Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Br J Pharmacol ; 172(12): 2961-73, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25631232

ABSTRACT

BACKGROUND AND PURPOSE: Hydrogen sulfide (H2 S), an endogenous volatile mediator with pleiotropic functions, promotes vasorelaxation, exerts anti-inflammatory actions and regulates angiogenesis. Previously, the SH-containing angiotensin-converting enzyme inhibitor (ACEI), zofenopril, was identified as being effective in preserving endothelial function and inducing angiogenesis among ACEIs. Based on the H2 S donor property of its active metabolite zofenoprilat, the objective of this study was to evaluate whether zofenoprilat-induced angiogenesis was due to increased H2 S availability. EXPERIMENTAL APPROACH: HUVECs were used for in vitro studies of angiogenesis, whereas the Matrigel plug assay was used for in vivo assessments. KEY RESULTS: Zofenoprilat-treated HUVECs showed an increase in all functional features of the angiogenic process in vitro. As zofenoprilat induced the expression of CSE (cystathionine-γ-lyase) and the continuous production of H2 S, CSE inhibition or silencing blocked the ability of zofenoprilat to induce angiogenesis, both in vitro and in vivo. The molecular mechanisms underlying H2 S/zofenoprilat-induced angiogenesis were dependent on Akt, eNOS and ERK1/2 cascades. ATP-sensitive potassium (KATP ) channels, the molecular target that mediates part of the vascular functions of H2 S, were shown to be involved in the upstream activation of Akt and ERK1/2. Moreover, the up-regulation of fibroblast growth factor-2 was dependent on CSE-derived H2 S response to H2 S and KATP activation. CONCLUSIONS AND IMPLICATIONS: Zofenoprilat induced a constant production of H2 S that stimulated the angiogenic process through a KATP channel/Akt/eNOS/ERK1/2 pathway. Thus, zofenopril can be considered as a pro-angiogenic drug acting through H2 S release and production, useful in cardiovascular pathologies where vascular functions need to be re-established and functional angiogenesis induced.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Captopril/analogs & derivatives , Hydrogen Sulfide/metabolism , Neovascularization, Physiologic/drug effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Captopril/pharmacology , Cystathionine gamma-Lyase/metabolism , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/metabolism , Human Umbilical Vein Endothelial Cells , KATP Channels/drug effects , KATP Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Up-Regulation/drug effects
2.
Nanotechnology ; 19(27): 275101, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-21828694

ABSTRACT

Plasma deposition was applied to deposit smooth and nanostructured fluorocarbon coatings on polyethylene terephthalate substrates, with the aim to obtain surfaces with identical chemical composition but different roughness to improve the endothelialization process on PET surfaces. We found that increased roughness was associated with enhanced endothelial cell response, as shown by the ability of cells to grow and adhere to nanostructures. We also observed specific interaction of filopodia protruding from the cell membrane with individual nanostructures, leading to increased cell attachment, spreading and cell viability. Among the modified surfaces, one termed PET-tfl90 emerged as the one capable of best sustaining the formation of a confluent monolayer of endothelial cells. In conclusion, PET modified by nanostructured fluorocarbon film represents an improved graft material, over conventional PET, for endothelial cell adhesion and growth.

3.
Clin Exp Metastasis ; 23(3-4): 187-201, 2006.
Article in English | MEDLINE | ID: mdl-17028923

ABSTRACT

Endothelial cells express two dependent intercellular adhesion molecules: vascular endothelial (VE)-cadherin, specific for endothelial cells, and N-cadherin, also present in neuronal, lens, skeletal and heart muscle cells, osteoblasts, pericytes and fibroblasts. While there exists a vast amount of evidence that VE-cadherin promotes angiogenesis, the role of N-cadherin still remains to be elucidated. We found that a soluble 90-kDa fragment N-cadherin promotes angiogenesis in the rabbit cornea assay and in the chorioallantoic assay when cleaved enzymatically from the extracellular domain of N-cadherin. Soluble N-cadherin stimulates migration of endothelial cells in the wound healing assay and stimulates phosphorylation of extracellular regulated kinase. In vitro experiments with PD173074 and knock-down of N-cadherin and fibroblast growth factor (FGF)-receptor, showed that the pro-angiogenic effect of soluble N-cadherin is N-cadherin- and FGF-receptor-dependent. Our results suggest that soluble N-cadherin stimulates migration of endothelial cells through the FGF-receptor.


