Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
Br J Cancer ; 112(3): 485-94, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25535734

ABSTRACT

BACKGROUND: Lung cancer remains the leading cause of cancer-related death, largely owing to the lack of effective treatments. A tumour vascular targeting strategy presents an attractive alternative; however, the molecular signature of the vasculature in lung cancer is poorly explored. This work aimed to identify novel tumour vascular targets in lung cancer. METHODS: Enzymatic digestion of fresh tissue followed by endothelial capture with Ulex lectin-coated magnetic beads was used to isolate the endothelium from fresh tumour specimens of lung cancer patients. Endothelial isolates from the healthy and tumour lung tissue were subjected to whole human genome expression profiling using microarray technology. RESULTS: Bioinformatics analysis identified tumour endothelial expression of angiogenic factors, matrix metalloproteases and cell-surface transmembrane proteins. Predicted novel tumour vascular targets were verified by RNA-seq, quantitative real-time PCR analysis and immunohistochemistry. Further detailed expression profiling of STEAP1 on 82 lung cancer patients confirmed STEAP1 as a novel target in the tumour vasculature. Functional analysis of STEAP1 using siRNA silencing implicates a role in endothelial cell migration and tube formation. CONCLUSIONS: The identification of cell-surface tumour endothelial markers in lung is of interest in therapeutic antibody and vaccine development.


Subject(s)
Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/blood supply , Lung Neoplasms/genetics , Molecular Targeted Therapy , Neovascularization, Pathologic/genetics , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Gene Expression Profiling , Genetic Association Studies/methods , Humans , Lung/blood supply , Lung/metabolism , Lung/pathology , Lung Neoplasms/drug therapy , Male , Microarray Analysis , Middle Aged , Neovascularization, Pathologic/drug therapy , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA
2.
Thromb Res ; 134(3): 693-703, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25064036

ABSTRACT

P2Y12 antagonism is a key therapeutic strategy in the management and prevention of arterial thrombosis. The objective of this study was to characterize the pharmacodynamic (PD) and pharmacokinetic (PK) properties of SAR216471, a novel P2Y12 receptor antagonist. SAR216471 blocks the binding of 2MeSADP to P2Y12 receptors in vitro (IC50=17 nM). This inhibition was shown to be reversible. It potently antagonized ADP-induced platelet aggregation in human and rat platelet-rich plasma (IC50=108 and 62 nM, respectively). It also inhibited platelet aggregation when blood was exposed to collagen or thromboxane A2. Its high selectivity was demonstrated against a large panel of receptors, enzymes, and ion channels. Despite its moderate bioavailability in rats, oral administration of SAR216471 resulted in a fast, potent, and sustained inhibition of platelet aggregation where the extent and duration of platelet inhibition were directly proportional to its circulating plasma levels. Pre-clinical study of SAR216471 in a rat shunt thrombosis model demonstrated a dose-dependent antithrombotic activity after oral administration (ED50=6.7 mg/kg). By comparison, ED50 values for clopidogrel, prasugrel and ticagrelor were 6.3, 0.35 and 2.6 mg/kg, respectively. Finally, the anti-hemostatic effect of SAR216471 and its competitors was investigated in a rat tail bleeding model, revealing a favorable safety profile of SAR216471. Together, these findings have established a reliable antiplatelet profile of SAR216471, and support its potential use in clinical practice as an alternative to currently available P2Y12 receptor antagonists.


Subject(s)
Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Indoles/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Pyridazines/pharmacology , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2/drug effects , Thrombosis/prevention & control , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/metabolism , Administration, Oral , Animals , Binding, Competitive , Biological Availability , Blood Platelets/metabolism , CHO Cells , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacokinetics , Fibrinolytic Agents/toxicity , Hemorrhage/chemically induced , Humans , Indoles/administration & dosage , Indoles/pharmacokinetics , Indoles/toxicity , Male , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/toxicity , Protein Binding , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Purinergic P2Y Receptor Antagonists/toxicity , Pyridazines/administration & dosage , Pyridazines/pharmacokinetics , Pyridazines/toxicity , Rats, Sprague-Dawley , Receptors, Purinergic P2/blood , Receptors, Purinergic P2Y12/blood , Signal Transduction/drug effects , Thionucleotides/metabolism , Thrombosis/blood , Transfection
3.
Oncogene ; 31(3): 293-305, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-21706054

ABSTRACT

Tumor endothelial markers (TEMs) that are highly expressed in human tumor vasculature compared with vasculature in normal tissue hold clear therapeutic potential. We report that the C-type lectin CLEC14A is a novel TEM. Immunohistochemical and immunofluorescence staining of tissue arrays has shown that CLEC14A is strongly expressed in tumor vasculature when compared with vessels in normal tissue. CLEC14A overexpression in tumor vessels was seen in a wide range of solid tumor types. Functional studies showed that CLEC14A induces filopodia and facilitates endothelial migration, tube formation and vascular development in zebrafish that is, CLEC14A regulates pro-angiogenic phenotypes. CLEC14A antisera inhibited cell migration and tube formation, suggesting that anti-CLEC14A antibodies may have anti-angiogenic activity. Finally, in endothelial cultures, expression of CLEC14A increased at low shear stress, and we hypothesize that low shear stress due to poor blood flow in the disorganized tumor vasculature induces expression of CLEC14A on tumor vessels and pro-angiogenic phenotypes.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/metabolism , Lectins, C-Type/metabolism , Neovascularization, Pathologic/metabolism , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement , Female , Humans , Lectins, C-Type/genetics , Liver Neoplasms/blood supply , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Neovascularization, Pathologic/genetics , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Pseudopodia/metabolism , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/metabolism , Zebrafish
4.
J Thromb Haemost ; 8(7): 1456-67, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20345705

