Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters










Publication year range
1.
Science ; 373(6554)2021 07 30.
Article in English | MEDLINE | ID: mdl-34326210

ABSTRACT

Aging is an established risk factor for vascular diseases, but vascular aging itself may contribute to the progressive deterioration of organ function. Here, we show in aged mice that vascular endothelial growth factor (VEGF) signaling insufficiency, which is caused by increased production of decoy receptors, may drive physiological aging across multiple organ systems. Increasing VEGF signaling prevented age-associated capillary loss, improved organ perfusion and function, and extended life span. Healthier aging was evidenced by favorable metabolism and body composition and amelioration of aging-associated pathologies including hepatic steatosis, sarcopenia, osteoporosis, "inflammaging" (age-related multiorgan chronic inflammation), and increased tumor burden. These results indicate that VEGF signaling insufficiency affects organ aging in mice and suggest that modulating this pathway may result in increased mammalian life span and improved overall health.


Subject(s)
Aging/physiology , Healthy Aging , Longevity , Vascular Endothelial Growth Factor A/metabolism , Adipose Tissue , Animals , Blood Vessels/physiology , Body Composition , Body Fat Distribution , Carbohydrate Metabolism , Carcinogenesis , Endothelium, Vascular/metabolism , Fatty Liver/pathology , Female , Inflammation/prevention & control , Liver/pathology , Male , Mice , Microvascular Density , Microvessels/physiology , Osteoporosis/prevention & control , Oxygen Consumption , Sarcopenia/prevention & control , Signal Transduction , Vascular Endothelial Growth Factor A/blood
3.
Brain Res ; 909(1-2): 1-7, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11478916

ABSTRACT

Expressional patterns of the endothelial and neuronal forms of nitric oxide synthase (NOS) in cerebral ischemia were studied utilizing a permanent middle cerebral artery occlusion (PMCAO) model. Motor performance and infarct volumes were determined in the rats. Immunohistochemical staining for eNOS, nNOS and neurofilament were performed at 1, 2, 3, 5, 7 and 14 days after PMCAO. Vascular endothelial growth factor (VEGF) expression was determined by in-situ hybridization. PMCAO caused a reproducible cortical infarct with motor deficits in the rats. Double immunohistochemical stainings indicated that eNOS and nNOS were induced in ischemic neurons. Most stained neurons were positive for both NOS forms but some reacted with only one NOS antibody. nNOS expression peaked at 24-48 h after PMCAO, stained mainly the cytoplasm of core neurons, and disappeared after the 3rd day. eNOS expression increased until the 7th day, stained mainly the cytoplasm and membrane of penumbral cells and disappeared by the 14th day after PMCAO. VEGF expression was significantly induced in the penumbral zone in a similar distribution to eNOS. The anatomical and temporal pattern of VEGF and eNOS induction in the brain after permanent ischemia suggest that these mediators may play a role in protecting penumbral tissue from additional ischemic damage.


Subject(s)
Brain Ischemia/enzymology , Cerebral Cortex/enzymology , Endothelial Growth Factors/genetics , Endothelium, Vascular/enzymology , Lymphokines/genetics , Neurons/enzymology , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Immunohistochemistry , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Neurons/pathology , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Br J Ophthalmol ; 85(8): 991-5, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11466260

ABSTRACT

AIMS: To test the feasibility of gene transfer into hyaloid blood vessels and into preretinal neovascularisation in a rat model of retinopathy of prematurity (ROP), using different viral vectors. METHODS: Newborn rats were exposed to alternating hypoxic and hyperoxic conditions in order to induce ocular neovascularisation (ROP rats). Adenovirus, herpes simplex, vaccinia, and retroviral (MuLV based) vectors, all carrying the beta galactosidase (beta-gal) gene, were injected intravitreally on postnatal day 18 (P18). Two sets of controls were also examined: P18 ROP rats injected with saline and P18 rats that were raised in room air before the viral vectors or saline were injected. Two days after injection, the rats were killed, eyes enucleated, and beta-gal expression was examined by X-gal staining in whole mounts and in histological sections. RESULTS: Intravitreal injection of the adenovirus and vaccinia vectors yielded marked beta-gal expression in hyaloid blood vessels in the rat ROP model. Retinal expression of beta-gal with these vectors was limited almost exclusively to the vicinity of the injection site. Injection of herpes simplex yielded a punctuate pattern of beta-gal expression in the retina but not in blood vessels. No significant beta-gal expression occurred in rat eyes injected with the retroviral vector. CONCLUSIONS: Adenovirus is an efficient vector for gene transfer into blood vessels in an animal model of ROP. This may be a first step towards utilising gene transfer as a tool for modulating ocular neovascularisation for experimental and therapeutic purposes.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Retinal Vessels , Retinopathy of Prematurity/therapy , Animals , Animals, Newborn , Disease Models, Animal , Feasibility Studies , Gene Expression , Genetic Vectors/administration & dosage , Humans , Infant, Newborn , Mastadenovirus/genetics , Rats , Rats, Inbred Strains , Retroviridae/genetics , Simplexvirus/genetics , Vaccinia virus/genetics , Vitreous Body , beta-Galactosidase/genetics
5.
Am J Physiol Cell Physiol ; 280(6): C1367-74, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11350731