Subject(s)
Cadherins/pharmacology , Endothelium, Vascular/drug effects , Neovascularization, Physiologic/drug effects , Animals , Cell Line , Cell Movement/drug effects , Cornea/blood supply , Humans , Models, Biological , Phosphorylation , Rabbits , Receptors, Fibroblast Growth Factor/physiology , Time Factors , Wound Healing
4.
Cell Death Differ ; 13(7): 1088-96, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16410806

ABSTRACT

Recent evidences suggest that Abeta peptides modulate endothelial cell (EC) functions. At low concentrations, Abeta1-40 enhances the pro-angiogenic activity of FGF-2, whereas deposition of excess Abeta causes EC dysfunction and cerebral amyloid angiopathy (CAA). We investigated whether FGF-2 attenuates EC dysfunction caused by pathological Abeta levels. We studied Abeta1-40 on EC survival, as well as on signals responsible of their angiogenic phenotype. At 5-50 microM Abeta1-40 reduced EC population, caused apoptosis, downregulated FGF-2 production, inhibited FGF-2 binding to heparin, and FGFR1 phosphorylation. Toxic effects were owing to lack of FGF-2 stimulation, as EC overexpressing FGF-2 displayed extraordinary resistance to Abeta1-40 injuries. The FGF-2 mechanism responsible for reversing damages, involves the downstream enhancement of Akt, a pathway independent of eNOS activation. In conclusion, we demonstrate that FGF-2 protects EC from the effects of excess Abeta1-40, suggesting that it may attenuate the consequences of Abeta deposition in pathologies as CAA.


Subject(s)
Amyloid beta-Peptides/pharmacology , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/metabolism , Peptide Fragments/pharmacology , Animals , Blotting, Western , CHO Cells , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Fibroblast Growth Factor 2/genetics , Humans , Mice , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/pharmacology
5.
Br J Cancer ; 88(9): 1484-91, 2003 May 06.
Article in English | MEDLINE | ID: mdl-12778081

ABSTRACT

The nitric oxide synthase (NOS) pathway has been clearly demonstrated to regulate angiogenesis. Increased levels of NO correlate with tumour growth and spreading in different experimental and human cancers. Drugs interfering with the NOS pathway may be useful in angiogenesis-dependent tumours. The aim of this study was to pharmacologically characterise certain ruthenium-based compounds, namely NAMI-A, KP1339, and RuEDTA, as potential NO scavengers to be used as antiangiogenic/antitumour agents. NAMI-A, KP1339 and RuEDTA were able to bind tightly and inactivate free NO in solution. Formation of ruthenium-NO adducts was documented by electronic absorption, FT-IR spectroscopy and (1)H-NMR. Pretreatment of rabbit aorta rings with NAMI-A, KP1339 or RuEDTA reduced endothelium-dependent vasorelaxation elicited by acetylcholine. This effect was reversed by 8-Br-cGMP. The key steps of angiogenesis, endothelial cell proliferation and migration stimulated by vascular endothelial growth factor (VEGF) or NO donor drugs, were blocked by NAMI-A, KP1339 and RuEDTA, these compounds being devoid of any cytotoxic activity. When tested in vivo, NAMI-A inhibited angiogenesis induced by VEGF. It is likely that the antitumour properties previously observed for ruthenium-based NO scavengers, such as NAMI-A, are related to their NO-related antiangiogenic properties.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/pharmacology , Endothelium, Vascular/physiology , Nitric Oxide/physiology , Organometallic Compounds/pharmacology , Ruthenium Compounds/pharmacology , Cell Line , Cell Survival/drug effects , Chemotaxis/drug effects , Coronary Vessels , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Free Radical Scavengers , Humans , Nitric Oxide Synthase/antagonists & inhibitors , Spectroscopy, Fourier Transform Infrared , Venules/drug effects , Venules/physiology
6.
J Vasc Res ; 38(4): 389-97, 2001.
Article in English | MEDLINE | ID: mdl-11455210

