Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Oncogene ; 34(20): 2609-20, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25023702

ABSTRACT

The presence of hypoxic regions in solid tumors is an adverse prognostic factor for patient outcome. Here, we show that hypoxia induces the expression of Ephrin-A3 through a novel hypoxia-inducible factor (HIF)-mediated mechanism. In response to hypoxia, the coding EFNA3 mRNA levels remained relatively stable, but HIFs drove the expression of previously unknown long noncoding (lnc) RNAs from EFNA3 locus and these lncRNA caused Ephrin-A3 protein accumulation. Ephrins are cell surface proteins that regulate diverse biological processes by modulating cellular adhesion and repulsion. Mounting evidence implicates deregulated ephrin function in multiple aspects of tumor biology. We demonstrate that sustained expression of both Ephrin-A3 and novel EFNA3 lncRNAs increased the metastatic potential of human breast cancer cells, possibly by increasing the ability of tumor cells to extravasate from the blood vessels into surrounding tissue. In agreement, we found a strong correlation between high EFNA3 expression and shorter metastasis-free survival in breast cancer patients. Taken together, our results suggest that hypoxia could contribute to metastatic spread of breast cancer via HIF-mediated induction of EFNA3 lncRNAs and subsequent Ephrin-A3 protein accumulation.


Subject(s)
Breast Neoplasms/metabolism , Genetic Loci , Hypoxia-Inducible Factor 1/metabolism , Neoplasm Proteins/metabolism , RNA, Long Noncoding/biosynthesis , RNA, Neoplasm/biosynthesis , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Hypoxia/genetics , Cell Line, Tumor , Ephrin-A3/genetics , Ephrin-A3/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1/genetics , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Zebrafish
2.
Oncogene ; 28(47): 4147-61, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19767774

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a multifunctional secreted glycoprotein that displays broad anti-tumor activity based on dual targeting of the tumor microenvironment (anti-angiogenic action) and the tumor cells (direct anti-tumor action). Here, we show that PEDF expression is high in melanocytes, but it is lost during malignant progression of human melanoma. Using a high-throughput analysis of the data from microarray studies of molecular profiling of human melanoma, we found that PEDF expression is lost in highly invasive melanomas. In paired cell lines established from the same lesion but representing the high and low extremes of malignant potential, abundant PEDF expression was restricted to the poorly aggressive counterparts. We used RNA interference to directly address the functional consequences of PEDF silencing. PEDF knockdown in poorly aggressive melanoma cell lines augmented migration, invasion and vasculogenic mimicry, which translated into an increased in vivo metastatic potential. PEDF interference also significantly enhanced the migratory and invasive capability of normal melanocytes and moderately increased their proliferative potential. Our results show that loss of PEDF enables melanoma cells to acquire an invasive phenotype and, therefore, modulation of this multifunctional factor could be critical for the malignant progression of human melanoma.


Subject(s)
Cell Movement , Eye Proteins/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Neoplasm Proteins/metabolism , Nerve Growth Factors/metabolism , Serpins/metabolism , Cell Line, Tumor , Eye Proteins/genetics , Gene Expression Profiling , Humans , Melanoma/genetics , Melanoma/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Nerve Growth Factors/genetics , Oligonucleotide Array Sequence Analysis , Serpins/genetics
3.
Curr Drug Targets ; 9(10): 851-62, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18855619

ABSTRACT

Thrombospondin-1 is the first and most studied naturally occurring protein inhibitor of angiogenesis. Its characteristic multi-domain structure determines thrombospondin-1 divergent functions, which include but are not limited to the regulation of angiogenesis. Below we overview the structural determinants and receptors expressed on the endothelial and other cell types, that are at the root of thrombospondin-1 striking ability to block neovascularization. We specifically emphasize thrombospondin-1 direct apoptotic action on the remodeling vascular endothelium and summarize current knowledge of its pro-apoptotic signaling and transcriptional networks. Further, we provide comprehensive survey of the thrombospondin-based anti-angiogenic strategies with special focus on the combination treatments. We convincingly illustrate how precise knowledge of the pro-apoptotic events and intermediates elicited by thrombospondin in the vascular endothelial cells facilitates the design of the most effective treatment combinations, where the efficacy of thrombospondin-derived compounds is maximized by the partner drug(s) ("complementation" strategies) and provide examples of such fine-tuning of the thrombospondin-based anti-angiogenic treatments.


