1201
|
Kabrun N, Bühring HJ, Choi K, Ullrich A, Risau W, Keller G. Flk-1 expression defines a population of early embryonic hematopoietic precursors. Development 1997; 124:2039-48. [PMID: 9169850 DOI: 10.1242/dev.124.10.2039] [Citation(s) in RCA: 227] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have investigated the expression pattern of the Flk-1 receptor tyrosine kinase in mouse embryonic and fetal hematopoietic tissues as well as on hematopoietic precursor cells derived from these tissues. RNA analysis indicated that flk-1 was expressed in the yolk sac at day 10 of gestation, in the whole embryo at day 10 and 12 of gestation, in the liver throughout fetal life and in embryoid bodies (EBs) generated from ES cells differentiated in culture. Flk-1 message was also detected in erythroid and macrophage colonies generated from precursors of yolk sac, fetal liver, adult marrow and EB origin. Using an antibody directed against the extracellular portion of the molecule we have found that up to 50% of cells from EBs differentiated for 4 days express Flk-1. Following the development of this early Flk-1+ population the number of receptor-positive cells declines progressively to represent less than 5% of the EBs by day 12 of differentiation. Kinetic analysis revealed that the establishment of the EB Flk-1+ population precedes the development of cells which express CD34, Ly6A (Sca-1) and AA4.1. Cell sorting experiments demonstrated that all day-4 EB-derived hematopoietic precursors are Flk-1+ whereas greater than 95% of those found within the day-12 EBs are Flk-1-, suggesting that the precursor population which expresses this receptor represents an early but transient wave of hematopoietic development. Analysis of yolk sac and whole embryos at day 8.5 of gestation revealed a small but distinct Flk-1+ population that contained hematopoietic precursors. Day-12.5 fetal liver contained few Flk-1+ cells that showed little hematopoietic potential. Together these findings indicate that Flk-1 is expressed on an early population of hematopoietic precursors that may represent the onset of embryonic hematopoiesis.
Collapse
Affiliation(s)
- N Kabrun
- National Jewish Medical and Research Center, Denver, CO 80206, USA
| | | | | | | | | | | |
Collapse
|
1202
|
Provias J, Claffey K, delAguila L, Lau N, Feldkamp M, Guha A. Meningiomas: role of vascular endothelial growth factor/vascular permeability factor in angiogenesis and peritumoral edema. Neurosurgery 1997; 40:1016-26. [PMID: 9149260 DOI: 10.1097/00006123-199705000-00027] [Citation(s) in RCA: 166] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE Vascular endothelial growth factor (VEGF)/vascular permeability factor (VPF) is a potent angiogenic growth factor implicated in the tumor angiogenesis/metastasis of a number of human cancers. Activation of receptors for VEGF/VPF is specifically mitogenic to endothelial cells and increases their permeability. Although extensive literature exists regarding VEGF/VPF in human astrocytomas, little is known about its potential biological role(s) in meningiomas. Our interest in meningiomas was initiated by the observation that some meningiomas are extremely vascular and are occasionally associated with a considerable degree of peritumoral brain edema, both potentially related to the biological attributes of VEGF/VPF. METHODS As a first test of this hypothesis, we examined a cohort of 18 meningiomas for expression of VEGF/VPF at the messenger ribonucleic acid and protein levels and correlated expression with pathological characteristics, vascularity, and degree of peritumoral edema. RESULTS The majority of meningiomas expressed VEGF/VPF at both the messenger ribonucleic acid and protein levels. Corresponding serial sections were stained with an endothelial cell marker to obtain a microvascular density count, which positively correlated (P = 0.0005) with expression of VEGF/VPF. Furthermore, meningiomas with a large amount of peritumoral edema, as determined from the preoperative computed tomographic scans or magnetic resonance imaging scans, had elevated expression of VEGF/VPF (P = 0.05). CONCLUSION These data suggest that VEGF/VPF may play a role in both meningioma vascularity and peritumoral edema.
Collapse
Affiliation(s)
- J Provias
- Toronto Hospital, Department of Pathology (Neuropathology), University of Toronto, Canada
| | | | | | | | | | | |
Collapse
|
1203
|
Ema M, Taya S, Yokotani N, Sogawa K, Matsuda Y, Fujii-Kuriyama Y. A novel bHLH-PAS factor with close sequence similarity to hypoxia-inducible factor 1alpha regulates the VEGF expression and is potentially involved in lung and vascular development. Proc Natl Acad Sci U S A 1997; 94:4273-8. [PMID: 9113979 PMCID: PMC20712 DOI: 10.1073/pnas.94.9.4273] [Citation(s) in RCA: 788] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We have isolated and characterized a cDNA for a novel Per-Arnt/AhR-Sim basic helix-loop-helix (bHLH-PAS) factor that interacts with the Ah receptor nuclear translocator (Arnt), and its predicted amino acid sequence exhibits significant similarity to the hypoxia-inducible factor 1alpha (HIF1alpha) and Drosophila trachealess (dTrh) gene product. The HIF1alpha-like factor (HLF) encoded by the isolated cDNA bound the hypoxia-response element (HRE) found in enhancers of genes for erythropoietin, vascular endothelial growth factor (VEGF), and various glycolytic enzymes, and activated transcription of a reporter gene harboring the HRE. Although transcription-activating properties of HLF were very similar to those reported for HIF1alpha, their expression patterns were quite different between the two factors; HLF mRNA was most abundantly expressed in lung, followed by heart, liver, and other various organs under normoxic conditions, whereas HIF1alpha mRNA was ubiquitously expressed at much lower levels. In lung development around parturition, HLF mRNA expression was markedly enhanced, whereas that of HIF1alpha mRNA remained apparently unchanged at a much lower level. Moreover, HLF mRNA expression was closely correlated with that of VEGF mRNA. Whole mount in situ hybridization experiments demonstrated that HLF mRNA was expressed in vascular endothelial cells at the middle stages (9.5 and 10.5 days postcoitus) of mouse embryo development, where HIF1alpha mRNA was almost undetectable. The high expression level of HLF mRNA in the O2 delivery system of developing embryos and adult organs suggests that in a normoxic state, HLF regulates gene expression of VEGF, various glycolytic enzymes, and others driven by the HRE sequence, and may be involved in development of blood vessels and the tubular system of lung.
Collapse
Affiliation(s)
- M Ema
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-77, Japan
| | | | | | | | | | | |
Collapse
|
1204
|
Barleon B, Totzke F, Herzog C, Blanke S, Kremmer E, Siemeister G, Marmé D, Martiny-Baron G. Mapping of the sites for ligand binding and receptor dimerization at the extracellular domain of the vascular endothelial growth factor receptor FLT-1. J Biol Chem 1997; 272:10382-8. [PMID: 9099677 DOI: 10.1074/jbc.272.16.10382] [Citation(s) in RCA: 148] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The vascular endothelial growth factor (VEGF) receptor FLT-1 has been shown to be involved in vasculogenesis and angiogenesis. The receptor is characterized by seven Ig-like loops within the extracellular domain. Upon VEGF binding FLT-1 becomes phosphorylated, which has been thought to be preceded by receptor dimerization. To further investigate high affinity binding of VEGF to FLT-1 and ligand-induced receptor dimerization, we expressed in Sf9 cells the entire extracellular domain comprising all seven Ig-like loops: sFLT-1(7) and several truncated mutants consisting of loop one, one and two, one to three, one to four, and one to five. The corresponding proteins, named sFLT-1(1), (2), (3), (4), and (5) were purified. Only mutants sFLT-1(3) to (7) were able to bind 125I-VEGF with high affinity. No binding of VEGF was observed with sFLT-1(1) and sFLT-1(2), indicating that the first three Ig-like loops are involved in high affinity binding of VEGF. The binding of VEGF to sFLT-1(3) could be competed with placenta growth factor (PlGF), a VEGF-related ligand, suggesting that high affinity binding of VEGF and PlGF is mediated by the same or closely related contact sites on sFLT-1. Deglycosylation of the sFLT-1(3), (4), (5), and (7) did not abolish VEGF binding. Furthermore, unglycosylated sFLT-1(3), expressed in Escherichia coli, was able to bind VEGF with similar affinity as sFLT-1(3) or sFLT-1(7), both expressed in Sf9 cells. This indicates that receptor glycosylation is not essential for high affinity binding. Dimerization of the extracellular domains of FLT-1 upon addition of VEGF was detected with all mutants containing the Ig-like loop four. Although sFLT-1(3) was able to bind VEGF, dimerization of this mutant was inefficient, indicating that sites on Ig-like loop four are essential to stabilize receptor dimers.
Collapse
Affiliation(s)
- B Barleon
- Institute of Molecular Medicine, Tumor Biology Center, D-79106 Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
1205
|
Mak KH, Topol EJ. Clinical trials to prevent restenosis after percutaneous coronary revascularization. Ann N Y Acad Sci 1997; 811:255-84; discussion 284-8. [PMID: 9186603 DOI: 10.1111/j.1749-6632.1997.tb52007.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- K H Mak
- Department of Cardiology, Joseph J. Jacobs Center for Vascular Biology, Cleveland Clinic Foundation, Ohio 44195, USA
| | | |
Collapse
|
1206
|
Kennedy M, Firpo M, Choi K, Wall C, Robertson S, Kabrun N, Keller G. A common precursor for primitive erythropoiesis and definitive haematopoiesis. Nature 1997; 386:488-93. [PMID: 9087406 DOI: 10.1038/386488a0] [Citation(s) in RCA: 429] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The generation of blood cells, haematopoiesis, in the mouse embryo begins with the development of primitive nucleated erythroid cells in the yolk sac followed by the appearance of precursors for multiple definitive haematopoietic lineages. The later developing lineages arise from multipotential stem cells, but the relationship of primitive erythroid cells to these other haematopoietic populations is unknown. Using an in vitro embryonic stem (ES) cell differentiation system, we show that primitive erythrocytes and other haematopoietic lineages arise from a common multipotential precursor that develops within embryoid bodies generated from differentiated ES cells. In response to vascular endothelial growth factor and c-kit ligand these precursors give rise to colonies containing immature cells (blasts) expressing marker genes characteristic of haematopoietic precursors. Many blast colonies also expressed betaH1 and beta major globins but not Brachyury, a mesodermal marker. Kinetic analysis demonstrated that the blast colony-forming cells represent a transient population, preceding the establishment of the primitive erythroid and other lineage-restricted precursors. This precursor population may represent the earliest stage of embryonic haematopoietic commitment.