ABSTRACT

Fibroblast growth factor-2 (FGF2) and vascular endothelial growth factor (VEGF) exert their angiogenic activity by interacting with endothelial cells in a distinct manner. In this study, we investigated the morphological features of endothelial cells of the chick embryo chorioallantoic membrane (CAM) microvasculature after stimulation with FGF2 or VEGF. In order to provide a continuous delivery of the growth factor, we utilized a recently developed gelatin sponge/CAM assay in which a limited number of FGF2- or VEGF-transfected cells were adsorbed onto gelatin sponges and applied on the top of the CAM on day 8 of development. Their angiogenic activity was compared to that exerted by a single bolus of the corresponding growth factor. All the angiogenic stimuli induced a comparable vasoproliferative response, as demonstrated by the appearance of similar numbers of immature blood vessels within the sponge on day 12. No angiogenic response was observed in CAMs implanted with the corresponding parental cell lines or vehicle. Electron microscopy demonstrated that VEGF-overexpressing cells modified the phenotype of the endothelium of the blood vessels at the boundary between the implant and the surrounding CAM mesenchyme. The endothelial lining of 30% of these vessels showed segmental attenuations, was frequently interrupted and became fenestrated, mimicking what is observed in tumor vasculature. In contrast, the vessels consisted of continuous endothelium sealed by tight junctions in all the other experimental conditions. These results indicate that FGF2 and VEGF interact with endothelial cells of the CAM in a distinct manner. Both growth factors induce a potent angiogenic response, but only VEGF delivered in a continuous manner by its transfectants can modify the phenotype of the otherwise quiescent endothelium of CAM blood microvessels. The gelatin sponge/CAM assay may constitute a new model to study the mechanisms leading to endothelial fenestration in tumor growth.


Subject(s)
Allantois/blood supply , Chorion/blood supply , Endothelial Growth Factors/administration & dosage , Endothelium, Vascular/cytology , Fibroblast Growth Factor 2/administration & dosage , Lymphokines/administration & dosage , Neovascularization, Physiologic , Adsorption , Animals , Aorta , Capillaries/physiology , Capillaries/ultrastructure , Cell Line , Cell Transplantation/methods , Chick Embryo , Endothelial Growth Factors/genetics , Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/genetics , Gelatin , Gene Expression , Lymphokines/genetics , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Time Factors , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Int J Biochem Cell Biol ; 33(4): 305-13, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11312101

ABSTRACT

The role of nitric oxide (NO) in the induction of angiogenesis was evaluated in a murine heart endothelioma cell line (H.end.FB) carrying the mT oncogene. Two clonal derivatives of H.end.FB, H80 and H73, exhibiting different NO synthase (NOS) activities were selected and used in the study. The relationship among NOS activity and tumor cell behaviour (growth, and angiogenic capacity) and the molecular control of gene expression were investigated. H.end.FB and H80 on one side and H73 on the other side exhibited the highest and lowest NOS activity, respectively. Cell growth was inversely correlated to the amount of NO produced by the cell lines. Conversely, in the avascular rabbit cornea assay, H.end.FB and H80 cells were strongly angiogenic, while H73 were poorly angiogenic, indicating that the ability of the cells to induce neovascularization was associated with the extent of NO produced. Consistently, systemic administration to rabbits of the NOS inhibitor N(w)-nitro-L-arginine methyl ester (L-NAME) significantly reduced the angiogenicity of H.end.FB cells. RT-PCR evidenced that H.end.FB expressed mRNA for TGF-beta1 and all VEGF isoforms, VEGF165 being predominantly expressed. NOS inhibition reduced the basal expression of VEGF isoforms, while it markedly potentiated TGF-beta1 expression. These results indicate that the endogenous production of NO in tumor cells can serve as an autocrine/paracrine signalling mechanism of progression, by controlling angiogenic factor/modulator expression.