Subject(s)
Apoptosis/physiology , Drug Design , Thrombospondins/administration & dosage , Thrombospondins/therapeutic use , Amino Acid Sequence , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/therapeutic use , Animals , Apoptosis/drug effects , Drug Therapy, Combination , Humans , Molecular Sequence Data , Peptide Fragments/administration & dosage , Peptide Fragments/chemical synthesis , Peptide Fragments/therapeutic use , Thrombospondins/chemical synthesis
4.
J Mol Med (Berl) ; 85(1): 15-22, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17106733

ABSTRACT

The design of new therapeutic strategies for cancer treatment is based on the combination of drugs directed against different tumor compartments, including the tumor cells themselves and components of the stroma, such as the tumor vasculature. Indeed, several antiangiogenic compounds have entered clinical trials for use alone or in combination with conventional cytotoxic drugs. Pigment epithelium-derived factor (PEDF) is a multifunctional natural peptide with complex neurotrophic, neuroprotective, antiangiogenic, and proapoptotic biological activities, any of which could potentially be exploited for therapeutic purposes. This review summarizes recent studies that reveal the antitumor potential of PEDF based on its antiangiogenic properties and its newly discovered direct antitumor effects, which involve the induction of differentiation or apoptosis in tumor cells. We also discuss possible therapeutic applications of PEDF, based on these mechanistic insights and on the identification of functional domains that retain specific biological activities.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Eye Proteins/therapeutic use , Neoplasms/therapy , Neovascularization, Pathologic/prevention & control , Nerve Growth Factors/therapeutic use , Serpins/therapeutic use , Animals , Apoptosis , Cell Differentiation , Humans , Neoplasms/blood supply , Neuroprotective Agents/pharmacology
5.
Cell Death Differ ; 12(6): 649-58, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15818399

ABSTRACT

Antiangiogenic thrombospondin-1 (TSP1) induces endothelial cell death via a CD95-mediated cascade. We used this signaling pathway, where CD95/Fas is a rate-limiting intermediate, as a target to optimize the efficacy of TSP1 active peptide, DI-TSP. Like TSP1, DI-TSP upregulated endothelial CD95L in vivo. To modulate CD95 levels, we chose chemotherapy agent doxorubicin (DXR). DXR caused sustained upregulation of CD95 in the activated endothelium at 1/100 of the maximal tolerated dose. DI-TSP and DXR synergistically induced endothelial apoptosis in vitro, and in vivo, in developing murine vessels. Fas decoy, TSP1 receptor antibody and Pifithrin, a p53 inhibitor, severely decreased apoptosis and restored angiogenesis by DXR-DI-TSP combination, evidencing critical roles of CD95 and TSP1. Combined therapy synergistically blocked neovascularization and progression of the bladder and prostate carcinoma. Such informed design of a complex antiangiogenic therapy based on the rate-limiting molecular targets is a novel concept, which may yield new approaches to cancer treatment.


Subject(s)
Doxorubicin/pharmacology , Membrane Glycoproteins/metabolism , Neovascularization, Pathologic/drug therapy , Thrombospondin 1/pharmacology , Up-Regulation/drug effects , fas Receptor/metabolism , Animals , Antigens, CD/metabolism , Apoptosis/drug effects , CD47 Antigen , Cells, Cultured , Disease Progression , Drug Synergism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fas Ligand Protein , Humans , Mice , Neoplasms/blood supply , Neoplasms/metabolism , Neoplasms/pathology , Peptide Fragments/pharmacology , Thrombospondin 1/chemistry , Tumor Suppressor Protein p53/metabolism , Umbilical Cord/cytology , Umbilical Cord/drug effects , Umbilical Cord/metabolism , Xenograft Model Antitumor Assays
6.
Horm Metab Res ; 35(6): 382-6, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12920663