Collapse
Affiliation(s)
- M Kennedy
- The National Jewish Medical and Research Center, Denver, Colorado 80206, USA
| | | | | | | | | | | | | |
Collapse
|
1207
|
Sumoy L, Keasey JB, Dittman TD, Kimelman D. A role for notochord in axial vascular development revealed by analysis of phenotype and the expression of VEGR-2 in zebrafish flh and ntl mutant embryos. Mech Dev 1997; 63:15-27. [PMID: 9178253 DOI: 10.1016/s0925-4773(97)00671-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The notochord is required for the differentiation of nearby tissues, including the neural tube and the floor plate. Because the dorsal aorta and axial vein are midline structures, their development might also be influenced by the notochord. To investigate this possibility, we cloned zebrafish VEGR-2, homologous to the earliest known marker of endothelial cells in mammals. In flh and ntl mutant embryos, which lack a notochord, we found a defect in axial blood vessel formation, and a delay in the fusion of VEGR-2 positive endothelial progenitor cells into the primary vascular plexus and a block in the establishment of mature vessels. Differences in the vascular phenotype between the two mutations correlated with the severity of their axial mesodermal defects. These observations support a role for the notochord in vasculogenesis.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Blood Vessels/embryology
- Blood Vessels/physiology
- Cell Differentiation
- Cloning, Molecular
- Embryo, Nonmammalian/physiology
- Embryonic Induction
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiology
- Gene Expression Regulation, Developmental
- Humans
- Molecular Sequence Data
- Mutation
- Notochord/physiology
- Phenotype
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptor Protein-Tyrosine Kinases/chemistry
- Receptor Protein-Tyrosine Kinases/genetics
- Receptors, Growth Factor/biosynthesis
- Receptors, Growth Factor/chemistry
- Receptors, Growth Factor/genetics
- Receptors, Vascular Endothelial Growth Factor
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/chemistry
- Sequence Alignment
- Sequence Homology, Amino Acid
- Stem Cells/physiology
- Zebrafish/embryology
- Zebrafish/genetics
Collapse
Affiliation(s)
- L Sumoy
- Department of Biochemistry, University of Washington, Seattle 98195-7350, USA
| | | | | | | |
Collapse
|
1208
|
|
1209
|
Flamme I, Fröhlich T, von Reutern M, Kappel A, Damert A, Risau W. HRF, a putative basic helix-loop-helix-PAS-domain transcription factor is closely related to hypoxia-inducible factor-1 alpha and developmentally expressed in blood vessels. Mech Dev 1997; 63:51-60. [PMID: 9178256 DOI: 10.1016/s0925-4773(97)00674-6] [Citation(s) in RCA: 282] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Transcription factors of the bHLH-PAS protein family are important regulators of developmental processes such as neurogenesis and tracheal development in invertebrates. Recently a bHLH-PAS protein, named trachealess (trl) was identified as a master regulator of tracheogenesis. Hypoxia-inducible factor, HIF-1 alpha, is a vertebrate relative of trl which is likely to be involved in growth of blood vessels by the induction of vascular endothelial growth factor (VEGF) in response to hypoxia. In the present study we describe mRNA cloning and mRNA expression pattern of mouse HIF-related factor (HRF), a novel close relative of HIF-1 alpha which is expressed most prominently in brain capillary endothelial cells and other blood vessels as well as in bronchial epithelium in the embryo and the adult. In addition, smooth muscle cells of the uterus, neurons, brown adipose tissue and various epithelial tissues express HRF mRNA as well. High expression levels of HRF mRNA in embryonic choroid plexus and kidney glomeruli, places where VEGF is highly expressed, suggest a role of this factor in VEGF gene activation similar to that of HIF-1 alpha. Given the similarity between morphogenesis of the tracheal system and the vertebrate vascular system, the expression pattern of HRF in the vasculature and the bronchial tree raises the possibility that this family of transcription factors may be involved in tubulogenesis.
Collapse
Affiliation(s)
- I Flamme
- Max-Planck-Institut für physiologische, Bad Nauheim, Germany
| | | | | | | | | | | |
Collapse
|
1210
|
Patterson C, Wu Y, Lee ME, DeVault JD, Runge MS, Haber E. Nuclear protein interactions with the human KDR/flk-1 promoter in vivo. Regulation of Sp1 binding is associated with cell type-specific expression. J Biol Chem 1997; 272:8410-6. [PMID: 9079666 DOI: 10.1074/jbc.272.13.8410] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The endothelial cell type-specific tyrosine kinase KDR/flk-1 is a receptor for vascular endothelial growth factor and a critical regulator of endothelial cell growth and development. To study mechanisms of endothelial cell differentiation and gene regulation, we have analyzed the topology of the proximal promoter of human KDR/flk-1. A protected sequence between base pairs -110 and -25 was defined by in vitro DNase I footprinting analysis in human umbilical vein endothelial cells (HUVECs). Purified Sp1 alone produced similar protection, and electrophoretic mobility shift assays demonstrated that Sp1 was indeed the major nuclear protein binding to this region. Despite the cell type specificity of KDR/flk-1 expression, no cell type differences were observed in DNA-protein interactions in vitro. In contrast, in vivo footprinting assays demonstrated marked differences in core promoter interactions between cell types. Protection of Sp1 binding sites was observed in HUVECs by in vivo DNase I footprinting, whereas in human fibroblasts and HeLa cells a pattern consistent with nucleosomal positioning was observed. In vivo dimethylsulfate footprinting confirmed that DNA-protein interactions occurred within Sp1 elements in HUVECs but not in nonendothelial cells. It is possible that distant elements coordinate Sp1 binding and chromatin structure to regulate cell type-specific expression of KDR/flk-1.
Collapse
Affiliation(s)
- C Patterson
- Division of Cardiology, University of Texas Medical Branch, Galveston, Texas 77555-1064, USA.
| | | | | | | | | | | |
Collapse
|
1211
|
Koblizek TI, Runting AS, Stacker SA, Wilks AF, Risau W, Deutsch U. Tie2 receptor expression and phosphorylation in cultured cells and mouse tissues. EUROPEAN JOURNAL OF BIOCHEMISTRY 1997; 244:774-9. [PMID: 9108247 DOI: 10.1111/j.1432-1033.1997.00774.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Accumulating experimental evidence indicates that endothelial cell growth and blood vessel morphogenesis are processes that are governed by the activity of specifically expressed receptor tyrosine kinases (RTKs). We have used two new rat monoclonal antibodies (mAbs) to study the expression and phosphorylation of one such receptor, mouse Tie2 (tyrosine kinase that contains immunoglobulin-like loops and epidermal-growth-factor-similar domains 2]), in transfected cells, endothelioma cell lines and mouse tissues. The Tie2 receptor was found to be constitutively autophosphorylated when over-expressed in COS7 cells. In contrast, the endogenous Tie2 protein was not phosphorylated in endothelioma cell lines. However, in these cell lines, Tie2 could be induced to become tyrosine phosphorylated, and this activation was found to be independent of Tie1. Studying Tie2 receptor activity during angiogenesis in mouse development, the receptor was only weakly phosphorylated in the early postnatal mouse brain whereas a stronger activation could be detected in mouse embryos at day 10.5 post coitum.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Brain/embryology
- Brain/metabolism
- COS Cells
- Cells, Cultured
- Female
- Gene Expression
- Mice
- Mice, Knockout
- Phosphorylation
- Pregnancy
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/immunology
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, TIE-1
- Receptor, TIE-2
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, TIE
- Tissue Distribution
- Transfection
Collapse
Affiliation(s)
- T I Koblizek
- Department of Molecular Cell Biology, Max Planck Institute for Physiological and Clinical Research, Bad Nauheim, Germany
| | | | | | | | | | | |
Collapse
|
1212
|
Okazaki T, Sharma HS, Aikawa M, Yamataka A, Nagai R, Miyano T, Tibboel D. Pulmonary expression of vascular endothelial growth factor and myosin isoforms in rats with congenital diaphragmatic hernia. J Pediatr Surg 1997; 32:391-4. [PMID: 9094000 DOI: 10.1016/s0022-3468(97)90588-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Abnormalities of the pulmonary vasculature are well documented in cases of congenital diaphragmatic hernia (CDH). Vascular endothelial growth factor (VEGF), an angiogenic factor, is a recently described endothelial cell-specific growth factor. Myosin heavy chain (MHC) isoforms such as SMemb, SM1 and SM2 are important molecular markers used to study vascular smooth muscle cell differentiation. SMemb is predominantly expressed in immature smooth muscle cells (SMC), and SM2 is expressed in mature SMCs. The authors investigated the expression of VEGF and SMC differentiation in pulmonary vessels in CDH rat lungs and in controls. The lungs of nitrofen-induced CDH rat fetuses (n = 16, gestational age 16, 18, 20, and 22 days) were stained immunohistochemically using antibodies against VEGF, SMemb and SM2, while alpha-actin was used as a general marker of vascular smooth muscle cells. In the CDH group VEGF expression was negative in pulmonary vessels before birth, and in the control group VEGF was positive in smooth muscle cells in vessel walls from 20 days both in vessels at the hilum and in pulmonary parenchyma. In both control and CDH groups, SMemb expression was positive from 16 days' gestation. SM2 expression was negative in vessel walls during the prenatal period in both groups. Alpha-actin was localized in both lungs obtained from control and CDH groups in the lung hilum from 16 days and around peripheral vessels from 18 days. Differences in vascular smooth muscle cell differentiation were not observed between control and CDH lung. These findings suggest that differences in pulmonary vascular development exist between control and CDH rats for VEGF expression, and maturational differences in smooth muscle cell differentiation are not present. This role of altered endothelial cell growth might be related to the different pulmonary vascular reactivity present in CDH lungs.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Disease Models, Animal
- Endothelial Growth Factors/metabolism
- Hernia, Diaphragmatic/complications
- Hernia, Diaphragmatic/embryology
- Hernia, Diaphragmatic/physiopathology
- Humans
- Hypertension, Pulmonary/complications
- Hypertension, Pulmonary/embryology
- Hypertension, Pulmonary/physiopathology
- Lymphokines/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myosin Heavy Chains/metabolism
- Rats
- Rats, Sprague-Dawley
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- T Okazaki
- Department of Pediatric Surgery, Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
1213
|
Abstract
Fibronectin (FN) is a cell adhesive extracellular matrix protein highly expressed in developing lungs. Although the role of FN in lung development remains to be elucidated, several observations suggest that it plays important roles in many developmental processes. In vitro, FN can affect the migration, proliferation, differentiation, and even apoptosis of various cell types, all considered necessary for organogenesis. The cellular effects of FN are primarily mediated by cell surface heterodimeric receptors of the integrin family. Ligand binding to these receptors triggers the activation of intracellular signaling events responsible for alterations in gene transcription and, ultimately, cell function. Immunohistochemical analysis of embryos revealed increased deposition of FN during the pseudoglandular stage of lung development, coinciding with the period of branching morphogenesis. This observation, together with the strategic location of FN around developing airways predominantly at cleft sites, suggests a role in airways formation. This hypothesis is supported by studies showing that reagents capable of preventing FN-FN receptor binding inhibit branching morphogenesis in murine lung explants. Less is known about the potential role of FN in other aspects of lung development. However, in vitro studies suggest that FN may affect vessel formation, alveolar epithelial cell differentiation and lung growth and maturation.
