Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Res ; 73(8): 2457-2467, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23423981

ABSTRACT

Most patients with ovarian cancer are diagnosed late in progression and often experience tumor recurrence and relapses due to drug resistance. Surface expression of matrix metalloprotease (MMP)-14 on ovarian cancer cells stimulates a tumor-stromal signaling pathway that promotes angiogenesis and tumor growth. In a cohort of 92 patients, we found that MMP-14 was increased in the serum of women with malignant ovarian tumors. Therefore, we investigated the preclinical efficacy of a MMP-14 monoclonal antibody that could inhibit the migratory and invasive properties of aggressive ovarian cancer cells in vitro. MMP-14 antibody disrupted ovarian tumor-stromal communication and was equivalent to Avastin in suppressing blood vessel growth in mice harboring Matrigel plugs. These effects on angiogenesis correlated with downregulation of several important angiogenic factors. Furthermore, mice with ovarian cancer tumors treated with anti-MMP-14 monotherapy showed a marked and sustained regression in tumor growth with decreased angiogenesis compared with immunoglobulin G (IgG)-treated controls. In a model of advanced peritoneal ovarian cancer, MMP-14-dependent invasion and metastasis was effectively inhibited by intraperitoneal administration of monoclonal MMP-14 antibody. Together, these studies provide a preclinical proof-of-concept for MMP-14 targeting as an adjuvant treatment strategy for advanced ovarian cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Tumor Burden/drug effects , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/blood , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Docetaxel , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Matrix Metalloproteinase 14/blood , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase Inhibitors/administration & dosage , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Ovarian Neoplasms/genetics , Taxoids/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
2.
EMBO Mol Med ; 3(7): 370-84, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21591259

ABSTRACT

Sepsis is a deadly disease characterized by the inability to regulate the inflammatory-coagulation response in which the endothelium plays a key role. The cause of this perturbation remains poorly understood and has hampered the development of effective therapeutics. Matrix metalloproteases (MMPs) are involved in the host response to pathogens, but can also cause uncontrolled tissue damage and contribute to mortality. We found that human sepsis patients had markedly elevated plasma proMMP-1 and active MMP-1 levels, which correlated with death at 7 and 28 days after diagnosis. Likewise, septic mice had increased plasma levels of the MMP-1 ortholog, MMP-1a. We identified mouse MMP-1a as an agonist of protease-activated receptor-1 (PAR1) on endothelial cells. MMP-1a was released from endothelial cells in septic mice. Blockade of MMP-1 activity suppressed endothelial barrier disruption, disseminated intravascular coagulation (DIC), lung vascular permeability as well as the cytokine storm and improved survival, which was lost in PAR1-deficient mice. Infusion of human MMP-1 increased lung vascular permeability in normal wild-type mice but not in PAR1-deficient mice. These findings implicate MMP-1 as an important activator of PAR1 in sepsis and suggest that therapeutics that target MMP1-PAR1 may prove beneficial in the treatment of sepsis.


Subject(s)
Capillary Permeability/physiology , Inflammation/metabolism , Matrix Metalloproteinase 1/blood , Receptor, PAR-1/metabolism , Sepsis/metabolism , Sepsis/mortality , Animals , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, PAR-1/genetics , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism
3.
Methods Mol Biol ; 704: 81-9, 2011.
Article in English | MEDLINE | ID: mdl-21161631

ABSTRACT

Nitric oxide produced from nitric oxide synthases mediates various physiological and pathological events in biological systems. However, quantitative assessment of nitric oxide from biological sources remains a difficult task. Here we describe a procedure for the quantification of low levels of nitric oxide using a nitric oxide - selective electrochemical sensor. Nitric oxide is oxidized to nitrite and/or nitrate and accumulated in the aqueous media. First, nitrate in biological fluids or culture media is converted to nitrite by an enzymatic method. Nitrite is then chemically converted to equimolar NO in an acidic iodide bath, where nitric oxide is detected by the sensor. Using this method, the present study demonstrates siRNA -mediated suppression of nitric oxide synthase 3 leading to a significant decline of basal nitric oxide production in human umbilical vein endothelial cells. Basal nitric oxide production from HUVECs is also shown to be inhibited by N (G)-nitro-L: -arginine methyl ester but not by N (G)-nitro-D: -arginine methyl ester (D-NAME) D-NAME . The analytical method presented here provides a sensitive and convenient tool for measuring basal and stimulated nitric oxide production from biological sources.


