therefore, other than differences in VEGF expression and growth in vivo, SF188/V- and SF188/V+ cells possess a similar phenotype, and can serve as models to evaluate the influence of VEGF on drug transport.
Collapse
Affiliation(s)
- J Ma
- Department Of Pharmacology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | |
Collapse
|
1107
|
Lymboussaki A, Partanen TA, Olofsson B, Thomas-Crusells J, Fletcher CD, de Waal RM, Kaipainen A, Alitalo K. Expression of the vascular endothelial growth factor C receptor VEGFR-3 in lymphatic endothelium of the skin and in vascular tumors. THE AMERICAN JOURNAL OF PATHOLOGY 1998; 153:395-403. [PMID: 9708800 PMCID: PMC1852985 DOI: 10.1016/s0002-9440(10)65583-6] [Citation(s) in RCA: 160] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
It is difficult to identify lymph vessels in tissue sections by histochemical staining, and thus a specific marker for lymphatic endothelial cells would be more practical in histopathological diagnostics. Here we have applied a specific antigenic marker for lymphatic endothelial cells in the human skin, the vascular endothelial growth factor receptor-3 (VEGFR-3), and show that it identifies a distinct vessel population both in fetal and adult skin, which has properties of lymphatic vessels. The expression of VEGFR-3 was studied in normal human skin by in situ hybridization, iodinated ligand binding, and immunohistochemistry. A subset of developing vessels expressed the VEGFR-3 mRNA in fetal skin as shown by in situ hybridization and radioiodinated vascular endothelial growth factor (VEGF)-C bound selectively to a subset of vessels in adult skin that had morphological characteristics of lymphatic vessels. Monoclonal antibodies against the extracellular domain of VEGFR-3 stained specifically endothelial cells of dermal lymph vessels, in contrast to PAL-E antibodies, which stained only blood vessel endothelia. In addition, staining for VEGFR-3 was strongly positive in the endothelium of cutaneous lymphangiomatosis, but staining of endothelial cells in cutaneous hemangiomas was weaker. These results establish the utility of anti-VEGFR-3 antibodies in the identification of lymphovascular channels in the skin and in the differential diagnosis of skin lesions involving lymphatic or blood vascular endothelium.
Collapse
Affiliation(s)
- A Lymboussaki
- Department of Pathology, Haartman Institute, University of Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
1108
|
Abstract
Vascular endothelial growth factor (VEGF) is a major inducer of angiogenesis and vasculogenesis. Two distinct receptors for VEGF, the tyrosine kinase receptors VEGFR-1 (Flt-1) and VEGFR-2 (Flk-1/KDR), have been identified. Transfection studies could demonstrate biological activities for the Flk-1/KDR-, but not for the Flt-1-receptor, which led to the hypothesis that Flt-1 is a decoy receptor for VEGF. However, Flt-1 is biologically active in non-endothelial cells, namely monocytes, which exclusively express this receptor. In addition, the Flt-1 ligand placenta growth factor (PlGF), which is unable to bind and activate Flk-1/KDR, elicits activities in both monocytes and endothelial cells. The implications of Flt-1 mediated monocyte transmigration through endothelial monolayers and induction of the procoagulant tissue factor on monocytes for the field of vascular medicine are discussed.
Collapse
Affiliation(s)
- M Clauss
- Physiology Clinic Forsch, Max-Planck Institute, Department of Molecular Biology, Parkstrasse 1, D-61231 WG Kerckhoff, Bad Nauheim, Germany
| |
Collapse
|
1109
|
Yang SP, Morita I, Murota SI. Eicosapentaenoic acid attenuates vascular endothelial growth factor-induced proliferation via inhibiting Flk-1 receptor expression in bovine carotid artery endothelial cells. J Cell Physiol 1998; 176:342-9. [PMID: 9648921 DOI: 10.1002/(sici)1097-4652(199808)176:2<342::aid-jcp12>3.0.co;2-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Eicosapentaenoic acid (EPA; 20:5, n-3) can restrain tumor growth and metastasis in vivo; however, the mechanism of its antitumor effect is still not fully understood. Angiogenesis is a crucial process for tumor growth and metastasis and inhibition of tumor angiogenesis can suppress tumor growth and metastasis in vivo. Vascular endothelial growth factor (VEGF) is an important angiogenic factor. In this study, we investigated the mechanisms of the inhibitory effect of EPA on VEGF-induced proliferation of bovine carotid artery endothelial (BAE) cells. BAE cells, treated with 0-5 microg/ml EPA for 48 h, displayed a dose-dependent suppression to VEGF (0.2 nM)-induced proliferation. Similar inhibitory effect was not found in BAE cells treated with arachidonic acid (AA; 20:4, n-6), or docasahexaenoic acid (DHA; 22:5, n-3). In contrast to its effect on VEGF-induced proliferation, EPA had no inhibition to basic fibroblast growth factor (bFGF, 0.2 nM)-induced proliferation in BAE cells. Both VEGF and bFGF activated mitogen-activated protein (MAP) kinase in BAE cells; however, EPA selectively inhibited VEGF-induced, but not bFGF-induced activation of MAP kinase. Flk-1 expression was inhibited dose-dependently in EPA-treated cells, whereas Flt-1 expression was increased in EPA treated cells. This in vitro inhibitory effect by EPA on Flk-1 receptor expression provides indirect evidence that one of the mechanisms of EPA for antitumor action in vivo maybe related to its antiangiogenic action.
Collapse
Affiliation(s)
- S P Yang
- Department of Physiological Chemistry, Postgraduate School, Tokyo Medical and Dental University, Japan
| | | | | |
Collapse
|
1110
|
Hata Y, Duh E, Zhang K, Robinson GS, Aiello LP. Transcription factors Sp1 and Sp3 alter vascular endothelial growth factor receptor expression through a novel recognition sequence. J Biol Chem 1998; 273:19294-303. [PMID: 9668119 DOI: 10.1074/jbc.273.30.19294] [Citation(s) in RCA: 104] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Kinase domain receptor (KDR) is a high affinity, endothelial cell-specific, autophosphorylating tyrosine kinase receptor for vascular endothelial growth factor. This transcriptionally regulated receptor is a critical mediator of endothelial cell (EC) growth and vascular development. In this study, we identify a DNA element modulating KDR promoter activity and evaluate the nuclear binding proteins accounting for a portion of the cell-type specificity of the region. KDR promoter luciferase activity was retained within -85/+296 and was 10-30-fold higher in EC than non-EC. Electrophoretic mobility shift assays demonstrated specific nuclear protein binding to -85/-64, and single point mutations suggested important binding nucleotides between -79/-68 with five critical bases between -74/-70 (5'-CTCCT-3'). DNA-protein complexes were displaced by Sp1 consensus sequence oligodeoxynucleotides and supershifted by Sp1- and Sp3-specific antibodies. Sp1 and Sp3 protein in EC nuclear extracts bound the -79/-68 region even when all surrounding classic Sp1 recognition sites were removed. Sp1 protein in nuclear extracts was 4-24-fold higher in EC than non-EC, whereas Sp3 was 3-7-fold higher. Sp1/Sp3 ratios in EC were 2-10-fold higher. Overexpression of Sp1 protein increased KDR promoter activity 3-fold in both EC and non-EC, whereas simultaneous co-expression of Sp3 attenuated this response. An Sp1 consensus sequence cis element "decoy" reduced EC KDR promoter activity and mRNA expression by 85 and 69%, respectively. An antisense phosphorothioate oligodeoxynucleotide to Sp1 inhibited Sp1 and KDR protein expression by 66 and 68%, respectively, without changing Sp3 protein expression. These data illustrate that Sp1 and Sp3 modulate KDR promoter activity through a novel recognition binding sequence. However, since Sp1-mediated promoter activation is attenuated by Sp3, endothelial selective KDR promoter activity may be partially regulated by variations in the Sp1/Sp3 ratio.
Collapse
Affiliation(s)
- Y Hata
- Research Division, Joslin Diabetes Center, Boston, Massachusetts 02215, USA
| | | | | | | | | |
Collapse
|
1111
|
|
1112
|
Yamaguchi R, Yano H, Iemura A, Ogasawara S, Haramaki M, Kojiro M. Expression of vascular endothelial growth factor in human hepatocellular carcinoma. Hepatology 1998; 28:68-77. [PMID: 9657098 DOI: 10.1002/hep.510280111] [Citation(s) in RCA: 270] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular endothelial growth factor (VEGF) is thought to take an important role in tumor angiogenesis. The present study examined VEGF expression immunohistochemically in hepatocellular carcinomas (HCCs) in various histological grades and sizes. In HCCs that were composed of cancerous tissues of single histological grade, VEGF expression was the highest in well-differentiated HCCs, followed by moderately differentiated HCCs, and then poorly differentiated HCCs. VEGF positivity gradually decreased with the increase in tumor size. In the nodules larger than 3.0 cm, 36.8% were VEGF-negative. In HCCs consisting of cancerous tissues of two different histological grades, the expression was less intensive in the higher-grade HCC component. VEGF was not expressed in sarcomatous areas, while VEGF was expressed in the surrounding HCC tissues. The expression was also remarkable in the noncancerous tissues in which inflammatory cell infiltration was apparent. VEGF expression was also examined in six HCC cell lines. In reverse-transcription polymerase chain reaction (RT-PCR) analysis, expressions of the two secretion types (VEGF121 and VEGF165) were the highest. Thus, VEGF protein in culture supernatant was measured by using enzyme-linked immunosorbent assay (ELISA) with or without inflammatory cytokines, i.e., interleukin (IL)-1beta, interferon (IFN)-alpha, IFN-gamma, and tumor necrosis factor (TNF)-alpha; and growth factors, i.e., epidermal growth factor (EGF), platelet-derived growth factor (PDGF)-BB, basic fibroblast growth factor (bFGF), and transforming growth factor (TGF)-alpha. As a result, secretion of VEGF from the cell lines was upregulated at various degrees. Based on these findings, VEGF expression in HCC tissues was thought to be related to the histological grade. The findings also indicate that various cytokines and growth factors could cooperatively act to enhance VEGF expressions in HCC.
