Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Am J Transplant ; 16(4): 1160-72, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26689983

ABSTRACT

Organ damage and innate immunity during heart transplantation may evoke adaptive immunity with serious consequences. Because lymphatic vessels bridge innate and adaptive immunity, they are critical in immune surveillance; however, their role in ischemia-reperfusion injury (IRI) in allotransplantation remains unknown. We investigated whether the lymphangiogenic VEGF-C/VEGFR3 pathway during cardiac allograft IRI regulates organ damage and subsequent interplay between innate and adaptive immunity. We found that cardiac allograft IRI, within hours, increased graft VEGF-C expression and lymphatic vessel activation in the form of increased lymphatic VEGFR3 and adhesion protein expression. Pharmacological VEGF-C/VEGFR3 stimulation resulted in early lymphatic activation and later increase in allograft inflammation. In contrast, pharmacological VEGF-C/VEGFR3 inhibition during cardiac allograft IRI decreased early lymphatic vessel activation with subsequent dampening of acute and chronic rejection. Genetic deletion of VEGFR3 specifically in the lymphatics of the transplanted heart recapitulated the survival effect achieved by pharmacological VEGF-C/VEGFR3 inhibition. Our results suggest that tissue damage rapidly changes lymphatic vessel phenotype, which, in turn, may shape the interplay of innate and adaptive immunity. Importantly, VEGF-C/VEGFR3 inhibition during solid organ transplant IRI could be used as lymphatic-targeted immunomodulatory therapy to prevent acute and chronic rejection.


Subject(s)
Endothelial Cells/metabolism , Graft Rejection/metabolism , Heart Transplantation , Reperfusion Injury/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Allografts , Animals , Endothelial Cells/pathology , Graft Rejection/pathology , Graft Survival/immunology , Inflammation/metabolism , Inflammation/pathology , Lymphangiogenesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Reperfusion Injury/physiopathology , Tissue Donors , Vascular Endothelial Growth Factor C/metabolism
2.
Acta Radiol ; 49(5): 530-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18568540

ABSTRACT

BACKGROUND: Cardiovascular magnetic resonance imaging (CMR) has become an established noninvasive method for evaluating ventricular function utilizing three-dimensional volumetry. Postprocessing of volumetric measurements is still tedious and time consuming. Stroke volumes obtained by flow quantification across the aortic root or pulmonary trunk could be utilized to increase both speed of workflow and accuracy. PURPOSE: To assess accuracy of stroke volume quantification using MR volumetric imaging compared to flow quantification in patients with various cardiac diseases. Strategies for the augmentation of accuracy in clinical routine were deduced. MATERIAL AND METHODS: 78 patients with various cardiac diseases-excluding intra- or extracardiac shunts, regurgitant valvular defects, or heart rhythm disturbance-underwent cardiac function analysis with flow measurements across the aortic root and cine imaging of the left ventricle. Forty-six patients additionally underwent flow measurements in the pulmonary trunk and cine imaging of the right ventricle. RESULTS: Left ventricular stroke volume (LVSV) and stroke volume of the aortic root (SVAo) correlated with r=0.97, and Bland-Altman analysis showed a mean difference of 0.11 ml and a standard error of estimation (SEE) of 4.31 ml. Ninety-two percent of the data were within the 95% limits of agreement. Right ventricular stroke volume (RVSV) and stroke volume of the pulmonary trunk (SVP) correlated with a factor of r=0.86, and mean difference in the Bland-Altman analysis was fixed at -2.62 ml (SEE 8.47 ml). For RVSV and SVP, we calculated r=0.82, and Bland-Altman analysis revealed a mean difference of 1.27 ml (SEE 9.89 ml). LVSV and RVSV correlated closely, with r=0.91 and a mean difference of 2.79 ml (SEE 7.17 ml). SVAo and SVP correlated with r=0.95 and a mean difference of 0.50 ml (SEE 5.56 ml). CONCLUSION: Flow quantification can be used as a guidance tool, providing accurate and reproducible stroke volumes of both ventricles. Combining both offers a highly accurate tool to gauge ventricular function in a routine clinical setting, increasing workflow speed.