ABSTRACT

Development of microvascular networks is set to meet the metabolic requirements of the tissue they perfuse. Accordingly, impairment of oxygen homeostasis, either due to increased oxygen consumption or as a result of blood vessel occlusion, triggers compensatory neovascularization. This feedback reaction is mediated by a hypoxia- and hypoglycemia-induced vascular endothelial growth factor (VEGF). VEGF accumulates under stress as a result of increased hypoxia-inducible factor-1alpha-mediated transcription, stabilization of the mRNA, and the function of a hypoxia-refractory internal ribosome entry site within its 5'-untranslated region. Matching of vascular density to the metabolic needs of the tissue may include a process of hyperoxia-induced vessel regression. Thus newly formed vascular networks may undergo a natural process of vascular pruning that takes place whenever VEGF, acting as a vascular survival factor, is downregulated below the level required to sustain immature vessels. Immature vessels are particularly vulnerable and are selectively obliterated upon withdrawal of VEGF. The plasticity window for vessel regression is determined by a delay in the recruitment of periendothelial cells to the preformed endothelial plexus. Thus fine-tuning of microvascular density takes place mostly in the newly formed plexus, but the mature system is refractory to episodic changes in tissue oxygenation. These regulatory links may malfunction in certain pathological settings.


Subject(s)
Endothelial Growth Factors/physiology , Homeostasis/physiology , Hypoxia/physiopathology , Lymphokines/physiology , Neovascularization, Physiologic/physiology , Oxygen/physiology , Animals , Humans , Hyperoxia/physiopathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
Development ; 128(9): 1531-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11290292

ABSTRACT

Normal cardiovascular development is exquisitely dependent on the correct dosage of the angiogenic growth factor and vascular morphogen vascular endothelial growth factor (VEGF). However, cardiac expression of VEGF is also robustly augmented during hypoxic insults, potentially mediating the well-established teratogenic effects of hypoxia on heart development. We report that during normal heart morphogenesis VEGF is specifically upregulated in the atrioventricular (AV) field of the heart tube soon after the onset of endocardial cushion formation (i.e. the endocardium-derived structures that build the heart septa and valves). To model hypoxia-dependent induction of VEGF in vivo, we conditionally induced VEGF expression in the myocardium using a tetracycline-regulated transgenic system. Premature induction of myocardial VEGF in E9.5 embryos to levels comparable with those induced by hypoxia prevented formation of endocardial cushions. When added to explanted embryonic AV tissue, VEGF fully inhibited endocardial-to-mesenchymal transformation. Transformation was also abrogated in AV explants subjected to experimental hypoxia but fully restored in the presence of an inhibitory soluble VEGF receptor 1 chimeric protein. Together, these results suggest a novel developmental role for VEGF as a negative regulator of endocardial-to-mesenchymal transformation that underlies the formation of endocardial cushions. Moreover, ischemia-induced VEGF may be the molecular link between hypoxia and congenital defects in heart septation.