Collapse
Affiliation(s)
- R Yamaguchi
- The First Department of Pathology, Kurume University School of Medicine, Fukuoka, Japan
| | | | | | | | | | | |
Collapse
|
1113
|
Flamme I, Krieg M, Plate KH. Up-regulation of vascular endothelial growth factor in stromal cells of hemangioblastomas is correlated with up-regulation of the transcription factor HRF/HIF-2alpha. THE AMERICAN JOURNAL OF PATHOLOGY 1998; 153:25-9. [PMID: 9665461 PMCID: PMC1852948 DOI: 10.1016/s0002-9440(10)65541-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hemangioblastomas, the most frequent manifestation of the hereditary von Hippel-Lindau disease (VHL), are highly vascularized tumors of the central nervous system. In previous studies, the endothelial-specific mitogen vascular endothelial growth factor (VEGF) was shown to be up-regulated in the stromal cells, the putative neoplastic cells in hemangioblastomas. Therefore, it was suggested that secretion of VEGF by stromal cells is the pathogenetic cause of the vascular lesions in hemangioblastomas. The novel basic helix loop helix transcription factor HRF/HIF-2alpha is a candidate regulator of VEGF expression during development. We therefore investigated expression of HRF/HIF-2alpha in hemangioblastomas and found the overexpression of VEGF mRNA in stromal cells to be highly correlated with elevated expression levels of HRF/HIF-2alpha mRNA. This finding is suggestive for a role of HRF in VEGF-dependent vascular growth in hemangioblastomas and could provide a link between transcriptional activation of the VEGF gene and loss of function of the VHL gene product.
Collapse
Affiliation(s)
- I Flamme
- Zentrum für Molekularbiologische Medizin, Universität zu Köln, Germany.
| | | | | |
Collapse
|
1114
|
Perkett EA, Klekamp JG. Vascular endothelial growth factor expression is decreased in rat lung following exposure to 24 or 48 hours of hyperoxia: implications for endothelial cell survival. Chest 1998; 114:52S-53S. [PMID: 9676627 DOI: 10.1378/chest.114.1_supplement.52s] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- E A Perkett
- Department of Pediatric Pulmonary Medicine, Vanderbilt University, Nashville, Tenn, USA
| | | |
Collapse
|
1115
|
Pepper MS, Mandriota SJ. Regulation of vascular endothelial growth factor receptor-2 (Flk-1) expression in vascular endothelial cells. Exp Cell Res 1998; 241:414-25. [PMID: 9637783 DOI: 10.1006/excr.1998.4072] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We have previously reported the existence of a synergistic interaction between vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in the induction of angiogenesis in vitro. Here we demonstrate that bFGF increases VEGF receptor-2 (VEGFR-2/Flk-1) expression: mRNA levels were increased by 4.5- to 8.0-fold and total protein by 2.0- to 3.5-fold, in bovine microvascular endothelial (BME), aortic endothelial (BAE), and transformed fetal aortic (GM7373) endothelial cells. VEGF itself did not affect VEGFR-2 expression, and neither bFGF nor VEGF altered expression of FGF receptor-1. We also show that synergism occurs at the level of proliferation when this is measured in a three-dimensional but not in a conventional two-dimensional assay. Differences in the level of VEGFR-2 expression were also observed when cells were grown on or within collagen gels under different conditions: mRNA levels were lowest under sparse conditions, increased 20- to 26-fold at confluence, and increased even further (57-fold) when cells were cultured in suspension in three-dimensional collagen gels. Finally, a synergistic increase was seen in the level of expression of urokinase and urokinase receptor mRNAs when cells were exposed to bFGF and VEGF for 4 days. These findings demonstrate that the level of VEGFR-2 expression can be modulated by environmental factors including cytokines and the geometry of the culture conditions and provide some insight into the mechanisms of synergism between bFGF and VEGF in the induction of angiogenesis in vitro.
Collapse
Affiliation(s)
- M S Pepper
- Department of Morphology, University Medical Center, Geneva 4, 1211, Switzerland.
| | | |
Collapse
|
1116
|
Rosenstein JM, Mani N, Silverman WF, Krum JM. Patterns of brain angiogenesis after vascular endothelial growth factor administration in vitro and in vivo. Proc Natl Acad Sci U S A 1998; 95:7086-91. [PMID: 9618543 PMCID: PMC22748 DOI: 10.1073/pnas.95.12.7086] [Citation(s) in RCA: 160] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a secreted endothelial cell mitogen that has been shown to induce vasculogenesis and angiogenesis in many organ systems and tumors. Considering the importance of VEGF to embryonic vascularization and survival, the effects of administered VEGF on developing or adult cerebrovasculature are unknown: can VEGF alter brain angiogenesis or mature cerebrovascular patterns? To examine these questions we exposed fetal, newborn, and adult rat cortical slice explants to graduated doses of recombinant VEGF. The effects of another known angiogenic factor, basic fibroblast growth factor (bFGF), were evaluated in a comparable manner. In addition, we infused VEGF via minipump into the adult cortex. Significant angiogenic effects were found in all VEGF experiments in a dose-responsive manner that were abolished by the addition of VEGF neutralizing antibody. Fetal and newborn explants had a highly complex network of branched vessels that immunoexpressed the flt-1 VEGF receptor, and flk-1 VEGF receptor expression was determined by reverse transcription-PCR. Adult explants had enlarged, dilated vessels that appeared to be an expansion of the existing network. All bFGF-treated explants had substantially fewer vascular profiles. VEGF infusions produced both a remarkable localized neovascularization and, unexpectedly, the expression of flt-1 on reactive astrocytes but not on endothelial cells. The preponderance of neovascularization in vitro and in vivo, however, lacked the blood-brain barrier (BBB) phenotype marker, GLUT-1, suggesting that in brain the angiogenic role of VEGF may differ from a potential BBB functional role, i.e., transport and permeability. VEGF may serve an important capacity in neovascularization or BBB alterations after brain injury.
Collapse
Affiliation(s)
- J M Rosenstein
- Department of Anatomy and Cell Biology, The George Washington University Medical Center, Washington, DC 20037, USA
| | | | | | | |
Collapse
|
1117
|
Roeckl W, Hecht D, Sztajer H, Waltenberger J, Yayon A, Weich HA. Differential binding characteristics and cellular inhibition by soluble VEGF receptors 1 and 2. Exp Cell Res 1998; 241:161-70. [PMID: 9633524 DOI: 10.1006/excr.1998.4039] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The FLT-1 and KDR genes encode transmembrane tyrosine kinases which function as high-affinity receptors for vascular endothelial growth factor (VEGF). We have used the baculovirus system to express the extracellular parts of the FLT-1 receptor and KDR receptor in soluble form (sFLT-1 and sKDR), for in vitro binding and competition assays. Here, we show that the binding of VEGF165 to sKDR but not sFLT-1 is dependent on heparin, regardless of whether VEGF165 or sKDR is immobilized. Further, only sFLT-1 acts as a receptor antagonist in solution and sKDR can neither compete with the binding of VEGF165 to human endothelial cells carrying both receptors nor block VEGF165 induced mitogenicity. Soluble KDR only partially inhibits cell migration even at high concentrations, in contrast to sFLT which can almost completely block (82%) VEGF-induced cell proliferation and migration. Taken together these results show that the two soluble VEGF receptor proteins, sFLT-1 and sKDR, despite binding the same ligand, behave very differently when immobilized with regard to their dependence on heparin for VEGF binding. In solution their respective ability to function as receptor antagonists is also strikingly different, possibly a reflection of their different dependency on heparin.
