Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Prostate ; 49(4): 293-305, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11746276

ABSTRACT

BACKGROUND: Prostate cells secrete many molecules capable of regulating angiogenesis; however, which of these actually function as essential regulators of neovascularization is not yet clear. METHODS: Functional angiogenic mediators secreted by normal and diseased prostate cells were identified using an in vitro angiogenesis assay. These factors were quantified by immunoblot or ELISA and localized in tissue by immunohistochemistry. RESULTS: Normal prostate epithelial cell secretions were anti-angiogenic due to inhibitory thrombospondin-1 (TSP-1) whereas this inhibitor was decreased in the pro-angiogenic secretions derived from benign prostatic hyperplasia (BPH) and cancer cells. This pro-angiogenic activity depended primarily on fibroblast growth factor-2 (FGF-2) and/or vascular endothelial growth factor (VEGF) whose secretion was increased. Immunolocalization studies confirmed that the changes detected in vitro also occurred in vivo. CONCLUSIONS: During disease progression in the prostate, production of TSP-1, the major inhibitor, is down-regulated while that of stimulatory FGF-2 and/or VEGF rise, leading to the induction of the new vessels necessary to support tumor growth.


Subject(s)
Endothelial Growth Factors/physiology , Fibroblast Growth Factor 2/physiology , Lymphokines/physiology , Neovascularization, Pathologic/physiopathology , Prostate/blood supply , Prostatic Neoplasms/blood supply , Thrombospondin 1/physiology , Adolescent , Adult , Blotting, Western , Endothelial Growth Factors/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Immunohistochemistry , Lymphokines/metabolism , Male , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/physiology , Prostate/metabolism , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/metabolism , Thrombospondin 1/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
2.
J Urol ; 165(6 Pt 1): 2075-81, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11371931

ABSTRACT

PURPOSE: Our previous studies defined thrombospondin-1 (TSP-1) and vascular endothelial growth factor (VEGF) as the primary mediators of angiogenesis in the bladder and the loss of inhibitory TSP-1 as a key event in the transition to an angiogenic phenotype during bladder cancer development. We evaluated the role of p53, which is commonly inactivated in bladder cancer, and hypoxia in the regulation of angiogenesis in the bladder. MATERIALS AND METHODS: The p53 status was modulated in normal urothelial and bladder cancer cells, and conditioned media was collected under normal oxygen or hypoxic (0.5% O2) conditions. Angiogenic activity was evaluated with the endothelial cell migration assay, and the levels of secreted TSP-1 and VEGF were determined by Western blot analysis and enzyme-linked immunosorbent assay, respectively. RESULTS: Retroviral mediated expression of the E6 oncoprotein reduced wild-type p53 levels in normal urothelial cells by greater than 90% but did not significantly alter TSP-1 or VEGF levels, while total inductive and inhibitory activities remained unchanged. Adenoviral mediated expression of wild-type p53 was confirmed in 4 bladder cancer cell lines by Western blot analysis for p53 and its downstream effector protein p21 (2.5 to 5.0-fold increase). TSP-1 levels remained unchanged but the levels of secreted VEGF in the high grade UMUC-3 and 253J cell lines were significantly decreased 5 to 50-fold and a corresponding decrease in net angiogenic activity was observed. However, (increased expression) of p53 had no effect on the angiogenic activity of the low grade RT4 or high grade HT1376 bladder cancer cells. Hypoxia converted normal urothelial cell derived conditioned media from anti-angiogenic to angiogenic and increased the angiogenic activity of bladder cancer cell derived conditioned media. This change was due to 2.5 to 6-fold hypoxic up-regulation of VEGF because the expression of inhibitory TSP-1 was not significantly altered. CONCLUSIONS: Our results suggest that p53 does not regulate angiogenesis in the bladder in the setting of an otherwise normal genome and gene therapy with wild-type p53, which is currently being studied for this cancer, may have only limited effects on angiogenesis. In contrast, hypoxia regulates angiogenesis in this system, primarily through its effects on VEGF.