Subject(s)
Chemistry Techniques, Analytical/methods , Electrochemistry/methods , Nitric Oxide/analysis , Cells, Cultured , Electrodes , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Nitrates/chemistry , Nitrates/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Nitrites/chemistry , Nitrites/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Transfection
4.
Methods Mol Biol ; 683: 259-75, 2011.
Article in English | MEDLINE | ID: mdl-21053136

ABSTRACT

G protein-coupled receptors (GPCR) are a superfamily of receptors that are vital in a wide array of physiological processes. Modulation of GPCR signaling has been an intensive area of therapeutic study, mainly due to the diverse pathophysiological significance of GPCRs. Pepducins are cell-penetrating lipidated peptides designed to target the intracellular loops of the GPCR of interest. Pepducins can function as agonists or antagonists of their cognate receptor, making them highly useful compounds for the study of GPCR signaling. Pepducins have been used to control platelet-dependent hemostasis and thrombosis, tumor growth, invasion, and angiogenesis, as well as to improve sepsis outcomes in mice. Pepducins have been successfully designed against a wide variety of GPCRs including the protease-activated receptors (PAR1, 2, 4), the chemokine receptors (CXCR1, 2, 4), the sphingosine-1-phosphate receptor (S1P3), the adrenergic receptor (ADRA1B), and have the potential to help reveal the functions of intractable GPCRs. Pharmacokinetic, pharmacodynamic, and biodistribution studies have showed that pepducins are widely distributed throughout the body except the brain and possess appropriate drug-like properties for use in vivo. Here, we discuss the delivery, pharmacology, and biodistribution of pepducins, as well as the effects of pepducins in models of inflammation, cardiovascular disease, cancer, and angiogenesis.


Subject(s)
Disease , Lipopeptides/pharmacology , Lipopeptides/pharmacokinetics , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Humans , Lipopeptides/chemistry , Lipopeptides/metabolism , Molecular Sequence Data , Protein Transport , Receptors, G-Protein-Coupled/chemistry
5.
Cancer Res ; 70(14): 5880-90, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20570895

ABSTRACT

Ovarian cancer is a lethal gynecologic malignancy that may benefit from new therapies that block key paracrine pathways involved in tumor-stromal interactions and tumor vascularity. It was recently shown that matrix metalloprotease-1 (MMP1) activation of the G protein-coupled receptor protease-activated receptor-1 (PAR1) is an important stimulator of angiogenesis and metastasis in peritoneal mouse models of ovarian cancer. In the present study, we tested the hypothesis that MMP1-PAR1 promotes angiogenesis through its paracrine control of angiogenic chemokine receptors. We found that MMP1-PAR1 activation induces the secretion of several angiogenic factors from ovarian carcinoma cells, most prominently interleukin (IL)-8, growth-regulated oncogene-alpha (GRO-alpha), and monocyte chemoattractant protein-1. The secreted IL-8 and GRO-alpha acts on endothelial CXCR1/2 receptors in a paracrine manner to cause robust endothelial cell proliferation, tube formation, and migration. A cell-penetrating pepducin, X1/2pal-i3, which targets the conserved third intracellular loop of both CXCR1 and CXCR2 receptors, significantly inhibited endothelial cell proliferation, tube formation, angiogenesis, and ovarian tumor growth in mice. Matrigel plugs mixed with MMP1-stimulated, OVCAR-4-conditioned media showed a dramatic 33-fold increase in blood vessel formation in mice. The X1/2pal-i3 pepducin completely inhibited MMP1-dependent angiogenesis compared with a negative control pepducin or vehicle. Conversely, a vascular endothelial growth factor-directed antibody, Avastin, suppressed angiogenesis in mice but, as expected, was unable to inhibit IL-8 and GRO-alpha-dependent endothelial tube formation in vitro. These studies identify a critical MMP1-PAR1-CXCR1/2 paracrine pathway that might be therapeutically targeted for ovarian cancer treatment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Matrix Metalloproteinase 1/metabolism , Ovarian Neoplasms/metabolism , Receptor, PAR-1/metabolism , Amino Acid Sequence , Animals , Cell Communication/physiology , Cell Line, Tumor , Chemokine CXCL1/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Interleukin-8/metabolism , Mice , Mice, Nude , Molecular Sequence Data , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Peptide Fragments/pharmacology , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Xenograft Model Antitumor Assays
6.
J Biol Chem ; 285(15): 11402-10, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20164183