ABSTRACT

SUMMARY: The glycoprotein VI (GPVI)-FcR gamma-chain complex initiates powerful activation of platelets by the subendothelial matrix proteins collagen and laminin through an immunoreceptor tyrosine-based activation motif (ITAM)-regulated signaling pathway. ITAMs are characterized by two YxxL sequences separated by 6-12 amino acids and are found associated with several classes of immunoglobulin (Ig) and C-type lectin receptors in hematopoietic cells, including Fc receptors. Cross-linking of the Ig GPVI leads to phosphorylation of two conserved tyrosines in the FcR gamma-chain ITAM by Src family tyrosine kinases, followed by binding and activation of the tandem SH2 domain-containing Syk tyrosine kinase and stimulation of a downstream signaling cascade that culminates in activation of phospholipase Cgamma2 (PLCgamma2). In contrast, the C-type lectin receptor CLEC-2 mediates powerful platelet activation through Src and Syk kinases, but regulates Syk through a novel dimerization mechanism via a single YxxL motif known as a hemITAM. CLEC-2 is a receptor for podoplanin, which is expressed at high levels in several tissues, including type 1 lung alveolar cells, lymphatic endothelial cells, kidney podocytes and some tumors, but is absent from vascular endothelial cells and platelets. In this article, we compare the mechanism of platelet activation by GPVI and CLEC-2 and consider their functional roles in hemostasis and other vascular processes, including maintenance of vascular integrity, angiogenesis and lymphogenesis.


Subject(s)
Hemostasis , Lectins, C-Type/physiology , Membrane Glycoproteins/physiology , Platelet Membrane Glycoproteins/physiology , Humans , Platelet Activation , Signal Transduction , Vascular Patency
5.
Br J Cancer ; 101(6): 957-66, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19738618

ABSTRACT

BACKGROUND: Considerable interest lies in the identification of novel anti-angiogenic compounds for cancer therapy. We have investigated whether dexrazoxane has anti-angiogenic properties and if so, the mechanism of the inhibition. METHODS: The phenotypic effects of dexrazoxane on endothelial cell behaviour was investigated both in vitro using human umbilical vein endothelial cells (HUVECs) in cell proliferation, migration, cell cycle and aortic ring assays; and in vivo using the mouse angiogenesis subcutaneous sponge assay. Custom angiogenesis pathway microarrays were used to identify differentially expressed genes in endothelial cells after treatment with dexrazoxane vs a control. The differentially expressed genes were validated using real-time RT-PCR and western blotting; and the functional effect of one induced gene was confirmed using siRNA technology. RESULTS: Treatment of endothelial cells with dexrazoxane resulted in a dose-response inhibition of cell growth lasting for up to 5 days after a single dose of the drug. Dexrazoxane was inhibitory in the aortic ring tube forming assay and strongly anti-angiogenic in vivo in the rodent subcutaneous sponge model. The anti-angiogenic effect in the sponge was seen after systemic injection into the tail vein as well as after direct injection of dexrazoxane into the sponge. Treatment of microvascular endothelial cells in vitro with subtoxic doses of dexrazoxane stimulated thrombospondin-1 (THBS-1) secretion. Knockdown of THBS-1 with siRNA removed the angiogenesis inhibition effect of dexrazoxane, which is consistent with the anti-angiogenic and vascular normalising properties of the drug being principally mediated by THBS-1. CONCLUSION: We show that dexrazoxane administered in small repeated doses is strongly anti-angiogenic and that this activity is mediated by induction of the anti-angiogenic THBS-1 in endothelial cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Razoxane/pharmacology , Thrombospondin 1/physiology , Animals , Endothelial Cells/drug effects , Humans , RNA, Small Interfering/genetics , Rats , Thrombospondin 1/antagonists & inhibitors
6.
Curr Cancer Drug Targets ; 8(6): 447-53, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18781891