ABSTRACT

Retinopathy is the most common microvascular diabetes complication and represents a major threat to the eyesight. The aim of this study was to address the role of pro- and anti-angiogenic molecules in diabetic retinopathy in the aqueous humor of the eye. Aqueous humor was collected at cataract surgery from 19 diabetic patients and from 13 age- and sex-matched normoglycemic controls. Levels of pro-angiogenic vascular endothelial growth factor (VEGF) and angiogenic inhibitor pigment epithelium-derived factor (PEDF) were determined. Angiogenic activity of the aqueous humor was quantified by measuring its effect on the migration of capillary endothelial cells. In the aqueous fluid, VEGF levels were increased in diabetics (mean values: 501 vs. 367 pg/ml; p = 0.05), compared to controls. PEDF was found to be decreased in diabetics (mean values: 2080 vs. 5780 ng/ml; p = 0.04) compared to controls. In seven diabetic patients with proliferative retinopathy, the most profound finding was a significant decrease of the PEDF level (mean value: 237 ng/ml), whereas VEGF levels were comparable to diabetic patients without proliferation (mean value: 3153; p = 0.003). Angiogenic activity in samples of patients from the control group was generally inhibitory due to PEDF, and inhibition was blocked by neutralizing antibodies to PEDF. Likewise, in diabetics without proliferation, angiogenic activity was also blocked by antibodies to PEDF. We will demonstrate here that the level of the natural ocular anti-angiogenic agent PEDF is inversely associated with proliferative retinopathy. PEDF is an important negative regulator of angiogenic activity of aqueous humor. Our data may have implications for the development of novel regimens for diabetic retinopathy.


Subject(s)
Angiogenesis Inhibitors , Aqueous Humor/chemistry , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Eye Proteins , Nerve Growth Factors , Proteins/analysis , Serpins/analysis , Aged , Aged, 80 and over , Eye/blood supply , Female , Glycated Hemoglobin/analysis , Humans , Male , Middle Aged , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/analysis
7.
Diabetologia ; 46(3): 394-400, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12687338

ABSTRACT

AIMS/HYPOTHESIS: Retinopathy is the most common microvascular complication of diabetes. Our aim was to address the predictive value of pro-angiogenic and anti-angiogenic markers for progression of retinopathy. METHODS: Aqueous humor was collected at cataract surgery from 32 diabetic patients who had no or very mild retinopathy (ETDRS stage 47B). This subgroup showed lower pigment epithelium-derived factor content when compared to non-progressors and control subjects. Migratory activity in samples of patients from the control group and in diabetic patients without progression was generally inhibitory due to pigment epithelium-derived factor. Inhibition was blocked by neutralizing antibodies to pigment epithelium-derived factor. In diabetic patients initial angiogenic activity was higher in those who later developed retinopathy (vs. controls p=0.00005; vs. no progressors p=0.0003). Both pigment epithelium-derived factor and migratory response predicted progression. CONCLUSION/INTERPRETATION: Pigment epithelium-derived factor is an important negative regulator of angiogenic activity of aqueous humor. Its content in the aqueous humor of diabetic patients strongly predicts who among them will develop progression of retinopathy.


Subject(s)
Angiogenesis Inhibitors/metabolism , Aqueous Humor/metabolism , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Eye Proteins , Nerve Growth Factors , Proteins/metabolism , Serpins/metabolism , Aged , Aged, 80 and over , Blotting, Western , Cell Movement , Disease Progression , Endothelial Cells/physiology , Female , Follow-Up Studies , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Vascular Endothelial Growth Factor A/metabolism
8.
Proc Natl Acad Sci U S A ; 98(23): 13055-9, 2001 Nov 06.
Article in English | MEDLINE | ID: mdl-11606769

ABSTRACT

Proliferin (PLF) is an angiogenic placental hormone. We now report that PLF gene expression can also occur in a progressive fibrosarcoma mouse tumor cell model. PLF mRNA and protein are detectable at very low levels in cell lines derived from the mild noninvasive stage of tumor development. Expression is greatly augmented in cell lines from the aggressively invasive stage of development, a stage at which the tumor becomes highly angiogenic, and PLF expression remains high in cell lines from the end stage of fibrosarcoma. Activator protein 1 factors present at high levels in the more invasive stages of the tumor may in part allow for increased PLF expression, as cells from the mild stage in which c-jun and junB are stably expressed secrete levels of PLF comparable to that of the advanced stages. Secreted PLF protein is functionally important in tumor cell angiogenic activity, as demonstrated by the reduction of angiogenic activity in fibrosarcoma cell culture medium by immunodepletion of PLF. These results suggest that an extraembryonic genetic program, which has evolved to support fetal growth, may be reactivated in certain tumors and contribute to tumor growth.