ABSTRACT

Protease-activated receptor-1 (PAR1) is a G protein-coupled receptor that plays critical roles in cancer, angiogenesis, inflammation, and thrombosis. Proteolytic cleavage of the extracellular domain of PAR1 generates a tethered ligand that activates PAR1 in an unusual intramolecular mode. The signal emanating from the irreversibly cleaved PAR1 is terminated by G protein uncoupling and internalization; however, the mechanisms of PAR1 signal shut off still remain unclear. Using a yeast two-hybrid screen, we identified Bicaudal D1 (BicD1) as a direct interactor with the C-terminal cytoplasmic domain of PAR1. BICD was originally identified as an essential developmental gene associated with mRNA and Golgi-endoplasmic reticulum transport. We discovered a novel function of BicD1 in the modulation of G protein signaling, cell proliferation, and endocytosis downstream of PAR1. BicD1 and its C-terminal CC3 domain inhibited PAR1 signaling to G(q)-phospholipase C-beta through coiled-coil interactions with the cytoplasmic 8th helix of PAR1. Unexpectedly, BicD1 was also found to be a potent suppressor of PAR1-driven proliferation of breast carcinoma cells. The growth-suppressing effects of BicD1 required the ability to interact with the 8th helix of PAR1. Silencing of BicD1 expression impaired endocytosis of PAR1, and BicD1 co-localized with PAR1 and tubulin, implicating BicD1 as an important adapter protein involved in the transport of PAR1 from the plasma membrane to endosomal vesicles. Together, these findings provide a link between PAR1 signal termination and internalization through the non-G protein effector, BicD1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , GTP-Binding Proteins/chemistry , Receptors, Proteinase-Activated/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , COS Cells , Cell Proliferation , Chlorocebus aethiops , Cytoplasm/metabolism , Female , HeLa Cells , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid , Signal Transduction
7.
Arterioscler Thromb Vasc Biol ; 28(11): 1989-95, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18772493

ABSTRACT

OBJECTIVE: Recently, we have shown that shear stress regulates the angiogenic potential of endothelial cells in vitro by an Angiopoietin-2 (Ang2)-dependent mechanism; however its pathophysiological significance in vivo was not clear. We hypothesized that Ang2 plays an important role in blood flow recovery after arterial occlusion in vivo by regulating angiogenesis and arteriogenesis. METHODS AND RESULTS: C57Bl/6J mice underwent femoral artery ligation and were injected with a specific Ang2 inhibitor, L1-10, or vehicle for 10 days. Ang2 mRNA was upregulated at day 2, and Ang2 protein was upregulated at day 2, 5, and 7 in the ligated hindlimb. L1-10 treatment significantly blunted blood flow recovery. L1-10 decreased smooth muscle cell coverage of neovessels without affecting capillary density, suggesting a specific role for Ang2 in arteriogenesis. Mechanistically, L1-10 decreased expression of intercellular and vascular cell adhesion molecules as well as infiltrating monocytes/macrophages in the ischemic tissue. Although L1-10 had no effect on the number of CD11b+ cells (monocytes/macrophages) mobilized in the bone marrow, it maintained elevated numbers of circulating CD11b+ cells in the peripheral blood. CONCLUSIONS: These results suggest that Ang2 induced in ischemic tissue plays a critical role in blood flow recovery by stimulating inflammation and arteriogenesis.


Subject(s)
Angiopoietin-2/metabolism , Arterial Occlusive Diseases/complications , Endothelial Cells/metabolism , Inflammation/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Angiopoietin-1/metabolism , Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/genetics , Animals , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/physiopathology , CD11b Antigen/blood , Cells, Cultured , Collateral Circulation , Disease Models, Animal , Endothelial Cells/drug effects , Femoral Artery/surgery , Humans , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/metabolism , Ischemia/etiology , Ischemia/physiopathology , Ligation , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , RNA, Messenger/metabolism , Receptor, TIE-2/metabolism , Recombinant Fusion Proteins/pharmacology , Regional Blood Flow , Time Factors , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism
8.
Arterioscler Thromb Vasc Biol ; 27(10): 2150-6, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17673702

ABSTRACT

OBJECTIVE: Fluid shear stress plays a role in angiogenesis. Laminar shear stress (LS) promotes endothelial cell (EC) quiescence, whereas oscillatory shear stress (OS) promotes EC turnover and dysfunction, which could lead to pathological angiogenesis. We hypothesized that LS inhibits EC migration and tubule formation, 2 functions important in angiogenesis, by inhibiting the secretion of proangiogenic factors. METHODS AND RESULTS: Human umbilical vein ECs (HUVECs), human microvascular ECs (HMECs), or bovine aortic ECs (BAECs) were subjected to either LS (15 dyn/cm2) or OS (+/-5 dyn/cm2) for 24 hours and used in Matrigel tubule formation or scratch migration assays. Exposure of HUVECs, HMECs, but not BAECs, to LS inhibited tubule formation compared with OS. LS also inhibited migration of HUVECs and BAECs compared with OS. Angiopoietin-2 (Ang2), a known angiogenic protein, was found to be downregulated by LS both in cultured ECs and mouse aortas. Using Ang2 siRNA, Ang2 knockdown blocked OS-mediated migration and tubule formation and the LS-inhibited tubule formation was partially rescued by recombinant Ang2. CONCLUSIONS: Our data suggests that Ang2 produced by OS in ECs plays a critical role in migration and tubule formation, and may play an important role in diseases with disturbed flow and angiogenesis.