ABSTRACT

Solid tumors are composed of the malignant cell itself (most commonly a carcinoma) and supporting cells that comprise the stroma. Significant stromal components include the extracellular matrix, supporting fibroblasts, vessels comprised of endothelium, pericytes and in some cases vascular smooth muscle, lymphatics and usually a major leukocyte infiltration. Indeed, macrophages may constitute up to 50% of the viable cells within the tumor. For many years, researchers have concentrated almost exclusively on the malignant carcinoma and looked for ways to either selectively kill or restrict its growth. In recent years the frustrating lack of advances in cytotoxic cancer therapy provoked a search for more novel strategies and foremost amongst these were anti-angiogenesis and vascular targeting. The purpose of this article is to illustrate how the stroma is now being pursued as an anti-cancer target. The article will briefly touch on anti-angiogenics that are now entering the clinic but concentrate on recent studies looking at vascular disrupting agents, stromal tumor fibroblasts and macrophages. Target identification is illustrated by the search for tumor endothelial markers. Finally, we draw attention to efforts to develop a cancer vaccine. The genetic instability and variation found in carcinoma cells made vaccination in the past a near impossibility. In contrast, genetically stable tumor endothelium with its unique accessibility to blood borne agents, together with recent advances in immunotherapy means that the possibility of a cancer vaccine now takes on a reality not previously recognised.


Subject(s)
Drug Delivery Systems , Neoplasms/drug therapy , Stromal Cells/pathology , Humans , Neoplasms/pathology
7.
J Thromb Haemost ; 6(10): 1697-706, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18647228

ABSTRACT

OBJECTIVE: In order to obtain a neutralizable antithrombotic, a chimeric molecule (SSR126517E) containing the sequence of a long-lasting antithrombin (AT)-dependent anti-factor Xa pentasaccharide, idraparinux, linked to a biotin molecule was synthesized and tested for anticoagulant and antithrombotic activity. METHODS: SSR126517E was tested in several models in vitro and in vivo for its pharmacological properties as well as its ability to be neutralized by avidin. RESULTS: SSR126517E displayed exactly the same properties as idraparinux. In vitro, SSR126517E had a very high affinity for AT (K(d) < 1 nm) and showed a potent anti-FXa effect and inhibition of thrombin generation with IC(50) values similar to those of idraparinux. Ex vivo, after intravenous administration to rats, SSR126517E produced a potent and long-lasting anti-FXa effect comparable to that obtained with idraparinux; as with idraparinux, the subcutaneous bioavailability was 100%. In vivo, SSR126517E was a potent antithrombotic in rat and mouse venous and arterial thrombosis models. Direct comparison in rats showed that SSR126517E was as active as idraparinux, when administered at the same molar dose. Furthermore, injection of avidin triggered the immediate elimination of SSR126517E from the bloodstream, resulting in complete neutralization of the antithrombotic activity of SSR126517E. CONCLUSIONS: These results show for the first time that coupling an oligosaccharide with biotin has no effect on the former's pharmacokinetic and pharmacologic properties and renders neutralization easy by injection of avidin.


Subject(s)
Anticoagulants/pharmacokinetics , Biotinylation , Fibrinolytic Agents/pharmacokinetics , Oligosaccharides/chemistry , Oligosaccharides/pharmacokinetics , Thrombosis/drug therapy , Animals , Antithrombin III/metabolism , Avidin/administration & dosage , Avidin/pharmacology , Factor Xa Inhibitors , Mice , Oligosaccharides/therapeutic use , Rats , Thrombin/biosynthesis
8.
Kidney Int ; 72(11): 1345-57, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17882151

ABSTRACT

Obesity is a major risk factor in the development of chronic renal failure. Rimonabant, a cannabinoid CB1 receptor antagonist, improves body weight and metabolic disorders; however, its effect on mortality and chronic renal failure associated with obesity is unknown. Obese Zucker rats received either rimonabant or vehicle for 12 months and were compared to a pair-fed but untreated group of obese rats. Mortality in the obese rats was significantly reduced by rimonabant along with a sustained decrease in body weight, transient reduction in food intake, and an increase in plasma adiponectin. This was associated with significant reduction in plasma total cholesterol, low-density lipoprotein cholesterol/high-density lipoprotein cholesterol ratio, triglycerides, glucose, norepinephrine, plasminogen activator inhibitor 1, and preservation of pancreatic weight and beta-cell mass index. The cannabinoid antagonist attenuated the increase in proteinuria, urinary N-acetylglucosaminidase excretion, plasma creatinine, and urea nitrogen levels while improving creatinine clearance. Renal hypertrophy along with glomerular and tubulointerstitial lesions were reduced by rimonabant. Although the drug did not modify hemodynamics, it normalized the pressor response to angiotensin II. Our study suggests that in a rat model of chronic renal failure due to obesity, rimonabant preserves renal function and increases survival.


Subject(s)
Kidney/physiology , Obesity/drug therapy , Obesity/metabolism , Piperidines/therapeutic use , Pyrazoles/therapeutic use , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Adiponectin/blood , Animals , Body Weight/drug effects , Body Weight/physiology , Disease Models, Animal , Eating/drug effects , Eating/physiology , Kidney/drug effects , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/prevention & control , Lipids/blood , Male , Obesity/complications , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Zucker , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/physiology , Rimonabant , Survival Analysis
9.
Diabetes Obes Metab ; 7(1): 65-72, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15642077