Subject(s)
Heart Diseases/physiopathology , Stroke Volume , Adolescent , Adult , Aged , Aged, 80 and over , Blood Flow Velocity , Child , Cohort Studies , Female , Heart Ventricles/physiopathology , Humans , Image Processing, Computer-Assisted , Imaging, Three-Dimensional/methods , Magnetic Resonance Imaging , Male , Middle Aged , Reproducibility of Results , Time Factors
3.
Eur Radiol ; 18(7): 1406-13, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18351347

ABSTRACT

The purpose of this study was to evaluate the accuracy of multi-detector row helical CT (MDCT) for detection and localization of acute upper and lower gastrointestinal (GI) hemorrhage or intraperitoneal bleeding. Thirty-six consecutive patients with clinical signs of acute bleeding underwent biphasic (16- or 40-channel) MDCT. MDCT findings were correlated with endoscopy, angiography or surgery. Among the 36 patients evaluated, 26 were examined for GI bleeding and 10 for intraperitoneal hemorrhage. Confirmed sites of GI bleeding were the stomach (n = 5), duodenum (n = 5), small bowel (n = 6), large bowel (n = 8) and rectum (n = 2). The correct site of bleeding was identifiable on MDCT in 24/26 patients with GI bleeding. In 20 of these 24 patients, active CM extravasation was apparent during the exam. Among the ten patients with intraperitoneal hemorrhage, MDCT correctly identified the bleeding source in nine patients. Our findings suggest that fast and accurate localization of acute gastrointestinal and intraperitoneal bleeding is achievable on MDCT.


Subject(s)
Gastrointestinal Hemorrhage/diagnostic imaging , Tomography, Spiral Computed/methods , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Contrast Media , Female , Gastrointestinal Hemorrhage/etiology , Humans , Iopamidol/analogs & derivatives , Male , Middle Aged
5.
Circ Res ; 88(6): 623-9, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11282897

ABSTRACT

The growth of blood and lymphatic vasculature is mediated in part by secreted polypeptides of the vascular endothelial growth factor (VEGF) family. The prototype VEGF binds VEGF receptor (VEGFR)-1 and VEGFR-2 and is angiogenic, whereas VEGF-C, which binds to VEGFR-2 and VEGFR-3, is either angiogenic or lymphangiogenic in different assays. We used an adenoviral gene transfer approach to compare the effects of these growth factors in adult mice. Recombinant adenoviruses encoding human VEGF-C or VEGF were injected subcutaneously into C57Bl6 mice or into the ears of nude mice. Immunohistochemical analysis showed that VEGF-C upregulated VEGFR-2 and VEGFR-3 expression and VEGF upregulated VEGFR-2 expression at 4 days after injection. After 2 weeks, histochemical and immunohistochemical analysis, including staining for the lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), the vascular endothelial marker platelet-endothelial cell adhesion molecule-1 (PECAM-1), and the proliferating cell nuclear antigen (PCNA) revealed that VEGF-C induced mainly lymphangiogenesis in contrast to VEGF, which induced only angiogenesis. These results have significant implications in the planning of gene therapy using these growth factors.


Subject(s)
Endothelial Growth Factors/physiology , Endothelium, Lymphatic/physiology , Neovascularization, Physiologic/physiology , Skin/blood supply , Adenoviridae/genetics , Animals , Cell Division , Cell Line , Endothelial Growth Factors/genetics , Endothelium, Lymphatic/chemistry , Endothelium, Lymphatic/cytology , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Gene Expression , Genetic Vectors/genetics , Glycoproteins/analysis , Humans , Immunohistochemistry , Lymphokines/genetics , Lymphokines/physiology , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Nude , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proliferating Cell Nuclear Antigen/analysis , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Skin/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3 , Vascular Endothelial Growth Factors , Vesicular Transport Proteins , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
6.
EMBO J ; 20(6): 1223-31, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11250889