Subject(s)
Fibrosarcoma/pathology , Gene Expression Regulation , Glycoproteins/genetics , Models, Biological , Neovascularization, Pathologic/genetics , Cells, Cultured , Disease Progression , Fibrosarcoma/genetics , Intercellular Signaling Peptides and Proteins , Prolactin , Transcription Factor AP-1/metabolism
9.
J Mol Med (Berl) ; 78(12): 663-72, 2001.
Article in English | MEDLINE | ID: mdl-11434719

ABSTRACT

Angiogenesis, the growth of new vasculature, is an absolute requirement for the maintenance and progression of the overwhelming majority of the solid tumors. Unraveling the mechanisms that govern this complex biological process has become a central issue not only for understanding of the molecular basis of cancer but also for developing new therapeutic approaches that interfere with neovascularization of the tumor mass. Here we discuss the survival and apoptosis of endothelial cells in the context of vessel formation and regression in response to mediators of angiogenesis produced by tumors. It is the balance between proangiogenic and antiangiogenic molecules in the microenvironment of a vessel in vivo that determines whether the existing vasculature will expand, remain the same, or regress. Here we propose that the vascular endothelial cells themselves interpret and respond to these environmental cues by integrating the activities of the survival and apoptotic pathways within the cell. Thus it is the survival or death of the vulnerable cells that venture out to form new vessels that is the ultimate arbiter of whether neovascularization, as well as the growth of a malignancy that depends on it, succeeds or fails.


Subject(s)
Growth Substances/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Animals , Apoptosis , Cell Survival , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Genes, Tumor Suppressor/genetics , Growth Substances/genetics , Humans , Lymphokines/genetics , Lymphokines/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Signal Transduction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Oncogene ; 20(26): 3443-8, 2001 Jun 07.
Article in English | MEDLINE | ID: mdl-11423995

ABSTRACT

Thrombospondin-1 (TSP-1) is a potent inhibitor of angiogenesis that acts directly on endothelial cells via the CD36 surface receptor molecule to halt their migration, proliferation, and morphogenesis in vitro and to block neovascularization in vivo. Here we show that inhibitory signals elicited by TSP-1 did not alter the ability of inducers of angiogenesis to activate p42 and p44 mitogen-activated protein kinase (MAPK). Rather, TSP-1 induced a rapid and transient activation of c-Jun N-terminal kinases (JNK). JNK activation by TSP-1 required engagement of CD36, as it was blocked by antagonistic CD36 antibodies and stimulated by short anti-angiogenic peptides derived from TSP-1 that act exclusively via CD36. TSP-1 inhibition of corneal neovascularization induced by bFGF was severely impaired in mice null for JNK-1, pointing to a critical role for this stress-activated kinase in the inhibition of neovascularization by TSP-1.


Subject(s)
Angiogenesis Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Physiologic/drug effects , Thrombospondin 1/pharmacology , Animals , Apoptosis , CD36 Antigens/physiology , Capillaries/cytology , Cells, Cultured/drug effects , Cornea/blood supply , Cysteine Proteinase Inhibitors/pharmacology , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Enzyme Activation/drug effects , Fibroblast Growth Factor 2/pharmacology , Flavonoids/pharmacology , JNK Mitogen-Activated Protein Kinases , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/deficiency , Mitogen-Activated Protein Kinases/genetics , Neovascularization, Pathologic/prevention & control , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Platelet-Derived Growth Factor/pharmacology , Thrombospondin 1/chemistry , Thrombospondin 1/therapeutic use , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Genes Dev ; 15(11): 1373-82, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11390357

ABSTRACT

Thrombospondin 1 (TSP1) is a multifunctional protein able to activate TGFbeta and to inhibit angiogenesis in vivo. Although usually thought of as an inhibitor of tumor growth, TSP1 may sometimes be present at high levels during tumor progression, suggesting that tumors can eventually overcome their anti-tumor effects. Using a tet-repressible expression system, we demonstrate that murine TSP1 delayed the onset of tumor growth when produced in the tumor bed by rat fibrosarcoma tumor cells or by stromal fibroblasts coinjected with unmodified C6 glioma tumor cells. Yet upon prolonged exposure to TSP1, tumors came to grow at the same rate in the presence as in the absence of TSP1 and transplantation experiments showed that they had become insensitive to inhibition by TSP1 in both syngeneic and immune compromised hosts. Tumor resistance to TSP1 developed as a result of the in vivo outgrowth of pre-existing tumor cell variants that (1) secreted increased amounts of angiogenic factors that counterbalanced the inhibitory effect of TSP1 on neovascularization and (2) grew more efficiently in the presence of TSP1-activated TGFbeta. These results indicate that prolonged and continuous local delivery of a single multifunctional angiogenesis inhibitor like TSP1 to fast-growing tumors can lead to tumor resistance in vivo by fostering the outgrowth of subpopulations that are a by-product of the genetic instability of the tumor cells themselves.