ABSTRACT

We investigated the effects of chronic treatment with the CB1 receptor antagonist rimonabant (10 mg/kg/day p.o. for 10 weeks) in mice with established obesity (5-month high-fat diet). Untreated obese mice showed a weight gain of 46% (45.0 +/- 0.6 g vs. 30.8 +/- 0.5 g) compared with age-matched animals fed a standard diet. Rimonabant treatment, commencing after 5-month high-fat diet, produced a marked and sustained decrease in body weight (34.5 +/- 0.8 g vs. 47.2 +/- 0.5 g in the high-fat vehicle group, p < 0.001). The anti-obesity effect of rimonabant was similar to that obtained by switching obese mice from high-fat diet to standard laboratory diet during 10 weeks (final weight 33.7 +/- 0.6 g) and was associated with only transient (14 days) reduction in energy intake. Serum leptin, insulin and glucose levels were markedly elevated in obese animals. Rimonabant treatment significantly reduced these elevations (leptin -81%, insulin -78%, glucose -67%, p < 0.001 in all cases vs. high-fat vehicle group). In addition, rimonabant treatment modestly but significantly increased serum adiponectin levels (+18%, p < 0.05 vs. high-fat vehicle group). Obese mice demonstrated abnormal serum lipid profiles. Although rimonabant did not modify high-density lipoprotein cholesterol (HDLc) and had modest effects on total cholesterol, it significantly reduced triglycerides and low-density lipoprotein cholesterol (LDLc) and, notably, increased the HDLc/LDLc ratio (12.4 +/- 0.8 vs. 7.9 +/- 0.2 in high-fat vehicle group, p < 0.001). Therefore, in a model of established obesity, chronic rimonabant treatment produces a marked and sustained decrease in body weight (equivalent to that achieved by dietary change) which is associated with favourable modifications in serum biochemical and lipid profiles.


Subject(s)
Anti-Obesity Agents/therapeutic use , Lipids/blood , Obesity/drug therapy , Piperidines/therapeutic use , Pyrazoles/therapeutic use , Adiponectin , Animals , Blood Glucose/analysis , Cholesterol/blood , Cholesterol, HDL/blood , Insulin/blood , Intercellular Signaling Peptides and Proteins/blood , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Rimonabant
10.
J Thromb Haemost ; 2(11): 2027-35, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15550035

ABSTRACT

BACKGROUND: The small bowel is a dose-limiting normal tissue in radiation therapy of malignancies in the abdomen and pelvis, as well as an important determinant of survival after non-therapeutic radiation exposure. Irradiation of normal tissues, including intestine, causes loss of vascular thromboresistance and upregulation of thrombin receptors. Radiation-induced endothelial dysfunction is thought to be involved in both early and delayed radiation responses. Hence, thrombin may be a potential target for ameliorating normal tissue radiation toxicity. OBJECTIVE: To assess direct thrombin inhibition as a protective strategy against small bowel radiation toxicity. METHODS: Rat small intestine was exposed to localized orthovoltage X-radiation. Recombinant hirudin, a direct thrombin inhibitor, or vehicle was infused from 2 days before irradiation to 14 days after irradiation. Structural, cellular, and molecular aspects of intestinal radiation injury were assessed at 2 weeks (early toxicity) and 26 weeks (chronic toxicity) after irradiation. RESULTS: Compared with unirradiated intestine, irradiated intestine showed increased expression of tissue factor, increased immunoreactivity for enzymatically active thrombin, and increased extravascular fibrin(ogen) deposition. Hirudin treatment significantly attenuated radiation-induced mucosal damage (P = 0.04), reactive intestinal wall thickening (P = 0.02), transforming growth factor-beta immunoreactivity levels (P = 0.0002), and collagen III deposition (P = 0.003). The differences between hirudin-treated and control rats were more pronounced at 2 weeks than at 26 weeks after irradiation. Hirudin treatment did not affect postradiation granulocyte infiltration. CONCLUSIONS: Short-term thrombin inhibition attenuates important aspects of intestinal radiation toxicity. Thrombin is a promising target for minimizing normal tissue injury after radiation therapy of cancer, as well as for protecting normal tissues from the adverse effects of non-therapeutic radiation exposure.


Subject(s)
Gamma Rays/adverse effects , Hirudins/pharmacology , Intestinal Diseases/drug therapy , Intestines/radiation effects , Radiation Tolerance/drug effects , Animals , Fibrin/analysis , Fibrin/genetics , Intestinal Diseases/etiology , Intestinal Diseases/pathology , Intestines/chemistry , Intestines/pathology , Male , RNA, Messenger/analysis , Radiation Injuries/drug therapy , Radiation-Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Thrombin/analysis , Thrombin/antagonists & inhibitors , Thrombin/genetics , Thromboplastin/analysis , Thromboplastin/genetics
11.
Gut ; 53(11): 1682-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15479692