Collapse
Affiliation(s)
- W Roeckl
- Department of Gene Regulation, GBF, Braunschweig, Germany
| | | | | | | | | | | |
Collapse
|
1118
|
Fairbrother WJ, Champe MA, Christinger HW, Keyt BA, Starovasnik MA. Solution structure of the heparin-binding domain of vascular endothelial growth factor. Structure 1998; 6:637-48. [PMID: 9634701 DOI: 10.1016/s0969-2126(98)00065-3] [Citation(s) in RCA: 147] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is an endothelial cell-specific mitogen and is a potent angiogenic and vascular permeabilizing factor. VEGF is also an important mediator of pathological angiogenesis associated with cancer, rheumatoid arthritis and proliferative retinopathy. The binding of VEGF to its two known receptors, KDR and Flt-1, is modulated by cell-surface-associated heparin-like glycosaminoglycans and exogenous heparin or heparan sulfate. Heparin binding to VEGF165, the most abundantly expressed isoform of VEGF, has been localized to the carboxy-terminal 55 residues; plasmin cleavage of VEGF165 yields a homodimeric 110-residue amino-terminal receptor-binding domain (VEGF110) and two 55-residue carboxy-terminal heparin-binding fragments. The endothelial cell mitogenic potency of VEGF110 is decreased significantly relative to VEGF165, indicating that the heparin-binding domains are critical for stimulating endothelial cell proliferation. RESULTS The solution structure of the 55-residue heparin-binding domain of VEGF165 has been solved using data from two-dimensional homonuclear and three-dimensional heteronuclear NMR spectroscopy. The structure has two subdomains, each containing two disulfide bridges and a short two-stranded antiparallel beta sheet; the carboxy-terminal subdomain also contains a short alpha helix. Hydrophobic interactions are limited to sidechains packing against the disulfide bridges. CONCLUSIONS The heparin-binding domain of VEGF has no significant sequence or structural similarity to any known proteins and thus represents a novel heparin-binding domain. Most of the positively charged amino acid sidechains are localized on one side of the carboxy-terminal subdomain or on an adjacent disordered loop in the amino-terminal subdomain. The observed distribution of surface charges suggests that these residues constitute a heparin interaction site.
Collapse
|
1119
|
Hippenstiel S, Krüll M, Ikemann A, Risau W, Clauss M, Suttorp N. VEGF induces hyperpermeability by a direct action on endothelial cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:L678-84. [PMID: 9612282 DOI: 10.1152/ajplung.1998.274.5.l678] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vascular endothelial growth factor (VEGF) is a key regulator of vasculo- and angiogenesis. Earlier studies demonstrated a permeability-increasing effect of VEGF in skin tests, leading to its other name, vascular permeability factor. We wondered whether VEGF-induced hyperpermeability was a direct effect of VEGF on endothelial cells and studied the permeability of human and porcine endothelial cell monolayers in a well-characterized in vitro system. VEGF increased the hydraulic conductivity up to 20-fold and simultaneously decreased the albumin reflection coefficient. This effect occurred after a delay of 150 min, although VEGF-induced early endothelial cell activation was verified by enhanced inositol phosphate accumulation within 5 min and increased P-selectin expression within 15 min. Platelet-derived growth factor and granulocyte-macrophage colony-stimulating factor, two endothelial cell nonspecific mitogens, also stimulated phosphatidylinositol metabolism and P-selectin expression; however, they had no effect on endothelial permeability. The increase in intracellular cyclic nucleotide levels of human endothelial monolayers abolished VEGF-induced endothelial hyperpermeability. In summary, VEGF increased endothelial permeability by a direct action on endothelial cells. Based on the pattern of endothelial cell activation by growth factors, VEGF appears to be a unique stimulus.
Collapse
Affiliation(s)
- S Hippenstiel
- Department of Internal Medicine, Justus-Liebig-University, Giessen, Germany
| | | | | | | | | | | |
Collapse
|
1120
|
Aitkenhead M, Christ B, Eichmann A, Feucht M, Wilson DJ, Wilting J. Paracrine and autocrine regulation of vascular endothelial growth factor during tissue differentiation in the quail. Dev Dyn 1998; 212:1-13. [PMID: 9603419 DOI: 10.1002/(sici)1097-0177(199805)212:1<1::aid-aja1>3.0.co;2-l] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The expression of vascular endothelial growth factor (VEGF) has been described to coincide both temporally and spatially with angiogenesis suggesting a role as a paracrine stimulator of endothelial cells. We have used digoxigenin labelled RNA probes to VEGF and the VEGF receptor-2 (Quek1) to investigate the relationship between VEGF expression and vascular events in quail embryos from day 1 to 13 of incubation. Furthermore, the effect of exogenously applied VEGF was studied in day 4 quail embryos using polyclonal anti-VEGF antibodies. Expression of VEGF mRNA was observed in day 1 and 2 embryos in regions of active angiogenesis and hemangiopoiesis. VEGF mRNA expression was found at high levels in the ventral aspect of the neural tube and Quek1 mRNA expression in the accompanying endothelial cells of day 3 embryos, suggesting a function in brain angiogenesis. However, in the neural tube, thyroid gland and cartilaginous skeleton VEGF mRNA was expressed at least 1 day before the ingrowth of vessels, suggesting that additional mechanisms are involved in control of angiogenesis. This is supported by the observation that application of VEGF165 into the midbrain induced dilatation of perineural vessels, while the intraneural vessels remained almost unaffected. Expression of VEGF mRNA was also observed at high levels in podocytes during all stages, indicative of its importance in glomerular development and function. The results of the present study indicate that as angiogenesis occurred in other tissues and organs (day 13 metanephros, dorsal third of the day 7 neural tube, skeletal muscle, and many mesodermal compartments), there was concurrent paracrine expression of VEGF mRNA and Quek1 mRNA. One noteable exception was the hepatocytes of the developing liver which appeared to remain VEGF-negative throughout the study. However, a small number of endothelial cells within liver sinuses, and additionally within the kidney and the elastic arteries, expressed VEGF mRNA. These results suggest that VEGF may also act as an autocrine mediator of angiogenesis, possibly as a result of localised tissue hypoxia.
Collapse
Affiliation(s)
- M Aitkenhead
- School of Biomedical Science/Anatomy, The Queen's University of Belfast, Northern Ireland.
| | | | | | | | | | | |
Collapse
|
1121
|
Nishikawa SI, Nishikawa S, Hirashima M, Matsuyoshi N, Kodama H. Progressive lineage analysis by cell sorting and culture identifies FLK1+VE-cadherin+ cells at a diverging point of endothelial and hemopoietic lineages. Development 1998; 125:1747-57. [PMID: 9521912 DOI: 10.1242/dev.125.9.1747] [Citation(s) in RCA: 543] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Totipotent murine ES cells have an enormous potential for the study of cell specification. Here we demonstrate that ES cells can differentiate to hemopoietic cells through the proximal lateral mesoderm, merely upon culturing in type IV collagen-coated dishes. Separation of the Flk1+ mesoderm from other cell lineages was critical for hemopoietic cell differentiation, whereas formation of the embryoid body was not. Since the two-dimensionally spreading cells can be monitored easily in real time, this culture system will greatly facilitate the study of the mechanisms involved in the cell specification to mesoderm, endothelial, and hemopoietic cells. In the culture of ES cells, however, lineages and stages of differentiating cells can only be defined by their own characteristics. We showed that a combination of monoclonal antibodies against E-cadherin, Flk1/KDR, PDGF receptor(alpha), VE-cadherin, CD45 and Ter119 was sufficient to define most intermediate stages during differentiation of ES cells to blood cells. Using this culture system and surface markers, we determined the following order for blood cell differentiation: ES cell (E-cadherin+Flk1-PDGFRalpha-), proximal lateral mesoderm (E-cadherin-Flk1+VE-cadherin-), progenitor with hemoangiogenic potential (Flk1+VE-cadherin+CD45-), hemopoietic progenitor (CD45+c-Kit+) and mature blood cells (c-Kit-CD45+ or Ter119+), though direct differentiation of blood cells from the Flk1+VE-cadherin- stage cannot be ruled out. Not only the VE-cadherin+CD45- population generated from ES cells but also those directly sorted from the yolk sac of 9.5 dpc embryos have a potential to give rise to hemopoietic cells. Progenitors with hemoangiogenic potential were identified in both the Flk1+VE-cadherin- and Flk1+VE-cadherin+ populations by the single cell deposition experiment. This line of evidence implicates Flk1+VE-cadherin+ cells as a diverging point of hemopoietic and endothelial cell lineages.
Collapse
Affiliation(s)
- S I Nishikawa
- Department of Molecular Genetics, Faculty of Medicine, Kyoto University, Kyoto, Japan.
| | | | | | | | | |
Collapse
|
1122
|
Yao TP, Oh SP, Fuchs M, Zhou ND, Ch'ng LE, Newsome D, Bronson RT, Li E, Livingston DM, Eckner R. Gene dosage-dependent embryonic development and proliferation defects in mice lacking the transcriptional integrator p300. Cell 1998; 93:361-72. [PMID: 9590171 DOI: 10.1016/s0092-8674(00)81165-4] [Citation(s) in RCA: 767] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The transcriptional coactivator and integrator p300 and its closely related family member CBP mediate multiple, signal-dependent transcriptional events. We have generated mice lacking a functional p300 gene. Animals nullizygous for p300 died between days 9 and 11.5 of gestation, exhibiting defects in neurulation, cell proliferation, and heart development. Cells derived from p300-deficient embryos displayed specific transcriptional defects and proliferated poorly. Surprisingly, p300 heterozygotes also manifested considerable embryonic lethality. Moreover, double heterozygosity for p300 and cbp was invariably associated with embryonic death. Thus, mouse development is exquisitely sensitive to the overall gene dosage of p300 and cbp. Our results provide genetic evidence that a coactivator endowed with histone acetyltransferase activity is essential for mammalian cell proliferation and development.