ABSTRACT

Vascular endothelial growth factor receptor-3 (VEGFR-3) has an essential role in the development of embryonic blood vessels; however, after midgestation its expression becomes restricted mainly to the developing lymphatic vessels. The VEGFR-3 ligand VEGF-C stimulates lymphangiogenesis in transgenic mice and in chick chorioallantoic membrane. As VEGF-C also binds VEGFR-2, which is expressed in lymphatic endothelia, it is not clear which receptors are responsible for the lymphangiogenic effects of VEGF-C. VEGF-D, which binds to the same receptors, has been reported to induce angiogenesis, but its lymphangiogenic potential is not known. In order to define the lymphangiogenic signalling pathway we have created transgenic mice overexpressing a VEGFR-3-specific mutant of VEGF-C (VEGF-C156S) or VEGF-D in epidermal keratinocytes under the keratin 14 promoter. Both transgenes induced the growth of lymphatic vessels in the skin, whereas the blood vessel architecture was not affected. Evidence was also obtained that these growth factors act in a paracrine manner in vivo. These results demonstrate that stimulation of the VEGFR-3 signal transduction pathway is sufficient to induce specifically lymphangiogenesis in vivo.


Subject(s)
Endothelial Growth Factors/metabolism , Lymphatic System/metabolism , Lymphokines/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Skin/metabolism , Animals , Half-Life , Hyperplasia , Lymphatic System/growth & development , Lymphatic System/pathology , Mice , Mice, Transgenic , Paracrine Communication , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Recombinant Proteins/metabolism , Signal Transduction , Skin/blood supply , Skin/growth & development , Skin/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor D , Vascular Endothelial Growth Factor Receptor-3 , Vascular Endothelial Growth Factors
7.
EMBO J ; 20(4): 672-82, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11179212

ABSTRACT

Metastasis is a frequent and lethal complication of cancer. Vascular endothelial growth factor-C (VEGF-C) is a recently described lymphangiogenic factor. Increased expression of VEGF-C in primary tumours correlates with dissemination of tumour cells to regional lymph nodes. However, a direct role for VEGF-C in tumour lymphangiogenesis and subsequent metastasis has yet to be demonstrated. Here we report the establishment of transgenic mice in which VEGF-C expression, driven by the rat insulin promoter (Rip), is targeted to beta-cells of the endocrine pancreas. In contrast to wild-type mice, which lack peri-insular lymphatics, RipVEGF-C transgenics develop an extensive network of lymphatics around the islets of Langerhans. These mice were crossed with Rip1Tag2 mice, which develop pancreatic beta-cell tumours that are neither lymphangiogenic nor metastatic. Double-transgenic mice formed tumours surrounded by well developed lymphatics, which frequently contained tumour cell masses of beta-cell origin. These mice frequently developed pancreatic lymph node metastases. Our findings demonstrate that VEGF-C-induced lymphangiogenesis mediates tumour cell dissemination and the formation of lymph node metastases.


Subject(s)
Endothelial Growth Factors/physiology , Lymphatic System/growth & development , Neoplasm Metastasis , Animals , DNA, Complementary , Endothelial Growth Factors/genetics , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Pancreas/ultrastructure , Vascular Endothelial Growth Factor C
8.
Circulation ; 102(18): 2262-8, 2000 Oct 31.
Article in English | MEDLINE | ID: mdl-11056103