Subject(s)
Endocardium/embryology , Endothelial Growth Factors/isolation & purification , Heart Defects, Congenital/etiology , Heart Septum/embryology , Heart Valves/embryology , Lymphokines/isolation & purification , Animals , Endocardium/cytology , Hypoxia/complications , In Vitro Techniques , Mesoderm/cytology , Mice , Mice, Transgenic , Morphogenesis , Tissue Distribution , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Brain Res Dev Brain Res ; 118(1-2): 135-45, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10611512

ABSTRACT

Astrocytes play a key role in the development of retinal vessels by detecting hypoxia in developing retina and secreting the hypoxia-induced angiogenic factor VEGF to induce vessel formation. The astrocytes which play this role are themselves spreading over the retina, just ahead of the growing vessels. To understand the mechanisms which keep astrocytes in this strategic 'just ahead' position we have studied the effects of hyperoxia and hypoxia on astrocyte differentiation and movement in situ in neonatal rat retina and in primary culture. Hyperoxia in situ inhibited the stellation of astrocytes, so that they persisted in a relatively unbranched form, which accumulated at the edge of their spreading population; hyperoxia permitted but did not accelerate migration. Conversely, hypoxia induced unstellated astrocytes to stellate within 6 h. If the hypoxia was abnormally severe, it caused the astrocytes to hyperstellate and slowed their spread. Astrocytes in primary culture did not change morphology or motility when challenged by hypoxia. When treated with medium conditioned by retina however, astrocytes became mobile and, if the medium was conditioned by hypoxic retina, became stellate. These results suggest that the oxygen released by retinal vessels maintains the mobility of astrocytes, via a diffusible factor released by other retinal cells. Conversely, naturally generated hypoxia of developing retina plays a triple role, inducing astrocytes to stellate, to end their migration and to produce VEGF, thereby inducing vessel formation. The induction of stellation is mediated by a diffusible factor released by other retinal cells. Thus hypoxia of the retina generated by neural maturation induces key events in both the differentiation of astrocytes and the formation of blood vessels.


Subject(s)
Aging/physiology , Astrocytes/cytology , Astrocytes/physiology , Oxygen/metabolism , Retina/physiology , Animals , Cell Differentiation/physiology , Cell Hypoxia/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Mitosis/physiology , Rats , Rats, Sprague-Dawley , Retina/cytology , Retina/growth & development
10.
Nat Med ; 5(10): 1135-42, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10502816

ABSTRACT

Cardiac rupture is a fatal complication of acute myocardial infarction lacking treatment. Here, acute myocardial infarction resulted in rupture in wild-type mice and in mice lacking tissue-type plasminogen activator, urokinase receptor, matrix metalloproteinase stromelysin-1 or metalloelastase. Instead, deficiency of urokinase-type plasminogen activator (u-PA-/-) completely protected against rupture, whereas lack of gelatinase-B partially protected against rupture. However, u-PA-/- mice showed impaired scar formation and infarct revascularization, even after treatment with vascular endothelial growth factor, and died of cardiac failure due to depressed contractility, arrhythmias and ischemia. Temporary administration of PA inhibitor-1 or the matrix metalloproteinase-inhibitor TIMP-1 completely protected wild-type mice against rupture but did not abort infarct healing, thus constituting a new approach to prevent cardiac rupture after acute myocardial infarction.


Subject(s)
Cardiac Output, Low/etiology , Heart Rupture/etiology , Metalloendopeptidases/antagonists & inhibitors , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Plasminogen Inactivators/therapeutic use , Protease Inhibitors/therapeutic use , Animals , Arrhythmias, Cardiac , Bone Marrow Transplantation , Cell Movement , Collagenases/metabolism , Gene Transfer Techniques , Leukocytes/cytology , Leukocytes/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 9 , Mice , Mice, Mutant Strains , Neovascularization, Physiologic/drug effects , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activators/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
11.
J Clin Invest ; 103(2): 159-65, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9916127

ABSTRACT

Features that distinguish tumor vasculatures from normal blood vessels are sought to enable the destruction of preformed tumor vessels. We show that blood vessels in both a xenografted tumor and primary human tumors contain a sizable fraction of immature blood vessels that have not yet recruited periendothelial cells. These immature vessels are selectively obliterated as a consequence of vascular endothelial growth factor (VEGF) withdrawal. In a xenografted glioma, the selective vulnerability of immature vessels to VEGF loss was demonstrated by downregulating VEGF transgene expression using a tetracycline-regulated expression system. In human prostate cancer, the constitutive production of VEGF by the glandular epithelium was suppressed as a consequence of androgen-ablation therapy. VEGF loss led, in turn, to selective apoptosis of endothelial cells in vessels devoid of periendothelial cells. These results suggest that the unique dependence on VEGF of blood vessels lacking periendothelial cells can be exploited to reduce an existing tumor vasculature.


