Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cancer Res ; 78(5): 1266-1274, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29301791

ABSTRACT

von Hippel-Lindau (VHL) disease is an autosomal-dominant tumor predisposition syndrome characterized by the development of highly vascularized tumors and cysts. LOH of the VHL gene results in aberrant upregulation of hypoxia-inducible factors (HIF) and has been associated with tumor formation. Hemangioblastomas of the central nervous system and retina represent the most prevalent VHL-associated tumors, but no VHL animal model has reproduced retinal capillary hemangioblastomas (RCH), the hallmark lesion of ocular VHL. Here we report our work in developing a murine model of VHL-associated RCH by conditionally inactivating Vhl in a hemangioblast population using a Scl-Cre-ERT2 transgenic mouse line. In transgenic mice carrying the conditional allele and the Scl-Cre-ERT2 allele, 64% exhibited various retinal vascular anomalies following tamoxifen induction. Affected Vhl-mutant mice demonstrated retinal vascular lesions associated with prominent vasculature, anomalous capillary networks, hemorrhage, exudates, and localized fibrosis. Histologic analyses showed RCH-like lesions characterized by tortuous, dilated vasculature surrounded by "tumorlet" cell cluster and isolated foamy stromal cells, which are typically associated with RCH. Fluorescein angiography suggested increased vascular permeability of the irregular retinal vasculature and hemangioblastoma-like lesions. Vhl deletion was detected in "tumorlet" cells via microdissection. Our findings provide a phenotypic recapitulation of VHL-associated RCH in a murine model that may be useful to study RCH pathogenesis and therapeutics aimed at treating ocular VHL.Significance: This study describes a model that phenotypically recapitulates a form of retinal pathogenesis that is driven by genetic loss of the VHL tumor suppressor, providing a useful tool for its study and therapeutic intervention. Cancer Res; 78(5); 1266-74. ©2018 AACR.


Subject(s)
Disease Models, Animal , Hemangioblastoma/pathology , Hemangioblasts/pathology , Retinal Neoplasms/pathology , Sequence Deletion , Von Hippel-Lindau Tumor Suppressor Protein/physiology , von Hippel-Lindau Disease/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Hemangioblastoma/genetics , Hemangioblastoma/metabolism , Hemangioblasts/metabolism , Male , Mice , Mice, Knockout , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/metabolism
2.
Cancer Res ; 74(20): 5808-18, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25320178

ABSTRACT

A genome-wide association study (GWAS) of bladder cancer identified a genetic marker rs8102137 within the 19q12 region as a novel susceptibility variant. This marker is located upstream of the CCNE1 gene, which encodes cyclin E, a cell-cycle protein. We performed genetic fine-mapping analysis of the CCNE1 region using data from two bladder cancer GWAS (5,942 cases and 10,857 controls). We found that the original GWAS marker rs8102137 represents a group of 47 linked SNPs (with r(2) ≥ 0.7) associated with increased bladder cancer risk. From this group, we selected a functional promoter variant rs7257330, which showed strong allele-specific binding of nuclear proteins in several cell lines. In both GWASs, rs7257330 was associated only with aggressive bladder cancer, with a combined per-allele OR = 1.18 [95% confidence interval (CI), 1.09-1.27, P = 4.67 × 10(-5)] versus OR = 1.01 (95% CI, 0.93-1.10, P = 0.79) for nonaggressive disease, with P = 0.0015 for case-only analysis. Cyclin E protein expression analyzed in 265 bladder tumors was increased in aggressive tumors (P = 0.013) and, independently, with each rs7257330-A risk allele (P(trend) = 0.024). Overexpression of recombinant cyclin E in cell lines caused significant acceleration of cell cycle. In conclusion, we defined the 19q12 signal as the first GWAS signal specific for aggressive bladder cancer. Molecular mechanisms of this genetic association may be related to cyclin E overexpression and alteration of cell cycle in carriers of CCNE1 risk variants. In combination with established bladder cancer risk factors and other somatic and germline genetic markers, the CCNE1 variants could be useful for inclusion into bladder cancer risk prediction models.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Cyclin E/genetics , Oncogene Proteins/genetics , Urinary Bladder Neoplasms/genetics , Case-Control Studies , Cyclin E/metabolism , Gene Expression , Gene Frequency , Genome-Wide Association Study , Haplotypes , HeLa Cells , Humans , Oncogene Proteins/metabolism , Polymorphism, Single Nucleotide , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
3.
J Immunother ; 35(6): 502-12, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22735808