ABSTRACT

BACKGROUND: Gene transfer to the vessel wall may provide new possibilities for the treatment of vascular disorders, such as postangioplasty restenosis. In this study, we analyzed the effects of adenovirus-mediated vascular endothelial growth factor (VEGF)-C gene transfer on neointima formation after endothelial denudation in rabbits. For comparison, a second group was treated with VEGF-A adenovirus and a third group with lacZ adenovirus. Clinical-grade adenoviruses were used for the study. METHODS AND RESULTS: Aortas of cholesterol-fed New Zealand White rabbits were balloon-denuded, and gene transfer was performed 3 days later. Animals were euthanized 2 and 4 weeks after the gene transfer, and intima/media ratio (I/M), histology, and cell proliferation were analyzed. Two weeks after the gene transfer, I/M in the lacZ-transfected control group was 0. 57+/-0.04. VEGF-C gene transfer reduced I/M to 0.38+/-0.02 (P:<0.05 versus lacZ group). I/M in VEGF-A-treated animals was 0.49+/-0.17 (P:=NS). The tendency that both VEGF groups had smaller I/M persisted at the 4-week time point, when the lacZ group had an I/M of 0.73+/-0.16, the VEGF-C group 0.44+/-0.14, and the VEGF-A group 0. 63+/-0.21 (P:=NS). Expression of VEGF receptors 1, 2, and 3 was detected in the vessel wall by immunocytochemistry and in situ hybridization. As an additional control, the effect of adenovirus on cell proliferation was analyzed by performing gene transfer to intact aorta without endothelial denudation. No differences were seen in smooth muscle cell proliferation or I/M between lacZ adenovirus and 0.9% saline-treated animals. CONCLUSIONS: Adenovirus-mediated VEGF-C gene transfer may be useful for the treatment of postangioplasty restenosis and vessel wall thickening after vascular manipulations.


Subject(s)
Adenoviridae/genetics , Angioplasty, Balloon/adverse effects , Aortic Valve Stenosis/prevention & control , Endothelial Growth Factors/pharmacology , Tunica Intima/drug effects , Adenoviridae/metabolism , Animals , Aortic Valve Stenosis/etiology , Aortic Valve Stenosis/metabolism , Cell Division/drug effects , Cells, Cultured , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Gene Transfer Techniques , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins/biosynthesis , Rabbits , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Cell Surface/biosynthesis , Receptors, Growth Factor/biosynthesis , Receptors, Vascular Endothelial Growth Factor , Transfection , Tunica Intima/metabolism , Tunica Intima/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factor Receptor-3
9.
Curr Opin Biotechnol ; 10(6): 528-35, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10600689

ABSTRACT

Endothelial growth factors and their receptors may provide important therapeutic tools for the treatment of pathological conditions characterised by defective or aberrant angiogenesis. Vascular endothelial growth factor (VEGF) is pivotal for vasculogenesis and for angiogenesis in normal and pathological conditions. VEGF-B and VEGF-C provide this gene family with additional functions, for example, VEGF-C also regulates lymphangiogenesis.


Subject(s)
Endothelial Growth Factors/physiology , Animals , Endothelial Growth Factors/chemistry , Endothelial Growth Factors/metabolism , Humans , Ligands , Neovascularization, Physiologic/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Signal Transduction , Vascular Endothelial Growth Factor B , Vascular Endothelial Growth Factor C
10.
J Cell Physiol ; 177(3): 439-52, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9808152

ABSTRACT

Vascular endothelial growth factor-C (VEGF-C) is a recently characterized member of the VEGF family of angiogenic polypeptides. We demonstrate here that VEGF-C is angiogenic in vitro when added to bovine aortic or lymphatic endothelial (BAE and BLE) cells but has little or no effect on bovine microvascular endothelial (BME) cells. As reported previously for VEGF, VEGF-C and basic fibroblast growth factor (bFGF) induced a synergistic in vitro angiogenic response in all three cells lines. Unexpectedly, VEGF and VEGF-C also synergized in the in vitro angiogenic response when assessed on BAE cells. Characterization of VEGF receptor (VEGFR) expression revealed that BME, BAE, and BLE cell lines express VEGFR-1 and -2, whereas of the three cell lines assessed, only BAE cells express VEGFR-3. We also demonstrate that VEGF-C increases plasminogen activator (PA) activity in the three bovine endothelial cell lines and that this is accompanied by a concomitant increase in PA inhibitor-1. Addition of alpha2-antiplasmin to BME cells co-treated with bFGF and VEGF-C partially inhibited collagen gel invasion. These results demonstrate, first, that by acting in concert with bFGF or VEGF, VEGF-C has a potent synergistic effect on the induction of angiogenesis in vitro and, second, that like VEGF and bFGF, VEGF-C is capable of altering endothelial cell extracellular proteolytic activity. These observations also highlight the notion of context, i.e., that the activity of an angiogenesis-regulating cytokine depends on the presence and concentration of other cytokines in the pericellular environment of the responding endothelial cell.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/pharmacology , Lymphokines/pharmacology , Neovascularization, Physiologic , Peptide Hydrolases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cattle , Cloning, Molecular , DNA, Complementary/genetics , Drug Synergism , Endothelium, Vascular/cytology , Molecular Sequence Data , Neovascularization, Physiologic/physiology , Plasminogen Activator Inhibitor 1/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/biosynthesis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3 , Vascular Endothelial Growth Factors
11.
Proc Natl Acad Sci U S A ; 95(20): 11709-14, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9751730