Subject(s)
Carcinoma, Transitional Cell/physiopathology , Neovascularization, Pathologic/physiopathology , Tumor Suppressor Protein p53/physiology , Urinary Bladder Neoplasms/physiopathology , Blotting, Western , Carcinoma, Transitional Cell/therapy , Cell Hypoxia/physiology , Down-Regulation , Endothelial Growth Factors/metabolism , Endothelial Growth Factors/physiology , Gene Expression Regulation , Genes, p53/physiology , Genetic Therapy , Humans , Lymphokines/metabolism , Lymphokines/physiology , Thrombospondin 1/metabolism , Tumor Cells, Cultured , Up-Regulation , Urinary Bladder Neoplasms/therapy , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
3.
Proc Natl Acad Sci U S A ; 97(17): 9624-9, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10944227

ABSTRACT

Smad4/DPC4 (deleted in pancreatic carcinoma, locus 4) is a tumor suppressor gene lost at high frequency in cancers of the pancreas and other gastrointestinal organs. Smad4 encodes a key intracellular messenger in the transforming growth factor beta (TGF-beta) signaling cascade. TGF-beta is a potent inhibitor of the growth of epithelial cells; thus, it has been assumed that loss of Smad4 during tumor progression relieves this inhibition. Herein, we show that restoration of Smad4 to human pancreatic carcinoma cells suppressed tumor formation in vivo, yet it did not restore sensitivity to TGF-beta. Rather, Smad4 restoration influenced angiogenesis, decreasing expression of vascular endothelial growth factor and increasing expression of thrombospondin-1. In contrast to the parental cell line and to control transfectants that produced rapidly growing tumors in vivo, Smad4 revertants induced small nonprogressive tumors with reduced vascular density. These data define the control of an angiogenic switch as an alternative, previously unknown mechanism of tumor suppression for Smad4 and identify the angiogenic mediators vascular endothelial growth factor and thrombospondin-1 as key target genes.


Subject(s)
Antineoplastic Agents/metabolism , DNA-Binding Proteins/metabolism , Genes, Tumor Suppressor , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Trans-Activators/metabolism , Animals , Cell Division/drug effects , Cell Movement , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Fibroblast Growth Factor 2/pharmacology , Genes, Tumor Suppressor/genetics , Humans , Lymphokines/genetics , Lymphokines/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad4 Protein , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Trans-Activators/genetics , Transfection , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Science ; 285(5425): 245-8, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10398599

ABSTRACT

In the absence of disease, the vasculature of the mammalian eye is quiescent, in part because of the action of angiogenic inhibitors that prevent vessels from invading the cornea and vitreous. Here, an inhibitor responsible for the avascularity of these ocular compartments is identified as pigment epithelium-derived factor (PEDF), a protein previously shown to have neurotrophic activity. The amount of inhibitory PEDF produced by retinal cells was positively correlated with oxygen concentrations, suggesting that its loss plays a permissive role in ischemia-driven retinal neovascularization. These results suggest that PEDF may be of therapeutic use, especially in retinopathies where pathological neovascularization compromises vision and leads to blindness.


Subject(s)
Eye Proteins , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Nerve Growth Factors , Proteins/pharmacology , Proteins/physiology , Retina/metabolism , Retinal Neovascularization/drug therapy , Serpins/pharmacology , Serpins/physiology , Animals , Animals, Newborn , Antibodies/immunology , Cattle , Cells, Cultured , Chemotaxis/drug effects , Culture Media, Conditioned , Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Eye/blood supply , Humans , Lymphokines/metabolism , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Oxygen/physiology , Proteins/genetics , Proteins/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Retina/pathology , Retinal Vessels/growth & development , Serpins/genetics , Serpins/immunology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
J Cell Sci ; 112 ( Pt 12): 2049-57, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10341222

ABSTRACT

Keratinocyte growth factor (KGF), also called fibroblast growth factor-7, is widely known as a paracrine growth and differentiation factor that is produced by mesenchymal cells and has been thought to act specifically on epithelial cells. Here it is shown to affect a new cell type, the microvascular endothelial cell. At subnanomolar concentrations KGF induced in vivo neovascularization in the rat cornea. In vitro it was not effective against endothelial cells cultured from large vessels, but did act directly on those cultured from small vessels, inducing chemotaxis with an ED50 of 0.02-0.05 ng/ml, stimulating proliferation and activating mitogen activated protein kinase (MAPK). KGF also helped to maintain the barrier function of monolayers of capillary but not aortic endothelial cells, protecting against hydrogen peroxide and vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) induced increases in permeability with an ED50 of 0.2-0.5 ng/ml. These newfound abilities of KGF to induce angiogenesis and to stabilize endothelial barriers suggest that it functions in microvascular tissue as it does in epithelial tissues to protect them against mild insults and to speed their repair after major damage.