Subject(s)
Neovascularization, Physiologic/physiology , Nitric Oxide/physiology , Oncogene Proteins/physiology , Animals , Cell Line, Transformed , Gene Expression Profiling , Gene Expression Regulation , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
10.
Blood ; 96(13): 4039-45, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11110671

ABSTRACT

Several chemokines have been shown to act as angiogenic molecules or to modulate the activity of growth factors such as fibroblast growth factor 2 (FGF-2) and vascular endothelial growth factor (VEGF). The detection of the CC chemokine receptor (CCR) 8 message in human umbilical vein endothelial cells (HUVECs) by reverse transcription- polymerase chain reaction (RT-PCR) and RNase protection assay (RPA), prompted us to investigate the potential role exerted by the CC chemokine I-309, a known ligand of such receptor, in both in vitro and in vivo angiogenesis assays. We show here that I-309 binds to endothelial cells, stimulates chemotaxis and invasion of these cells, and enhances HUVEC differentiation into capillary-like structures in an in vitro Matrigel assay. Furthermore, I-309 is an inducer of angiogenesis in vivo in both the rabbit cornea and the chick chorioallantoic membrane assay (CAM).


Subject(s)
Chemokines, CC/pharmacology , Endothelium, Vascular/drug effects , Neovascularization, Physiologic/drug effects , Allantois/blood supply , Allantois/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured/drug effects , Chemokine CCL1 , Chemokines, CC/metabolism , Chemotaxis/drug effects , Chick Embryo , Chorion/blood supply , Chorion/drug effects , Collagen , Cornea/blood supply , Cornea/drug effects , Drug Combinations , Endothelium, Vascular/cytology , Gene Expression Regulation/drug effects , Humans , Laminin , Male , Proteoglycans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rabbits , Receptors, CCR8 , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics , Umbilical Veins
11.
J Inorg Biochem ; 81(4): 229-37, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11065187

ABSTRACT

With the aim of improving the compatibility of biomaterials to be used for the construction of cardiovascular prosthesis, we have designed bioactive macromolecules resulting from chemical modifications of hyaluronic acid (Hyal). The stability constants of Cu(II) and Zn(II) complexes with the sulphated derivative of hyaluronic acid (HyalS3.5) were evaluated. Two different complexes have been found for each metal ion, CuL, Cu(OH)2L and ZnL, Zn(OH)2L (L means the disaccharide unit of the ligands) in aqueous solution at 37 degrees C. The dihydroxo Cu(II) complex was present in high percentage at pH=7.4. On the contrary, the Zn(II) ion was present with a relatively low percentage of both complexes. The ability to stimulate endothelial cell adhesion and migration was evaluated for Hyal, HyalS3.5 and their complexes with Cu(II) and Zn(II) ions. The results revealed that Hyal and [Cu(OH)2HyalS3.5](4.5)- induced cell adhesion, while [ZnHyalS3.5](2.5)- and [Zn(OH)2HyalS3.5](4.5)- inhibited the process. The chemotactic activity of increasing concentrations of the above complexes was also evaluated, demonstrating that [Cu(OH)2HyalS3.5](4.5)- complex at 1 microM concentration was the most active in inducing cell migration. These results have been also strengthened by analysing adherent cell migration in agarose. In conclusion, sulphated hyaluronic acid coordinated to Cu(II) seems to be a promising matrix molecule for the construction of cardiovascular prosthesis.


Subject(s)
Biocompatible Materials/pharmacology , Copper/pharmacology , Endothelium, Vascular/drug effects , Hyaluronic Acid/pharmacology , Zinc/pharmacology , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Chemotaxis/drug effects , Copper/chemistry , Drug Stability , Endothelium, Vascular/cytology , Hyaluronic Acid/chemistry , Hydrogen-Ion Concentration , Materials Testing , Mice , Mice, Inbred Strains , Sulfates/chemistry , Sulfates/pharmacology , Zinc/chemistry
12.
Blood ; 96(7): 2568-73, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11001913

ABSTRACT

Because of its immunosuppressive properties, interleukin-10 (IL-10) is thought to play an important role in a number of human disease states, including inflammation, autoimmunity, and transplant rejection. In this study, we demonstrate that introduction of human or viral IL-10 genes into Burkitt's lymphoma cells markedly reduced their ability to grow as subcutaneous (sc) tumors in SCID mice. In vivo assays for angiogenesis revealed an inhibition of the angiogenic capacity of the IL-10-transfected lines. Recombinant human IL-10 abolished and viral IL-10 reduced vascular endothelial growth factor (VEGF)-165-induced neovascularization. Furthermore, IL-10 blocked the VEGF- and fibroblast growth factor (FGF)-2-induced proliferation of microvascular endothelial cells in vitro. The current observations suggest a direct role for IL-10 in the prevention of angiogenesis in human lymphoid malignancies.