Subject(s)
Blood Vessels/metabolism , Endothelial Growth Factors/metabolism , Glioma/blood supply , Lymphokines/metabolism , Neoplasms, Experimental/blood supply , Androgens/metabolism , Animals , Apoptosis/physiology , Down-Regulation/genetics , Endothelial Growth Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioma/pathology , Humans , In Situ Nick-End Labeling , Lymphokines/genetics , Male , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , Tetracycline/pharmacology , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Proc Natl Acad Sci U S A ; 95(18): 10820-5, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9724788

ABSTRACT

The sequence of events that leads to tumor vessel regression and the functional characteristics of these vessels during hormone-ablation therapy are not known. This is because of the lack of an appropriate animal model and monitoring technology. By using in vivo microscopy and in situ molecular analysis of the androgen-dependent Shionogi carcinoma grown in severe combined immunodeficient mice, we show that castration of these mice leads to tumor regression and a concomitant decrease in vascular endothelial growth factor (VEGF) expression. Androgen withdrawal is known to induce apoptosis in Shionogi tumor cells. Surprisingly, tumor endothelial cells begin to undergo apoptosis before neoplastic cells, and rarefaction of tumor vessels precedes the decrease in tumor size. The regressing vessels begin to exhibit normal phenotype, i.e., lower diameter, tortuosity, vascular permeability, and leukocyte adhesion. Two weeks after castration, a second wave of angiogenesis and tumor growth begins with a concomitant increase in VEGF expression. Because human tumors often relapse following hormone-ablation therapy, our data suggest that these patients may benefit from combined anti-VEGF therapy.


Subject(s)
Androgens/physiology , Castration , Cell Death , Endothelial Growth Factors/physiology , Endothelium, Vascular/pathology , Lymphokines/physiology , Neoplasms, Hormone-Dependent/pathology , Animals , Blotting, Northern , Endothelial Growth Factors/genetics , Endothelium, Vascular/physiopathology , Humans , Lymphokines/genetics , Male , Mice , Mice, SCID , Neoplasms, Hormone-Dependent/blood supply , Neovascularization, Pathologic , RNA, Messenger/genetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
Nature ; 394(6692): 485-90, 1998 Jul 30.
Article in English | MEDLINE | ID: mdl-9697772

ABSTRACT

As a result of deprivation of oxygen (hypoxia) and nutrients, the growth and viability of cells is reduced. Hypoxia-inducible factor (HIF)-1alpha helps to restore oxygen homeostasis by inducing glycolysis, erythropoiesis and angiogenesis. Here we show that hypoxia and hypoglycaemia reduce proliferation and increase apoptosis in wild-type (HIF-1alpha+/+) embryonic stem (ES) cells, but not in ES cells with inactivated HIF-1alpha genes (HIF-1alpha-/-); however, a deficiency of HIF-1alpha does not affect apoptosis induced by cytokines. We find that hypoxia/hypoglycaemia-regulated genes involved in controlling the cell cycle are either HIF-1alpha-dependent (those encoding the proteins p53, p21, Bcl-2) or HIF-1alpha-independent (p27, GADD153), suggesting that there are at least two different adaptive responses to being deprived of oxygen and nutrients. Loss of HIF-1alpha reduces hypoxia-induced expression of vascular endothelial growth factor, prevents formation of large vessels in ES-derived tumours, and impairs vascular function, resulting in hypoxic microenvironments within the tumour mass. However, growth of HIF-1alpha tumours was not retarded but was accelerated, owing to decreased hypoxia-induced apoptosis and increased stress-induced proliferation. As hypoxic stress contributes to many (patho)biological disorders, this new role for HIF-1alpha in hypoxic control of cell growth and death may be of general pathophysiological importance.