Collapse
Affiliation(s)
- J Roman
- Department of Medicine, Pulmonary & Critical Care Division, Atlanta Veterans Affairs Medical Center, GA 30033, USA
| |
Collapse
|
1214
|
Fouquet B, Weinstein BM, Serluca FC, Fishman MC. Vessel patterning in the embryo of the zebrafish: guidance by notochord. Dev Biol 1997; 183:37-48. [PMID: 9119113 DOI: 10.1006/dbio.1996.8495] [Citation(s) in RCA: 227] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We have cloned the zebrafish homolog of the receptor tyrosine kinase flk-1 to provide us with a tool to study normal vascular pattern formation in the developing zebrafish embryo and to compare it to mutants in which vascular pattern is perturbed. We find that during normal development the first angioblasts arise laterally in the mesoderm and then migrate medially to form the primordia of the large axial vessels, the dorsal aorta (axial artery) and the axial vein. Lumen formation occurs shortly before onset of circulation at 24 hr postfertilization. We examined the specification of vascular progenitors in the mutant cloche, which fails to form both vessels and blood. cloche lacks all flk-expressing cells and therefore appears to lack angioblasts. The axial vessels of the trunk form in close proximity to notochord and endoderm, which may provide cues for their formation. The dorsal aorta is normally just ventral to the notochord; the axial vein is just below the dorsal aorta and above the endoderm. floating head (flh) and no tail (ntl) mutants both have defects in the formation of notochord. Both are cell-autonomous lesions, flh abolishing notochord and ntl preventing its differentiation. In both mutants the dorsal aorta fails to form, while formation of the axial vein is less affected. Mosaic analysis of mutant embryos shows that transplanted wild-type cells can become notochord in mutant flh embryos. In these mosaic embryos flh cells expressing flk assemble at the midline, beneath the wild-type notochord, and form an aortic primordium. This suggests that signals from the notochord may guide angioblasts in the fashioning of the dorsal aorta. The notochord seems to be less important for the formation of the vein.
Collapse
Affiliation(s)
- B Fouquet
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown 02129, USA
| | | | | | | |
Collapse
|
1215
|
Cunningham SA, Stephan CC, Arrate MP, Ayer KG, Brock TA. Identification of the extracellular domains of Flt-1 that mediate ligand interactions. Biochem Biophys Res Commun 1997; 231:596-9. [PMID: 9070853 DOI: 10.1006/bbrc.1997.6156] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Vascular Endothelial Growth Factor (VEGF) mediates its actions through the Flt-1 and KDR(Flk-1) receptor tyrosine kinases. To localize the extracellular region of Flt-1 that is involved in ligand interactions, we prepared secreted fusion proteins between various combinations of its seven extracellular IgG-like folds. Ligand binding studies show that in combination, domains one and two (amino acids 1-234) are sufficient to achieve VEGF165 interactions. Either domain alone is insufficient to achieve this effect. However, Scatchard analysis reveals that despite the binding capabilities of this construct, the Kd is five fold lower than ligand binding to the full extracellular domain. We find that addition of domain three to this minimal site restores high affinity receptor binding. Further, we show that domains one and two are sufficient to achieve interactions of Flt-1 with Placental Growth Factor (PIGF-1).
Collapse
Affiliation(s)
- S A Cunningham
- Department of Pharmacology, Texas Biotechnology Corporation, Houston 77030, USA
| | | | | | | | | |
Collapse
|
1216
|
van der Zee R, Murohara T, Luo Z, Zollmann F, Passeri J, Lekutat C, Isner JM. Vascular endothelial growth factor/vascular permeability factor augments nitric oxide release from quiescent rabbit and human vascular endothelium. Circulation 1997; 95:1030-7. [PMID: 9054767 DOI: 10.1161/01.cir.95.4.1030] [Citation(s) in RCA: 278] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF)/ vascular permeability factor (VPF) is an endothelial cell (EC) mitogen. This feature is considered central to the documented role of VEGF/VPF in promoting angiogenesis. More recent evidence suggests that VEGF/VPF may also serve a "maintenance" function, modulating various aspects of EC biology. In the present study, we sought to determine the extent to which VEGF/VPF may stimulate the release of NO from normal ECs. METHODS AND RESULTS VEGF/VPF produced a dose-dependent rise in NO concentration ([NO]) from vascular segments of rabbit thoracic aorta, pulmonary artery, and inferior vena cava. In comparison to stimulation with acetylcholine, the onset of increased [NO] after administration of VEGF/VPF was slower, reaching a maximum value after 8 minutes. Preincubation of the aortic segments with L-arginine raised by twofold both baseline [NO] and [NO] stimulated by addition of 2.5 micrograms/mL VEGF/VPF. Removal of CaCl2 from the Krebs solution, disruption of the endothelium, and administration of NG-monomethyl-L-arginine abrogated the stimulatory effect of 10 micrograms/mL VEGF/VPF. Similar findings were documented with an NO-specific polarographic electrode to measure NO released from cultured human umbilical vein ECs. CONCLUSIONS VEGF/VPF stimulates production of NO from rabbit and human ECs. This finding (1) constitutes inferential evidence for the presence of functional VEGF/VPF receptors on quiescent endothelium of the adult rabbit as well as human ECs and (2) supports the notion that putative maintenance functions of VEGF/VPF may include regulation of baseline synthesis and/or release of EC NO.
Collapse
Affiliation(s)
- R van der Zee
- Department of Biomedical Research, St Elizabeth's Medical Center, Tufts University, School of Medicine, Boston, Mass 02135-2997, USA
| | | | | | | | | | | | | |
Collapse
|
1217
|
Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science 1997; 275:964-7. [PMID: 9020076 DOI: 10.1126/science.275.5302.964] [Citation(s) in RCA: 6334] [Impact Index Per Article: 226.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Putative endothelial cell (EC) progenitors or angioblasts were isolated from human peripheral blood by magnetic bead selection on the basis of cell surface antigen expression. In vitro, these cells differentiated into ECs. In animal models of ischemia, heterologous, homologous, and autologous EC progenitors incorporated into sites of active angiogenesis. These findings suggest that EC progenitors may be useful for augmenting collateral vessel growth to ischemic tissues (therapeutic angiogenesis) and for delivering anti- or pro-angiogenic agents, respectively, to sites of pathologic or utilitarian angiogenesis.