Subject(s)
Burkitt Lymphoma/pathology , Interleukin-10/genetics , Interleukin-10/physiology , Neovascularization, Pathologic/prevention & control , Animals , Burkitt Lymphoma/immunology , Burkitt Lymphoma/prevention & control , Cell Division , Cell Line , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Fibroblast Growth Factor 2/pharmacology , Gene Expression , Interleukin-10/pharmacology , Killer Cells, Natural/immunology , Lymphokines/pharmacology , Mice , Mice, SCID , Neoplasm Transplantation , Rabbits , Recombinant Proteins/pharmacology , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
Br J Cancer ; 83(1): 63-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10883669

ABSTRACT

Vascular endothelial growth factor (VEGF) is known to occur as at least six differentially spliced variants, giving rise to mature isoforms containing 121, 145, 165, 183, 189 and 206 amino acids. However, little is yet known concerning the in vivo activities of this differential splicing. Stably transfected MCF-7 breast carcinoma cells were constructed that secreted comparable amounts of the 121, 165 or 189 isoforms. Rabbit corneal angiogenesis assays showed the VEGF121 transfectant to have much greater angiogenic activity than the 165 or 189 expressing MCF-7 cells. While the VEGF121-expressing MCF-7 cells were reproducibly more tumorigenic than the control transfectants, this was not the case with the VEGF165- or VEGF189-expressing cells. More surprising was the observation that VEGF189 located to the nucleus, consistent with the presence of a highly conserved nuclear localization sequence in exon 6a that is expressed in VEGF189 but not 121 or 165. It was concluded that the VEGF121 isoform is both more angiogenic and tumorigenic than are the 165 and 189 isoforms. This is probably due to the ability of the 121 isoform, unlike the 165 and 189 isoforms, to freely diffuse from the cells producing it.


Subject(s)
Alternative Splicing , Endothelial Growth Factors/physiology , Lymphokines/physiology , Neovascularization, Pathologic/etiology , Protein Isoforms/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cells, Cultured/drug effects , Cornea/blood supply , Diffusion , Endothelial Growth Factors/chemistry , Endothelial Growth Factors/genetics , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Exons/genetics , Female , Humans , Lymphokines/chemistry , Lymphokines/genetics , Lymphokines/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Molecular Weight , Neoplasm Transplantation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/pharmacology , Rabbits , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/transplantation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Pharmacol Res ; 42(1): 9-19, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10860629

ABSTRACT

Vascular endothelium - lining the inner side of blood bessels - is one of the largest secretory tissues of the body. Therefore, understanding the cellular and molecular biology of the endothelial cells is essential for the development of new approaches for both the prevention and therapy of cardiovascular diseases. To this aim, in vitro cultures of endothelial cells provide a valuable technical resource. This review focuses on some of the critical phases of the endothelial cells culturing methodology such as: i) isolation and growing of endothelial cells; ii) identification of endothelial cells by morphological, biochemical and cellular markers; iii) studying endothelial cells in function of vascular pharmacology, vasomotor tone, vessel remodelling (angiogenesis/apoptosis), blood haemostasis, inflammatory reactions, and molecular engineering. Practical suggestions for culturing endothelial cells are presented while pros and cons of each method are discussed.