Subject(s)
Angiopoietin-2/metabolism , Cell Movement , Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Angiopoietin-2/genetics , Animals , Cattle , Cells, Cultured , Collagen/metabolism , Drug Combinations , Gene Expression Profiling , Humans , Laminin/metabolism , Neovascularization, Pathologic/physiopathology , Oligonucleotide Array Sequence Analysis , Protein Array Analysis , Proteoglycans/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Recombinant Proteins/metabolism , Stress, Mechanical , Time Factors
9.
Nitric Oxide ; 16(2): 306-12, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17056288

ABSTRACT

Nitric oxide (NO) produced from NO synthase(s) (NOS) is an important cell signaling molecule in physiology and pathophysiology. It remains challenging, however, to measure NO accurately and reproducibly in many cell types producing relatively low levels of NO from the enzymes such as endothelial NO synthase (eNOS). In the present study, we describe a very sensitive and convenient analytical method that affords measurement of 1 to 2 nM concentration of NO(x) (nitrite plus nitrate) in culture media. In the present study, we used an ultra-sensitive NO-selective electrochemical sensor (AmiNO700) in combination with a highly efficient nitrate conversion method, which coupled the nitrate reductase step with the glucose-6-phosphate dehydrogenase system. An aliquot of conditioned culture media was first treated with nitrate reductase, NADPH, glucose-6-phosphate dehydrogenase and glucose-6-phosphate to convert nitrate to nitrite quantitatively. The nitrite (that is present originally plus the reduced nitrate) was then reduced to equimolar NO in an acidic iodide bath while NO was being detected by the sensor. With this analytical method, we can quantitatively and reliably measure basal and stimulated NO release from cultured endothelial cells. We believe this improved assay should be useful in measuring a wide range of NO levels, especially the low but physiologically relevant levels, in many cell types.


Subject(s)
Electrochemistry/instrumentation , Nitrates/analysis , Nitric Oxide/chemistry , Nitrites/analysis , Animals , Calibration , Cattle , Cells, Cultured , Culture Media , Nitrates/blood , Nitrites/blood
10.
Circ Res ; 95(8): 773-9, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15388638

ABSTRACT

Atherosclerosis is an inflammatory disease occurring preferentially in arterial regions exposed to disturbed flow conditions including oscillatory shear stress (OS). OS exposure induces endothelial expression of bone morphogenic protein 4 (BMP4), which in turn may activate intercellular adhesion molecule-1 (ICAM-1) expression and monocyte adhesion. OS is also known to induce monocyte adhesion by producing reactive oxygen species (ROS) from reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, raising the possibility that BMP4 may stimulate the inflammatory response by ROS-dependent mechanisms. Here we show that ROS scavengers blocked ICAM-1 expression and monocyte adhesion induced by BMP4 or OS in endothelial cells (ECs). Similar to OS, BMP4 stimulated H2O2 and O2- production in ECs. Next, we used ECs obtained from p47phox-/- mice (MAE-p47-/-), which do not produce ROS in response to OS, to determine the role of NADPH oxidases. Similar to OS, BMP4 failed to induce monocyte adhesion in MAE-p47-/-, but it was restored when the cells were transfected with p47phox plasmid. Moreover, OS-induced O2- production was blocked by noggin (a BMP antagonist), suggesting a role for BMP. Furthermore, OS increased gp91phox (nox2) and nox1 mRNA levels while decreasing nox4. In contrast, BMP4 induced nox1 mRNA expression, whereas nox2 and nox4 were decreased or not affected, respectively. Also, OS-induced monocyte adhesion was blocked by knocking down nox1 with the small interfering RNA (siRNA). Finally, BMP4 siRNA inhibited OS-induced ROS production and monocyte adhesion. Together, these results suggest that BMP4 produced in ECs by OS stimulates ROS release from the nox1-dependent NADPH oxidase leading to inflammation, a critical early atherogenic step.


Subject(s)
Bone Morphogenetic Proteins/physiology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Gene Expression Regulation , Monocytes/cytology , NADH, NADPH Oxidoreductases/metabolism , Reactive Oxygen Species/metabolism , Stress, Mechanical , Animals , Antioxidants/pharmacology , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , Cell Adhesion , Cells, Cultured/metabolism , Humans , Hydrogen Peroxide/metabolism , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Mice , Mice, Inbred C57BL , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , NADPH Oxidases , Phosphoproteins/deficiency , Phosphoproteins/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/pharmacology , Rheology , Superoxides/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...