Collapse
MESH Headings
- Animals
- Antigens, CD34/analysis
- Biomarkers/analysis
- Cell Differentiation
- Cell Separation
- Cells, Cultured
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/cytology
- Flow Cytometry
- Hindlimb/blood supply
- Humans
- Ischemia/physiopathology
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Mice, Transgenic
- Neovascularization, Physiologic
- Nitric Oxide Synthase/analysis
- Rabbits
- Receptor Protein-Tyrosine Kinases/analysis
- Receptors, Growth Factor/analysis
- Receptors, Vascular Endothelial Growth Factor
- Stem Cells/chemistry
- Stem Cells/cytology
Collapse
Affiliation(s)
- T Asahara
- Department of Medicine (Cardiology), St. Elizabeth's Medical Center, Tufts University School of Medicine, 736 Cambridge Street, Boston, MA 02135, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1218
|
Oberg-Welsh C, Sandler S, Andersson A, Welsh M. Effects of vascular endothelial growth factor on pancreatic duct cell replication and the insulin production of fetal islet-like cell clusters in vitro. Mol Cell Endocrinol 1997; 126:125-32. [PMID: 9089650 DOI: 10.1016/s0303-7207(96)03977-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We have previously shown that the tyrosine kinase receptor Flk-1 and its ligand, vascular endothelial growth factor (VEGF), may play a role in the development of fetal rat islet-like structures in vitro, possibly by stimulating the maturation of endocrine precursor cells in the pancreatic ductal epithelium. In order to further assess this, adult rat pancreatic ducts and fetal porcine islet-like cell clusters (ICC) were cultured in the presence of VEGF. In ducts, VEGF stimulated the mitogenesis in the epithelium. Culture of ICC in the presence of VEGF significantly enhanced their insulin content, but decreased the insulin accumulation to the culture medium. Glucose-stimulated acute insulin release was not affected by VEGF. Northern blot analysis after partial pancreatectomy in adult rats revealed induction of VEGF mRNA 3 days after the operation. Immunohistochemistry of fetal rat pancreas showed staining mainly in the islets of Langerhans. We conclude that VEGF directly stimulates the replication of the ductal epithelium, a possible prerequisite for beta-cell formation. This could require local production of VEGF, which may alter in response to physiological demands.
Collapse
Affiliation(s)
- C Oberg-Welsh
- Department of Medical Cell Biology, Uppsala University, Sweden.
| | | | | | | |
Collapse
|
1219
|
Arbiser JL, Moses MA, Fernandez CA, Ghiso N, Cao Y, Klauber N, Frank D, Brownlee M, Flynn E, Parangi S, Byers HR, Folkman J. Oncogenic H-ras stimulates tumor angiogenesis by two distinct pathways. Proc Natl Acad Sci U S A 1997; 94:861-6. [PMID: 9023347 PMCID: PMC19604 DOI: 10.1073/pnas.94.3.861] [Citation(s) in RCA: 356] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/1996] [Indexed: 02/03/2023] Open
Abstract
The switch from a quiescent tumor to an invasive tumor is accompanied by the acquisition of angiogenic properties. This phenotypic change likely requires a change in the balance of angiogenic stimulators and angiogenic inhibitors. The nature of the angiogenic switch is not known. Here, we show that introduction of activated H-ras into immortalized endothelial cells is capable of activating the angiogenic switch. Angiogenic switching is accompanied by up-regulation of vascular endothelial growth factor and matrix metalloproteinase (MMP) bioactivity and downregulation of tissue inhibitor of MMP. Furthermore, we show that inhibition of phosphatidylinositol-3-kinase leads to partial inhibition of tumor angiogenesis, thus demonstrating that activated H-ras activates tumor angiogenesis through two distinct pathways. Finally, we show evidence for two forms of tumor dormancy.
Collapse
Affiliation(s)
- J L Arbiser
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1220
|
Masood R, Cai J, Zheng T, Smith DL, Naidu Y, Gill PS. Vascular endothelial growth factor/vascular permeability factor is an autocrine growth factor for AIDS-Kaposi sarcoma. Proc Natl Acad Sci U S A 1997; 94:979-84. [PMID: 9023368 PMCID: PMC19625 DOI: 10.1073/pnas.94.3.979] [Citation(s) in RCA: 217] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Kaposi sarcoma (KS) is the most common tumor associated with HIV-1 infection and develops in nearly 30% of cases. The principal features of this tumor are abnormal vascularization and the proliferation of endothelial cells and spindle (tumor) cells. KS-derived spindle cells induce vascular lesions and display enhanced vascular permeability when inoculated subcutaneously in the nude mouse. This finding suggests that angiogenesis and capillary permeability play a central role in the development and progression of KS. In this study, we show that AIDS-KS cell lines express higher levels of vascular endothelial growth factor/vascular permeability factor (VEGF/VGF) than either human umbilical vein endothelial cells or human aortic smooth muscle cells. AIDS-KS cells and primary tumor tissues also expressed high levels of Flt-1 and KDR, the receptors for VEGF, while the normal skin of the same patients did not show any expression. We further demonstrate that VEGF antisense oligonucleotides AS-1 and AS-3 specifically block VEGF mRNA and protein production and inhibit KS cell growth in a dose-dependent manner. Furthermore, growth of KS cells in nude mice was specifically inhibited by VEGF antisense oligonucleotides. These results show that VEGF is an autocrine growth factor for AIDS-KS cells. To our knowledge, this is the first report that shows that VEGF acts as a growth stimulator in a human tumor. Inhibitors of VEGF or its cognate receptors may thus be candidates for therapeutic intervention.
Collapse
MESH Headings
- AIDS-Related Opportunistic Infections/genetics
- AIDS-Related Opportunistic Infections/pathology
- Animals
- Endothelial Growth Factors/genetics
- Endothelium, Vascular
- Gene Expression Regulation, Neoplastic/physiology
- Humans
- Lymphokines/genetics
- Mice
- Mice, Nude
- Muscle, Smooth, Vascular
- Neoplasm Transplantation
- Oligonucleotides, Antisense
- Proto-Oncogene Proteins/genetics
- Receptor Protein-Tyrosine Kinases/genetics
- Receptors, Growth Factor/genetics
- Receptors, Vascular Endothelial Growth Factor
- Sarcoma, Kaposi/genetics
- Sarcoma, Kaposi/pathology
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factor Receptor-1
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- R Masood
- University of Southern California, School of Medicine, Los Angeles 90033, USA
| | | | | | | | | | | |
Collapse
|
1221
|
|
1222
|
Sandner P, Wolf K, Bergmaier U, Gess B, Kurtz A. Induction of VEGF and VEGF receptor gene expression by hypoxia: divergent regulation in vivo and in vitro. Kidney Int 1997; 51:448-53. [PMID: 9027720 DOI: 10.1038/ki.1997.60] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
This study examined the expression of EPO, VEGF and VEGF receptor gene under conditions of reduced oxygen supply in primary cultures of rat hepatocytes, and compared it with the expression of these genes in hypoxic rat livers in vivo. To this end we exposed male Sprague-Dawley rats to hypoxia (10% and 8% O2), carbon monoxide (0.1% CO) or injected cobalt chloride (60 mg/kg CoCl2) subcutaneously. For the in vitro experiments we used primary cultures of rat hepatocytes which were kept at high (20% O2) and low (1% O2) oxygen tensions for three hours. The EPO mRNA was up-regulated by hypoxia in vitro and in vivo about 10-fold. The VEGF mRNA was up-regulated fivefold in the hepatocytes only, whereas the in vivo mRNA levels remained unchanged. The mRNA levels of flt-1 were up-regulated threefold by 8% O2 in livers, dependent on the strength of hypoxia (10% caused no changes in flt-1 gene expression) and on the kind of hypoxic stimulus (8% O2 was as effective as 0.1% CO and more effective than cobalt). The mRNA levels of flk-1/KDR and flt-4 remained unchanged in the liver. In vitro there were no changes in the mRNA levels of flt-1, flt-4 and flk-1/KDR. Consequently, the in vivo regulation of VEGF, which might be modulated by induction of flt-1 receptor gene expression, differs from the in vitro cell culture situation and might be different from the EPO regulation in vivo.
Collapse
Affiliation(s)
- P Sandner
- Institut für Physiologie, Universität Regensburg, Germany.
| | | | | | | | | |
Collapse
|
1223
|
Affiliation(s)
- N Ferrara
- Department of Cardiovascular Research, Genentech, Inc., South San Francisco, California 94080, USA
| | | |
Collapse
|
1224
|
Gupta K, Ramakrishnan S, Browne PV, Solovey A, Hebbel RP. A novel technique for culture of human dermal microvascular endothelial cells under either serum-free or serum-supplemented conditions: isolation by panning and stimulation with vascular endothelial growth factor. Exp Cell Res 1997; 230:244-51. [PMID: 9024783 DOI: 10.1006/excr.1996.3421] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Several physiological and pathophysiological events involving vascular endothelium occur at the microvascular level. Studies on human microvasculature require homogenous primary cultures of microvascular endothelial cells. However, procedures available for isolating and culturing human dermal microvascular cells (HDMEC) result in significant contamination with fibroblasts. To eliminate contamination with fibroblasts or other cells, we developed a procedure to isolate HDMEC from neonatal human foreskin by panning the cells using EN4, an anti-endothelial cell monoclonal antibody. Panned cells uniformly expressed von Willebrand factor and CD36, confirming their microvascular endothelial characteristics, whereas cells cultured without panning showed a significant degree of contamination with fibroblasts. In the presence of vascular endothelial growth factor (VEGF), HDMEC could be cultured under serum-free conditions. VEGF stimulated the growth of HDMEC in a dose-dependent manner in serum-free medium or in media supplemented with either human serum or newborn calf serum. Since differences exist between large vessel endothelial cells and microvascular endothelial cells, we compared the response to VEGF stimulation of HDMEC with human umbilical vein endothelial cells (HUVEC). The dose response of the two cell types to VEGF was different. This effect of VEGF on endothelial cells may be mediated by the VEGF receptor kdr, since mRNA for kdr was detected using RT-PCR in both HDMEC and HUVEC. The procedure described in this study will make possible the culture of highly enriched HDMEC without contamination with fibroblasts and facilitate studies with these cells under defined assay conditions in a serum-free environment.