ABSTRACT

BACKGROUND/AIM: Several lines of evidence incriminate the serine proteinase thrombin in liver fibrogenesis either through its procoagulant function or its signaling via cell-surface receptors. The aim of this study was therefore to evaluate the effect of thrombin inhibition on experimental liver fibrosis. METHODS: Fibrosis was induced in rats by administration of CCl4 for either three or seven weeks. Oral administration of the thrombin antagonist SSR182289 started one week after the start of CCl4 intoxication. Fibrosis and the area occupied by alpha smooth muscle actin (ASMA) positive cells were quantified with histomorphometry. Expression of fibrosis related genes was measured by real time RT-PCR. RESULTS: After three weeks of CCl4, treatment with SSR182289 did not significantly decrease the area of fibrosis but significantly decreased the area of ASMA positive cells by 22% (p = 0.03) and the expression of TIMP-1 mRNA by 52% (p = 0.02). There was no effect on gene expression of collagen I, MMP-2, or TIMP-2. After seven weeks of CCl4, treatment with SSR182289 resulted in a significant decrease in fibrosis (-30%, p = 0.04) and ASMA positive areas (-35%, p = 0.05). SSR182289 alone had no effect on the measured parameters. Additionally, it did not alleviate the acute toxicity of CCl4 as shown by measuring levels of serum aminotransferases and the area of necrosis. CONCLUSIONS: These data provide evidence that thrombin antagonism can reduce liver fibrogenesis. The early effect of SSR182289 on ASMA and TIMP-1 expression suggests that it is beneficial in reducing fibrogenic cell activation.


Subject(s)
Liver Cirrhosis, Experimental/physiopathology , Thrombin/physiology , Actins/metabolism , Aminopyridines/therapeutic use , Animals , Blood Coagulation/drug effects , Carbon Tetrachloride , Gene Expression Regulation/drug effects , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/drug therapy , Liver Cirrhosis, Experimental/pathology , Male , RNA, Messenger/genetics , Rats , Rats, Wistar , Sulfonamides/therapeutic use , Thrombin/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics
12.
Am J Physiol Heart Circ Physiol ; 287(1): H340-50, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15210453

ABSTRACT

A marked increase in plasma TNF-alpha has been described in patients with chronic heart failure (CHF). Nevertheless, little is known about the direct role of this cytokine early after myocardial infarction (MI) and its possible effects on the subsequent development of CHF. Wistar rats were subjected to permanent in vivo coronary artery ligation. At 5, 7, and 9 days after MI, cardiac function, passive compliance of the left ventricle (LV), and cardiac geometry were evaluated. The same model was used to perform pharmacological studies 7 days and 10 wk after MI in rats treated with monomeric recombinant human soluble TNF-alpha receptor type II (sTNF-RII, 40 microg/kg iv) or a placebo on day 3. Maximal alterations of cardiac function and geometry occurred 7 days after MI, which correlated chronologically with a peak of cardiac and serum TNF-alpha, as shown by immunohistochemistry and ELISA, respectively. sTNF-RII improved LV end-diastolic pressure under basal conditions and after volume overload 7 days and 10 wk after MI. Moreover, a significant leftward shift of the pressure-volume curve in the sTNF-RII-treated group 7 days after MI indicated a preservation of LV volume. Infarct expansion index was also significantly improved by sTNF-RII 7 days after MI (P < 0.01). Nevertheless, 10 wk after MI, geometric indexes and passive pressure-volume curves were not significantly improved by the treatment. In conclusion, TNF-alpha plays a major role in cardiac alterations 7 days after MI in rats and contributes to hemodynamic derangement, but not to cardiac remodeling, in subsequent CHF.


Subject(s)
Cardiac Output, Low/etiology , Heart/physiopathology , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antigens, CD/pharmacology , Blood Volume , Hemodynamics , Immunohistochemistry , Male , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Wistar , Receptors, Tumor Necrosis Factor , Receptors, Tumor Necrosis Factor, Type II , Tissue Distribution , Ventricular Function, Left
13.
J Thromb Haemost ; 2(6): 925-30, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15140128

ABSTRACT

The activity of SanOrg123781A, a new synthetic antithrombotic drug inhibiting both factor Xa and thrombin through antithrombin (AT), was compared to that of unfractionated heparin (UFH) and of the synthetic pentasaccharide (fondaparinux, SP) in an ex vivo arterial thrombosis model in the pig. Six groups of four pigs were administered intravenously with SanOrg123781A (1, 3, 10 and 30 nmol kg(-1)), UFH (30 nmol kg(-1)) or SP (30 nmol kg(-1)). In this arterial model in which platelet thrombus was formed on a thrombogenic surface under a constant high shear rate, UFH and SP had moderate antithrombotic effects while SanOrg123781A exhibited a strong, dose-dependent inhibitory activity on platelet adhesion and platelet thrombus formation. In contrast to UFH, SanOrg123781A did not modify the activated partial thromboplastin time (aPTT) even at 30 nmol kg(-1), but strongly inhibited thrombin generation. At the same dose, despite a lower antithrombotic activity than SanOrg123781A, UFH significantly affected all the coagulation parameters. Taken together, these results show that SanOrg123781A, due to its potent and selective antifactor Xa and antifactor IIa activities is a promising new antithrombotic agent even in arterial setting.