ABSTRACT

Angiogenesis and inflammation are important therapeutic targets in non-small cell lung cancer (NSCLC). It is well known that proteolysis mediated by matrix metalloproteinases (MMPs) promotes angiogenesis and inflammation in the tumor microenvironment. Here, the effects of the MMP inhibitor TIMP-2 on NSCLC inflammation and angiogenesis were evaluated in TIMP-2-deficient (timp2-/-) mice injected subcutaneously (SC) with Lewis lung carcinoma cells and compared with the effects on tumors in wild-type mice. TIMP-2-deficient mice demonstrated increased tumor growth, enhanced expression of angiogenic marker αvß3 in tumor and endothelial cells, and significantly higher serum vascular endothelial growth factor-A levels. Tumor-bearing timp2-/- mice showed a significant number of inflammatory cells in their tumors, upregulation of inflammation mediators, nuclear factor-kappaB, and Annexin A1, as well as higher levels of serum interleukin (IL)-6. Phenotypic analysis revealed an increase in myeloid-derived suppressor cell (MDSC) cells (CD11b+ and Gr-1+) that coexpressed vascular-endothelial-growth factor receptor 1 (VEGF-R1) and elevated MMP activation present in tumors and spleens from timp2-/- mice. Furthermore, TIMP-2-deficient tumors upregulated expression of the immunosuppressing genes controlling MDSC growth, IL-10, IL-13, IL-11, and chemokine ligand (CCL-5/RANTES), and decreased interferon-γ and increased CD40L. Moreover, forced TIMP-2 expression in human lung adenocarcinoma A-549 resulted in a significant reduction of MDSCs recruited into tumors, as well as suppression of angiogenesis and tumor growth. The increase in MDSCs has been linked to cancer immunosuppression and angiogenesis. Therefore, this study supports TIMP-2 as a negative regulator of MDSCs with important implications for the immunotherapy and/or antiangiogenic treatment of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Myeloid Cells/metabolism , Neovascularization, Pathologic , Tissue Inhibitor of Metalloproteinase-2/metabolism , Animals , Annexin A1/biosynthesis , CD40 Antigens/biosynthesis , Carcinoma, Lewis Lung , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Chemokine CCL5/biosynthesis , Female , Humans , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-11/biosynthesis , Interleukin-13/biosynthesis , Interleukin-6/blood , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Metalloproteases/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , NF-kappa B/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/deficiency , Tissue Inhibitor of Metalloproteinase-2/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
4.
Peptides ; 32(9): 1840-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21871510

ABSTRACT

Tissue inhibitor of metalloproteinases-2 (TIMP-2) inhibits angiogenesis by several mechanisms involving either MMP inhibition or direct endothelial cell binding. The primary aim of this study was to identify the TIMP-2 region involved in binding to the previously identified receptor integrin α3ß1, and to determine whether synthetic peptides derived from this region retained angio-inhibitory and tumor suppressor activity. We demonstrated that the N-terminal domain of TIMP-2 (N-TIMP-2) binds to α3ß1 and inhibits vascular endothelial growth factor-stimulated endothelial cell growth in vitro, suggesting that both the α3ß1-binding domain and the growth suppressor activity of TIMP-2 localize to the N-terminal domain. Using a peptide array approach we identify a 24 amino acid region of TIMP-2 primary sequence, consisting of residues Ile43-Ala66, which shows α3ß1-binding activity. Subsequently we demonstrate that synthetic peptides from this region compete for TIMP-2 binding to α3ß1 and suppress endothelial growth in vitro. We define a minimal peptide sequence (peptide 8-9) that possesses both angio-inhibitory and, using a murine xenograft model of Kaposi's sarcoma, anti-tumorigenic activity in vivo. Thus, both the α3ß1-binding and the angio-inhibitory activities co-localize to a solvent exposed, flexible region in the TIMP-2 primary sequence that is unique in amino acid sequence compared with other members of the TIMP family. Furthermore, comparison of the TIMP-2 and TIMP-1 protein 3-D structures in this region also identified unique structural differences. Our findings demonstrate that the integrin binding, tumor growth suppressor and in vivo angio-inhibitory activities of TIMP-2 are intimately associated within a unique sequence/structural loop (B-C loop).