Subject(s)
Capillary Permeability/drug effects , Cornea/drug effects , Endothelium, Vascular/drug effects , Fibroblast Growth Factors , Growth Substances/pharmacology , Keratinocytes , Neovascularization, Physiologic/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Cornea/cytology , Endothelium, Vascular/cytology , Female , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Humans , Rats , Rats, Inbred F344 , Recombinant Proteins/pharmacology
6.
Mol Pharmacol ; 55(2): 332-8, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9927626

ABSTRACT

Mal II, a 19-residue peptide derived from the second type 1 properdin-like repeat of the antiangiogenic protein thrombospondin-1 (TSP-1), was inactive in angiogenesis assays. Yet the substitution of any one of three L-amino acids by their D-enantiomers conferred on this peptide a potent antiangiogenic activity approaching that of the intact 450-kDa TSP-1. Substituted peptides inhibited the migration of capillary endothelial cells with an ED50 of 8.5 nM for the D-Ile-15 substitution, 10 nM for the D-Ser-4 substitution, and 0.75 nM for the D-Ser-5 substitution. A peptide with D-Ile at position 15 could be shortened to its last seven amino acids with little loss in activity. Like whole TSP-1, the Mal II D-Ile derivative inhibited a broad range of angiogenic inducers, was selective for endothelial cells, and required CD36 receptor binding for activity. A variety of end modifications further improved peptide potency. An ethylamide-capped heptapeptide was also active systemically in that when injected i.p. it rendered mice unable to mount a corneal angiogenic response, suggesting the potential usefulness of such peptides as antiangiogenic therapeutics.


Subject(s)
Amino Acids/chemistry , Neovascularization, Pathologic/prevention & control , Oligopeptides/pharmacology , Thrombospondin 1/pharmacology , Acetylation , Amino Acid Sequence , Animals , Cell Movement/drug effects , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 2/pharmacology , Interleukin-8/pharmacology , Isoleucine/chemistry , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oligopeptides/chemistry , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Sprague-Dawley , Repetitive Sequences, Nucleic Acid , Sensitivity and Specificity , Serine/chemistry , Stereoisomerism , Thrombospondin 1/chemistry , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
Cancer Res ; 58(23): 5551-8, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9850093

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is an aggressive malignancy in which multiple independent lesions develop over time throughout the mucosa of the upper aerodigestive tract. Therefore, the comprehensive treatment of this neoplasm must include a chemopreventive arm to hold premalignant lesions in check, a role well-suited to antiangiogenic agents. Retinoic acid (RA) and interferon alpha (IFN-alpha), drugs with known biological activity against HNSCC when used individually, are also inhibitors of angiogenesis. Here we show that they are remarkably synergistic antiangiogenic agents able to inhibit both the growth and the neovascularization of HNSCC injected into the floor of the mouth of nude mice. The mechanism of action of these drugs as antiangiogenic agents was 2-fold. They decreased the angiogenic activity of the tumor cells, and they caused the endothelial cells to become refractory to inducers of angiogenesis. When tumor cells were treated in vitro with IFN-alpha A/D, there was a dramatic drop in their secretion of interleukin-8, the major angiogenic factor produced by these tumors. When combined with RA, which causes tumor cells to secrete an inhibitor of angiogenesis, there was a synergistic inhibition of both tumor cell growth and secreted angiogenic activity. The combination of RA and IFN-alpha also acted synergistically on endothelial cells by reducing their responsiveness to both interleukin-8 and tumor conditioned media. Doses of each drug could be reduced by two logs without loss of activity. When animals bearing human HNSCC tumor cells were treated systemically with a combination of RA and IFN-alpha A/D at doses that were ineffective when used alone, dramatic decreases in both tumor growth and tumor angiogenesis were seen. These data suggest that the use of antiangiogenic mixtures may be a particularly effective way to design future chemoprevention protocols against HNSCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/therapy , Neovascularization, Pathologic/therapy , Animals , Carcinoma, Squamous Cell/prevention & control , Cattle , Cell Division/drug effects , Cells, Cultured , Chemoprevention , Drug Synergism , Endothelium, Vascular/drug effects , Female , Head and Neck Neoplasms/prevention & control , Humans , Interferon Type I/administration & dosage , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/prevention & control , Rats , Rats, Inbred F344 , Recombinant Proteins , Tretinoin/administration & dosage , Tumor Cells, Cultured
8.
J Exp Med ; 188(6): 1039-46, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9743522