ABSTRACT

The vascular endothelial growth factor (VEGF) family has recently expanded by the identification and cloning of three additional members, namely VEGF-B, VEGF-C, and VEGF-D. In this study we demonstrate that VEGF-B binds selectively to VEGF receptor-1/Flt-1. This binding can be blocked by excess VEGF, indicating that the interaction sites on the receptor are at least partially overlapping. Mutating the putative VEGF receptor-1/Flt-1 binding determinants Asp63, Asp64, and Glu67 to alanine residues in VEGF-B reduced the affinity to VEGF receptor-1 but did not abolish binding. Mutational analysis of conserved cysteines contributing to VEGF-B dimer formation suggest a structural conservation with VEGF and platelet-derived growth factor. Proteolytic processing of the 60-kDa VEGF-B186 dimer results in a 34-kDa dimer containing the receptor-binding epitopes. The binding of VEGF-B to its receptor on endothelial cells leads to increased expression and activity of urokinase type plasminogen activator and plasminogen activator inhibitor 1, suggesting a role for VEGF-B in the regulation of extracellular matrix degradation, cell adhesion, and migration.


Subject(s)
Endothelial Growth Factors/metabolism , Plasminogen Activators/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Animals , Baculoviridae/genetics , Base Sequence , Binding Sites/genetics , Cattle , Cells, Cultured , Cysteine/genetics , DNA Primers/genetics , Endothelial Growth Factors/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Mice , Mutagenesis, Site-Directed , Plasminogen Activator Inhibitor 1/biosynthesis , Protein Processing, Post-Translational , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera , Transfection , Urokinase-Type Plasminogen Activator/biosynthesis , Vascular Endothelial Growth Factor B , Vascular Endothelial Growth Factor Receptor-1
12.
Proc Natl Acad Sci U S A ; 95(2): 548-53, 1998 Jan 20.
Article in English | MEDLINE | ID: mdl-9435229

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.


Subject(s)
Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Growth Factor/metabolism , Adult , Amino Acid Sequence , Humans , Ligands , Molecular Sequence Data , Receptors, Mitogen/metabolism , Receptors, Vascular Endothelial Growth Factor , Sequence Alignment , Substrate Specificity , Vascular Endothelial Growth Factor D , Vascular Endothelial Growth Factor Receptor-3
14.
J Biol Chem ; 272(40): 25176-83, 1997 Oct 03.
Article in English | MEDLINE | ID: mdl-9312130

ABSTRACT

We report here the cloning and characterization of human and mouse genes for vascular endothelial growth factor C (VEGF-C), a newly isolated member of the vascular endothelial growth factor/platelet-derived growth factor (VEGF/PDGF) family. Both VEGF-C genes comprise over 40 kilobase pairs of genomic DNA and consist of seven exons, all containing coding sequences. The VEGF homology domain of VEGF-C is encoded by exons 3 and 4. Exons 5 and 7 encode cysteine-rich motifs of the type C6C10CRC, and exon 6 encodes additional C10CXCXC motifs typical of a silk protein. A putative alternatively spliced rare RNA form lacking exon 4 was identified in human fibrosarcoma cells, and a major transcription start site was located in the human VEGF-C gene 523 base pairs upstream of the translation initiation codon. The upstream promoter sequences contain conserved putative binding sites for Sp-1, AP-2, and NF-kappaB transcription factors but no TATA box, and they show promoter activity when transfected into cells. The VEGF-C gene structure is thus assembled from exons encoding propeptides and distinct cysteine-rich domains in addition to the VEGF homology domain, and it shows both similarities and distinct differences in comparison with other members of the VEGF/PDGF gene family.