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Integrin alpha3beta1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Amino Acid Sequence , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Mice , Mice, Nude , Microarray Analysis , Molecular Sequence Data , Neovascularization, Pathologic/metabolism , Peptides/administration & dosage , Peptides/chemical synthesis , Peptides/pharmacology , Protein Binding , Sarcoma, Kaposi/pathology , Tissue Inhibitor of Metalloproteinase-1/chemistry , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/chemistry , Vascular Endothelial Growth Factor A/pharmacology
5.
Ophthalmology ; 117(9): 1782-91, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20605212

ABSTRACT

PURPOSE: To determine the in vivo effect of doxycycline on choroidal angiogenesis and pterygium growth by using a choroidal neovascular (CNV) murine model, a directed in vivo angiogenesis assay (DIVAA) and a pterygium murine model. DESIGN: Experimental study. PARTICIPANTS: Three murine models were investigated with 4 mice minimum per group and 22 maximum per group. METHODS: Mice received water with or without doxycycline. For the CNV, the neovascular lesion volume was determined in choroid-retinal pigment epithelial flat mounts using confocal microscopy 7 days after laser induction. For DIVAA, silicone capsules containing 10,000 human pterygium epithelial cells were implanted in the flanks of mice subcutaneously. After 11 days, neovascularization (NV) was quantified using spectrofluorometry after murine tail-vein injection of fluorescein isothiocyanate-labeled dextran. A pterygium epithelial cell model was developed by injecting 10,000 human pterygium epithelial cells in the nasal subconjunctival space in athymic nude mice. Doxycycline was started on day 6 at 50 mg/kg per day; corneal lesions that resulted from the injections were compared at days 6 and 15. MAIN OUTCOME MEASURES: The Student t-test was used to evaluate the data for the CNV and DIVAA models and histologic preparations were used to evaluate pterygia lesions. RESULTS: There was significantly less NV and lesion volume with doxycycline taken in drinking water versus plain water. With doxycycline treatment, the laser-induced CNV showed a maximal 66% decrease in choroidal blood vessel volume (P< or =0.008) and the DIVAA showed a 30% reduction of blood vessel growth and migration (P<0.004). Histologic preparations demonstrated that pterygium cell lesions regressed when mice were administered doxycycline for 9 days. CONCLUSIONS: Doxycycline significantly inhibited angiogenesis in 3 murine models. The most dramatic effect was found in the CNV model followed by the pterygia epithelial cell DIVAA model. The anterior segment pterygium model also showed regression histologically. This suggests that doxycycline may be successful as an adjunctive treatment for CNV and pterygia in humans; clinical trials would be necessary to determine if there is a benefit.


Subject(s)
Anti-Bacterial Agents/pharmacology , Choroidal Neovascularization/prevention & control , Corneal Neovascularization/prevention & control , Disease Models, Animal , Doxycycline/pharmacology , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Cell Movement/drug effects , Cell Transplantation , Cells, Cultured , Doxycycline/administration & dosage , Endothelium, Vascular/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Microscopy, Confocal , Pterygium/drug therapy , Pterygium/pathology , Spectrometry, Fluorescence
7.
J Natl Cancer Inst ; 97(2): 132-41, 2005 Jan 19.
Article in English | MEDLINE | ID: mdl-15657343