Subject(s)
Polysaccharides/pharmacokinetics , Thrombosis/drug therapy , Animals , Blood Platelets/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Factor Xa Inhibitors , Fondaparinux , Heparin/administration & dosage , Heparin/pharmacokinetics , Heparin/pharmacology , Partial Thromboplastin Time , Polysaccharides/administration & dosage , Polysaccharides/pharmacology , Prothrombin/antagonists & inhibitors , Swine , Thrombosis/prevention & control
14.
J Thromb Haemost ; 2(4): 629-36, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15102019

ABSTRACT

BACKGROUND AND OBJECTIVES: The purpose of this work was to investigate whether thrombolysis induced by recombinant tissue plasminogen activator (rt-PA) or streptokinase (SK) was enhanced in different rabbit models of thrombolysis by SSR182289A, a novel synthetic direct thrombin inhibitor which has been shown to possess potent antithrombotic properties in several experimental animal models. METHODS AND RESULTS: Human rt-PA alone (0.125 mg kg(-1) h(-1) for 2 h) induced a significant thrombolysis (18%, P < 0.05) of a venous-type thrombus in the rabbit jugular vein. Under these conditions, SSR182289A (3 mg kg(-1) i.v. bolus) inhibited 125I-fibrin accretion onto a preformed thrombus in the rabbit jugular vein by 72%, but was unable to induce thrombolysis on its own. However, coadministration of SSR182289A and rt-PA strongly enhanced rt-PA-induced thrombolysis (38.4%, P < 0.01). The effect of SSR182289A was further assessed in a model of thrombolysis of an electrical injury-induced, stable (occlusion duration > 2 h) thrombus formed in the rabbit femoral artery. Whereas local intra-arterial infusion of high doses of SSR182289A (3 mg kg(-1) h(-1) for 1 h) alone was able to restore flow, SK (12,000 U kg(-1) h(-1)) and a low dose of SSR182289A (0.3 mg kg(-1) h(-1)) were ineffective. However, intra-arterial coadministration of SSR182289A (0.3 mg kg(-1) h(-1)) and SK (12,000 U kg(-1) h(-1)) induced significant reflow (time to reflow was shortened by 34.7 +/- 7.5 min, P < 0.05). In the same model, systemic i.v. administration of high doses of SSR182289A (10 mg kg(-1) i.v. bolus) and rt-PA (1 mg kg(-1) h(-1)) alone did not induce any thrombolysis. However, the association of both compounds quickly (30 +/- 6 min) restored and maintained flow (duration > 2 h) in all animals. CONCLUSIONS: The present results show that bolus i.v. injection of SSR182289A is able to potentiate thrombolysis induced by two fibrinolytic agents whether the thrombus is of venous or arterial origin, thus suggesting that SSR182289A may be of use as an adjunct to thrombolysis.


Subject(s)
Aminopyridines/pharmacology , Fibrinolytic Agents/pharmacology , Sulfonamides/pharmacology , Thrombolytic Therapy/methods , Thrombosis/drug therapy , Aminopyridines/therapeutic use , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Femoral Artery , Fibrinolytic Agents/therapeutic use , Jugular Veins , Male , Rabbits , Streptokinase/pharmacology , Streptokinase/therapeutic use , Sulfonamides/therapeutic use , Tissue Plasminogen Activator/pharmacology , Tissue Plasminogen Activator/therapeutic use
15.
J Thromb Haemost ; 2(1): 135-46, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14717977

ABSTRACT

The P2Y12 ADP receptor is one of the major regulators of platelet activation and the target of antithrombotic thienopyridines (ticlopidine and clopidogrel). It has been recently cloned but the signaling pathways triggered by this receptor are still poorly documented. Here, we show that stimulation of the human P2Y12 receptor stably expressed in Chinese hamster ovary cells activates two major intracellular signaling mechanisms leading either to cell proliferation or to actin cytoskeleton reorganization. Both effects were blocked by the active metabolite of clopidogrel, a specific antagonist of P2Y12. The P2Y12-mediated stimulation of proliferation required the pertussis toxin-sensitive activation of PI3-kinase/Akt upstream of MAP-kinases. A partial contribution of a transactivation mechanism, through the tyrosine kinase receptor platelet-derived growth factor (PDGF)-R-beta, was also observed. Conversely, the P2Y12-mediated reorganization of the actin cytoskeleton was Gi-independent, requiring activation of RhoA and Rho-kinase. Our results provide new insights into the molecular basis of P2Y12-mediated intracellular signaling. These data may prove to be useful for a better understanding of the physiological role of P2Y12, particularly in platelets and glial cells which express this important therapeutic target.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Membrane Proteins/metabolism , Receptors, Purinergic P2/metabolism , Actins/metabolism , Adenosine Diphosphate/pharmacology , Animals , CHO Cells , Cell Division/drug effects , Cricetinae , Humans , MAP Kinase Signaling System , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Purinergic P2Y12 , Recombinant Proteins/metabolism , Signal Transduction , Thionucleotides/pharmacology , rhoA GTP-Binding Protein/metabolism
16.
Biochem Biophys Res Commun ; 310(3): 785-90, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-14550272

ABSTRACT

The peripheral benzodiazepine receptor (PBR), has been recently shown to play a key role in the regulation of the mitochondrial process leading to apoptosis, which occurs during cardiac ischemia. The present work shows that SSR180575, a novel PBR ligand of potential interest in pathological cardiovascular indications, irreversibly and specifically binds with high affinity on both rat heart mitochondria and on a cell line transfected with the human PBR (K(d)=1.95+/-0.22 and 4.58+/-0.83nM, respectively). In conclusion, SSR180575 is a specific and potent PBR ligand which irreversible binding to PBR appears of high interest in various therapeutic indications where apoptosis occurs.