Subject(s)
Alternative Splicing , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cloning, Molecular , Cysteine , DNA Primers , Exons , Fibrosarcoma , Genetic Variation , Genomic Library , Humans , Introns , Mice , Molecular Sequence Data , Multigene Family , Polymerase Chain Reaction , Protein Biosynthesis , Restriction Mapping , Sequence Alignment , Sequence Homology, Nucleic Acid , Transcription Factors/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor C
15.
Dev Biol ; 188(1): 96-109, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9245515

ABSTRACT

The lymphangiogenic potency of endothelial growth factors has not been studied to date. This is partially due to the lack of in vivo lymphangiogenesis assays. We have studied the lymphatics of differentiated avian chorioallantoic membrane (CAM) using microinjection of Mercox resin, semi- and ultrathin sectioning, immunohistochemical detection of fibronectin and alpha-smooth muscle actin, and in situ hybridization with VEGFR-2 and VEGFR-3 probes. CAM is drained by lymphatic vessels which are arranged in a regular pattern. Arterioles and arteries are accompanied by a pair of interconnected lymphatics and form a plexus around bigger arteries. Veins are also associated with lymphatics, particularly larger veins, which are surrounded by a lymphatic plexus. The lymphatics are characterized by an extremely thin endothelial lining, pores, and the absence of a basal lamina. Patches of the extracellular matrix can be stained with an antibody against fibronectin. Lymphatic endothelial cells of differentiated CAM show ultrastructural features of this cell type. CAM lymphatics do not possess mediae. In contrast, the lymphatic trunks of the umbilical stalk are invested by a single but discontinuous layer of smooth muscle cells. CAM lymphatics express VEGFR-2 and VEGFR-3. Both the regular pattern and the typical structure of these lymphatics suggest that CAM is a suitable site to study the in vivo effects of potential lymphangiogenic factors. We have studied the effects of VEGF homo- and heterodimers, VEGF/PlGF heterodimers, and PlGF and VEGF-C homodimers on Day 13 CAM. All the growth factors containing at least one VEGF chain are angiogenic but do not induce lymphangiogenesis. PlGF-1 and PlGF-2 are neither angiogenic nor lymphangiogenic. VEGF-C is the first lymphangiogenic factor and seems to be highly chemoattractive for lymphatic endothelial cells. It induces proliferation of lymphatic endothelial cells and development of new lymphatic sinuses which are directed immediately beneath the chorionic epithelium. Our studies show that VEGF and VEGF-C are specific angiogenic and lymphangiogenic growth factors, respectively.


Subject(s)
Chorion/cytology , Endothelial Growth Factors/pharmacology , Lymphatic System/embryology , Lymphokines/pharmacology , Neovascularization, Physiologic , Actins/analysis , Animals , Cell Differentiation , Cell Division , Chick Embryo , Chorion/blood supply , Chorion/chemistry , Coturnix , DNA Probes/genetics , DNA Probes/metabolism , Endothelial Growth Factors/genetics , Fibronectins/analysis , Immunohistochemistry , In Situ Hybridization , Lymphatic System/cytology , Lymphokines/genetics , Microcirculation , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Cell Surface/analysis , Receptors, Cell Surface/genetics , Receptors, Growth Factor/analysis , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3 , Vascular Endothelial Growth Factors
16.
EMBO J ; 16(13): 3898-911, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9233800

ABSTRACT

The recently identified vascular endothelial growth factor C (VEGF-C) belongs to the platelet-derived growth factor (PDGF)/VEGF family of growth factors and is a ligand for the endothelial-specific receptor tyrosine kinases VEGFR-3 and VEGFR-2. The VEGF homology domain spans only about one-third of the cysteine-rich VEGF-C precursor. Here we have analysed the role of post-translational processing in VEGF-C secretion and function, as well as the structure of the mature VEGF-C. The stepwise proteolytic processing of VEGF-C generated several VEGF-C forms with increased activity towards VEGFR-3, but only the fully processed VEGF-C could activate VEGFR-2. Recombinant 'mature' VEGF-C made in yeast bound VEGFR-3 (K[D] = 135 pM) and VEGFR-2 (K[D] = 410 pM) and activated these receptors. Like VEGF, mature VEGF-C increased vascular permeability, as well as the migration and proliferation of endothelial cells. Unlike other members of the PDGF/VEGF family, mature VEGF-C formed mostly non-covalent homodimers. These data implicate proteolytic processing as a regulator of VEGF-C activity, and reveal novel structure-function relationships in the PDGF/VEGF family.