Subject(s)
Apoptosis/physiology , Cell Division/physiology , DNA-Binding Proteins/physiology , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/etiology , Nuclear Proteins/physiology , Oxygen/physiology , Transcription Factors , Animals , CHO Cells , Cell Hypoxia , Cell Line , Cricetinae , DNA-Binding Proteins/genetics , Endothelial Growth Factors/physiology , Gene Targeting , Glucose/physiology , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Lymphokines/physiology , Mice , Mice, Nude , Nuclear Proteins/genetics , Regional Blood Flow , Stem Cells , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Blood ; 92(3): 939-45, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9680362

ABSTRACT

Plasminogen-activator inhibitor type I (PAI-1), the primary inhibitor of urinary-type plasminogen activator, is thought to play an important role in the control of stroma invasion by both endothelial and tumor cells. Using an in vitro angiogenesis model of capillary extension through a preformed monolayer, in conjunction with in situ hybridization analysis, we showed that PAI-1 mRNA is specifically induced in cells juxtaposed next to elongating sprouts. To further establish that PAI-1 expression is induced as a consequence of a direct contact with endothelial cells, coculture experiments were performed. PAI-1 mRNA was induced exclusively in fibroblasts (L-cells) contacting endothelial cell (LE-II) colonies. Reporter gene constructs driven by a PAI-1 promoter and stably transfected into L-cells were used to establish that both mouse and rat PAI-1 promoters mediate apposition-dependent regulation. This mode of PAI-1 regulation is not mediated by plasmin, as an identical spatial pattern of expression was detected in cocultures treated with plasmin inhibitors. Because endothelial cells may establish direct contacts with fibroblasts only during angiogenesis, we propose that focal induction of PAI-1 at the site of heterotypic cell contacts provides a mechanism to negate excessive pericellular proteolysis associated with endothelial cell invasion.


Subject(s)
Endothelium, Vascular/cytology , Gene Expression Regulation , Neovascularization, Physiologic/physiology , Plasminogen Activator Inhibitor 1/biosynthesis , Animals , Aorta/cytology , Capillaries/cytology , Cell Communication , Coculture Techniques , Fibrinolysin/antagonists & inhibitors , Fibrinolysin/pharmacology , Genes, Reporter , In Situ Hybridization , L Cells/metabolism , Mice , Plasminogen Activator Inhibitor 1/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , Rats , Recombinant Fusion Proteins/biosynthesis , Species Specificity , Transfection
15.
Mol Cell Biol ; 18(6): 3112-9, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9584152

ABSTRACT

Vascular endothelial growth factor (VEGF) is a hypoxia-inducible angiogenic growth factor that promotes compensatory angiogenesis in circumstances of oxygen shortage. The requirement for translational regulation of VEGF is imposed by the cumbersome structure of the 5' untranslated region (5'UTR), which is incompatible with efficient translation by ribosomal scanning, and by the physiologic requirement for maximal VEGF production under conditions of hypoxia, where overall protein synthesis is compromised. Using bicistronic reporter gene constructs, we show that the 1,014-bp 5'UTR of VEGF contains a functional internal ribosome entry site (IRES). Efficient cap-independent translation is maintained under hypoxia, thereby securing efficient production of VEGF even under unfavorable stress conditions. To identify sequences within the 5'UTR required for maximal IRES activity, deletion mutants were analyzed. Elimination of the majority (851 nucleotides) of internal 5'UTR sequences not only maintained full IRES activity but also generated a significantly more potent IRES. Activity of the 163-bp long "improved" IRES element was abrogated, however, following substitution of a few bases near the 5' terminus as well as substitutions close to the translation start codon. Both the full-length 5'UTR and its truncated version function as translational enhancers in the context of a monocistronic mRNA.


Subject(s)
Endothelial Growth Factors/genetics , Lymphokines/genetics , Oxygen/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomes/metabolism , 3T3 Cells , Animals , Cell Hypoxia , Cells, Cultured , Endothelial Growth Factors/biosynthesis , Humans , Lymphokines/biosynthesis , Mice , Neovascularization, Pathologic/genetics , Rats , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Development ; 125(9): 1591-8, 1998 May.
Article in English | MEDLINE | ID: mdl-9521897