ABSTRACT

BACKGROUND: Human cripto-1 (CR-1) promotes cell transformation and increases migration and invasion of various mouse and human epithelial cell lines. We investigated whether CR-1 also stimulates angiogenesis. METHODS: We used human umbilical vein endothelial cells (HUVECs) to measure in vitro migration with fibronectin-coated Boyden chambers, invasion with Matrigel-coated Boyden chambers, proliferation with a tetrazolium salt, and differentiation with an in vitro Matrigel assay. We investigated new blood vessel formation in vivo by use of Matrigel-filled silicone cylinders implanted under the skin of nude mice and by use of a breast cancer xenograft model with CR-1-transfected or control Neo-transfected MCF-7 human breast cancer cells. We also used a blocking anti-CR-1 monoclonal antibody to investigate the role of CR-1 in angiogenesis in vivo and in vitro. All statistical tests were two-sided. RESULTS: CR-1 stimulated HUVEC proliferation, migration, and invasion and induced HUVEC differentiation into vascular-like structures on Matrigel. In vivo, recombinant CR-1 protein induced microvessel formation in Matrigel-filled silicone cylinders, and microvessel formation was statistically significantly inhibited with a blocking anti-CR-1 monoclonal antibody (CR-1 and antibody = 127% of microvessel formation compared with that in untreated control cylinders and CR-1 alone = 259%; difference = 132%, 95% confidence interval [CI] = 123% to 140%; P<.001). Tumors formed by CR-1-transfected MCF-7 cells in the cleared mammary fat pad of nude mice had higher microvessel density than tumors formed by control Neo-transfected MCF-7 cells (CR-1-transfected cells = 4.66 vessels per field and Neo-transfected cells = 2.33 vessels per field; difference = 2.33 vessels per field, 95% CI = 1.2 to 2.8; P = .004). CONCLUSION: CR-1 appears to have an important role in the multistep process of angiogenesis.


Subject(s)
Breast Neoplasms/blood supply , Endothelial Cells/metabolism , Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Animals , Antibodies, Monoclonal , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Collagen , Drug Combinations , Epidermal Growth Factor/immunology , Epidermal Growth Factor/pharmacology , Female , Fibroblast Growth Factor 2/analysis , GPI-Linked Proteins , Growth Substances/metabolism , Humans , Immunoassay , Intercellular Signaling Peptides and Proteins , Laminin , Membrane Glycoproteins/immunology , Membrane Glycoproteins/pharmacology , Mice , Mice, Nude , Neoplasm Proteins/immunology , Neoplasm Proteins/pharmacology , Proteoglycans , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transplantation, Heterologous , Umbilical Veins , Vascular Endothelial Growth Factor A/analysis
8.
Blood ; 105(4): 1660-8, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15479729

ABSTRACT

Tissue inhibitor of metalloproteinase 1 (TIMP-1) is a stromal factor with multiple functions. Overexpression of TIMP-1 correlates with aggressive clinical behavior of a spectrum of tumors. Here, for the first time, we address the role of TIMP-1 in the pathogenesis of B-cell lymphomas. An Epstein-Barr virus (EBV)-negative Burkitt lymphoma cell line with ectopic TIMP-1 expression (TIMP-1JD38) was used to identify genes induced/repressed by TIMP-1. Differentially expressed genes were analyzed by cDNA microarray, and they were validated by immunohistochemistry, flow cytometry, and Western blotting. Analysis revealed changes of genes coding for B-cell growth/differentiation, transcription, and cell cycle regulators. TIMP-1 repressed expression of germinal center (GC) markers CD10, Bcl-6, PAX-5 and up-regulated plasma cell-associated antigens CD138, MUM-1/IRF-4, XBP-1, and CD44, suggesting a plasma cell differentiation. This is accompanied by activation of signal transducer and activator of transcription 3 (STAT-3) and switch to cyclin D2 expression. However, TIMP-1JD38 cells expressed an inactive form of XBP-1, lacking antibody production/secretion. This incomplete plasmacytic differentiation occurs without altering cell proliferation, and despite c-Myc deregulation, indicating an arrested plasmacytic/plasmablastic stage of differentiation. Further validation in human lymphoma cell lines and in primary B-cell tumors demonstrated a predominant TIMP-1 expression in tumors with plasmacytic/plasmablastic phenotypes, including multiple myelomas. These findings strongly support TIMP-1 as an important factor in the pathogenesis of plasmacytic/plasmablastic tumors.