ABSTRACT

Interleukin (IL)-4, a crucial modulator of the immune system and an active antitumor agent, is also a potent inhibitor of angiogenesis. When incorporated at concentrations of 10 ng/ml or more into pellets implanted into the rat cornea or when delivered systemically to the mouse by intraperitoneal injection, IL-4 blocked the induction of corneal neovascularization by basic fibroblast growth factor. IL-4 as well as IL-13 inhibited the migration of cultured bovine or human microvascular cells, showing unusual dose-response curves that were sharply stimulatory at a concentration of 0.01 ng/ml but inhibitory over a wide range of higher concentrations. Recombinant cytokine from mouse and from human worked equally well in vitro on bovine and human endothelial cells and in vivo in the rat, showing no species specificity. IL-4 was secreted at inhibitory levels by activated murine T helper (TH0) cells and by a line of carcinoma cells whose tumorigenicity is known to be inhibited by IL-4. Its ability to cause media conditioned by these cells to be antiangiogenic suggested that the antiangiogenic activity of IL-4 may play a role in normal physiology and contribute significantly to its demonstrated antitumor activity.


Subject(s)
Immunosuppressive Agents/pharmacology , Interleukin-4/physiology , Neovascularization, Physiologic/immunology , Adenocarcinoma , Animals , Cell Movement/drug effects , Cell Movement/immunology , Cells, Cultured , Cornea/blood supply , Cornea/drug effects , Cornea/immunology , Culture Media, Conditioned/chemistry , Culture Media, Serum-Free/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Humans , Injections, Intraperitoneal , Interleukin-4/administration & dosage , Mammary Neoplasms, Experimental , Mice , Mice, Inbred BALB C , Neovascularization, Physiologic/drug effects , Rats , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 95(11): 6343-8, 1998 May 26.
Article in English | MEDLINE | ID: mdl-9600967

ABSTRACT

Concomitant tumor resistance refers to the ability of some large primary tumors to hold smaller tumors in check, preventing their progressive growth. Here, we demonstrate this phenomenon with a human tumor growing in a nude mouse and show that it is caused by secretion by the tumor of the inhibitor of angiogenesis, thrombospondin-1. When growing subcutaneously, the human fibrosarcoma line HT1080 induced concomitant tumor resistance, preventing the growth of experimental B16/F10 melanoma metastases in the lung. Resistance was due to the production by the tumor cells themselves of high levels of thrombospondin-1, which was present at inhibitory levels in the plasma of tumor-bearing animals who become unable to mount an angiogenic response in their corneas. Animals carrying tumors formed by antisense-derived subclones of HT1080 that secreted low or no thrombospondin had weak or no ability to control the growth of lung metastases. Although purified human platelet thrombospondin-1 had no effect on the growth of melanoma cells in vitro, when injected into mice it was able to halt the growth of their experimental metastases, providing clear evidence of the efficacy of thrombospondin-1 as an anti-tumor agent.


Subject(s)
Fibrosarcoma/blood supply , Fibrosarcoma/pathology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/physiopathology , Thrombospondin 1/physiology , Animals , Fibrosarcoma/physiopathology , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasms, Experimental/physiopathology , Neovascularization, Pathologic/prevention & control , Thrombospondin 1/pharmacology
10.
Clin Immunol Immunopathol ; 86(2): 199-208, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9473383

ABSTRACT

Leukocyte extravasation into the synovium is important in rheumatoid arthritis (RA). Thrombospondin (TSP)-1 mediates cell adhesion and migration and inhibits angiogenesis, and it has been implicated in RA. However, little information is available on the role of TSP-1 in arthritis-associated inflammation and neovascularization. Therefore, we analyzed the effects of TSP-1 in adjuvant-induced arthritis (AIA), a rat model for RA. Hydron pellets containing TSP-1 were implanted in one ankle of AIA rats post-adjuvant injection, while the contralateral ankle received sham implants. Body weight loss and joint swelling were determined in comparison to nonimplanted AIA controls. In addition, synovial vessel counts were obtained in TSP-1-versus sham-implanted ankles of the same rat. The implantation of TSP-1 pellets into one ankle resulted in an enhancement of swelling in both ankles. Furthermore, TSP-1 exhibited a biphasic modulatory effect on synovial vessel counts (P < 0.05). In conclusion, TSP-1 implanted into one ankle of AIA rats may augment the severity of the disease. One possible explanation, among others, for the modulating effect of TSP-1 on inflammation may be its effect on arthritis-related angiogenesis.