ABSTRACT

Little is known about how the initial endothelial plexus is remodelled into a mature and functioning vascular network. Studying postnatal remodelling of the retina vasculature, we show that a critical step in vascular maturation, namely pericyte recruitment, proceeds by outmigration of cells positive for (alpha)-smooth muscle actin from arterioles and that coverage of primary and smaller branches lags many days behind formation of the endothelial plexus. The transient existence of a pericyte-free endothelial plexus coincides temporally and spatially with the process of hyperoxia-induced vascular pruning, which is a mechanism for fine tuning of vascular density according to available oxygen. Acquisition of a pericyte coating marks the end of this plasticity window. To substantiate that association with pericytes stabilizes the vasculature, endothelial-pericyte associations were disrupted by intraocular injection of PDGF-BB. Ectopic PDGF-BB caused the detachment of PDGF-beta receptor-positive pericytes from newly coated vessels, presumably through interference with endogenous cues, but had no effect on mature vessels. Disruption of endothelial-pericyte associations resulted in excessive regression of vascular loops and abnormal remodelling. Conversely, intraocular injection of VEGF accelerated pericyte coverage of the preformed endothelial plexus, thereby revealing a novel function of this pleiotropic angiogenic growth factor. These findings also provide a cellular basis for clinical observations that vascular regression in premature neonates subjected to oxygen therapy [i.e. in retinopathy of prematurity] drops precipitously upon maturation of retina vessels and a mechanistic explanation to our previous findings that VEGF can rescue immature vessels from hyperoxia-induced regression.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/growth & development , Lymphokines/pharmacology , Neovascularization, Physiologic/physiology , Plant Lectins , Platelet-Derived Growth Factor/pharmacology , Retinal Vessels/growth & development , Actins/analysis , Animals , Animals, Newborn , Arterioles/cytology , Becaplermin , Cell Movement , Endothelium, Vascular/cytology , Humans , Hyperoxia , Infant, Newborn , Lectins/analysis , Proto-Oncogene Proteins c-sis , Rats , Receptors, Platelet-Derived Growth Factor/analysis , Retinopathy of Prematurity/physiopathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
FEBS Lett ; 425(3): 441-7, 1998 Apr 03.
Article in English | MEDLINE | ID: mdl-9563510

ABSTRACT

Heparin-binding epidermal growth factor-like growth factor (HB-EGF), a potent mitogen and migration factor for vascular smooth muscle cells (SMC), promoted neovascularization in vivo in the rabbit cornea. MRI demonstrated quantitatively the angiogenic effect of HB-EGF when introduced subcutaneously into nude mice. HB-EGF is not directly mitogenic to endothelial cells but it induced the migration of bovine endothelial cells and release of endothelial cell mitogenic activity from bovine vascular SMC. This mitogenic activity was specifically blocked by neutralizing anti-vascular endothelial growth factor (VEGF) antibodies. In contrast, EGF or transforming growth factor-alpha (TGF-alpha) had almost no effect on release of endothelial mitogenicity from SMC. In addition, RT-PCR analysis demonstrated that VEGF165 mRNA levels were increased in vascular SMC 4-10-fold by 0.35-2 nM of HB-EGF, respectively. Our data suggest that HB-EGF, as a mediator of intercellular communication, may play a new important role in supporting wound healing, tumor progression and atherosclerosis by stimulating angiogenesis.


Subject(s)
Cell Communication/physiology , Cornea/blood supply , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Epidermal Growth Factor/pharmacology , Lymphokines/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Cattle , Cell Movement/drug effects , Cells, Cultured , Corneal Neovascularization/blood , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins , Magnetic Resonance Imaging , Mice , Mice, Nude , Neovascularization, Physiologic/drug effects , RNA, Messenger/metabolism , Rabbits , Skin/blood supply , Transforming Growth Factor alpha/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
Ophthalmology ; 105(3): 412-6, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9499769

ABSTRACT

BACKGROUND AND OBJECTIVE: Vascular endothelial growth factor (VEGF), a key mediator of intraocular neovascularization, is triggered by hypoxia and has been shown in the eyes of animal models of central retinal vein occlusion (CRVO). However, there is little information on CRVO in humans, in particular, the identity of VEGF-producing cells. STUDY DESIGN: The study design was molecular localization of the site of VEGF production in the eyes of patients with CRVO. PARTICIPANTS: Ten formaldehyde solution-fixed and paraffin-embedded eyes removed surgically from patients with CRVO and neovascular glaucoma were studied. Five eyes with uveal melanoma and no neovascularization served as control specimens. METHODS: Thin whole-eye sections were hybridized in situ with a VEGF-specific probe to identify cells producing VEGF messenger RNA (mRNA). RESULTS: All ten eyes with CRVO showed evidence of intraretinal expression of VEGF mRNA. In all eyes, the inner nuclear layer showed VEGF-upregulated expression. Upregulation of VEGF mRNA was identified in four eyes in the ganglion cell layer and in two eyes with retinal detachment in the outer nuclear layer as well. CONCLUSIONS: The population of VEGF-producing retinal cells in each eye is likely to represent cells residing in ischemic regions of the retina. Hypoxia-induced VEGF is, most likely, the linking factor between retinal ischemia and iris and retinal neovascularization in CRVO.