Subject(s)
Burkitt Lymphoma/pathology , Cell Differentiation/physiology , Plasma Cells/pathology , Tissue Inhibitor of Metalloproteinase-1/physiology , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , B-Lymphocytes/metabolism , Burkitt Lymphoma/enzymology , Burkitt Lymphoma/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Cell Proliferation , Down-Regulation/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/physiology , Humans , Lymphoid Tissue/enzymology , Lymphoid Tissue/pathology , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Oligonucleotide Array Sequence Analysis , Repressor Proteins/physiology , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transcription, Genetic/physiology , Up-Regulation/genetics
9.
Blood ; 105(7): 2836-44, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15576472

ABSTRACT

CD97, a membrane protein expressed at high levels on inflammatory cells and some carcinomas, is a member of the adhesion G protein-coupled receptor family, whose members have bipartite structures consisting of an extracellular peptide containing adhesion motifs noncovalently coupled to a class B 7-transmembrane domain. CD97alpha, the extracellular domain of CD97, contains 3 to 5 fibrillin class 1 epidermal growth factor (EGF)-like repeats, an Arg-Gly-Asp (RGD) tripeptide, and a mucin stalk. We show here that CD97alpha promotes angiogenesis in vivo as demonstrated with purified protein in a directed in vivo angiogenesis assay (DIVAA) and by enhanced vascularization of developing tumors expressing CD97. These data suggest that CD97 can contribute to angiogenesis associated with inflammation and tumor progression. Strong integrin alpha5beta1 interactions with CD97 have been identified, but alpha v beta3 also contributes to cell attachment. Furthermore, soluble CD97 acts as a potent chemoattractant for migration and invasion of human umbilical vein endothelial cells (HUVECs), and this function is integrin dependent. CD97 EGF-like repeat 4 is known to bind chondroitin sulfate. It was found that coengagement of alpha5beta1 and chondroitotin sulfate proteoglycan by CD97 synergistically initiates endothelial cell invasion. Integrin alpha5beta1 is the first high-affinity cellular counterreceptor that has been identified for a member within this family of adhesion receptors.


Subject(s)
Endothelium, Vascular/metabolism , Integrin alpha5beta1/metabolism , Membrane Glycoproteins/metabolism , Neoplasms/metabolism , Neovascularization, Physiologic/physiology , Animals , Antigens, CD , Cell Adhesion/physiology , Chondroitin Sulfates/metabolism , Endothelium, Vascular/cytology , Epidermal Growth Factor/genetics , HT29 Cells , Humans , Integrin alphaVbeta3/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , NIH 3T3 Cells , Neoplasms/blood supply , Neoplasms/physiopathology , Oligopeptides/genetics , Phenotype , Protein Structure, Tertiary , Rats , Receptors, G-Protein-Coupled , Umbilical Veins/cytology
10.
Cancer Res ; 64(18): 6489-94, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15374959

ABSTRACT

We have found through ex vivo and in vivo angiogenesis models that the adrenomedullin gene-related peptide, proadrenomedullin NH2-terminal 20 peptide (PAMP), exhibits a potent angiogenic potential at femtomolar concentrations, whereas classic angiogenic factors such as vascular endothelial growth factor and adrenomedullin mediate a comparable effect at nanomolar concentrations. We found that human microvascular endothelial cells express PAMP receptors and respond to exogenous addition of PAMP by increasing migration and cord formation. Exposure of endothelial cells to PAMP increases gene expression of other angiogenic factors such as adrenomedullin, vascular endothelial growth factor, basic fibroblast growth factor, and platelet-derived growth factor C. In addition, the peptide fragment PAMP(12-20) inhibits tumor cell-induced angiogenesis in vivo and reduces tumor growth in xenograft models. Together, our data demonstrate PAMP to be an extremely potent angiogenic factor and implicate this peptide as an attractive molecular target for angiogenesis-based antitumor therapy.