Subject(s)
Endothelial Growth Factors/metabolism , Protein Processing, Post-Translational , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Amino Acid Sequence , Animals , Antibodies/metabolism , COS Cells , Dimerization , Disulfides , Endopeptidases/metabolism , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelium, Vascular/metabolism , Humans , Molecular Sequence Data , Phosphorylation , Rabbits , Receptors, Vascular Endothelial Growth Factor , Tumor Cells, Cultured , Vascular Endothelial Growth Factor C
17.
Science ; 276(5317): 1423-5, 1997 May 30.
Article in English | MEDLINE | ID: mdl-9162011

ABSTRACT

No growth factors specific for the lymphatic vascular system have yet been described. Vascular endothelial growth factor (VEGF) regulates vascular permeability and angiogenesis, but does not promote lymphangiogenesis. Overexpression of VEGF-C, a ligand of the VEGF receptors VEGFR-3 and VEGFR-2, in the skin of transgenic mice resulted in lymphatic, but not vascular, endothelial proliferation and vessel enlargement. Thus, VEGF-C induces selective hyperplasia of the lymphatic vasculature, which is involved in the draining of interstitial fluid and in immune function, inflammation, and tumor metastasis. VEGF-C may play a role in disorders involving the lymphatic system and may be of potential use in therapeutic lymphangiogenesis.


Subject(s)
Endothelial Growth Factors/physiology , Lymphatic System/pathology , Animals , Cell Division , Cloning, Molecular , Endothelial Growth Factors/genetics , Endothelium, Lymphatic/physiology , Endothelium, Lymphatic/ultrastructure , Endothelium, Vascular/physiology , Humans , Hyperplasia , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Molecular Sequence Data , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Skin/pathology , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3
18.
Development ; 122(12): 3829-37, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9012504

ABSTRACT

The vascular endothelial growth factor family has recently been expanded by the isolation of two new VEGF-related factors, VEGF-B and VEGF-C. The physiological functions of these factors are largely unknown. Here we report the cloning and characterization of mouse VEGF-C, which is produced as a disulfide-linked dimer of 415 amino acid residue polypeptides, sharing an 85% identity with the human VEGF-C amino acid sequence. The recombinant mouse VEGF-C protein was secreted from transfected cells as VEGFR-3 (Flt4) binding polypeptides of 30-32x10(3) Mr and 22-23x10(3) Mr which preferentially stimulated the autophosphorylation of VEGFR-3 in comparison with VEGFR-2 (KDR). In in situ hybridization, mouse VEGF-C mRNA expression was detected in mesenchymal cells of postimplantation mouse embryos, particularly in the regions where the lymphatic vessels undergo sprouting from embryonic veins, such as the perimetanephric, axillary and jugular regions. In addition, the developing mesenterium, which is rich in lymphatic vessels, showed strong VEGF-C expression. VEGF-C was also highly expressed in adult mouse lung, heart and kidney, where VEGFR-3 was also prominent. The pattern of expression of VEGF-C in relation to its major receptor VEGFR-3 during the sprouting of the lymphatic endothelium in embryos suggests a paracrine mode of action and that one of the functions of VEGF-C may be in the regulation of angiogenesis of the lymphatic vasculature.


Subject(s)
Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Lymphatic System/embryology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Growth Factor/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , In Situ Hybridization , Ligands , Mice , Molecular Sequence Data , Phosphorylation , Protein Binding , RNA, Messenger/analysis , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution , Vascular Endothelial Growth Factor C , Vascular Endothelial Growth Factor Receptor-3
SELECTION OF CITATIONS
SEARCH DETAIL