Collapse
Affiliation(s)
- K Gupta
- Department of Medicine, University of Minnesota Medical School, Minneapolis 55455, USA
| | | | | | | | | |
Collapse
|
1225
|
Möhle R, Green D, Moore MA, Nachman RL, Rafii S. Constitutive production and thrombin-induced release of vascular endothelial growth factor by human megakaryocytes and platelets. Proc Natl Acad Sci U S A 1997; 94:663-8. [PMID: 9012841 PMCID: PMC19570 DOI: 10.1073/pnas.94.2.663] [Citation(s) in RCA: 524] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We have shown that coculture of bone marrow microvascular endothelial cells with hematopoietic progenitor cells results in proliferation and differentiation of megakaryocytes. In these long-term cultures, bone marrow microvascular endothelial cell monolayers maintain their cellular integrity in the absence of exogenous endothelial growth factors. Because this interaction may involve paracrine secretion of cytokines, we evaluated megakaryocytic cells for secretion of cytokines, we evaluated megakaryocytic cells for secretion of vascular endothelial growth factor (VEGF). Megakaryocytes (CD41a+) were generated by ex vivo expansion of hematopoietic progenitor cells with kit-ligand and thrombopoietin for 10 days and further purified with immunomagnetic microbeads. Using reverse transcription-PCR, we showed that megakaryocytic cell lines (Dami, HEL) and purified megakaryocytes expressed mRNA of the three VEGF isoforms (121, 165, and 189 amino acids). Large quantities of VEGF (> 1 ng/10(6) cells/3 days) were detected in the supernatant of Dami cells, ex vivo-generated megakaryocytes, and CD41a+ cells isolated from bone marrow. The constitutive secretion of VEGF by CD41a+ cells was stimulated by growth factors of the megakaryocytic lineage (interleukin 3, thrombopoietin). Western blotting of heparin-Sepharose-enriched supernatant mainly detected the isoform VEGF165. In addition, immunohistochemistry showed intracytoplasmic VEGF in polyploid megakaryocytes. Thrombin stimulation of megakaryocytes and platelets resulted in rapid release of VEGF within 30 min. We conclude that human megakaryocytes produce and secrete VEGF in an inducible manner. Within the bone marrow microenvironment, VEGF secreted by megakaryocytes may contribute to the proliferation of endothelial cells. VEGF delivered to sites of vascular injury by activated platelets may initiate angiogenesis.
Collapse
Affiliation(s)
- R Möhle
- Developmental Hematopoiesis Laboratory, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
1226
|
Magovern CJ, Mack CA, Zhang J, Rosengart TK, Isom OW, Crystal RG. Regional angiogenesis induced in nonischemic tissue by an adenoviral vector expressing vascular endothelial growth factor. Hum Gene Ther 1997; 8:215-27. [PMID: 9017425 DOI: 10.1089/hum.1997.8.2-215] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The feasibility of a single administration of a replication-deficient adenovirus (Ad) vector encoding the cDNA for human vascular endothelial growth factor (VEGF) (AdCMV.VEGF) to induce neovascularization in vivo in normal tissue was evaluated in retroperitoneal adipose tissue. Following administration of AdCMV.VEGF (10(9) pfu/50 microliters), maximal VEGF cDNA expression was observed at 2-5 days in the injected adipose tissue. No VEGF protein was detected at > or = 10 days in injected adipose tissue, and there was no increase in serum VEGF levels at any time. In vivo quantification of the number of blood vessels using 30x visualization of the adipose tissue demonstrated an increase in vessel number by 10 days, plateauing by 30 days with a 123% increase in vessel number compared to the control vector AdCMV.Null, despite the fact that no VEGF protein was detected after 5 days. Consistent with the in vivo data, histologic quantification of capillary number demonstrated an increase by day 5, reaching a 38% increase over AdCMV.Null by day 30. These observations demonstrate that an Ad vector carrying the VEGF cDNA is capable of inducing the growth of new blood vessels in a regional fashion in a relatively avascular, normal organ. This suggests in vivo Ad-mediated gene transfer may be useful for therapeutic angiogenesis in the treatment of ischemic cardiovascular disease.
Collapse
Affiliation(s)
- C J Magovern
- Division of Pulmonary and Critical Care Medicine, New York Hospital-Cornell Medical Center, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
1227
|
Torry DS, Torry RJ. Angiogenesis and the expression of vascular endothelial growth factor in endometrium and placenta. Am J Reprod Immunol 1997; 37:21-9. [PMID: 9138450 DOI: 10.1111/j.1600-0897.1997.tb00189.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PROBLEM The demand for increased angiogenesis and microvascular permeability during cyclical changes in the endometrium and during placentation raises the possibility that aberrations in these events could lead to suboptimal reproductive performance. However, relatively little is presently known regarding the regulation of vascular growth and permeability in these tissues. METHOD OF STUDY This review of current literature focuses on the expression, regulation, and potential physiological effects of vascular endothelial growth factor (VEGF) within endometrial and placental tissue. RESULTS Spatial and temporal expression of VEGF as well as its restricted specificity, essential role in vasculogenesis/angiogenesis, and ability to induce vascular permeability makes VEGF an attractive regulator of vascular growth and permeability in the endometrium and placenta. CONCLUSION A better understanding of the production, regulation, and physiological responses of the vasculature to angiogenic growth factors may lead to new therapeutic strategies for reproductive disturbances secondary to vascular insufficiencies within the female reproductive tract.
Collapse
Affiliation(s)
- D S Torry
- Department of OB/GYN, University of Tennessee, Graduate School of Medicine, Knoxville 37920, USA
| | | |
Collapse
|
1228
|
Abstract
In many organs and tissues, the cellular response to injury is associated with a reiteration of specific developmental processes. Studies have shown that, in response to injury, vascular wall cells in adult organisms express genes or gene products characteristic of earlier developmental states. Other genes, expressed preferentially in adult cells in vivo, are down-regulated following injurious stimuli. Complicating matters, however, are recent observations demonstrating that the vascular wall is comprised of phenotypically heterogeneous subpopulations of endothelial cells, smooth muscle cells, and fibroblasts. It is unclear how specific subsets of cells respond to injury and thus contribute to the vascular remodeling that characterizes chronic pulmonary hypertension. This review discusses vascular development in the lung and the cellular responses occurring in pulmonary hypertension; special attention is given to heterogeneity of responses within cell populations and reiteration of developmental processes.
Collapse
Affiliation(s)
- K R Stenmark
- University of Colorado Health Sciences Center, Developmental Lung Biology Laboratory, Denver 80262, USA
| | | |
Collapse
|
1229
|
Westerband A, Mills JL, Marek JM, Heimark RL, Hunter GC, Williams SK. Immunocytochemical determination of cell type and proliferation rate in human vein graft stenoses. J Vasc Surg 1997; 25:64-73. [PMID: 9013909 DOI: 10.1016/s0741-5214(97)70322-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Vascular reconstructions are prone to fail as a result of the development of stenotic lesions, which have historically been attributed to myointimal hyperplasia. In animal models, these lesions are associated with marked proliferative smooth muscle cell (SMC) response to vascular injury. However, recent studies using sensitive immunocytochemical techniques in human lesions have generally failed to detect significant cellular proliferation. To clarify the role of cellular proliferation in humans, we characterized the cellular composition and proliferative index of 14 early infrainguinal vein graft stenoses. METHODS All infrainguinal vein grafts at our institution are prospectively enrolled in a duplex surveillance protocol, the details of which have been previously reported. Among 98 grafts placed within the last year, 11 patients were identified with 14 progressive, focal, high-grade lesions that met previously established threshold criteria for prophylactic revision to prevent graft thrombosis. Lesions were first detected from 1 week to 7 months after surgery and were removed and replaced with segmental interposition grafts (1.5 to 10 months). Freshly excised lesions were placed in Methyl Carnoy's fixative, paraffin embedded, and serially sectioned. The cellular composition of each lesion was determined with cell-specific immunochemical reagents: alpha SMC actin, von Willebrand factor (endothelial cell), CD 68 (macrophage), and CD 45RB (monocyte). Actively proliferating cells were identified using antibody to proliferating cell nuclear antigen (PCNA). The identity of PCNA-positive cells was determined by double-label immunocytochemical staining, and the proliferative index (PCNA-positive cells/total cells x 100) was calculated by computer-assisted counts of representative gridded cross-sections of each lesion. RESULTS All excised lesions demonstrated marked thickening with severe luminal encroachment and were highly cellular, with a predominance of alpha SMC actin+. Endothelial cells on the blood flow surface were present to a variable degree, and seven lesions exhibited striking numbers of macrophages and monocytes. The latter cell types were most abundant near microvessels in the deep neointima and adventitia. Active cellular proliferation was identified primarily in SMCs, with a mean PCNA index of 1.34%. However, significant PCNA reactivity was not limited to SMCs, but was also identified in macrophages and monocytes, particularly in lesions greater than 3 months old. CONCLUSIONS Previous immunocytochemical studies of human coronary restenosis atherectomy specimens have generally detected low rates of cellular proliferation (0.5%), but these lesions may not truly represent myointimal hyperplasia, rather a mixture of atherosclerosis, thrombosis, and "restenosis." In contrast, the present study of early human vein graft lesions detected by duplex surveillance indicates that significant cellular proliferation occurs, although rates are lower than those obtained in animals such as the rat carotid injury model. In addition, although SMCs are the predominant proliferating cell type in human vein grafts, our identification of proliferating monocytes and macrophages raises the question of the contribution of an inflammatory component to the development of human lesions. The present study represents the first report of PCNA determination in a series of human infrainguinal vein grafting procedures.