Subject(s)
Acetamides/chemistry , Acetamides/pharmacokinetics , Benzodiazepines/chemistry , Indoles/chemistry , Indoles/pharmacokinetics , Animals , Blotting, Western , Cell Line , Humans , Inhibitory Concentration 50 , Ischemia , Kinetics , Ligands , Male , Mitochondria/metabolism , Myocardium/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Time Factors , Transfection
17.
Blood Coagul Fibrinolysis ; 14(6): 587-91, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12960613

ABSTRACT

On a previous model using Wessler's principle in the rat, we have demonstrated that a partial ligature of the inferior vena cava leading to a 40% increase in up-stream venous pressure was thrombogenic only in association with the infusion of low dose of thromboplastin (90 microg/kg). In these thrombogenic conditions, the infusion of pentasaccharide (Arixtra, fondaparinux) should lead to a strong inhibition of thrombus formation. Therefore, we performed on five groups of 10 rats: stasis alone (group S) with a 40% increase in up-stream venous pressure; stasis and thromboplastin (group ST90); and stasis, thromboplastin and pentasaccharide (groups SPT50, SPT100 and SPT250) at three different dosages (50, 100 and 250 microg/kg). The efficacy of pentasaccharide was measured according to the variations in up-stream venous pressure, thrombus weight and thrombin-antithrombin complexes levels. Only 250 microl/kg pentasaccharide significantly reduced the thrombus weight in comparison with group ST90 (5 mg versus 23.8 mg, P = 0.01) but it was not sufficient to induce a return to the basic state (5 mg versus 0.2 mg in group S, P = 0.049). Thrombin-antithrombin complex levels measured at the end of the experiment were significantly reduced in comparison with group ST90 (16.7 versus 57.8 mg, P = 0.01) and were not statistically different from group S (16.1 versus 16.6 mg, P = 0.65). In conclusion, in a very borderline model toward thrombogenesis, pentasaccharide was able to reduce thrombus weight and abolished biological hypercoagulability.


Subject(s)
Blood Pressure , Hemostasis , Polysaccharides/pharmacology , Thrombosis/prevention & control , Animals , Antithrombin III/analysis , Disease Models, Animal , Dose-Response Relationship, Drug , Fondaparinux , Male , Peptide Hydrolases/analysis , Polysaccharides/therapeutic use , Rats , Rats, Sprague-Dawley , Thrombophilia/drug therapy , Thromboplastin/pharmacology , Thrombosis/drug therapy , Treatment Outcome
18.
J Thromb Haemost ; 1(9): 1959-65, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12941037

ABSTRACT

Factor (F)Xa and thrombin bound to the clot during its formation contribute to the propensity of thrombi to activate the coagulation system. The aim of this work was to study the inhibition of clot-bound FXa and clot-bound thrombin by SanOrg123781A, a synthetic hexadecasaccharide that enhances the inhibition of thrombin and FXa by antithrombin (AT). SanOrg123781A, designed to exhibit low non-specific binding to proteins other than AT, was compared with heparin. In buffer, heparin and SanOrg123781A inhibited FXa and thrombin at similar concentrations [concentration inhibiting 50% (IC50) of Xa and IIa activity were, respectively: heparin 120 +/- 7 and 3 +/- 1 ng mL-1; SanOrg123781A 77 +/- 5 and 4 +/- 1 ng mL-1]. In human plasma, the activity of both compounds was reduced, although the activity of heparin was much more affected than that of SanOrg123781A (IC50 values for inhibition of FXa and FIIa activity were, respectively: heparin 100 +/- 5 and 800 +/- 40 ng mL-1; SanOrg123781A 10 +/- 5 and 30 +/- 3 ng mL-1). We demonstrated, in agreement with our previous results, that the procoagulant activity of the clot is essentially due to clot-bound FXa and to some extent to clot-bound thrombin. We showed that heparin and SanOrg123781A were able to inhibit fragment F1+2 generation induced by clot-bound FXa with IC50 values of 2 +/- 0.5 micro g mL-1 and 0.6 +/- 0.2 micro g mL-1, respectively. Both compounds also inhibited clot-bound thrombin activity, the IC50 values of heparin and SanOrg123781A being 1 +/- 0.01 micro g mL-1 and 0.1 +/- 0.1 micro g mL-1, respectively. Moreover, both heparin and SanOrg123781A significantly inhibited fibrinopeptide A generated by the action of clot-bound thrombin on fibrinogen but also by free thrombin generated from prothrombin by clot-bound FXa with IC50 values of 4 +/- 0.6 and 1 +/- 0.1 micro g mL-1, respectively. As with clot-bound enzymatic activities, SanOrg123781A was three times more active than heparin in vivo on fibrinogen accretion onto a pre-existing thrombus and as activators of recombinant tissue-type plasminogen activator-induced thrombolysis. In conclusion, due to the specific activities of SanOrg123781A, this compound is much more active than heparin in the presence of plasma proteins, on clot-bound enzymes and in in vivo models of thrombosis/thrombolysis.


