Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
Biosens Bioelectron ; 78: 58-66, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26594887

ABSTRACT

Detection of thrombin in plasma raises timely challenges to enable therapeutic management of thrombosis in patients under vital threat. Thrombin binding aptamers represent promising candidates as sensing elements for the development of real-time thrombin biosensors; however implementation of such biosensor requires the clear understanding of thrombin-aptamer interaction properties in real-like environment. In this study, we used Surface Plasmon Resonance technique to answer the questions of specificity and sensitivity of thrombin detection by the thrombin-binding aptamers HD1, NU172 and HD22. We systematically characterized their properties in the presence of thrombin, as well as interfering molecular species such as the thrombin precursor prothrombin, thrombin in complex with some of its natural inhibitors, nonspecific serum proteins, and diluted plasma. Kinetic experiments show the multiple binding modes of HD1 and NU172, which both interact with multiple sites of thrombin with low nanomolar affinities and show little specificity of interaction for prothrombin vs. thrombin. HD22, on the other hand, binds specifically to thrombin exosite II and has no affinity to prothrombin at all. While thrombin in complex with some of its inhibitors could not be recognized by any aptamer, the binding of HD1 and NU172 properties is compromised by thrombin inhibitors alone, as well as with serum albumin. Finally, the complex nature of plasma was overwhelming for HD1, but we define conditions for the thrombin detection at 10nM range in 100-fold diluted plasma by HD22. Consequently HD22 showed key advantage over HD1 and NU172, and appears as the only alternative to design an aptasensor.


Subject(s)
Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Thrombin/isolation & purification , Thrombosis/diagnosis , Animals , Binding Sites , Humans , Mice , Plasma/chemistry , Protein Binding , Serum Albumin/chemistry , Surface Plasmon Resonance , Thrombin/chemistry , Thrombosis/pathology
3.
J Biol Chem ; 290(6): 3405-17, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25384978

ABSTRACT

The formation of new vessels in the tumor, termed angiogenesis, is essential for primary tumor growth and facilitates tumor invasion and metastasis. Hypoxia has been described as one trigger of angiogenesis. Indeed, hypoxia, which is characterized by areas of low oxygen levels, is a hallmark of solid tumors arising from an imbalance between oxygen delivery and consumption. Hypoxic conditions have profound effects on the different components of the tumoral environment. For example, hypoxia is able to activate endothelial cells, leading to angiogenesis but also thereby initiating a cascade of reactions involving neutrophils, smooth muscle cells, and fibroblasts. In addition, hypoxia directly regulates the expression of many genes for which the role and the importance in the tumoral environment remain to be completely elucidated. In this study, we used a method to selectively label sialoglycoproteins to identify new membrane and secreted proteins involved in the adaptative process of endothelial cells by mass spectrometry-based proteomics. We used an in vitro assay under hypoxic condition to observe an increase of protein expression or modifications of glycosylation. Then the function of the identified proteins was assessed in a vasculogenesis assay in vivo by using a morpholino strategy in zebrafish. First, our approach was validated by the identification of sialoglycoproteins such as CD105, neuropilin-1, and CLEC14A, which have already been described as playing key roles in angiogenesis. Second, we identified several new proteins regulated by hypoxia and demonstrated for the first time the pivotal role of GLUT-1, TMEM16F, and SDF4 in angiogenesis.


Subject(s)
Neovascularization, Physiologic , Protein Processing, Post-Translational , Sialoglycoproteins/metabolism , Adaptation, Physiological , Animals , Anoctamins , Antigens, CD/genetics , Antigens, CD/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Hypoxia , Endoglin , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Human Umbilical Vein Endothelial Cells , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Proteome/chemistry , Proteome/metabolism , Proteomics/methods , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sialoglycoproteins/genetics , Zebrafish
4.
J Cell Physiol ; 230(1): 43-51, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24760775

ABSTRACT

Tumor angiogenesis is accompanied by vasculogenesis, which is involved in the differentiation and mobilization of human bone marrow cells. In order to further characterize the role of vasculogenesis in the tumor growth process, the effects of FGF2 on the differentiation of human bone marrow AC133(+) cells (BM-AC133(+)) into vascular precursors were studied in vitro. FGF2, like VEGFA, induced progenitor cell differentiation into cell types with endothelial cell characteristics. SSR128129E, a newly discovered specific FGFR antagonist acting by allosteric interaction with FGFR, abrogated FGF2-induced endothelial cell differentiation, showing that FGFR signaling is essential during this process. To assess the involvement of the FGF/FRGR signaling in vivo, the pre-clinical model of Lewis lung carcinoma (LL2) in mice was used. Subcutaneous injection of LL2 cells into mice induced an increase of circulating EPCs from peripheral blood associated with tumor growth and an increase of intra-tumoral vascular index. Treatment with the FGFR antagonist SSR128129E strongly decreased LL2 tumor growth as well as the intra-tumoral vascular index (41% and 50% decrease vs. vehicle-treated mice respectively, P < 0.01). Interestingly, SSR128129E treatment significantly decreased the number of circulating EPCs from the peripheral blood (53% inhibition vs. vehicle-treated mice, P < 0.01). These results demonstrate for the first time that the blockade of the FGF/FGFR pathway by SSR128129E reduces EPC recruitment during angiogenesis-dependent tumor growth. In this context, circulating EPCs could be a reliable surrogate marker for tumor growth and angiogenic activity.