Collapse
Affiliation(s)
- T P Yao
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1123
|
Lyons MS, Bell B, Stainier D, Peters KG. Isolation of the zebrafish homologues for the tie-1 and tie-2 endothelium-specific receptor tyrosine kinases. Dev Dyn 1998; 212:133-40. [PMID: 9603430 DOI: 10.1002/(sici)1097-0177(199805)212:1<133::aid-aja12>3.0.co;2-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Several characteristics of the zebrafish embryo make it an attractive model in which to study the development of the cardiovascular system. The utility of the zebrafish as a model of mammalian vascular development will depend on the conservation of molecular and morphogenetic mechanisms of vessel growth. Here, we report the cloning of the zebrafish homologues of the endothelium-specific receptor tyrosine kinases tie-1 and tie-2. The Z tie-2 clone represents the first report of a full-length zebrafish endothelium-specific gene. The zebrafish tie family members have significant structural homology with their murine and human counterparts. In addition, like the murine tie-1 and tie-2 genes, expression was found predominantly in endothelial cells. At 24-hr postfertilization (HPF), Z tie-1 was expressed in all observed populations of endothelial cells. Interestingly, Z tie-2 exhibited a similar, although slightly more restricted, expression pattern. Taken together, these data strongly suggest that mechanisms of vascular development are highly conserved across species and that zebrafish will continue to be a useful model for the investigation of vertebrate embryonic vascular development.
Collapse
Affiliation(s)
- M S Lyons
- Duke University Medical Center, Department of Medicine, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
1124
|
Williams B. Vascular permeability/vascular endothelial growth factors: a potential role in the pathogenesis and treatment of vascular diseases. Vasc Med 1998; 1:251-8. [PMID: 9552580 DOI: 10.1177/1358863x9600100403] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- B Williams
- Department of Medicine and Therapeutics, University of Leicester School of Medicine, UK
| |
Collapse
|
1125
|
Abstract
Angiogenic cytokines constitute a potentially novel form of therapy for patients with cardiovascular disease. The feasibility of using recombinant formulations of angiogenic growth factors to expedite and/or augment collateral artery development in animal models of myocardial and hindlimb ischemia--'therapeutic angiogenesis'--has now been well established. These studies have suggested that two angiogenic growth factors in particular--basic fibroblast growth factor and vascular endothelial growth factor--are sufficiently potent to merit further investigation. More recently, experiments performed in our laboratory have indicated that, in the case of vascular endothelial growth factor--a secreted protein--similar results may be achieved by percutaneous arterial gene transfer. Further laboratory and clinical studies may yield promising insights into the fundamental basis for native as well as therapeutic angiogenesis, and at the same time more explicitly define the manner in which therapeutic angiogenesis may be successfully incorporated into clinical practice.
Collapse
Affiliation(s)
- J M Isner
- Department of Medicine (Cardiology), St Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135-2997, USA
| |
Collapse
|
1126
|
Hatzopoulos AK, Folkman J, Vasile E, Eiselen GK, Rosenberg RD. Isolation and characterization of endothelial progenitor cells from mouse embryos. Development 1998; 125:1457-68. [PMID: 9502726 DOI: 10.1242/dev.125.8.1457] [Citation(s) in RCA: 153] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The cardiovascular system develops early in embryogenesis from cells of mesodermal origin. To study the molecular and cellular processes underlying this transition, we have isolated mesodermal cells from murine embryos at E7.5 with characteristic properties of endothelial progenitors by using a combination of stromal cell layers and growth conditions. The isolated embryonic cells displayed unlimited stem-cell-like growth potential and a stable phenotype in culture. RNA analysis revealed that the embryonic cells express the endothelial-specific genes tie-2 and thrombomodulin (TM) as well as the early mesodermal marker fgf-3. The GSL I-B4 isolectin, a marker of early endothelial cells, specifically binds to the isolated cells. The in vitro differentiation with retinoic acid and cAMP led to a 5- to 10-fold induction of flk-1, von Willebrand Factor (vWF), TM, GATA-4 and GATA-6. Electron microscopy revealed that in vitro differentiation is associated with increased amounts of rER and Golgi, and a dramatic increase in secretory vesicles packed with vWF. When cultured in Matrigel, the embryonic cells assume the characteristic endothelial cobblestone morphology and form tubes. Injection into chicken embryos showed incorporation of the embryonic cells in the endocardium and the brain vasculature. The expression of TM, tie-2, GATA-4 and GATA-6 suggests that the isolated embryonic endothelial cell progenitors are derived from the proximal lateral mesoderm where the pre-endocardial tubes form. The properties of the endothelial cell progenitors described here provide a novel approach to analyze mediators, signaling pathways and transcriptional control in early vascular development.
Collapse
|
1127
|
Kong HL, Hecht D, Song W, Kovesdi I, Hackett NR, Yayon A, Crystal RG. Regional suppression of tumor growth by in vivo transfer of a cDNA encoding a secreted form of the extracellular domain of the flt-1 vascular endothelial growth factor receptor. Hum Gene Ther 1998; 9:823-33. [PMID: 9581905 DOI: 10.1089/hum.1998.9.6-823] [Citation(s) in RCA: 120] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Vascular endothelial growth factor (VEGF), a potent angiogenic mediator, is overexpressed in most solid tumors. On the basis of the knowledge that solid tumor growth beyond a small volume is critically dependent on angiogenesis, and that adenovirus (Ad) vectors can mediate efficient in vivo gene transfer and expression, we hypothesized that Ad-mediated transfer of a secreted form of the extracellular domain of the flt-1 VEGF receptor (Adsflt) would suppress tumor growth on a regional basis. To evaluate this concept, three tumor models were examined using a murine colon carcinoma cell line and syngeneic BALB/c mice. First, mice with preestablished splenic CT26.CL25 tumors and liver metastases were given Adsflt on AdNull intravenously and, after 15 days, spleens and livers were harvested to quantify tumor burden. Adslft-treated animals had minimal residual splenic tumors and liver metastases; in contrast, control animals had bulky splenic tumors and extensive liver metastases (p < 0.003). Second, mice with preestablished lung metastases showed a significant reduction in pulmonary metastases with regionally administered Adslft (intratracheal, p < 0.02) but not when the vector was systemically administered (intravenous, p > 0.9). Finally, mice with primary subcutaneous tumors treated with intratumoral administration of Adslft showed significant tumor suppression (p < 0.05) not observed in AdNull-treated mice or mice given Adslft intravenously (p > 0.3). We conclude that Ad-mediated in vivo regional delivery of a secreted form of the extracellular domain of the flt-1 VEGF receptor can effectively inhibit regional tumor growth, a strategy that may provide a means to control tumor growth within the treated organ without the risk of systemic antiangiogenesis.
Collapse
Affiliation(s)
- H L Kong
- Division of Pulmonary and Critical Care Medicine, The New York Hospital-Cornell Medical Center, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
1128
|
Paradis H, Arceci RJ, Adams LC, Gendron RL. Differentiation responses of embryonic endothelium to leukemia inhibitory factor. Exp Cell Res 1998; 240:7-15. [PMID: 9570916 DOI: 10.1006/excr.1998.4006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The IEM cell line is a murine embryonic endothelial cell line that responds to combinations of basic fibroblast growth factor (bFGF) and leukemia inhibitory factor (LIF) by undergoing proliferation and vasculogenic differentiation in vitro and in vivo. Exposure to LIF and bFGF in vitro permits the IEM cells to specifically chimerize endothelium in vivo and recapitulate normal endothelial development after blastocyst injection. We report here that unmanipulated IEM cells form vascular neoplasias when injected into immunodeficient nude mice. Examination of IEM neoplasia following exposure in vitro to bFGF and LIF before injection into nude mice profoundly reduced or completely suppressed the neoplastic growth of IEM cells. Furthermore, this suppression was observed by treatment with LIF alone, while bFGF treatment did not significantly alter IEM neoplasia and did not modify the LIF-mediated suppression. Characterization of the IEM responses to LIF revealed that the LIF suppression of IEM neoplasia depended on how long the cells were exposed to LIF in vitro. The IEM cell response to LIF was associated with the specific activation of the transcription factor Stat3. Stat1 activation could not be detected in response to LIF, although it is expressed in IEM cells. Our results demonstrate that the LIF-induced differentiation of IEM cells involves suppression of IEM-derived neoplasia and is associated with the specific activation of Stat3.