Subject(s)
Arthritis, Experimental/etiology , Arthritis, Rheumatoid/etiology , Neovascularization, Pathologic/etiology , Thrombospondin 1/pharmacology , Animals , Arthritis, Experimental/complications , Arthritis, Rheumatoid/complications , Body Weight , Disease Models, Animal , Dose-Response Relationship, Drug , Joints/pathology , Rats , Rats, Inbred Lew , Tarsus, Animal
11.
Cancer Res ; 58(6): 1298-304, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9515819

ABSTRACT

Bladder tumors are characterized by markedly increased angiogenesis when compared to the normal urothelium (NU) from which they are derived. Here, we use both cultured cells and immunohistochemistry to demonstrate a primary regulatory role for thrombospondin-1 (TSP-1), a potent inhibitor of angiogenesis, in the development of bladder tumor angiogenesis. Secretions from bladder cancer (CA) cells stimulated endothelial cell migration and corneal neovascularization, whereas those from NU cells were inhibitory. The antiangiogenic activity of NU cells was primarily due to secreted TSP-1 because neutralizing antibodies completely relieved the inhibition. Neutralizing antibodies to several putative angiogenesis inducers identified vascular endothelial growth factor (VEGF) and, to a lesser extent, basic fibroblast growth factor as the primary inducers secreted by bladder cancer cells. The secretion of TSP-1 by low- and high-grade cancer cells was reduced >94% when compared to NU cells, and this loss of inhibitory TSP-1 accounted for the development of an angiogenic phenotype because both NU cells and cancer cells secreted similar levels of total stimulatory activity and VEGF. Immunohistochemistry showed that TSP-1 was significantly reduced in all grades of bladder cancer when compared to NU, whereas VEGF staining remained relatively constant. Taken together, these data suggest that down-regulation of TSP-1 secretion is a key event in the switch from an antiangiogenic to an angiogenic phenotype, which occurs early in the development of bladder cancer.


Subject(s)
Endothelial Growth Factors/physiology , Fibroblast Growth Factor 2/physiology , Lymphokines/physiology , Neovascularization, Pathologic , Thrombospondin 1/physiology , Urinary Bladder Neoplasms/blood supply , Cells, Cultured , Down-Regulation , Humans , Tumor Cells, Cultured , Urothelium/cytology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
J Cell Biol ; 138(3): 707-17, 1997 Aug 11.
Article in English | MEDLINE | ID: mdl-9245797

ABSTRACT

Thrombospondin-1 (TSP-1) is a naturally occurring inhibitor of angiogenesis that is able to make normal endothelial cells unresponsive to a wide variety of inducers. Here we use both native TSP-1 and small antiangiogenic peptides derived from it to show that this inhibition is mediated by CD36, a transmembrane glycoprotein found on microvascular endothelial cells. Both IgG antibodies against CD36 and glutathione-S-transferase-CD36 fusion proteins that contain the TSP-1 binding site blocked the ability of intact TSP-1 and its active peptides to inhibit the migration of cultured microvascular endothelial cells. In addition, antiangiogenic TSP-1 peptides inhibited the binding of native TSP-1 to solid phase CD36 and its fusion proteins, as well as to CD36-expressing cells. Additional molecules known to bind CD36, including the IgM anti-CD36 antibody SM, oxidized (but not unoxidized) low density lipoprotein, and human collagen 1, mimicked TSP-1 by inhibiting the migration of human microvascular endothelial cells. Transfection of CD36-deficient human umbilical vein endothelial cells with a CD36 expression plasmid caused them to become sensitive to TSP-1 inhibition of their migration and tube formation. This work demonstrates that endothelial CD36, previously thought to be involved only in adhesion and scavenging activities, may be essential for the inhibition of angiogenesis by thrombospondin-1.