Collapse
Affiliation(s)
- A Westerband
- Section of Vascular Surgery and Vascular Biology, University of Arizona Health Sciences Center, Tuscon 85724, USA
| | | | | | | | | | | |
Collapse
|
1230
|
Engels K, Fox SB, Harris AL. Angiogenesis as a biologic and prognostic indicator in human breast carcinoma. EXS 1997; 79:113-56. [PMID: 9002231 DOI: 10.1007/978-3-0348-9006-9_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In this review we describe angiogenesis pathways involved in the development of breast carcinoma. Different assessment techniques for angiogenesis and their optimisation are discussed. Angiogenesis is an important factor for prognosis and will be increasingly important in therapeutic decisions.
Collapse
Affiliation(s)
- K Engels
- Department of Cellular Science, University of Oxford, John Radcliffe Hospital, UK
| | | | | |
Collapse
|
1231
|
Brown LF, Detmar M, Claffey K, Nagy JA, Feng D, Dvorak AM, Dvorak HF. Vascular permeability factor/vascular endothelial growth factor: a multifunctional angiogenic cytokine. EXS 1997; 79:233-69. [PMID: 9002222 DOI: 10.1007/978-3-0348-9006-9_10] [Citation(s) in RCA: 137] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
VPF/VEGF is a multifunctional cytokine that contributes to angiogenesis by both direct and indirect mechanisms. On the one hand, VPF/VEGF stimulates the endothelial cells lining nearby microvessels to proliferate, to migrate and to alter their pattern of gene expression. On the other hand, VPF/VEGF renders these same microvascular endothelial cells hyperpermeable so that they spill plasma proteins into the extravascular space, leading to profound alterations in the extracellular matrix that favor angiogenesis. These same principles apply in tumors, in several examples of non-neoplastic pathology, and in physiological processes that involve angiogenesis and new stroma generation. In all of these examples, microvascular hyperpermeability and the introduction of a provisional, plasma-derived matrix precede and accompany the onset of endothelial cell division and new blood vessel formation. It would seem, therefore, that tumors have made use of fundamental pathways that developed in multicellular organisms for purposes of tissue defense, renewal and repair. VPF/VEGF, therefore, has taught us something new about angiogenesis; namely, that vascular hyperpermeability and consequent plasma protein extravasation are important--perhaps essential--elements in its generation. However, this finding raises a paradox. While VPF/VEGF induces vascular hyperpermeability, other potent angiogenic factors apparently do not, at least in sub-toxic concentrations that are more than sufficient to induce angiogenesis (Connolly et al., 1989a). Nonetheless, wherever angiogenesis has been studied, the newly generated vessels have been found to be hyperpermeable. How, therefore, do angiogenic factors other than VPF/VEGF lead to the formation of new and leaky blood vessels? We do not as yet have a complete answer to this question. One possibility is that at least some angiogenic factors mediate their effect by inducing or stimulating VPF/VEGF expression. In fact, there are already clear example of this. A number of putative angiogenic factors including small molecules (e.g. prostaglandins, adenosine) as well as many cytokines (e.g. TGF-alpha, bFGF, TGF-beta, TNF-alpha, KGF, PDGF) have all been shown to upregulate VPF/VEGF expression. Further studies that elucidate the crosstalk among various angiogenic factors are likely to contribute significantly to a better understanding of the mechanisms by which new blood vessels are formed in health and in disease.
Collapse
Affiliation(s)
- L F Brown
- Department of Pathology, Beth Israel Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
1232
|
Achen MG, Gad JM, Stacker SA, Wilks AF. Placenta growth factor and vascular endothelial growth factor are co-expressed during early embryonic development. Growth Factors 1997; 15:69-80. [PMID: 9401819 DOI: 10.3109/08977199709002113] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We have used the polymerase chain reaction to identify mouse proteins similar in primary structure to the endothelial cell mitogen Vascular Endothelial Growth Factor (VEGF). One amplified product encoded mouse Placenta Growth Factor (PIGF). The pattern of PIGF gene expression in mouse embryos was studied by in situ hybridization. Transcripts encoding mouse PIGF were abundant in trophoblastic giant cells associated with the parietal yolk sac at early stages of embryogenesis. VEGF transcripts were also detected in trophoblastic giant cells raising the possibility that these cells may secrete heterodimers consisting of one PIGF subunit and one VEGF subunit. The secretion of PIGF and VEGF by trophoblastic giant cells is likely to be the signal which initiates and co-ordinates vascularization in the deciduum and placenta during early embryogenesis.
Collapse
Affiliation(s)
- M G Achen
- Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Victoria, Australia.
| | | | | | | |
Collapse
|
1233
|
Ferrara N, Keyt B. Vascular endothelial growth factor: basic biology and clinical implications. EXS 1997; 79:209-32. [PMID: 9002234 DOI: 10.1007/978-3-0348-9006-9_9] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- N Ferrara
- Department of Cardiovascular Research, Genentech, Inc., South San Francisco, California 94080, USA
| | | |
Collapse
|
1234
|
Kaplan JB, Sridharan L, Zaccardi JA, Dougher-Vermazen M, Terman BI. Characterization of a soluble vascular endothelial growth factor receptor-immunoglobulin chimera. Growth Factors 1997; 14:243-56. [PMID: 9386989 DOI: 10.3109/08977199709021523] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To investigate the interaction between vascular endothelial growth factor (VEGF) and its receptor, we have constructed a chimeric protein consisting of the extracellular ligand-binding domain of the human VEGF receptor subtype KDR fused to a human IgG1 Fc domain (KDR-Fc). KDR-Fc was expressed in human 293 kidney epithelial cells as a 300-kDa secreted, dimeric glycoprotein that bound 125I-VEGF165 with high affinity (Kd = 150 pM). Unlike the full length cellular receptor, KDR-Fc did not require heparin for 125I-VEGF165 binding, although heparin did stimulate 125I-VEGF165 binding approximately 50 to 100%. Similar results were observed for KDR-Fc expressed in yeast cells. Since yeast do not synthesize heparan sulfate proteoglycans, we conclude that cellular heparan sulfates do not account for the lack of a heparin requirement for 125I-VEGF165 binding to KDR-Fc. The polycationic protein protamine, which inhibits (IC50 = 1 microgram/ml) 125I-VEGF165 binding to bovine aortic endothelial cells and other KDR-expressing cells by blocking heparin interactions, had no effect on the heparin independent component of 125I-VEGF165 binding to KDR-Fc. Protamine does inhibit (IC50 = 1 microgram/ml) the heparin dependent component of 125I-VEGF165 binding to KDR-Fc. KDR-Fc bound VEGF121 with the same affinity as VEGF165. Heparin had no effect on 125I-VEGF121 binding to KDR-Fc, indicating that heparin interaction with the 44 amino acids contained in VEGF165 but not VEGF121 allow for maximal VEGF165 binding. Deletion analysis of KDR-Fc demonstrated that the determinants required for high affinity VEGF binding are located in the three aminoterminal Ig-domains of the protein. Heparin had no effect on 125I-VEGF165 binding to the three Ig-domain receptor, suggesting that there are heparin binding determinants located in KDR Ig-domains 4 to 7.
Collapse
Affiliation(s)
- J B Kaplan
- Oncology Section, Wyeth-Ayerst Research, Pearl River, New York 10965, USA
| | | | | | | | | |
Collapse
|
1235
|
Stratmann A, Machein MR, Plate KH. Anti-angiogenic gene therapy of malignant glioma. ACTA NEUROCHIRURGICA. SUPPLEMENT 1997; 68:105-10. [PMID: 9233424 DOI: 10.1007/978-3-7091-6513-3_20] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Glioblastoma, one of the best vascularized tumours in humans, appears well suited for an antiangiogenic therapy. VEGF (vascular endothelial growth factor), the most important angiogenesis factor identified to date, is highly expressed in glioblastoma. VEGF is particulary upregulated in palisading cells adjacent to necroses and has subsequently been shown to be hypoxia-inducible in glioma cells in vitro. VEGF-receptor tyrosine kinases, VEGF-R1 (flt-1) and VEGF-R2 (flk-1), are induced in a tumour stage dependent manner during glioma progression and are exclusively expressed in tumour vascular endothelial cells. These observations suggest that VEGF-receptors are promising targets for tumour endothelial cell specific therapy. The ability to block VEGF-signalling by the VEGF-R2 dominant-negative mutant identifies the VEGF/VEGF-R2 system as a major regulator of glioma angiogenesis. Several experimental approaches demonstrate that in rat gliomas tumour growth can be prevented by the inhibition of angiogenesis. These findings are of pivotal importance for the development of anti-angiogenic therapies in glioblastoma patients.