Collapse
Affiliation(s)
- H Paradis
- Department of Pediatrics, Children's Hospital Research Foundation, Cincinnati, Ohio 45229-3039, USA
| | | | | | | |
Collapse
|
1129
|
Takahashi T, Huynh-Do U, Daniel TO. Renal microvascular assembly and repair: power and promise of molecular definition. Kidney Int 1998; 53:826-35. [PMID: 9551388 DOI: 10.1111/j.1523-1755.1998.00822.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Developmental assembly of the renal microcirculation is a precise and coordinated process now accessible to experimental scrutiny. Although definition of the cellular and molecular determinants is incomplete, recent findings have reframed concepts and questions about the origins of vascular cells in the glomerulus and the molecules that direct cell recruitment, specialization and morphogenesis. New findings illustrate principles that may be applied to defining critical steps in microvascular repair following glomerular injury. Developmental assembly of endothelial, mesangial and epithelial cells into glomerular capillaries requires that a coordinated, temporally defined series of steps occur in an anatomically ordered sequence. Recent evidence shows that both vasculogenic and angiogenic processes participate. Local signals direct cell migration, proliferation, differentiation, cell-cell recognition, formation of intercellular connections, and morphogenesis. Growth factor receptor tyrosine kinases on vascular cells are important mediators of many of these events. Cultured cell systems have suggested that basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), and vascular endothelial growth factor (VEGF) promote endothelial cell proliferation, migration or morphogenesis, while genetic deletion experiments have defined an important role for PDGF beta receptors and platelet-derived growth factor (PDGF) B in glomerular development. Receptor tyrosine kinases that convey non-proliferative signals also contribute in kidney and other sites. The EphB1 receptor, one of a diverse class of Eph receptors implicated in neural cell targeting, directs renal endothelial migration, cell-cell recognition and assembly, and is expressed with its ligand in developing glomeruli. Endothelial TIE2 receptors bind angiopoietins (1 and 2), the products of adjacent supportive cells, to signals direct capillary maturation in a sequence that defines cooperative roles for cells of different lineages. Ultimately, definition of the cellular steps and molecular sequence that direct microvascular cell assembly promises to identify therapeutic targets for repair and adaptive remodeling of injured glomeruli.
Collapse
Affiliation(s)
- T Takahashi
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | |
Collapse
|
1130
|
Bennett DC, Trayner ID, Piao X, Easty DJ, Klüppel M, Alexander WS, Wagner EF, Bernstein A. recessive spotting: a linked locus that interacts with W/Kit but is not allelic. Genes Cells 1998; 3:235-44. [PMID: 9663658 DOI: 10.1046/j.1365-2443.1998.00184.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The murine coat-colour mutation recessive spotting (rs) maps very closely to the W/Kit locus, encoding the proto-oncoprotein Kit, the protein tyrosine kinase receptor for stem cell factor. Kit is important in the development of melanocytes, germ cells, interstitial cells of Cajal (ICC) and haemopoietic lineages, including mast cells. rs has never been genetically separated from Kit, and interacts with Kit mutations, suggesting that it is a recessive allele of Kit. Here we have tested this possibility. We have shown previously that diploid rs/rs melanocytes proliferated more slowly than did +/+ melanocytes, as did an immortal line of rs/rs melanocytes, melan-rs. RESULTS The Kit mRNA level in rs/rs melanocytes was indistinguishable from that of other melanocyte lines. The Kit cDNA sequence from rs/rs melanocytes and the kinase activity of Kit in rs/rs mast cells appeared to be normal. No deficiency of mast cells or ICC was observed in rs/rs mice. Moreover, following the overexpression of a normal Kit cDNA, proliferation of rs/rs melanocytes was retarded further, but that of +/+ melanocytes was increased, indicating an intracellular interaction between rs and Kit. Of other closely linked tyrosine kinase genes, melanocytes and melanoblasts did not express mRNA for Pdgfra, Flk-1 or Txk, but both expressed Tec, encoding a nonreceptor kinase that interacts with Kit. CONCLUSIONS rs is not a mutation in Kit, although we have confirmed that rs interacts with Kit. It seems unlikely that rs affects Pdgfra, Flk-1 or Txk, but Tec remains a candidate for rs.
Collapse
Affiliation(s)
- D C Bennett
- St. George's Hospital Medical School, London, UK.
| | | | | | | | | | | | | | | |
Collapse
|
1131
|
Otani A, Takagi H, Suzuma K, Honda Y. Angiotensin II potentiates vascular endothelial growth factor-induced angiogenic activity in retinal microcapillary endothelial cells. Circ Res 1998; 82:619-28. [PMID: 9529167 DOI: 10.1161/01.res.82.5.619] [Citation(s) in RCA: 167] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiotensin II (Ang II) plays a role in the development of many vascular diseases. In the present study, we have investigated the effect of Ang II on vascular endothelial growth factor (VEGF) receptor expression and VEGF-induced angiogenic activity in bovine retinal microcapillary endothelial cells (BRECs). Ang II induced a significant increase of kinase domain-containing receptor/total liver kinase (KDR/Flk-1) mRNA in a time- and dose-dependent manner, with a maximal 4.3+/-0.8-fold increase after a 4-hour stimulation. Ang II increased the rate of KDR gene transcription by 5.4-fold, whereas the half-life of KDR mRNA was not increased significantly. The increase depended partially on new protein synthesis. The Ang II-induced KDR mRNA increase was inhibited by either [Sar1,Ile8]angiotensin or angiotensin type 1 receptor antagonists but was not significantly altered by angiotensin type 2 receptor antagonists. The PKC inhibitor reduced Ang II-induced KDR mRNA expression by 70+/-15%. The tyrosine kinase inhibitor reduced the Ang II- and phorbol 12-myristate 13-acetate-induced KDR mRNA increases by 35+/-8% and 44+/-26%, respectively. Ang II increased by 3.1-fold the 35S-labeled KDR/Flk-1 immunoprecipitated by a specific antibody to KDR/Flk-1. Scatchard analysis demonstrated that Ang II induced a significant increase of binding sites without changing binding affinity. Ang II enhanced VEGF-induced cell growth and tube formation. Ang II itself had no effect on cell growth, tube formation, or mRNA levels of VEGF and tms-like tyrosine kinase (Flt-1) in BRECs. These findings suggest that Ang II might potentiate VEGF-induced angiogenic activity through an increase of the VEGF receptor KDR/Flk-1.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Aorta/cytology
- Capillaries/cytology
- Capillaries/drug effects
- Capillaries/enzymology
- Cattle
- Cell Division/drug effects
- Cells, Cultured
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Gene Expression/drug effects
- Neovascularization, Physiologic/drug effects
- Protein Kinase C/metabolism
- Protein-Tyrosine Kinases/metabolism
- RNA, Messenger/metabolism
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Growth Factor/genetics
- Receptors, Growth Factor/metabolism
- Receptors, Mitogen/genetics
- Receptors, Mitogen/metabolism
- Receptors, Vascular Endothelial Growth Factor
- Retina/cytology
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Transcription, Genetic/drug effects
Collapse
Affiliation(s)
- A Otani
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Japan
| | | | | | | |
Collapse
|
1132
|
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; 92:735-45. [PMID: 9529250 DOI: 10.1016/s0092-8674(00)81402-6] [Citation(s) in RCA: 1864] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vascular endothelial growth factor (VEGF), a major regulator of angiogenesis, binds to two receptor tyrosine kinases, KDR/Flk-1 and Flt-1. We now describe the purification and the expression cloning from tumor cells of a third VEGF receptor, one that binds VEGF165 but not VEGF121. This isoform-specific VEGF receptor (VEGF165R) is identical to human neuropilin-1, a receptor for the collapsin/semaphorin family that mediates neuronal cell guidance. When coexpressed in cells with KDR, neuropilin-1 enhances the binding of VEGF165 to KDR and VEGF165-mediated chemotaxis. Conversely, inhibition of VEGF165 binding to neuropilin-1 inhibits its binding to KDR and its mitogenic activity for endothelial cells. We propose that neuropilin-1 is a novel VEGF receptor that modulates VEGF binding to KDR and subsequent bioactivity and therefore may regulate VEGF-induced angiogenesis.