Subject(s)
CD36 Antigens/physiology , Endothelium, Vascular/drug effects , Membrane Glycoproteins/pharmacology , Neovascularization, Physiologic/drug effects , Amino Acid Sequence , Animals , CD36 Antigens/genetics , CD36 Antigens/immunology , CD36 Antigens/metabolism , Cattle , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/physiology , Humans , Ligands , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Recombinant Fusion Proteins/pharmacology , Thrombospondins , Transfection
13.
Carcinogenesis ; 18(2): 329-38, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9054625

ABSTRACT

This study was undertaken to investigate the mechanisms by which Syrian hamster buccal pouch keratinocytes treated in vivo with 7,12-dimethylbenz[a]anthracene (DMBA), switch from an angio-inhibitory to an angiogenic phenotype. Cells were cultured from pouches at various times after exposure to carcinogen and their angiogenic activity assessed. The angio-inhibitory activity present in conditioned media from normal cells was lost as early as 3 weeks after carcinogen treatment, resulting in weak expression of angiogenic activity. By 5 weeks, cells had become strongly angiogenic due to the secretion of high levels of TGFbeta-1, a potent angiogenic factor. Because the switch to high levels of secreted TGFbeta-1 occurred at the same time as the activation of the H-ras oncogene, non-angiogenic cell lines lacking an activated H-ras oncogene were stably transfected with mutant H-ras and their transformed and angiogenic phenotypes were evaluated. Although ras transfection drove two of the three cultured cell lines to anchorage independence and modestly increased their ability to clone in low serum, it had no effect on the angiogenic phenotype or on the level of secreted active TGFbeta-1. These results demonstrate that the angiogenic phenotype in the hamster buccal pouch model of oral carcinogenesis develops in a step-wise fashion with an early decrease in the production of an inhibitor of angiogenesis and a subsequent marked increase in the secretion of the inducer TGFbeta-1. Although the activation of the H-ras oncogene contributed to anchorage independence, it did not affect the expression of the angiogenic phenotype in this model system.


Subject(s)
Gene Expression Regulation , Genes, ras/genetics , Keratinocytes/drug effects , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/genetics , Transforming Growth Factor beta/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogens , Cells, Cultured , Cheek/blood supply , Cricetinae , Genes, ras/physiology , Keratinocytes/metabolism , Male , Mesocricetus , Phenotype , Transfection
14.
Cancer Res ; 56(24): 5684-91, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8971176

ABSTRACT

Glioblastoma multiforme is distinguished from its less malignant astrocytoma precursors by intense angiogenesis and frequent loss of tumor suppressor genes on chromosome 10. Here we link these traits by showing that when a wild-type chromosome 10 was returned to any of three human glioblastoma cell lines U251, U87, or LG11, they lost their ability to form tumors in nude mice and switched to an antiangiogenic phenotype, as measured by the inhibition of capillary endothelial cell migration and of corneal neovascularization. This change in angiogenesis was directly due to the increased secretion of a potent inhibitor of angiogenesis, thrombospondin-1, because: (a) neutralizing thrombospondin completely relieved the inhibition; (b) the inhibitory activity of thrombospondin was not dependent on transforming growth factor beta; and (c) chromosome 10 introduction did not alter secreted inducing activity. The inducing activity was dependent on vascular endothelial cell growth factor and had an ED50 of 10 microg/ml in media conditioned by parental cells and 9-13 microg/ml in media conditioned by chromosome 10 revertants. Normal human astrocytes were also antiangiogenic due to secreted thrombospondin. The effect of chromosome 10 on thrombospondin production in vitro was reflected in patient material. Normal brain and lower grade astrocytomas known to retain chromosome 10 stained strongly for thrombospondin, but 12 of 13 glioblastomas, the majority of which lose chromosome 10, did not. These data indicate that the loss of tumor suppressors on chromosome 10 contributes to the aggressive malignancy of glioblastomas in part by releasing constraints on angiogenesis that are maintained by thrombospondin in lower grade tumors.


Subject(s)
Brain Neoplasms/blood supply , Chromosomes, Human, Pair 10/genetics , Glioblastoma/blood supply , Membrane Glycoproteins/genetics , Neovascularization, Pathologic/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Endothelial Growth Factors/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Lymphokines/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Phenotype , Rats , Thrombospondins , Transfection , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Tumor Stem Cell Assay , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
J Clin Invest ; 98(3): 671-9, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8698858