Subject(s)
Angiogenesis Inhibitors/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Thrombospondin 1/metabolism , Transforming Growth Factor beta/metabolism , Animals , Blotting, Northern , Fibrosarcoma/blood supply , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Glioblastoma/blood supply , Glioblastoma/metabolism , Glioblastoma/pathology , Immunoblotting , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/blood supply , Neoplasms/pathology , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
12.
J Exp Med ; 193(5): 607-20, 2001 Mar 05.
Article in English | MEDLINE | ID: mdl-11238591

ABSTRACT

Cyclosporin A (CsA) is an immunosuppressive drug that inhibits the activity of transcription factors of the nuclear factor of activated T cells (NFAT) family, interfering with the induction of cytokines and other inducible genes required for the immune response. Here we show that CsA inhibits migration of primary endothelial cells and angiogenesis induced by vascular endothelial growth factor (VEGF); this effect appears to be mediated through the inhibition of cyclooxygenase (Cox)-2, the transcription of which is activated by VEGF in primary endothelial cells. Consistent with this, we show that the induction of Cox-2 gene expression by VEGF requires NFAT activation. Most important, the CsA-mediated inhibition of angiogenesis both in vitro and in vivo was comparable to the Cox-2 inhibitor NS-398, and reversed by prostaglandin E(2). Furthermore, the in vivo corneal angiogenesis induced by VEGF, but not by basic fibroblast growth factor, was selectively inhibited in mice treated with CsA systemically. These findings involve NFAT in the regulation of Cox-2 in endothelial cells, point to a role for this transcription factor in angiogenesis, and may provide a novel mechanism underlying the beneficial effects of CsA in angiogenesis-related diseases such as rheumatoid arthritis and psoriasis.


Subject(s)
Cyclosporine/pharmacology , DNA-Binding Proteins/metabolism , Endothelial Growth Factors/antagonists & inhibitors , Isoenzymes/metabolism , Lymphokines/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Nuclear Proteins , Prostaglandin-Endoperoxide Synthases/metabolism , Transcription Factors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Cell Migration Inhibition , Cell Movement/drug effects , Cells, Cultured , Cornea/blood supply , Cornea/drug effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Endothelial Growth Factors/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Lymphokines/metabolism , Lymphokines/pharmacology , Membrane Proteins , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , NFATC Transcription Factors , Nitrobenzenes/pharmacology , Promoter Regions, Genetic , Prostaglandin-Endoperoxide Synthases/genetics , Sulfonamides/pharmacology , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
J Cell Sci ; 114(Pt 24): 4421-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11792807

ABSTRACT

Neuroblastoma is notable for its cellular heterogeneity and unpredictable outcome. Tumors are a variable mixture of primitive malignant neuroblasts, more differentiated ganglionic cells, Schwann and endothelial cells. Although often fatal, neuroblastomas can spontaneously regress, possibly due to favorable autocrine and paracrine interactions among these cells. Here, pigment epithelium-derived factor (PEDF), a potent inhibitor of angiogenesis and inducer of neural differentiation, is shown to be produced by ganglionic cells and Schwann cells, but not by more primitive tumor cells. Although undifferentiated neuroblastoma tumor cell secretions were angiogenic primarily due to vascular endothelial growth factor, secretions of Schwann cells were anti-angiogenic due to PEDF. In addition, PEDF was the major factor responsible for Schwann cell's ability to induce tumor cell differentiation in vitro and recombinant PEDF had the same effect in vitro and in vivo. Both the growth and the survival of Schwann cells were enhanced by PEDF. Thus PEDF may serve as a multifunctional antitumor agent in neuroblastomas, inhibiting angiogenesis while promoting the numbers of Schwann cells and differentiated tumor cells that in turn produce PEDF, suggesting that its clinical administration could stimulate a multifaceted antitumor feedback loop with the potential to limit and possibly regress tumor growth.