Collapse
Affiliation(s)
- A Stratmann
- Department of Neuropathology, University of Freiburg Medical School, Federal Republic of Germany
| | | | | |
Collapse
|
1236
|
|
1237
|
|
1238
|
Robbins SG, Conaway JR, Ford BL, Roberto KA, Penn JS. Detection of vascular endothelial growth factor (VEGF) protein in vascular and non-vascular cells of the normal and oxygen-injured rat retina. Growth Factors 1997; 14:229-41. [PMID: 9386988 DOI: 10.3109/08977199709021522] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Vascular endothelial growth factor (VEGF) is a potent and specific endothelial cell cytokine that can be up-regulated by hypoxia. There is evidence that VEGF is a significant mediator in retinal neovascular diseases and other disorders in which hypoxia is believed to influence the pathogenesis. Here we demonstrate the spatial relationships among areas of retinal non-perfusion, VEGF protein and vascular endothelial cells throughout the retina, and relate these results to cellular distribution of VEGF in cross section. Newborn albino rats were oxygen-injured by cycles of alternating 50% and 10% oxygen for 14 days and then placed in room air. On days 16, 21 and 26, oxygen-injured and control (raised in room air) rats were sacrificed, enucleated and retinas were dissected and fixed for whole mount immunostaining for VEGF or embedding in glycol methacrylate for VEGF immunohistochemistry. Intact eyes taken on days 16 and 20 were processed similarly. Vascular endothelial cells were demonstrated by staining whole-mounted retinas for adenosine diphosphatase (ADPase) activity. Preretinal neovascular growths (i.e., abnormal vessels extending from the retina into the vitreous) were VEGF-positive. There was also a pan-retinal distribution of non-endothelial cells that were VEGF-positive in both room air and oxygen-injured rats, with stronger immunostaining in day 16 oxygen-injured retinas. In cross-section, VEGF staining was confirmed in preretinal growths, normal retinal vessels, cells in the inner nuclear layer (primarily Müller cells) and ganglion cells. Retinas which had been incubated with nonimmune IgG or absorbed anti-VEGF antibody showed little or no staining. In conclusion, we have identified cells of the inner retina which express VEGF. The production of VEGF by these cells--in particular, Müller cells--may promote preretinal neovascularization in oxygen-injured eyes. We have found, moreover, that the combination of immunohistochemistry and ADPase staining of whole mount preparations is a unique and powerful tool for evaluating relationships between presumed areas of retinal ischemia, VEGF (and other cytokines) and retinal blood vessels, within an entire retina. This approach can be used to study any proliferative retinal disorders in which VEGF is a potential component of the pathogenesis.
Collapse
Affiliation(s)
- S G Robbins
- Arkansas Center for Eye Research, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock 72205-7199, USA
| | | | | | | | | |
Collapse
|
1239
|
Flk-1, a receptor for vascular endothelial growth factor (VEGF), is expressed by retinal progenitor cells. J Neurosci 1996. [PMID: 8815891 DOI: 10.1523/jneurosci.16-19-06089.1996] [Citation(s) in RCA: 156] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Throughout development of the vertebrate retina, progenitor cells are multipotential, producing a variety of distinctive cell types. Little is known of the molecular mechanisms directing the determination of cell fate. We have examined retinal progenitor cells for expression of receptor tyrosine kinases in an attempt to define receptors that could allow a progenitor to respond to its environment. We found that the receptor tyrosine kinase Flk-1, previously shown to be expressed in endothelial cells, is also expressed in neural progenitor cells of the mouse retina. Flk-1 RNA expression in the retinal progenitors commences with the onset of neuronal differentiation and persists throughout retinal neurogenesis. Flk-1 RNA and protein levels in the retina vary temporally during development, as shown by in situ hybridization and Western blot analysis. Patterns of beta-galactosidase expression in mice containing the lacZ gene in place of the Flk-1 gene are consistent with Flk-1 being expressed in retinal progenitors. In addition, we show that the ligand of Flk-1, vascular endothelial growth factor (VEGF), is expressed in the developing retina by differentiated cells and that a chimeric ligand of VEGF fused to alkaline phosphatase binds to proliferating retinal progenitors. Furthermore, the neural retina-derived Flk-1 protein kinase is activated by VEGF in vitro. Thus, the Flk-1 receptor protein kinase is expressed on the surface of neural progenitors in mouse retina and may play a critical role in neurogenesis as well as in vasculogenesis.
Collapse
|
1240
|
Fruttiger M, Calver AR, Krüger WH, Mudhar HS, Michalovich D, Takakura N, Nishikawa S, Richardson WD. PDGF mediates a neuron-astrocyte interaction in the developing retina. Neuron 1996; 17:1117-31. [PMID: 8982160 DOI: 10.1016/s0896-6273(00)80244-5] [Citation(s) in RCA: 184] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Astrocytes invade the developing retina from the optic nerve head, over the axons of retinal ganglion cells (RGCs). RGCs express the platelet-derived growth factor A-chain (PDGF-A) and retinal astrocytes the PDGF alpha-receptor (PDGFR alpha), suggesting that PDGF mediates a paracrine interaction between these cells. To test this, we inhibited PDGF signaling in the eye with a neutralizing anti-PDGFR alpha antibody or a soluble extracellular fragment of PDGFR alpha. These treatments inhibited development of the astrocyte network. We also generated transgenic mice that overexpress PDGF-A in RGCs. This resulted in hyperproliferation of astrocytes, which in turn induced excessive vasculogenesis. Thus, PDGF appears to be a link in the chain of cell-cell interactions responsible for matching numbers of neurons, astrocytes, and blood vessels during retinal development.
Collapse
Affiliation(s)
- M Fruttiger
- MRC Laboratory for Molecular Cell Biology, University College London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
1241
|
Affiliation(s)
- N Ferrara
- Department of Cardiovascular Research, Genetech, Inc., South San Francisco, California 94080, USA
| |
Collapse
|
1242
|
Kalkanis SN, Carroll RS, Zhang J, Zamani AA, Black PM. Correlation of vascular endothelial growth factor messenger RNA expression with peritumoral vasogenic cerebral edema in meningiomas. J Neurosurg 1996; 85:1095-101. [PMID: 8929501 DOI: 10.3171/jns.1996.85.6.1095] [Citation(s) in RCA: 110] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Intracranial meningiomas are often complicated by peritumoral vasogenic cerebral edema, which appears to result from increased microvascular permeability and extravasation of proteinaceous and plasma fluid into the adjacent peritumoral space. The source of such edema has long been mysterious. The contents of this paper support the concept that vascular endothelial growth factor (VEGF) production plays a significant role in edema formation. Vascular endothelial growth factor messenger RNA expression has been found in a wide range of intracranial neoplasms, including malignant gliomas, metastatic melanomas, meningiomas, and other benign tumors. Several studies have confirmed the importance of VEGF in tumorigenesis, neovascularization, and edema production. This study tests the hypothesis that the presence of peritumoral edema in meningiomas is positively correlated with increased expression of VEGF mRNA. To investigate this hypothesis, 31 meningioma specimens were subjected to Northern blot analysis, hybridization with a complementary DNA VEGF probe, and laser densitometry to determine the relative levels of VEGF mRNA expression. Magnetic resonance imaging was then used in a double-blind fashion to correlate the neuropathological tissue samples with the presence of preoperative peritumoral edema. Of 31 patients studied, 14 exhibited no edema and 17 exhibited some level of peritumoral fluid accumulation. There was a marked increase in VEGF expression in patients with edema (p = 0.0004, Wilcoxon-Mann-Whitney rank-sum test). Meningiomas with peritumoral edema exhibited 3.4 times the level of VEGF mRNA as those without edema. These data demonstrate a strong link between VEGF mRNA expression and peritumoral edema and indicate that VEGF expression is an important factor in the etiology of edema around meningiomas.
Collapse
Affiliation(s)
- S N Kalkanis
- Neurosurgical Service, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
1243
|
Wakiya K, Begue A, Stehelin D, Shibuya M. A cAMP response element and an Ets motif are involved in the transcriptional regulation of flt-1 tyrosine kinase (vascular endothelial growth factor receptor 1) gene. J Biol Chem 1996; 271:30823-8. [PMID: 8940064 DOI: 10.1074/jbc.271.48.30823] [Citation(s) in RCA: 117] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The flt-1 gene encodes a transmembrane tyrosine kinase, Flt-1, a receptor for vascular endothelial growth factor. The expression of flt-1 gene is restricted to endothelial cells in vivo. To understand the molecular mechanism underlying endothelial-specific expression of this gene, we studied the functional significance of transcriptional motifs in the 200-base pair region of the human flt-1 gene promoter, which has been identified to confer cell type specificity. By point mutation analysis using chloramphenicol acetyltransferase plasmids in 293E1 cells, which express significant levels of flt-1 mRNA, we found that an Ets motif, E4, at -54 to -51 and a cAMP response element (CRE) at -83 to -76 are involved in the transcriptional regulation of this gene. Disruption of either this CRE or E4 within the promoter sequence of 90 base pairs resulted in a decrease in chloramphenicol acetyltransferase activity of 90%, indicating that co-existence of both of CRE and Ets motif E4 is necessary for transcription of the flt-1 gene. Co-transfection of an expression vector containing c-ets-1, c-ets-2, or c-erg cDNA with this 90-base pair sequence yielded a 5-8-fold elevation of chloramphenicol acetyltransferase activity, further supporting the idea that Ets family protein(s) participates in the regulation of the flt-1 gene. Gel shift assays using nuclear extracts of 293E1 and endothelial cells demonstrated the existence of protein factor(s) that specifically binds to CRE and Ets motif E4, respectively. Taken together, our results strongly suggest cooperation of a CRE and an Ets motif for the function of the flt-1 gene promoter.