Subject(s)
Neovascularization, Physiologic/drug effects , Peptides/antagonists & inhibitors , Adrenomedullin , Animals , Calcium/metabolism , Cell Division/drug effects , Cell Line, Tumor , Chick Embryo , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasms/blood supply , Neoplasms/pathology , Peptide Fragments/pharmacology , Peptides/pharmacology , Peptides/physiology , Xenograft Model Antitumor Assays
11.
Cell ; 114(2): 171-80, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12887919

ABSTRACT

Tissue inhibitors of metalloproteinases (TIMPs) suppress matrix metalloproteinase (MMP) activity critical for extracellular matrix turnover associated with both physiologic and pathologic tissue remodeling. We demonstrate here that TIMP-2 abrogates angiogenic factor-induced endothelial cell proliferation in vitro and angiogenesis in vivo independent of MMP inhibition. These effects require alpha 3 beta 1 integrin-mediated binding of TIMP-2 to endothelial cells. Further, TIMP-2 induces a decrease in total protein tyrosine phosphatase (PTP) activity associated with beta1 integrin subunits as well as dissociation of the phosphatase SHP-1 from beta1. TIMP-2 treatment also results in a concomitant increase in PTP activity associated with tyrosine kinase receptors FGFR-1 and KDR. Our findings establish an unexpected, MMP-independent mechanism for TIMP-2 inhibition of endothelial cell proliferation in vitro and reveal an important component of the antiangiogenic effect of TIMP2 in vivo.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Matrix Metalloproteinases/metabolism , Neovascularization, Physiologic/physiology , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Angiogenesis Inhibitors/genetics , Animals , Binding, Competitive , Cell Division/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian/cytology , Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/metabolism , Fibroblasts/metabolism , Humans , Integrin alpha3beta1/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Neovascularization, Physiologic/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/drug effects , Protein Tyrosine Phosphatases/metabolism , Receptor Protein-Tyrosine Kinases/pharmacology , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor , Recombinant Proteins/pharmacology , Tissue Inhibitor of Metalloproteinase-2/genetics , Vanadates/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Am J Pathol ; 162(5): 1431-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12707026

ABSTRACT

One of the major problems in angiogenesis research remains the lack of suitable methods for quantifying the angiogenic response in vivo. We describe the development and application of the directed in vivo angiogenesis assay (DIVAA) and demonstrated that it is reproducible and quantitative. This assay consists of subcutaneous implantation of semiclosed silicone cylinders (angioreactors) into nude mice. Angioreactors are filled with only 18 micro l of extracellular matrix premixed with or without angiogenic factors. Vascularization within angioreactors is quantified by the intravenous injection of fluorescein isothiocyanate (FITC)-dextran before their recovery, followed by spectrofluorimetry. Angioreactors examined by immunofluorescence show cells and invading angiogenic vessels at different developmental stages. The minimally detectable angiogenic response requires 9 days after implantation and >/=50 ng/ml (P < 0.01) of either fibroblast growth factor-2 or vascular endothelial growth factor. Characterization of this assay system demonstrates that the FITC-labeled dextran quantitation is highly reproducible and that levels of FITC-dextran are not significantly influenced by vascular permeability. DIVAA allows accurate dose-response analysis and identification of effective doses of angiogenesis-modulating factors in vivo. TNP-470 potently inhibits angiogenesis (EC(50) = 88 pmol/L) induced by 500 ng/ml of fibroblast growth factor-2. This inhibition correlates with decreased endothelial cell invasion. DIVAA efficiently detects differences in anti-angiogenic potencies of thrombospondin-1 peptides (25 micro mol/L) and demonstrates a partial inhibition of angiogenesis ( approximately 40%) in a matrix metalloprotease (MMP)-2-deficient mouse compared with that in wild-type animals. Zymography of angioreactors from MMP-deficient and control animals reveals quantitative changes in MMP expression. These results support DIVAA as an assay to compare potencies of angiogenic factors or inhibitors, and for profiling molecular markers of angiogenesis in vivo.


Subject(s)
Endothelium, Vascular/pathology , Neovascularization, Pathologic/physiopathology , Animals , Biological Assay , Capillary Permeability/physiology , Endothelial Growth Factors/pharmacology , Gelatinases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphokines/pharmacology , Mice , Neovascularization, Pathologic/pathology , Recombinant Proteins/pharmacology , Reproducibility of Results , Thrombospondin 1/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
Br J Haematol ; 120(3): 457-63, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12580960