Subject(s)
Antineoplastic Agents/metabolism , Eye Proteins , Nerve Growth Factors , Neuroblastoma/metabolism , Neuroblastoma/prevention & control , Pigment Epithelium of Eye/physiology , Proteins/physiology , Schwann Cells/physiology , Serpins/physiology , Angiogenesis Inhibitors/metabolism , Animals , Antineoplastic Agents/pharmacology , Cattle , Cell Differentiation/drug effects , Cell Division/physiology , Cell Survival/physiology , Cells, Cultured , Culture Media, Conditioned/metabolism , Endothelial Growth Factors/metabolism , Female , Growth Substances/metabolism , Humans , Injections, Subcutaneous , Lymphokines/metabolism , Mice , Mice, Nude , Neuroblastoma/blood supply , Neuroblastoma/pathology , Proteins/administration & dosage , Proteins/metabolism , Rats , Rats, Inbred F344 , Recombinant Proteins/administration & dosage , Schwann Cells/metabolism , Serpins/administration & dosage , Serpins/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
FASEB J ; 14(14): 2255-65, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053247

ABSTRACT

Recent studies have identified factors responsible for angiogenesis within developing tumors, but mediators of vessel formation at sites of trauma, injury, and wound healing are not clearly established. Here we show that sphingosine 1-phosphate (S1P) released by platelets during blood clotting is a potent, specific, and selective endothelial cell chemoattractant that accounts for most of the strong endothelial cell chemotactic activity of blood serum, an activity that is markedly diminished in plasma. Preincubation of endothelial cells with pertussis toxin inhibited this effect of S1P, demonstrating the involvement of a Galphai-coupled receptor. After S1P-induced migration, endothelial cells proliferated avidly and differentiated forming multicellular structures suggestive of early blood vessel formation. S1P was strikingly effective in enhancing the ability of fibroblast growth factor to induce angiogenesis in the avascular mouse cornea. Our results show that blood coagulation initiates endothelial cell angiogenic responses through the release of S1P, a potent endothelial cell chemoattractant that exerts its effects by activating a receptor-dependent process.


Subject(s)
Blood Platelets/metabolism , Endothelium, Vascular/drug effects , Hemostasis/drug effects , Lysophospholipids , Neovascularization, Physiologic/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Animals , Blood Coagulation , Blood Proteins/drug effects , Blood Proteins/metabolism , Cell Line , Charcoal/pharmacology , Chemotaxis/drug effects , Chromatography, Thin Layer , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Growth Substances/blood , Hemostasis/physiology , Humans , Kinetics , Lipids/blood , Lipids/pharmacology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Sphingosine/blood
15.
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
16.
Cell Growth Differ ; 11(4): 201-10, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10775037

ABSTRACT

The ability of neoplastic cells to recruit blood vasculature is crucial to their survival in the host organism. However, the evidence linking dominant oncogenes to the angiogenic switch remains incomplete. We demonstrate here that Myc, an oncoprotein implicated in many human malignancies, stimulates neovascularization. As an experimental model, we used Rat-1A fibroblasts that form vascular tumors upon transformation by Myc in immunocompromised mice. Our previous work and the use of neutralizing antibodies reveal that in these cells, the angiogenic switch is achieved via down-modulation of thrombospondin-1, a secreted inhibitor of angiogenesis, whereas the levels of vascular endothelial growth factor, a major activator of angiogenesis, remain high and unaffected by Myc. Consistent with this finding, overexpression of Myc confers upon the conditioned media the ability to promote migration of adjacent endothelial cells in vitro and corneal neovascularization in vivo. Furthermore, mobilization of estrogen-dependent Myc in vivo with the appropriate steroid provokes neovascularization of cell implants embedded in Matrigel. These data suggest that Myc is fully competent to trigger the angiogenic switch in vivo and that secondary events may not be required for neovascularization of Myc-induced tumors.