Collapse
Affiliation(s)
- K Wakiya
- Department of Genetics, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108, Japan
| | | | | | | |
Collapse
|
1244
|
Warren RS, Yuan H, Matli MR, Ferrara N, Donner DB. Induction of vascular endothelial growth factor by insulin-like growth factor 1 in colorectal carcinoma. J Biol Chem 1996; 271:29483-8. [PMID: 8910616 DOI: 10.1074/jbc.271.46.29483] [Citation(s) in RCA: 174] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic hormone that is produced by and supports the growth of many types of malignancies. The present study shows that insulin-like growth factor 1 (IGF-I), a mitogen that promotes the propagation of cancers through autocrine and paracrine mechanisms, increases the expression of mRNA for VEGF and production of VEGF protein by COLO 205 colon carcinoma cells. IGF-I also induces expression of VEGF mRNA in SW620, LSLiM6, and HCT15 colon carcinoma cells showing that this is a common response to IGF-I. Whereas IGF-I induced VEGF mRNA in each cell line examined (2.3-12-fold), it induced proliferation only in COLO 205 and LSLiM6 cells. Thus, the proliferative response induced by IGF-I and its ability to induce VEGF occur through distinguishable mechanisms. IGF-I increases the cellular content of VEGF mRNA by increasing the rate of transcription (5-fold after 4 h) and also by increasing the half-life of VEGF mRNA (0.6 +/- 0.07 h in control cells to 2.0 +/- 0.37 h in IGF-I-treated cells). Monoclonal antibody (alphaIR3) directed against the type 1 IGF receptor significantly attenuated the ability of IGF-I to promote expression of VEGF mRNA. Interestingly, by itself alphaIR3 acted as a weak agonist and induced a modest increase in the cellular content of VEGF mRNA. alphaIR3 also promoted tyrosine phosphorylation of the beta subunit of the IGF-I receptor, and the magnitude of this response was comparable with that induced by IGF-I. These observations point to a nonlinear relationship between activation of the IGF-I receptor and induction of VEGF mRNA. Thus, in addition to its direct, growth stimulatory effect on transformed cells, IGF-I induces the expression of VEGF which can promote the progression of cancer by regulating the development of new blood vessels.
Collapse
Affiliation(s)
- R S Warren
- Department of Surgery, University of California School of Medicine, San Francisco, California 94143, USA.
| | | | | | | | | |
Collapse
|
1245
|
Sonstein WJ, Kader A, Michelsen WJ, Llena JF, Hirano A, Casper D. Expression of vascular endothelial growth factor in pediatric and adult cerebral arteriovenous malformations: an immunocytochemical study. J Neurosurg 1996; 85:838-45. [PMID: 8893722 DOI: 10.3171/jns.1996.85.5.0838] [Citation(s) in RCA: 125] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Children and adults may differ with respect to their cerebral vasculature in both normal and pathological states. The authors have identified four pediatric patients in whom a cerebral arteriovenous malformation (AVM) recurred after surgery for removal of the AVM and in whom a normal postoperative angiogram had been obtained. This phenomenon has not been observed in adults. The propensity to regrow a cerebral AVM may reflect a less mature cerebral vasculature and a disregulated angiogenic process. Recently, attention has focused on vascular endothelial growth factor (VEGF) as a possible general mediator of angiogenesis in development and neoplasia. A retrospective immunocytochemical analysis of VEGF expression in AVM tissue was conducted to test the hypothesis that VEGF expression may be found in association with the regrowth of AVMs. The results demonstrate a high degree of astrocytic VEGF expression in four (100%) of four specimens from the initial operation in the children with recurrent AVMs as compared to one (14%) of seven nonrecurrent AVMs in the pediatric and two (25%) of eight adult specimens. All of the specimens from the first operation of the recurrent group demonstrate a clear association of cellular immunoreactivity to the abnormal blood vessels, a relationship that was not observed in the specimens from the nonrecurrent groups. These observations indicate that a humoral mechanism mediated by VEGF may play a role in AVM recurrence.
Collapse
Affiliation(s)
- W J Sonstein
- Department of Neurological Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | | | | | | | | |
Collapse
|
1246
|
Xia P, Aiello LP, Ishii H, Jiang ZY, Park DJ, Robinson GS, Takagi H, Newsome WP, Jirousek MR, King GL. Characterization of vascular endothelial growth factor's effect on the activation of protein kinase C, its isoforms, and endothelial cell growth. J Clin Invest 1996; 98:2018-26. [PMID: 8903320 PMCID: PMC507645 DOI: 10.1172/jci119006] [Citation(s) in RCA: 395] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a potent endothelial cell mitogen which mediates its effects by binding to tyrosine kinase receptors. We have characterized the VEGF-activated intracellular signal transduction pathway in bovine aortic endothelial cells and correlated this to its mitogenic effects. VEGF induced concentration- and time-dependent increases in protein kinase C (PKC) activation with a maximum of 2.2-fold above the basal level at 5 x 10(-10) M within 10 min as measured both by in situ and translocation assays. Immunoblotting analysis of PKC isoforms in cytosolic and membrane fractions indicated that after VEGF stimulation the content of Ca(2+)-sensitive PKC isoforms (alpha and betaII) was increased in the membrane fractions, whereas no changes were observed for PKC isoforms delta and epsilon. The stimulation of PKC activity by VEGF was preceded by the activation of phospholipase Cgamma (PLCgamma). This was demonstrated by parallel increases in PLCgamma tyrosine phosphorylation, [3H]inositol phosphate production, and [3H]arachidonic acid-labeled diacylglycerol formation in bovine aortic endothelial cells. In addition, VEGF increased phosphatidylinositol 3-kinase activity 2.1-fold which was inhibited by wortmannin, a phosphatidylinositol 3-kinase inhibitor, without decreasing the VEGF-induced increase in PKC activity or endothelial cell growth. Interestingly, genistein, a tyrosine kinase inhibitor, and GFX or H-7, PKC inhibitors, abolished both VEGF-induced PKC activation and endothelial cell proliferation. VEGF's mitogenic effect was inhibited by a PKC isoform beta-selective inhibitor, LY333531, in a concentration-dependent manner. In contrast, antisense PKC-alpha oligonucleotides enhanced VEGF-stimulated cell growth with a simultaneous decrease of 70% in PKC-alpha protein content. Thus, VEGF appears to mediate its mitogenic effects partly through the activation of the PLCgamma and PKC pathway, involving predominately PKC-beta isoform activation in endothelial cells.
Collapse
Affiliation(s)
- P Xia
- Research Division, Joslin Diabetes Center, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1247
|
Affiliation(s)
- W Schaper
- Max-Planck-Institute for Physiological and Clinical Research, Department of Experimental Cardiology, Bad Nauheim, Germany
| | | |
Collapse
|
1248
|
Lindner V, Reidy MA. Expression of VEGF receptors in arteries after endothelial injury and lack of increased endothelial regrowth in response to VEGF. Arterioscler Thromb Vasc Biol 1996; 16:1399-405. [PMID: 8911280 DOI: 10.1161/01.atv.16.11.1399] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Vascular endothelial growth factor (VEGF) is an endothelial cell-specific factor with angiogenic effects in vivo and mitogenic effects in vitro. Administration of VEGF has been reported to stimulate endothelial growth in denuded arteries and new blood vessel formation in models of induced tissue ischemia. In the present study, expression of VEGF and its receptors flk-l and flt-l was determined in injured aortas and carotid arteries of rats and mice. Neither VEGF nor flk-l mRNA was detectable in vascular cells. mRNA levels for flt-l were dramatically upregulated at the leading edge of a growing endothelial monolayer in vivo; however, these cells did not demonstrate increased replication after VEGF infusion. Furthermore, all doses and treatment protocols of VEGF failed to promote reendothelialization in denuded arteries. At sites of flt-l expression, VEGF increased permeability. These areas revealed a loss of endothelial contacts at the ultrastructural level. These findings suggest that VEGF is not a direct mitogen for large-vessel endothelium in vivo and that VEGF may play a role in abolishing contact inhibition, which may be a prerequisite for endothelial proliferation.
Collapse
Affiliation(s)
- V Lindner
- Department of Surgery, Maine Medical Center Research Institute, South Portland 04106, USA.
| | | |
Collapse
|
1249
|
The expression of antisense vascular endothelial growth factor (VEGF) sequences inhibits intracranial C6 glioma growth in vivo by suppressing tumour angiogenesis. J Clin Neurosci 1996; 3:366-72. [DOI: 10.1016/s0967-5868(96)90035-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/1995] [Accepted: 02/20/1996] [Indexed: 11/23/2022]
|
1250
|
Kovács Z, Ikezaki K, Samoto K, Inamura T, Fukui M. VEGF and flt. Expression time kinetics in rat brain infarct. Stroke 1996; 27:1865-72; discussion 1872-3. [PMID: 8841346 DOI: 10.1161/01.str.27.10.1865] [Citation(s) in RCA: 217] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND PURPOSE Vascular endothelial growth/vascular permeability factor (VEGF) is a candidate for an angiogenic and hyperpermeability inducing factor in an infarct because it is a secretable mitogen specific for endothelial cells and is upregulated by hypoxia. Our study attempts to clarify the chronological expression of VEGF and its receptor (flt) system in experimental cerebral infarction. METHODS With the use of a reproducible middle cerebral artery occlusion model in rats, VEGF expression was identified by Western blotting with anti-VEGF antibody. The chronological expression of the VEGF/flt system was analyzed semiquantitatively by immunohistochemical means in infarcts with different time courses from 3 hours to 3 weeks. RESULTS VEGF and flt were expressed exclusively in the ischemic brain. The bands obtained on the immunoblot at 38 and 45 kD are related to those of VEGF121 and VEGF165 isoforms. Macrophages, neurons, and glial cells chronologically expressed VEGF immunoreactivity in a different fashion. Both VEGF (bound) and flt were detected in endothelial cells along with the development of angiogenesis. CONCLUSIONS In the ischemic brain the macrophages, neurons, and glial cells appear to contain VEGF. The VEGF receptor flt was induced in endothelial cells along with the progression of angiogenesis in infarct. The VEGF/flt system is thus considered to be involved in the healing process of brain infarct.
Collapse
Affiliation(s)
- Z Kovács
- Department of Neurosurgery, Kyushu University Faculty of Medicine, Fukuoka, Japan
| | | | | | | | | |
Collapse
|