Subject(s)
Blood Coagulation , Factor Xa/metabolism , Polysaccharides/pharmacology , Thrombin/metabolism , Animals , Antithrombin III/physiology , Factor Xa Inhibitors , Fibrinopeptide A/antagonists & inhibitors , Heparin/pharmacology , Humans , Inhibitory Concentration 50 , Male , Peptide Fragments/antagonists & inhibitors , Prothrombin/antagonists & inhibitors , Rabbits , Thrombin/antagonists & inhibitors , Thrombolytic Therapy , Thrombosis/drug therapy
19.
J Thromb Haemost ; 1(9): 2008-13, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12941044

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is a serious secondary event encountered in the clinical use of heparin. HIT results from the consumption of platelets that are immunologically activated by antibodies directed against complexes formed by platelet factor 4 (PF4) and sulfated polysaccharides that activate platelet aggregation, leading to paradoxical, life-threatening thrombosis. There is strong evidence that the ability of heparin and related compounds to induce HIT is closely linked to the structure of the polysaccharide, and particularly to its negative charge and to the length of the molecule. To test this hypothesis, we synthesized two sulfated oligosaccharides: SanOrg123781, a 16-mer, presenting two terminal charged domains separated by a 7-mer neutral linker, and SR121903, a highly sulfated 17-mer. Both of them displayed strong anti-factor (F) Xa and anti-FIIa activities but their affinities for PF4 were markedly different. SR121903 displaced PF4-bound heparin, whereas SanOrg123781 did not, underlining the importance of the charge of the molecule for the interaction with PF4. Platelet studies, in the presence of HIT serum, showed that SR121903 induced the secretion of platelet-dense granules (measured by the release of serotonin) whereas SanOrg123781 did not, a result in accordance with an absence of affinity of this molecule for PF4. These results were confirmed by measurements of platelet activation by flow cytometry (measured by annexin V binding, CD62 detection and activation of the GpIIb-IIIa complexes). In conclusion, we have demonstrated the importance of the charge of the polysaccharides in the HIT-induced platelet reactions measured by diverse methods, of which some are described for this purpose for the first time.


Subject(s)
Heparin/adverse effects , Oligosaccharides/pharmacology , Platelet Activation/drug effects , Thrombocytopenia/etiology , Cell Degranulation/drug effects , Dose-Response Relationship, Drug , Heparin/chemistry , Humans , Platelet Factor 4/metabolism , Polysaccharides/pharmacology , Static Electricity , Structure-Activity Relationship , Thrombocytopenia/blood , Thrombocytopenia/chemically induced
20.
J Pharmacol Exp Ther ; 304(2): 567-74, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538808

ABSTRACT

N-[3-[[[(1S)-4-(5-Amino-2-pyridinyl)-1-[[4-difluoromethylene)-1-piperidinyl]carbonyl]butyl]amino]sulfonyl][1,1'-biphenyl]-2-yl]acetamide hydrochloride (SSR182289A) is a novel, potent, and selective thrombin inhibitor. We have examined the antithrombotic properties of SSR182289A administered by i.v. and p.o. routes in several different animal thrombosis models in comparison with reference antithrombotic agents. Oral administration of SSR182289A produced dose-related antithrombotic effects in the following models; rat venous thrombosis (ED(50) 0.9 mg/kg p.o.), rat silk thread arterio-venous (AV) shunt (ED(50) 3.8 mg/kg p.o.), rat thromboplastin-induced AV shunt (ED(50) 3.1 mg/kg p.o.), rat carotid artery thrombosis (ED(200) 5.9 mg/kg p.o.), and rabbit venous thrombosis (ED(50) 7.5 mg/kg p.o.). Administered as an i.v. bolus, SSR182289A showed antithrombotic activity in the above models with ED(50)/ED(200) values in the range of 0.2 to 1.9 mg/kg i.v. SSR182289A increased rat tail transection bleeding time at doses > or =10 mg/kg p.o. In the rat thromboplastin-induced AV shunt model, SSR182289A 10 mg/kg p.o. produced marked antithrombotic effects at 30, 60, 120, and 240 min after administration. Hence, SSR182289A demonstrates potent oral antithrombotic properties in animal venous, AV-shunt, and arterial thrombosis models.


Subject(s)
Aminopyridines/pharmacology , Fibrinolytic Agents/pharmacology , Sulfonamides/pharmacology , Thrombin/antagonists & inhibitors , Administration, Oral , Aminopyridines/therapeutic use , Animals , Bleeding Time/statistics & numerical data , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrinolytic Agents/therapeutic use , Male , Rabbits , Rats , Rats, Sprague-Dawley , Sulfonamides/therapeutic use , Thrombin/physiology , Thrombosis/drug therapy , Thrombosis/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...