Collapse
MESH Headings
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Cell Line/chemistry
- Cell Line/cytology
- Cell Line/metabolism
- Chemotaxis/physiology
- Cloning, Molecular
- Endothelial Growth Factors/chemistry
- Endothelial Growth Factors/metabolism
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/cytology
- Exons/physiology
- Gene Expression
- Humans
- Isomerism
- Lymphokines/chemistry
- Lymphokines/metabolism
- Molecular Sequence Data
- Neovascularization, Physiologic/physiology
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neuropilin-1
- RNA, Messenger/metabolism
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/isolation & purification
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptors, Growth Factor/genetics
- Receptors, Growth Factor/isolation & purification
- Receptors, Growth Factor/metabolism
- Receptors, Mitogen/genetics
- Receptors, Mitogen/isolation & purification
- Receptors, Mitogen/metabolism
- Receptors, Vascular Endothelial Growth Factor
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Umbilical Cord/cytology
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- S Soker
- Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
1133
|
Abstract
Vascular endothelial growth factor (VEGF) was expressed in developing respiratory epithelial cells under control of the promoter from the human surfactant protein C (SP-C) gene. SP-C-VEGF transgenic mice did not survive after birth. When obtained by hysterectomy on embryonic day 15 (E15) or 17 (E17), abnormalities in the transgenic mice were confined to the lung and were correlated with the expression of transgene mRNA as revealed by in situ hybridization. On E15 and E17, marked abnormalities in lung morphogenesis were observed in transgenic mice. Lungs consisted of large dilated tubules with increased peritubular vascularity. The mRNA levels of the VEGF receptor, Flk-1, and the endothelial cell specific receptor tyrosine kinase, Tie-1, were increased in lung mesenchyme of the transgenic mice. The numbers of acinar tubules and the abundance of mesenchyme were decreased. Endogenous VEGF mRNA was expressed in the respiratory epithelial cells of the developing lungs, and the levels of VEGF mRNA were increased in the SP-C-VEGF transgenic mice. Although the normal pattern of immunostaining for SP-C and Clara cell secretory protein (CCSP) indicated that epithelial cell differentiation was relatively unaltered by the transgene, electron microscopic analysis revealed a lack of alveolar Type I cell differentiation at E18. Expression of VEGF in the developing respiratory epithelium of transgenic mice increased growth of the pulmonary blood vessels, disrupted branching morphogenesis of the lung and inhibited Type I cell differentiation.
Collapse
Affiliation(s)
- X Zeng
- Children's Hospital Medical Center, Division of Pulmonary Biology, Cincinnati, Ohio 45229-3039, USA
| | | | | | | | | |
Collapse
|
1134
|
Banks RE, Forbes MA, Kinsey SE, Stanley A, Ingham E, Walters C, Selby PJ. Release of the angiogenic cytokine vascular endothelial growth factor (VEGF) from platelets: significance for VEGF measurements and cancer biology. Br J Cancer 1998; 77:956-64. [PMID: 9528841 PMCID: PMC2150108 DOI: 10.1038/bjc.1998.158] [Citation(s) in RCA: 433] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a potent angiogenic factor with a key role in several pathological processes, including tumour vascularization. Our preliminary observations indicated higher VEGF concentrations in serum samples than in matched plasma samples. We have now demonstrated that this difference is due to the presence of VEGF within platelets and its release upon their activation during coagulation. In eight healthy volunteers, serum VEGF concentrations ranged from 76 to 854 pg ml(-1) and were significantly higher (P < 0.01) than the matched citrated plasma VEGF concentrations, which ranged from < 9 to 42 pg ml(-1). Using platelet-rich plasma, mean (s.d.) platelet VEGF contents of 0.56 (0.36) pg of VEGF 10(-6) platelets were found. Immunocytochemistry demonstrated the cytoplasmic presence of VEGF within megakaryocytes and other cell types within the bone marrow. From examination of the effects of blood sample processing on circulating VEGF concentrations, it is apparent that for accurate measurements, citrated plasma processed within 1 h of venepuncture should be used. Serum is completely unsuitable. The presence of VEGF within platelets has implications for processes involving platelet and endothelial cell interactions. e.g. wound healing, and in tumour metastasis, when platelets adhering to circulating tumour cells may release VEGF at points of adhesion to endothelium, leading to hyperpermeability and extravasation of cells.
Collapse
Affiliation(s)
- R E Banks
- ICRF Cancer Medicine Research Unit, St James's University Hospital, Leeds UK
| | | | | | | | | | | | | |
Collapse
|
1135
|
Wysocki SJ, Zheng MH, Smith A, Norman PE. Vascular endothelial growth factor (VEGF) expression during arterial repair in the pig. Eur J Vasc Endovasc Surg 1998; 15:225-30. [PMID: 9587335 DOI: 10.1016/s1078-5884(98)80180-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Vascular endothelial growth factor (VEGF) is reported to be a potent and specific mitogen for endothelial cells (EC) and an inducer of angiogenesis in vivo. Originally called vascular permeability factor (VPF), VEGF also increases permeability of microvessels to circulating macromolecules. The aim of this study was to examine whether the VEGF gene was expressed in porcine arteries following denudation of EC. DESIGN Experimental animal model with mechanical injury to large arteries. METHODS The right iliac artery of juvenile pigs was de-endothelialised using an inflated balloon catheter. At a number of time-points after injury, these arteries were harvested together with uninjured contralateral arteries. Sections of arteries were used for RNA analysis by Northern blots and for protein localisation studies by immunohistochemistry. RESULTS Two VEGF transcripts (2.0 kb, 4.5 kb) were markedly elevated in pig arteries soon after injury. Newly synthesised VEGF protein was located in smooth muscle cells (SMC) throughout the media of injured arteries. CONCLUSIONS The elevated expression of VEGF by SMC in denuded porcine arteries is evidence that this cytokine plays a role in the injury response of large arteries. Since several biological activities have been identified for VEGF, the function of this cytokine in the arterial repair process remains to be determined.
Collapse
Affiliation(s)
- S J Wysocki
- University Department of Surgery, Fremantle Hospital, Western Australia
| | | | | | | |
Collapse
|
1136
|
Esser S, Wolburg K, Wolburg H, Breier G, Kurzchalia T, Risau W. Vascular endothelial growth factor induces endothelial fenestrations in vitro. J Cell Biol 1998; 140:947-59. [PMID: 9472045 PMCID: PMC2141756 DOI: 10.1083/jcb.140.4.947] [Citation(s) in RCA: 433] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is an important regulator of vasculogenesis, angiogenesis, and vascular permeability. In contrast to its transient expression during the formation of new blood vessels, VEGF and its receptors are continuously and highly expressed in some adult tissues, such as the kidney glomerulus and choroid plexus. This suggests that VEGF produced by the epithelial cells of these tissues might be involved in the induction or maintenance of fenestrations in adjacent endothelial cells expressing the VEGF receptors. Here we describe a defined in vitro culture system where fenestrae formation was induced in adrenal cortex capillary endothelial cells by VEGF, but not by fibroblast growth factor. A strong induction of endothelial fenestrations was observed in cocultures of endothelial cells with choroid plexus epithelial cells, or mammary epithelial cells stably transfected with cDNAs for VEGF 120 or 164, but not with untransfected cells. These results demonstrate that, in these cocultures, VEGF is sufficient to induce fenestrations in vitro. Identical results were achieved when the epithelial cells were replaced by an epithelial-derived basal lamina-type extracellular matrix, but not with collagen alone. In this defined system, VEGF-mediated induction of fenestrae was always accompanied by an increase in the number of fused diaphragmed caveolae-like vesicles. Caveolae, but not fenestrae, were labeled with a caveolin-1-specific antibody both in vivo and in vitro. VEGF stimulation led to VEGF receptor tyrosine phosphorylation, but no change in the distribution, phosphorylation, or protein level of caveolin-1 was observed. We conclude that VEGF in the presence of a basal lamina-type extracellular matrix specifically induces fenestrations in endothelial cells. This defined in vitro system will allow further study of the signaling mechanisms involved in fenestrae formation, modification of caveolae, and vascular permeability.
Collapse
Affiliation(s)
- S Esser
- Max Planck Institut für Physiologische und Klinische Forschung, W.G. Kerckhoff Institut, Abteilung Molekulare Zellbiologie, D-61231 Bad Nauheim, Germany
| | | | | | | | | | | |
Collapse
|
1137
|
|
1138
|
Choi K, Kennedy M, Kazarov A, Papadimitriou JC, Keller G. A common precursor for hematopoietic and endothelial cells. Development 1998; 125:725-32. [PMID: 9435292 DOI: 10.1242/dev.125.4.725] [Citation(s) in RCA: 885] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Embryonic stem cell-derived embryoid bodies contain a unique precursor population which, in response to vascular endothelial growth factor, gives rise to blast colonies in semi-solid medium. Upon transfer to liquid culture with appropriate cytokines, these blast colonies generate both hematopoietic and adherent, stromal-type cells. Cells within the adherent population display characteristics of endothelial lineage including the expression of CD31, flk-1, flt-1, tie-2, the capacity to take up acetylated LDL and the presence of cytoplasmic Weibel-Palade bodies. Mixing studies demonstrated that the hematopoietic and endothelial precursors within the blast colonies develop from the same cell, the blast colony-forming cell. Kinetic analysis showed that the blast colony-forming cell represents a transient cell population that develops early and is lost quickly during embryoid body development. These findings provide strong evidence that the blast colony-forming cell represents the long-hypothesized hemangioblast, the common precursor of the hematopoietic and endothelial lineages.