ABSTRACT

Overexpression of manganese superoxide dismutase (MnSOD) has been postulated as one possible mechanism of protection from oxidative damage and free radicals. Doxorubicin treatment induces oxygen free radicals, leading to cytotoxicity and myelosuppression. The present study was performed to determine whether over-expression of MnSOD may play a role in resistance to doxorubicin. Retroviral constructs having the human MNSOD gene in the sense orientation and the neomycin phosphotransferase gene (NEOR) as a selectable marker were transduced into the human melanoma cell line A375 and the human histiocytic lymphoma cell line U937. Stably transduced A375 and U937 cells were subjected to 10-100 ng/ml doxorubicin for 24 h and compared with doxorubicin-treated A375 and U937 cells transduced with vector only. A colony forming assay was used to determine cell viability in semi-solid medium. Results demonstrated that wild-type A375 and U937 cells display low levels of endogenous MnSOD mRNA and protein, and are sensitive to doxorubicin treatment. In contrast, A375 and U937 cells transduced with the MNSOD gene consistently demonstrate increased colony formation in the presence of increasing concentrations of doxorubicin. MnSOD-transduced A375 and U937 cells also demonstrate increased MnSOD mRNA and protein levels when compared with wild type or those cells transduced with vector only. These results indicate that overexpression of MnSOD can enhance resistance to doxorubicin treatment.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Melanoma/enzymology , Superoxide Dismutase/metabolism , U937 Cells/enzymology , Blotting, Northern , Blotting, Western , Cell Survival/drug effects , Dose-Response Relationship, Drug , Free Radical Scavengers/metabolism , Gene Transfer Techniques , Genetic Vectors , Humans , Melanoma/pathology , Retroviridae/genetics , Superoxide Dismutase/genetics , Tumor Cells, Cultured , U937 Cells/pathology
14.
J Natl Cancer Inst ; 94(16): 1226-37, 2002 Aug 21.
Article in English | MEDLINE | ID: mdl-12189226

ABSTRACT

BACKGROUND: Adrenomedullin is a secreted peptide hormone with multiple activities. Several reports have indicated that adrenomedullin may be involved in tumor survival, but this has not been directly shown. Here we evaluate the in vitro and in vivo effects of adrenomedullin overexpression in human breast cancer cells. METHODS: The human breast cancer cell lines T47D and MCF7, both of which express low basal levels of adrenomedullin, were stably transfected with an expression construct that contained the coding region of the human adrenomedullin gene or with empty expression vector. Properties of the transfected cells were assessed by proliferation and apoptosis assays, in vitro and in vivo angiogenesis assays, cell migration experiments, and xenograft implants. The effect of synthetic adrenomedullin on human ovarian (ECV) cancer cell motility was also tested. Western blot analysis was used to compare expression levels of several genes whose products are associated with cell growth and regulation of apoptosis. RESULTS: T47D and MCF7 cells transfected with the adrenomedullin construct both expressed high levels of adrenomedullin mRNA and protein. Compared with cells transfected with empty vector, cells that overexpressed adrenomedullin displayed a more pleiotropic morphology, an increased angiogenic potential both in vitro and in vivo, and less apoptosis after serum deprivation. T47D and MCF7 cells did not display measurable motility, but ECV ovarian cancer cells treated with synthetic adrenomedullin were more motile than saline-treated ECV cells. Adrenomedullin-overexpressing T47D cells had higher levels of proteins involved in oncogenic signal transduction pathways (such as Ras, Raf, PKC, and MAPKp49) and lower levels of pro-apoptotic proteins (such as Bax, Bid, and caspase 8) than T47D cells transfected with empty vector. In a preliminary in vivo experiment, three of 10 nude mice injected with adrenomedullin-overexpressing T47D cells developed xenograft tumors, whereas none of the 10 nude mice injected with cells carrying the empty plasmid developed tumors. CONCLUSIONS: These results further support the role of adrenomedullin as a survival factor for tumors. Development of physiologically efficient inhibitors of adrenomedullin may prove useful in the clinical management of cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Peptides/metabolism , Adrenomedullin , Aorta , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Breast Neoplasms/blood supply , Cell Cycle , Cell Division , Cell Survival , DNA, Neoplasm/metabolism , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Neovascularization, Pathologic/metabolism , Peptides/genetics , Plasmids , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Transfection , Tumor Cells, Cultured , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...