Subject(s)
Endothelium, Vascular/cytology , Animals , Apoptosis , Blood Coagulation , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Humans , Muscle, Smooth, Vascular/cytology , Neoplasm Metastasis , Neovascularization, Physiologic , Stress, Mechanical
17.
Int J Oncol ; 17(1): 75-81, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10853021

ABSTRACT

The vMIPs are chemokine-like proteins expressed by the Kaposi's sarcoma-associated herpesvirus (KSHV/HHV8) during the lytic phase of viral infection. vMIP-I activates CCR8, a chemokine receptor expressed by Th2 lymphocytes and cultured monocytes. vMIP-II is an agonist for CCR3, a receptor expressed by eosinophils, and an antagonist for several other chemokine receptors. Both are highly angiogenic in the chick chorio-allantoic membrane. We designed and tested three 26-mer peptides, derived from vMIP-I (pK-I), from vMIP-II (pK-II) and from the control MIP-1alpha (pM), spanning key residues of chemokines. pK-I, pK-II and pM all were able to activate a strong chemotactic response in monocytes, higher than parental vMIP-I and II. This corresponded to induction of calcium fluxes in these cells, typical of chemokines. Interestingly, pK-II and pM were also active on PMN neutrophils. In vivo studies (matrigel sponge and rabbit cornea models) showed that pK-I retains the strong angiogenic potential exerted by vMIP-I, while pK-II and pM induced an inflammatory response, probably mediated by PMN recruitment. Our observations indicate that chemokine-derived peptides can show biological activity at pharmacological concentrations. pK-I, in particular, displays the angiogenic activity of full-length vMIP-I, while all peptides appear to have acquired additional properties, stimulating new cellular targets.


Subject(s)
Calcium Channels/physiology , Chemotaxis, Leukocyte/drug effects , Herpesvirus 8, Human/physiology , Macrophage Inflammatory Proteins/pharmacology , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Receptors, Chemokine/physiology , Viral Proteins , Allantois/blood supply , Amino Acid Sequence , Animals , Calcium/blood , Calcium Channels/drug effects , Chemokines, CC/physiology , Chick Embryo , Chorion/blood supply , Cornea/blood supply , Granulocytes/drug effects , Granulocytes/physiology , Herpesvirus 8, Human/genetics , In Vitro Techniques , Macrophage Inflammatory Proteins/chemistry , Macrophage Inflammatory Proteins/genetics , Molecular Sequence Data , Neovascularization, Pathologic , Neutrophils/drug effects , Neutrophils/physiology , Rabbits , Receptors, CCR8
18.
J Neurooncol ; 50(1-2): 139-48, 2000.
Article in English | MEDLINE | ID: mdl-11245273

ABSTRACT

The steps required for new vessel growth are biologically complex and require coordinate regulation of contributing components, including modifications of cell--cell interactions, proliferation and migration of endothelial cells and matrix degradation. The observation that in vivo angiogenesis is accompanied by vasodilation, that many angiogenesis effectors possess vasodilating properties and that tumor vasculature is in a persistent state of vasodilation, support the existence of a molecular/biochemical link between vasodilation and angiogenesis. Several pieces of evidence converge in the indication of a role for nitric oxide (NO), the factor responsible for vasodilation, in physiological and pathological angiogenesis. Data originated in different labs indicate that NO can act both as an 'actor' of angiogenesis and as a 'director of angiogenesis', both functions being equally expressed during physiological and pathological processes. NO significantly contributes to the prosurvival/proangiogenic program of capillary endothelium by triggering and transducing cell growth and differentiation via endothelial-constitutive NO synthase (ec-NOS) activation, cyclic GMP (cGMP) elevation, mitogen activated kinase (MAPK) activation and fibroblast growth factor-2 (FGF-2) expression. Re-establishment of a balanced NO production in the central nervous system results in a reduction of cell damage during inflammatory and vascular diseases. Elevation of NOS activity in correlation with angiogenesis and tumor progression has been extensively reported in experimental and human tumors. In the brain, tumor expansion and edema formation are sensitive to NOS inhibition. On this basis, the nitric oxide pathway appears to be a promising target for consideration in pro- and anti-angiogenic therapeutic strategies. The use of NOS inhibitors seems appropriate to reduce edema, block angiogenesis and facilitate antitumor drug delivery.