Collapse
Affiliation(s)
- K Choi
- Department of Pathology, University of Maryland at Baltimore, Baltimore, MD 21201, USA.
| | | | | | | | | |
Collapse
|
1139
|
Davis-Smyth T, Presta LG, Ferrara N. Mapping the charged residues in the second immunoglobulin-like domain of the vascular endothelial growth factor/placenta growth factor receptor Flt-1 required for binding and structural stability. J Biol Chem 1998; 273:3216-22. [PMID: 9452434 DOI: 10.1074/jbc.273.6.3216] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Flt-1 is one of two receptor tyrosine kinases through which the angiogenic factor vascular endothelial growth factor (VEGF) functions. Placenta growth factor (PlGF) is an additional ligand for Flt-1. The second immunoglobulin-like domain in the extracellular domain of Flt-1 has previously been identified as the region containing the critical ligand-binding determinants. We analyzed the contribution of charged residues within the first three domains of Flt-1 to ligand binding by alanine-scanning mutagenesis. Domain 2 residues Arg159, Glu208 and His223-Arg224 (together) affect both VEGF and PlGF binding, while Glu137, Lys171, His223, and Arg224 affect PlGF but not VEGF. Several charged residues, especially Asp187, are important in maintaining the structural integrity of domain 2. In addition, some residues in domain 3 contribute to binding (Asp231) or provide for additional discrimination between ligands (Arg280-Asp283).
Collapse
Affiliation(s)
- T Davis-Smyth
- Departments of Cardiovascular Research, Genentech, Inc., South San Francisco, California 94080, USA
| | | | | |
Collapse
|
1140
|
Sibilia M, Steinbach JP, Stingl L, Aguzzi A, Wagner EF. A strain-independent postnatal neurodegeneration in mice lacking the EGF receptor. EMBO J 1998; 17:719-31. [PMID: 9450997 PMCID: PMC1170421 DOI: 10.1093/emboj/17.3.719] [Citation(s) in RCA: 245] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mice lacking the epidermal growth factor receptor (EGFR) exhibit strain-dependent phenotypes ranging from placental to postnatal skin, lung and brain defects. After birth, all mutant mice develop a progressive neurodegeneration in the frontal cortex, olfactory bulb and thalamus, characterized by massive apoptosis and upregulation of c-fos. These defects occur in a strain-independent manner, since neither rescue of the placental phenotype by aggregation of diploid 129/Sv EGFR mutant and tetraploid wild-type embryos, nor promotion of lung maturation by transplacental dexamethasone administration alters the course of neurodegeneration. VEGF is not induced during the degenerative process, excluding hypoxia and ischemia as causes of cell death. A migratory disorder is detected in the hippocampus with nests of ectopic neurons, which are also apoptotic. Cerebral cortices from EGFR mutants contain lower numbers of GFAP positive astrocytes, which display reduced proliferation in vitro. Since EGFR is expressed in the affected cell-types, these results define a specific function for EGFR in the proliferation and/or differentiation of astrocytes and in the survival of postmitotic neurons.
Collapse
Affiliation(s)
- M Sibilia
- Research Institute of Molecular Pathology (IMP), Dr Bohr-Gasse 7, A-1030 Vienna, Austria
| | | | | | | | | |
Collapse
|
1141
|
Jensen RL. Growth factor-mediated angiogenesis in the malignant progression of glial tumors: a review. SURGICAL NEUROLOGY 1998; 49:189-95; discussion 196. [PMID: 9457270 DOI: 10.1016/s0090-3019(97)00218-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND We review the role of peptide growth factors in angiogenesis and progression of low grade glial tumors to higher grade glioblastoma multiforme (GBM). METHODS Vascular pathology is a key feature of glioblastoma multiforme characterized by hypervascularity, vascular permeability, and hypercoagulability. RESULTS Vascular endothelial growth factor (VEGF) can mediate all of these effects, but by itself does not promote malignant growth. Epidermal growth factor (EGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), and transforming growth factor beta (TGF-beta) are implicated in the angiogenesis of a number of tumors including those of glial origin. CONCLUSIONS These growth factors are suggested to play a role in autocrine and/or paracrine mediated tumorogenesis of astrocytic tumors. VEGF secretion might be the product of induction by physiologic concentrations of other growth factors with VEGF being the common pathway of neovascularization and progression to GBM.
Collapse
Affiliation(s)
- R L Jensen
- Department of Neurosurgery, Loyola University of Chicago Medical Center, Maywood, IL 60153, USA
| |
Collapse
|
1142
|
Wen Y, Edelman JL, Kang T, Zeng N, Sachs G. Two functional forms of vascular endothelial growth factor receptor-2/Flk-1 mRNA are expressed in normal rat retina. J Biol Chem 1998; 273:2090-7. [PMID: 9442048 DOI: 10.1074/jbc.273.4.2090] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is an important mediator of ocular neovascularization by exerting its endothelial specific mitogenic effects through high affinity tyrosine kinase receptors. By screening a rat retina cDNA library, we have isolated a clone encoding the full-length prototypic form of the rat VEGF receptor-2/Flk-1, as well as a short form of the mRNA that encodes the complete seven N-terminal immunoglobulin-like extracellular ligand-binding domains, transmembrane region, NH2-terminal half of the intracellular kinase domain, and kinase insert domain but does not encode the COOH-terminal half of the intracellular kinase domain and carboxyl-terminal region. Both forms of mRNA are detected in rat retina, although the short form is expressed at a lower level. VEGF induced a biphasic increase of cytoplasmic calcium with both forms in HK 293 transfected cells, indicating that both forms of the VEGF receptor-2/Flk-1 are functional and that the COOH-terminal half of the intracellular kinase domain and carboxyl region of VEGF receptor-2/Flk-1 are not strictly necessary for either ligand binding or this biological activity.
Collapse
Affiliation(s)
- Y Wen
- Department of Medicine, West Los Angeles VA Medical Center, California, USA
| | | | | | | | | |
Collapse
|
1143
|
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 U S A 1998; 95:548-53. [PMID: 9435229 PMCID: PMC18457 DOI: 10.1073/pnas.95.2.548] [Citation(s) in RCA: 859] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We have identified a member of the VEGF family by computer-based homology searching and have designated it VEGF-D. VEGF-D is most closely related to VEGF-C by virtue of the presence of N- and C-terminal extensions that are not found in other VEGF family members. In adult human tissues, VEGF-D mRNA is most abundant in heart, lung, skeletal muscle, colon, and small intestine. Analyses of VEGF-D receptor specificity revealed that VEGF-D is a ligand for both VEGF receptors (VEGFRs) VEGFR-2 (Flk1) and VEGFR-3 (Flt4) and can activate these receptors. However. VEGF-D does not bind to VEGFR-1. Expression of a truncated derivative of VEGF-D demonstrated that the receptor-binding capacities reside in the portion of the molecule that is most closely related in primary structure to other VEGF family members and that corresponds to the mature form of VEGF-C. In addition, VEGF-D is a mitogen for endothelial cells. The structural and functional similarities between VEGF-D and VEGF-C define a subfamily of the VEGFs.
Collapse
Affiliation(s)
- M G Achen
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
1144
|
Murohara T, Horowitz JR, Silver M, Tsurumi Y, Chen D, Sullivan A, Isner JM. Vascular endothelial growth factor/vascular permeability factor enhances vascular permeability via nitric oxide and prostacyclin. Circulation 1998; 97:99-107. [PMID: 9443437 DOI: 10.1161/01.cir.97.1.99] [Citation(s) in RCA: 355] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF), an endothelial cell mitogen that promotes angiogenesis, was initially identified as a vascular permeability factor (VPF). Abundant evidence suggests that angiogenesis is preceded and/or accompanied by enhanced microvascular permeability. The mechanism by which VEGF/VPF increases vascular permeability (VP), however, has remained enigmatic. Accordingly, we used an in vivo assay of VP (Miles assay) to study the putative mediators of VEGF/VPF-induced permeability. METHODS AND RESULTS VEGF/VPF and positive controls (platelet-activating factor [PAF], histamine, and bradykinin) all increased vascular permeability. Prior administration of the tyrosine kinase inhibitors genistein or herbimycin A prevented VEGF/VPF-induced permeability. Placenta growth factor, which binds to Flt-1/VEGF-R1 but not Flk-1/KDR/VEGF-R2 receptor tyrosine kinase, failed to increase permeability. Other growth factors such as basic fibroblast growth factor (FGF), acidic FGF, platelet-derived growth factor-BB, transforming growth factor-beta, scatter factor, and granulocyte macrophage-colony stimulating factor (8 to 128 ng) failed to increase permeability. VEGF/VPF-induced permeability was significantly attenuated by the nitric oxide (NO) synthase inhibitors N(omega)-nitro-L-arginine (10 mg/kg) or N(omega)-nitro-L-arginine methyl ester (20 mg/kg) and the cyclooxygenase inhibitor indomethacin (5 mg/kg). The inactive enantiomer N(omega)-nitro-D-arginine methyl ester (20 mg/kg) did not inhibit VEGF/VPF-induced permeability. In vitro studies confirmed that VEGF/VPF stimulates synthesis of NO and prostaglandin metabolites in microvascular endothelial cells. Finally, NO donors and the prostacyclin analogue taprostene administered together but not alone reproduced the increase in permeability observed with VEGF/VPF. CONCLUSIONS These results implicate NO and prostacyclin produced by the interaction of VEGF/VPF with its Flk-1/KDR/VEGF-R2 receptor as mediators of VEGF/VPF-induced vascular permeability. Moreover, this property appears unique to VEGF/VPF among angiogenic cytokines.