Subject(s)
Carcinoma, Lewis Lung/blood supply , Hematopoietic Stem Cells/cytology , Indolizines/pharmacology , Neovascularization, Pathologic/pathology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , ortho-Aminobenzoates/pharmacology , AC133 Antigen , Animals , Antigens, CD/biosynthesis , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Differentiation/drug effects , Cell Line , Cell Movement , Endothelial Cells/cytology , Fibroblast Growth Factor 2/metabolism , Glycoproteins/biosynthesis , Humans , Mice , Mice, Inbred C57BL , Peptides , RNA, Messenger/biosynthesis , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction
5.
J Med Chem ; 57(17): 7293-316, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25075638

ABSTRACT

In the search of a potential backup for clopidogrel, we have initiated a HTS campaign designed to identify novel reversible P2Y12 antagonists. Starting from a hit with low micromolar binding activity, we report here the main steps of the optimization process leading to the identification of the preclinical candidate SAR216471. It is a potent, highly selective, and reversible P2Y12 receptor antagonist and by far the most potent inhibitor of ADP-induced platelet aggregation among the P2Y12 antagonists described in the literature. SAR216471 displays potent in vivo antiplatelet and antithrombotic activities and has the potential to differentiate from other antiplatelet agents.


Subject(s)
Indoles/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2 Receptor Antagonists/pharmacology , Pyridazines/pharmacology , Receptors, Purinergic P2Y12/metabolism , Acute Coronary Syndrome/prevention & control , Adenosine Diphosphate/pharmacology , Administration, Oral , Animals , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , Humans , Indoles/chemical synthesis , Indoles/metabolism , Injections, Intravenous , Male , Models, Chemical , Molecular Structure , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Aggregation Inhibitors/metabolism , Purinergic P2 Receptor Antagonists/chemical synthesis , Purinergic P2 Receptor Antagonists/metabolism , Pyridazines/chemical synthesis , Pyridazines/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y12/genetics , Thrombosis/prevention & control
6.
Cancers (Basel) ; 6(1): 472-90, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24589997

ABSTRACT

Myeloid derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) represent prominent components in cancer progression. We previously showed that inhibition of the VEGFR-3 pathway by SAR131675 leads to reduction of TAM infiltration and tumor growth. Here, we found that treatment with SAR131675 prevents the accumulation of immunosuppressive blood and splenic MDSCs which express VEGFR-3, in 4T1 tumor bearing mice. Moreover we showed that soluble factors secreted by tumor cells promote MDSCs proliferation and differentiation into M2 polarized F4/80+ macrophages. In addition, cell sorting and transcriptomic analysis of tumor infiltrating myeloid cells revealed the presence of a heterogeneous population that could be divided into 3 subpopulations: (i) immature cells with a MDSC phenotype (GR1+/CD11b+/F4/80-); (ii) "immuno-incompetent" macrophages (F4/80high/CD86neg/MHCIILow) strongly expressing M2 markers such as Legumain, CD206 and Mgl1/2 and (iii) "immuno-competent"-M1 like macrophages (F4/80Low/CD86+/MHCIIHigh). SAR131675 treatment reduced MDSCs in lymphoid organs as well as F4/80High populations in tumors. Interestingly, in the tumor SAR131675 was able to increase the immunocompetent M1 like population (F4/80low). Altogether these results demonstrate that the specific VEGFR-3 inhibitor SAR131675 exerts its anti tumoral activity by acting on different players that orchestrate immunosuppression and cancer progression in a tumoral context: MDSCs in peripheral lymphoid organs and TAMs infiltrating the tumor.

7.
J Med Chem ; 56(23): 9441-56, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24175584

ABSTRACT

Compound 15 (SAR107375), a novel potent dual thrombin and factor Xa inhibitor resulted from a rational optimization process. Starting from compound 14, with low factor Xa and modest anti-thrombin inhibitory activities (IC50's of 3.5 and 0.39 µM, respectively), both activities were considerably improved, notably through the incorporation of a neutral chlorothiophene P1 fragment and tuning of P2 and P3-P4 fragments. Final optimization of metabolic stability with microsomes led to the identification of 15, which displays strong activity in vitro vs factor Xa and thrombin (with Ki's of 1 and 8 nM, respectively). In addition 15 presents good selectivity versus related serine proteases (roughly 300-fold), including trypsin (1000-fold), and is very active (0.39 µM) in the thrombin generation time (TGT) coagulation assay in human platelet rich plasma (PRP). Potent in vivo activity in a rat model of venous thrombosis following iv and, more importantly, po administration was also observed (ED50 of 0.07 and 2.8 mg/kg, respectively). Bleeding liability was reduced in the rat wire coil model, more relevant to arterial thrombosis, with 15 (blood loss increase of 2-fold relative to the ED80 value) compared to rivaroxaban 2 and dabigatran etexilate 1a.