Subject(s)
Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Retina/metabolism , Retinal Vein Occlusion/metabolism , Up-Regulation , Aged , Aged, 80 and over , Endothelial Growth Factors/genetics , Eye Enucleation , Female , Glaucoma, Neovascular/metabolism , Glaucoma, Neovascular/pathology , Humans , In Situ Hybridization , Lymphokines/genetics , Male , RNA, Messenger/metabolism , Retinal Vein Occlusion/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Proc Natl Acad Sci U S A ; 94(24): 13203-8, 1997 Nov 25.
Article in English | MEDLINE | ID: mdl-9371824

ABSTRACT

We show here that elevated levels of gonadotropins (luteinizing hormone and follicle stimulating hormone), as found in menopause or after ovariectomy, promote growth of human ovarian carcinoma by induction of tumor angiogenesis. Human epithelial ovarian cancer tumors progressed faster in ovariectomized mice. This induced growth could be attributed to the elevated levels of gonadotropins associated with loss of ovarian function because direct administration of gonadotropins also was effective in promoting tumor progression in vivo. On the other hand, gonadotropins had no direct effect on the proliferation of human ovarian cancer cells in vitro. Using MRI, we demonstrated that ovariectomy significantly (P < 0.02) induces neovascularization of human ovarian carcinoma spheroids implanted in nude mice. Moreover, conditioned medium of gonadotropin-treated human ovarian carcinoma cells showed increased mitogenic activity to bovine endothelial cells, and this activity could be blocked by neutralizing antibodies against luteinizing hormone and against vascular endothelial growth factor. Accordingly, gonadotropin stimulation resulted in a dose-dependent-induced expression of vascular endothelial growth factor in monolayer culture as well as in the outer proliferating cells of human ovarian cancer spheroids. These results demonstrate the significance of the elevated levels of gonadotropins, as found in menopause and in all ovarian cancer patients, on the progression of ovarian cancer and could explain the protective effect of estrogen replacement therapy. Based on these results, we suggest that hormonal therapy aimed at lowering the circulating levels of gonadotropins may possibly prolong remission in ovarian cancer by extending tumor dormancy.


Subject(s)
Neovascularization, Pathologic/physiopathology , Ovarian Neoplasms/blood supply , Ovary/physiopathology , Animals , Endothelial Growth Factors/genetics , Female , Follicle Stimulating Hormone/metabolism , Humans , In Situ Hybridization , Luteinizing Hormone/metabolism , Lymphokines/genetics , Magnetic Resonance Imaging , Menopause , Mice , Mice, Nude , Ovarian Neoplasms/physiopathology , Ovariectomy , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
Proc Natl Acad Sci U S A ; 94(16): 8761-6, 1997 Aug 05.
Article in English | MEDLINE | ID: mdl-9238051

ABSTRACT

We have recently shown that VEGF functions as a survival factor for newly formed vessels during developmental neovascularization, but is not required for maintenance of mature vessels. Reasoning that expanding tumors contain a significant fraction of newly formed and remodeling vessels, we examined whether abrupt withdrawal of VEGF will result in regression of preformed tumor vessels. Using a tetracycline-regulated VEGF expression system in xenografted C6 glioma cells, we showed that shutting off VEGF production leads to detachment of endothelial cells from the walls of preformed vessels and their subsequent death by apoptosis. Vascular collapse then leads to hemorrhages and extensive tumor necrosis. These results suggest that enforced withdrawal of vascular survival factors can be applied to target preformed tumor vasculature in established tumors. The system was also used to examine phenotypes resulting from over-expression of VEGF. When expression of the transfected VEGF cDNA was continuously "on," tumors became hyper-vascularized with abnormally large vessels, presumably arising from excessive fusions. Tumors were significantly less necrotic, suggesting that necrosis in these tumors is the result of insufficient angiogenesis.


Subject(s)
Endothelial Growth Factors/physiology , Gene Expression Regulation, Neoplastic , Glioma/blood supply , Hemangioblastoma/pathology , Lymphokines/physiology , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic , Animals , Cell Line , Gene Expression Regulation, Neoplastic/drug effects , Mice , Mice, Nude , Protein Synthesis Inhibitors/pharmacology , Tetracycline/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...