Collapse
Affiliation(s)
- T Murohara
- Department of Medicine, St Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135, USA
| | | | | | | | | | | | | |
Collapse
|
1145
|
Lelkes PI, Hahn KA, Karmiol S, Schmidt DH. Hypoxia/Reoxygenation Enhances Tube Formation of Cultured Human Microvascular Endothelial Cells: the Role of Reactive Oxygen Species. Angiogenesis 1998. [DOI: 10.1007/978-1-4757-9185-3_31] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
1146
|
Fuchs M, Wang H, Ciossek T, Chen Z, Ullrich A. Differential expression of MAM-subfamily protein tyrosine phosphatases during mouse development. Mech Dev 1998; 70:91-109. [PMID: 9510027 DOI: 10.1016/s0925-4773(97)00179-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The MAM-subfamily of type II transmembrane protein tyrosine phosphatases (PTPases) currently comprises the enzymes PTPkappa, PTPmu and PCP2. In an effort to elucidate the individual physiological roles of these closely related proteins we performed a detailed analysis of their mRNA transcript distributions at different stages of mouse embryogenesis and postnatal brain development. Our in situ hybridization studies revealed distinct and complementary expression patterns of PTPkappa, PTPmu and PCP2 transcripts. Based on our results and previous reports we discuss MAM-PTPases as a new class of morphoregulatory molecules.
Collapse
Affiliation(s)
- M Fuchs
- Department of Molecular Biology, Max-Planck-Institut für Biochemie, Martinsried, Germany
| | | | | | | | | |
Collapse
|
1147
|
Noiri E, Lee E, Testa J, Quigley J, Colflesh D, Keese CR, Giaever I, Goligorsky MS. Podokinesis in endothelial cell migration: role of nitric oxide. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:C236-44. [PMID: 9458733 DOI: 10.1152/ajpcell.1998.274.1.c236] [Citation(s) in RCA: 139] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previously, we demonstrated the role of nitric oxide (NO) in transforming epithelial cells from a stationary to locomoting phenotype [E. Noiri, T. Peresleni, N. Srivastava, P. Weber, W.F. Bahou, N. Peunova, and M. S. Goligorsky. Am. J. Physiol. 270 (Cell Physiol. 39): C794-C802, 1996] and its permissive function in endothelin-1-stimulated endothelial cell migration (E. Noiri, Y. Hu, W. F. Bahou; C. Keese, I. Giaever, and M. S. Goligorsky, J. Biol: Chem. 272: 1747-1753, 1997). In the present study, the role of functional NO synthase in executing the vascular endothelial growth factor (VEGF)-guided program of endothelial cell migration and angiogenesis was studied in two independent experimental settings. First, VEGF, shown to stimulate NO release from simian virus 40-immortalized microvascular endothelial cells, induced endothelial cell transwell migration, whereas NG-nitro-L-arginine methyl ester (L-NAME) or antisense oligonucleotides to endothelial NO synthase suppressed this effect of VEGF. Second, in a series of experiments on endothelial cell wound healing, the rate of VEGF-stimulated cell migration was significantly blunted by the inhibition of NO synthesis. To gain insight into the possible mode of NO action, we next addressed the possibility that NO modulates cell matrix adhesion by performing impedance analysis of endothelial cell monolayers subjected to NO. The data showed the presence of spontaneous fluctuations of the resistance in ostensibly stationary endothelial cells. Spontaneous oscillations were induced by NO, which also inhibited cell matrix adhesion. This process we propose to term "podokinesis" to emphasize a scalar from of micromotion that, in the presence of guidance cues, e.g., VEGF, is transformed to a vectorial movement. In conclusion, execution of the program for directional endothelial cell migration requires two coexisting messages: NO-induced podokinesis (scalar motion) and guidance cues, e.g., VEGF, which imparts a vectorial component to the movement. Such a requirement for the dual signaling may explain a mismatch in the demand and supply with newly formed vessels in different pathological states accompanied by the inhibition of NO synthase.
Collapse
Affiliation(s)
- E Noiri
- Department of Medicine, State University of New York, Stony Brook 11794-8152, USA
| | | | | | | | | | | | | | | |
Collapse
|
1148
|
Robbins SG, Rajaratnam VS, Penn JS. Evidence for upregulation and redistribution of vascular endothelial growth factor (VEGF) receptors flt-1 and flk-1 in the oxygen-injured rat retina. Growth Factors 1998; 16:1-9. [PMID: 9777366 DOI: 10.3109/08977199809017487] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
There is considerable evidence that vascular endothelial growth factor (VEGF) is important in the pathogenesis of retinal neovascular diseases. The effects of this endothelial cell-specific mitogen are mediated by specific cell surface receptors. In this study we probed for the two VEGF receptors (VEGFRs) known to have highest affinity in the rat--flt-1 and flk-1. Using a well-characterized rat model of the neovascular disease retinopathy of prematurity (ROP), we performed immunohistochemical assays on methacrylate sections of eyes from normal and oxygen-injured animals at the time neovascularization is first observed (16 days of age) and at its peak (day 20). In day 16 room air retinas there was light, diffuse labeling of the inner nuclear layer and outer plexiform layer. In contrast, in 4 of 5 oxygen-injured eyes on day 16, there was specific labeling of small neovascular growths and normal retinal vessels, and the outermost (sclerad) limit of the label had shifted inward to the vitread border of the inner nuclear layer and the inner plexiform layer. Day 20 room air eyes showed a pattern similar to day 16, although with stronger labeling. However, in oxygen-injured eyes on day 20 the labeling pattern had shifted toward the vitreous, with extremely strong labeling of the preretinal neovascular growths. As on day 16 there was also labeling of the inner plexiform layer and the inner portion of the inner nuclear layer, but not the outer plexiform layer. Comparison of VEGF protein immunolabel with both of the VEGFR immunolabels revealed overlap and strong similarity on day 20 in the oxygen-injured eyes. This is the first report of VEGF receptor protein being concentrated in preretinal neovascular growths in a model of ROP. These results lend themselves to further investigation of the roles of VEGFRs in preretinal neovascularization in ROP and other retinal diseases and suggest avenues of research toward therapies using VEGFR antagonists.
Collapse
Affiliation(s)
- S G Robbins
- Arkansas Center for Eye Research, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock 72205, USA
| | | | | |
Collapse
|
1149
|
Abstract
This chapter describes the earlier stages of development of the vertebrate metanephric kidney. It focuses on the mouse and descriptive morphology is used for considering both molecular mechanisms, underpinning kidney morphogenesis and differentiation, and the ways in which these processes can go awry and lead to congenital kidney disorders—particularly in humans. The mature kidney is a fairly complex organ attached to an arterial input vessel and two output vessels, the vein and the ureter. Inside, the artery and vein are connected by a complex network of capillaries that invade a large number of glomeruli, the proximal entrance to nephrons, which are filtration units that link to an arborized collecting-duct system that drains into the ureter. The ability of the kidney and isolated metanephrogenic mesenchyme, to develop in culture means that the developing tissues can be subjected to a wide variety of experimental procedures designed to investigate their molecular and cellular properties and to test hypotheses about developmental mechanisms.
Collapse
Affiliation(s)
- J A Davies
- Centre for Developmental Biology, University of Edinburgh, United Kingdom
| | | |
Collapse
|
1150
|
Hidai C, Zupancic T, Penta K, Mikhail A, Kawana M, Quertermous EE, Aoka Y, Fukagawa M, Matsui Y, Platika D, Auerbach R, Hogan BL, Snodgrass R, Quertermous T. Cloning and characterization of developmental endothelial locus-1: an embryonic endothelial cell protein that binds the alphavbeta3 integrin receptor. Genes Dev 1998; 12:21-33. [PMID: 9420328 PMCID: PMC529342 DOI: 10.1101/gad.12.1.21] [Citation(s) in RCA: 192] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We have taken advantage of an enhancer trap event in a line of transgenic mice to identify a unique developmentally regulated endothelial cell locus (Del1). The protein encoded in this locus contains three EGF-like repeats homologous to those in Notch and related proteins, including an EGF-like repeat that contains an RGD motif, and two discoidin I-like domains. Del1 is shown to be a matrix protein and to promote adhesion of endothelial cells through interaction with the alphavbeta3 integrin receptor. Embryonic endothelial-like yolk sac cells expressing recombinant Del1 protein, or grown on an extracellular matrix containing Del1 protein, are inhibited from forming vascular-like structures. Expression of Del1 protein in the chick chorioallantoic membrane leads to loss of vascular integrity and promotes vessel remodeling. Del1 is thus a new ligand for the alphavbeta3 integrin receptor and may function to regulate vascular morphogenesis or remodeling in embryonic development.
Collapse
Affiliation(s)
- C Hidai
- Tokyo Women's Medical College, Tokyo 162, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|