ABSTRACT

Captopril, an inhibitor of angiotensin converting enzyme, is widely used clinically to manage hypertension and congestive heart failure. Here captopril is shown to be an inhibitor of angiogenesis able to block neovascularization induced in the rat cornea. Captopril acted directly and specifically on capillary endothelial cells, inhibiting their chemotaxis with a biphasic dose-response curve showing an initial decrease at clinically achievable doses under 10 microM and a further slow decline in the millimolar range. Captopril inhibition of endothelial cell migration was not mediated by angiotensin converting enzyme inhibition, but was suppressed by zinc. Direct inhibition by captopril of zinc-dependent endothelial cell-derived 72-and 92-kD metalloproteinases known to be essential for angiogenesis was also seen. When used systemically on rats captopril inhibited corneal neovascularization and showed the antitumor activity expected of an inhibitor of angiogenesis, decreasing the number of mitoses present in carcinogen-induced foci of preneoplastic liver cells and slowing the growth rate of an experimental fibrosarcoma whose cells were resistant to captopril in vitro. These data define this widely used drug as a new inhibitor of neovascularization and raise the possibility that patients on long term captopril therapy may derive unexpected benefits from its antiangiogenic activities.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Captopril/pharmacology , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/prevention & control , Animals , Cattle , Cell Movement/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Metalloendopeptidases/antagonists & inhibitors , Rats , Rats, Inbred F344
16.
Am J Pathol ; 149(1): 247-58, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8686749

ABSTRACT

Retinoids have shown great promise as chemopreventive against the development of squamous cell carcinomas of the upper aerodigestive tract. However, the exact mechanism by which they block new tumors from arising is unknown. Here, we report that 13-cis- and all-trans-retinoic acid, used at clinically achievable doses of 10(-6) mol/L or less, can directly and specifically affect cell lines cultured from oral squamous cell carcinomas, inducing them to switch from an angiogenic to an anti-angiogenic phenotype. Although retinoic-acid-treated and untreated tumor cells make the same amount of interleukin-8, the major inducer of neovascularization produced by such tumor lines, they vary in production of inhibitory activity. Only the retinoic-acid-treated cells produce a potent angio-inhibitory activity that is able to block in vitro migration of endothelial cells toward tumor cell conditioned media and to halt neovascularization induced by such media in the rat cornea. Anti-angiogenic activity is induced in the tumor cells by low doses of retinoids in the absence of toxicity with a kinetics that suggest that it could be contributing to the effectiveness of the retinoids as chemopreventive agents.


Subject(s)
Carcinoma, Squamous Cell/physiopathology , Neovascularization, Pathologic/physiopathology , Tongue Neoplasms/physiopathology , Tretinoin/pharmacology , Animals , Breast Neoplasms/physiopathology , Colonic Neoplasms/physiopathology , Cornea/blood supply , Cornea/drug effects , Endothelium, Vascular/drug effects , Female , Fibrosarcoma/physiopathology , Humans , Interleukin-8/analysis , Keratinocytes/physiology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Neutralization Tests , Phenotype , Rats , Rats, Inbred F344 , Transforming Growth Factor beta/analysis , Tretinoin/metabolism , Tumor Cells, Cultured/drug effects
17.
Lab Invest ; 74(2): 476-83, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8780165

ABSTRACT

Retinoic acid (RA) is a multifunctional drug that is particularly effective at preventing the development of multiple primary oral squamous cell carcinomas. A portion of this activity is due to the inhibition of tumor angiogenesis. It has been thought that RA influences tumor angiogenesis only via its interactions with the tumor cells themselves. Here, we test the hypothesis that the drug can also block neovascularization by directly inhibiting the angiogenic activity of normal endothelial cells. Clinically achievable doses of RA rapidly caused large- and small-vessel endothelial cells to become refractory to stimulation of migration either by tumor-conditioned media or purified angiogenic factors (a-fibroblast growth factor (aFGF), bFGF, vascular endothelial GF, platelet-derived GF, TGF beta-1, and IL-8). However, RA had little effect on their proliferation. Inhibition of migration was complete within 3 hours and was reversed 36 hours after drug removal. The migration of human oral keratinocytes was not sensitive to RA, whereas the migration of fibroblasts and vascular smooth muscle cells was inhibited. To determine if systemic RA affected neovascularization, rats were given 1 mg/kg/day of all-trans RA and their angiogenic potential was tested by implanting pellets of tumor-conditioned media into their avascular corneas. This treatment rendered the rats unable to mount a neovascular response in their corneas. These data demonstrate that RA directly affects endothelial cells, rapidly and reversibly inhibiting their ability to migrate toward a variety of stimuli in vitro and halting the formation of new vessels in vivo. These direct effects on vascular cells seem likely to contribute to the success of RA as a chemopreventive agent for oral squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Endothelium, Vascular/drug effects , Neovascularization, Pathologic , Tretinoin/pharmacology , Animals , Cattle , Cell Division/drug effects , Cell Line , Cell Movement/drug effects , Cornea/blood supply , Cornea/drug effects , Culture Media, Conditioned/pharmacology , Drug Implants , Endothelium, Vascular/pathology , Humans
18.
Cancer Genet Cytogenet ; 73(2): 109-21, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8174085