Subject(s)
Cell Transformation, Neoplastic/genetics , Proto-Oncogene Proteins c-myc/physiology , Animals , Cattle , Cell Line , Cell Line, Transformed , Cell Movement/drug effects , Cells, Cultured , Cornea/drug effects , Cornea/pathology , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblasts/cytology , Fibroblasts/transplantation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/genetics , Phenotype , Proto-Oncogene Proteins c-myc/drug effects , Proto-Oncogene Proteins c-myc/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
18.
Nat Med ; 6(2): 196-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10655109

ABSTRACT

Maspin, a unique member of the serpin family, is a secreted protein encoded by a class II tumor suppressor gene whose downregulation is associated with the development of breast and prostate cancers. Overexpression of maspin in breast tumor cells limits their growth and metastases in vivo. In this report we demonstrate that maspin is an effective inhibitor of angiogenesis. In vitro, it acted directly on cultured endothelial cells to stop their migration towards basic fibroblast growth factor and vascular endothelial growth factor and to limit mitogenesis and tube formation. In vivo, it blocked neovascularization in the rat cornea pocket model. Maspin derivatives mutated in the serpin reactive site lost their ability to inhibit the migration of fibroblasts, keratinocytes, and breast cancer cells but were still able to block angiogenesis in vitro and in vivo. When maspin was delivered locally to human prostate tumor cells in a xenograft mouse model, it blocked tumor growth and dramatically reduced the density of tumor-associated microvessels. These data suggest that the tumor suppressor activity of maspin may depend in large part on its ability to inhibit angiogenesis and raise the possibility that maspin and similar serpins may be excellent leads for the development of drugs that modulate angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Proteins/pharmacology , Serpins/pharmacology , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cattle , Cell Movement/drug effects , Cornea/blood supply , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Genes, Tumor Suppressor , Humans , Male , Mice , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Rats , Tumor Cells, Cultured
19.
Cancer Metastasis Rev ; 19(1-2): 87-92, 2000.
Article in English | MEDLINE | ID: mdl-11191069

ABSTRACT

Angiogenesis is a process of capillary formation from pre-existing blood vessels. It is tightly controlled by the balance between positive and negative environmental signals--inducers and inhibitors of angiogenesis in such a way that predominance of inducers results in angiogenesis and predominance of inhibitors--in vascular quiescence. Here we discuss the ability of the angiogenic stimuli to promote survival and the pathways they may utilize. We also summarize information available on the signaling events elicited in the endothelial cells by a naturally occurring inhibitor of angiogenesis Thrombospondin-1 (TSP-1), that result in the endothelial cell apoptosis and inhibition of angiogenesis in vivo. This ability to cause programmed cell death in vascular endothelium is not unique to TSP-1. A substantial number of known angiogenesis inhibitors can also trigger apoptosis in the activated endothelial cells. This fact argues for the possibility of apoptosis to be a common denominator for a major fraction of anti-angiogenic molecules. If this is the case, it is equally possible that the ratio between environmental factors that control angiogenesis is interpreted within individual endothelial cell as a balance between pro-apoptotic and survival signals. Thus the relative strength of the death and survival signal or signals determines the fate of endothelial cell and therefore the fate of remodeling vessel.


Subject(s)
Angiogenesis Inhibitors , Cell Survival/drug effects , Endothelium, Vascular/cytology , Neovascularization, Physiologic/physiology , Thrombospondin 1/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Endothelium, Vascular/drug effects , Humans , Models, Biological , Neovascularization, Physiologic/drug effects
20.
Nat Med ; 6(1): 41-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10613822

ABSTRACT

Thrombospondin-1 (TSP-1) is a naturally occurring inhibitor of angiogenesis that limits vessel density in normal tissues and curtails tumor growth. Here, we show that the inhibition of angiogenesis in vitro and in vivo and the induction of apoptosis by thrombospondin-1 all required the sequential activation of CD36, p59fyn, caspase-3 like proteases and p38 mitogen-activated protein kinases. We also detected increased endothelial cell apoptosis in situ at the margins of tumors in mice treated with thrombospondin-1. These results indicate that thrombospondin-1, and possibly other broad-spectrum natural inhibitors of angiogenesis, act in vivo by inducing receptor-mediated apoptosis in activated microvascular endothelial cells.


Subject(s)
Apoptosis/physiology , CD36 Antigens/physiology , Endothelium, Vascular/physiology , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins/physiology , Thrombospondin 1/pharmacology , Angiostatins , Animals , Apoptosis/drug effects , CD36 Antigens/genetics , Caspases/metabolism , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Knockout , Microcirculation , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Plasminogen/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fyn , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...