Subject(s)
Neovascularization, Physiologic/physiology , Nitric Oxide/physiology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , MAP Kinase Signaling System , Mice , Models, Biological , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Rabbits , Rats , Signal Transduction , Stromal Cells/pathology , Vasodilation/physiology
19.
Proc Natl Acad Sci U S A ; 96(17): 9671-6, 1999 Aug 17.
Article in English | MEDLINE | ID: mdl-10449752

ABSTRACT

c-fos-induced growth factor/vascular endothelial growth factor D (Figf/Vegf-D) is a secreted factor of the VEGF family that binds to the vessel and lymphatic receptors VEGFR-2 and VEGFR-3. Here we report that Figf/Vegf-D is a potent angiogenic factor in rabbit cornea in vivo in a dose-dependent manner. In vitro Figf/Vegf-D induces tyrosine phosphorylation of VEGFR-2 and VEGFR-3 in primary human umbilical cord vein endothelial cells (HUVECs) and in an immortal cell line derived from Kaposi's sarcoma lesion (KS-IMM). The treatment of HUVECs with Figf/Vegf-D induces dose-dependent cell growth. Figf/VEGF-D also induces HUVEC elongation and branching to form an extensive network of capillary-like cords in three-dimensional matrix. In KS-IMM cells Figf/Vegf-D treatment results in dose-dependent mitogenic and motogenic activities. Taken together with the previous observations that Figf/Vegf-D expression is under the control of the nuclear oncogene c-fos, our data uncover a link between a nuclear oncogene and angiogenesis, suggesting that Figf/Vegf-D may play a critical role in tumor cell growth and invasion.


Subject(s)
Endothelial Growth Factors/pharmacology , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins c-fos/pharmacology , Animals , CHO Cells , Cell Division/drug effects , Cell Line , Chemotaxis/drug effects , Cornea/blood supply , Cricetinae , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Mice , Phosphorylation , Rabbits , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Tyrosine/metabolism , Vascular Endothelial Growth Factor D , Vascular Endothelial Growth Factor Receptor-3
20.
Cancer Res ; 59(10): 2417-24, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10344752

ABSTRACT

Angiogenesis has been identified as an important target for antineoplastic therapy. The use of purine analogue antimetabolites in combination chemotherapy of solid tumors has been proposed. To assess the possibility that selected purine analogues may affect tumor neovascularization, 6-methylmercaptopurine riboside (6-MMPR), 6-methylmercaptopurine, 2-aminopurine, and adenosine were evaluated for the capacity to inhibit angiogenesis in vitro and in vivo. 6-MMPR inhibited fibroblast growth factor-2 (FGF2)-induced proliferation and delayed the repair of mechanically wounded monolayer in endothelial GM 7373 cell cultures. 6-MMPR also inhibited the formation of solid sprouts within fibrin gel by FGF2-treated murine brain microvascular endothelial cells and the formation of capillary-like structures on Matrigel by murine aortic endothelial cells transfected with FGF2 cDNA. 6-MMPR affected FGF2-induced intracellular signaling in murine aortic endothelial cells by inhibiting the phosphorylation of extracellular signal-regulated kinase-2. The other molecules were ineffective in all of the assays. In vivo, 6-MMPR inhibited vascularization in the chick embryo chorioallantoic membrane and prevented blood vessel formation induced by human endometrial adenocarcinoma specimens grafted onto the chorioallantoic membrane. Also, topical administration of 6-MMPR caused the regression of newly formed blood vessels in the rabbit cornea. Thus, 6-MMPR specifically inhibits both the early and the late phases of the angiogenesis process in vitro and exerts a potent anti-angiogenic activity in vivo. These results provide a new rationale for the use of selected purine analogues in combination therapy of solid cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Methylthioinosine/pharmacology , Neovascularization, Physiologic/drug effects , 2-Aminopurine/pharmacology , Adenocarcinoma/blood supply , Adenosine/pharmacology , Allantois/blood supply , Allantois/drug effects , Animals , Antineoplastic Agents/pharmacology , Aorta/cytology , Brain/blood supply , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cattle , Cells, Cultured , Chick Embryo , Chorion/blood supply , Chorion/drug effects , Cornea/blood supply , Cornea/drug effects , Depression, Chemical , Endometrial Neoplasms/blood supply , Endothelium, Vascular/drug effects , Female , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/pharmacology , Humans , Mercaptopurine/analogs & derivatives , Mercaptopurine/pharmacology , Mice , Mitogen-Activated Protein Kinase 1 , Neoplasm Transplantation , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rabbits , Recombinant Fusion Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...