ABSTRACT

Loss or inactivation of tumor suppressor genes has been implicated by indirect methods in the etiology of most human cancers. In the functional studies presented here, tumor suppressors on human chromosome 1 were investigated using microcell-mediated chromosome transfer. Translocated chromosomes from normal human cells representing most of 1q, or all of 1p and a small portion of 1q translocated onto the region of the X chromosome encoding HPRT, were transferred into human fibrosarcoma cell line HT1080. Analysis of HT1080 microcell hybrids showed a tumor suppressor activity associated with 1q. All HT1080 cells carrying transferred 1q in a ratio of 1:1 with the HT1080 genome showed a more flattened morphology and a reduced ability to form tumors in nude mice compared to parental HT1080 cells. Diploid HT1080 cells carrying a single extra 1q also had a longer population doubling time and showed a loss of ability to clone in soft agar. Tumors arose from 1q-containing clones with a longer latency period, and a large majority of the cells comprising these tumors had lost the transferred chromosome. These results indicate the presence on chromosome 1q23-qter of a tumor suppressor gene or genes that can act to suppress transformation of a human fibrosarcoma cell line.


Subject(s)
Chromosomes, Human, Pair 1 , Fibrosarcoma/genetics , Genes, Tumor Suppressor/genetics , Cell Line, Transformed , Fibrosarcoma/pathology , Humans , Hybrid Cells , Karyotyping , Neoplasm Transplantation , Phenotype , Tumor Cells, Cultured , X Chromosome
19.
Carcinogenesis ; 14(6): 1209-14, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8389673

ABSTRACT

To determine if the tumor suppressor gene active in BHK hamster cells acts to maintain the normal phenotype by influencing oncogene transformation, careful, quantitative transfections with a variety of oncogenes were performed on four closely related BHK subclones. Two of the clones had an active suppressor gene (sup+ clones) and two of them had lost the suppressor (sup- clones) yet remained anchorage dependent. Both sup+ and sup- clones could be transformed to anchorage independence by ras, src, mos, neu, polyoma mT and SV40 suggesting that neither the presence nor the absence of the suppressor gene in BHK limits the transforming ability of these common oncogenes. All lines were resistant to transformation by N-myc, E1A and c-sis, oncogenes that may perform redundant functions in the immortal, fast growing BHK cell. SV40 small t antigen which has previously been considered unable to transform cultured cells by itself, was nevertheless able to transform sup+ BHK lines to anchorage independence in the absence of the viral large T antigen. Clones of sup- cells expressing high levels of small t antigen protein could be isolated, but they remained anchorage dependent and in tumorigenicity assays retained the long latent period characteristic of normal BHK cells. Such lines should enable the identification of cellular targets vital to the transforming function of SV40 small t.


Subject(s)
Cell Transformation, Neoplastic , Cell Transformation, Viral , Genes, Tumor Suppressor , Animals , Antigens, Viral, Tumor/pharmacology , Cell Line , Cricetinae , Mice , Mice, Nude , Simian virus 40/genetics , Transfection
20.
Exp Cell Res ; 205(2): 232-9, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8482334

ABSTRACT

Despite their familiar sensitivity to transformation by dominant-acting ras oncogenes, NIH/3T3 cells carry a ras suppressor. When tested by cell fusion they were able to suppress the anchorage-independent phenotype of both mouse and human cells transformed by activated H-ras or N-ras. This suppression occurred without a decrease in expression of the activated ras oncogene. Ras-transformed NIH/3T3 clones cured of their oncogene by benzamide treatment reverted to a nontransformed phenotype, but had lost the ability to suppress other ras transformants, indicating that their initial transformation was accompanied by suppressor loss. In hamster cells an active ras oncogene increased the rate of chromosome segregation by > 100-fold. These results suggest that in vitro transformation of NIH/3T3 cells by ras may be more similar to multistep in vivo tumor development than previously suspected, involving not only expression of an active oncogene but also loss of a suppressor activity, perhaps induced by the clastogenic oncogene.


Subject(s)
3T3 Cells/cytology , Cell Transformation, Neoplastic/pathology , Genes, ras , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cell Adhesion , Cell Division , Genes, Tumor Suppressor , Mice , Mutagens
SELECTION OF CITATIONS
SEARCH DETAIL
...