Subject(s)
Anticoagulants/chemical synthesis , Factor Xa Inhibitors , Fibrinolytic Agents/chemical synthesis , Piperazines/chemical synthesis , Sulfonamides/chemical synthesis , Thrombin/antagonists & inhibitors , Animals , Anticoagulants/pharmacology , Crystallography, X-Ray , Drug Design , Fibrinolytic Agents/pharmacology , Humans , Piperazines/pharmacology , Piperazines/therapeutic use , Rats , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Venous Thrombosis/drug therapy
8.
J Lab Autom ; 18(5): 411-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23813914

ABSTRACT

Angiogenesis is the biological process of generating new capillary blood vessels. It is a fundamental component of a number of normal (reproduction and wound healing) and pathological processes (diabetic retinopathy, rheumatoid arthritis, tumor growth, and metastasis). In vitro angiogenesis assays provide a platform for evaluating the effects of pro- or antiangiogenic compounds. One of the most informative assays is the endothelial cells capillary tube formation assay performed on a biological matrix. This assay is based on quantification of the stimulatory and inhibitory effects of various agents, which is estimated through the measurement of the pseudo-tubules network length. This standard measurement is usually carried out manually by trained operators but requires time, attention, and dedication to achieve a reasonable degree of accuracy. Moreover, the screening is operator dependent. In this article, we propose an automated procedure to evaluate the pseudo-tubule network lengths. We propose a series of image analysis procedures developed using a freely available image analysis software library. More than 800 images from 12 experiments were analyzed automatically and manually, and their results were compared to improve and validate the proposed image analysis procedure. The resulting image analysis software is currently running on a dedicated server, with comparable accuracy to manual measurements. Using this new automated procedure, we are able to treat 540 images, or three complete assays per hour.


Subject(s)
Cytological Techniques/methods , Endothelial Cells/drug effects , Image Processing, Computer-Assisted/methods , Neovascularization, Physiologic/drug effects , Humans , Software
9.
Cancer Cell ; 23(4): 477-88, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23597562

ABSTRACT

Receptor tyrosine kinases (RTK) are targets for anticancer drug development. To date, only RTK inhibitors that block orthosteric binding of ligands and substrates have been developed. Here, we report the pharmacologic characterization of the chemical SSR128129E (SSR), which inhibits fibroblast growth factor receptor (FGFR) signaling by binding to the extracellular FGFR domain without affecting orthosteric FGF binding. SSR exhibits allosteric properties, including probe dependence, signaling bias, and ceiling effects. Inhibition by SSR is highly conserved throughout the animal kingdom. Oral delivery of SSR inhibits arthritis and tumors that are relatively refractory to anti-vascular endothelial growth factor receptor-2 antibodies. Thus, orally-active extracellularly acting small-molecule modulators of RTKs with allosteric properties can be developed and may offer opportunities to improve anticancer treatment.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Small Molecule Libraries/pharmacology , Allosteric Regulation , Animals , Antibodies, Monoclonal/pharmacology , Arthritis, Experimental/drug therapy , Bone Resorption/drug therapy , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction , Xenograft Model Antitumor Assays
10.
Cancer Cell ; 23(4): 489-501, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23597563

ABSTRACT

The fibroblast growth factor (FGF)/fibroblast growth factor receptor (FGFR) signaling network plays an important role in cell growth, survival, differentiation, and angiogenesis. Deregulation of FGFR signaling can lead to cancer development. Here, we report an FGFR inhibitor, SSR128129E (SSR), that binds to the extracellular part of the receptor. SSR does not compete with FGF for binding to FGFR but inhibits FGF-induced signaling linked to FGFR internalization in an allosteric manner, as shown by crystallography studies, nuclear magnetic resonance, Fourier transform infrared spectroscopy, molecular dynamics simulations, free energy calculations, structure-activity relationship analysis, and FGFR mutagenesis. Overall, SSR is a small molecule allosteric inhibitor of FGF/FGFR signaling, acting via binding to the extracellular part of the FGFR.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Allosteric Regulation/drug effects , Binding, Competitive , Cell Growth Processes/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Conformation/drug effects , Protein Structure, Tertiary , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
11.
Mol Cancer Ther ; 11(8): 1637-49, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22584122

ABSTRACT

SAR131675 is a potent and selective VEGFR-3 inhibitor. It inhibited VEGFR-3 tyrosine kinase activity and VEGFR-3 autophosphorylation in HEK cells with IC(50) values of 20 and 45 nmol/L, respectively. SAR131675 dose dependently inhibited the proliferation of primary human lymphatic cells, induced by the VEGFR-3 ligands VEGFC and VEGFD, with an IC(50) of about 20 nmol/L. SAR131675 was found to be highly selective for VEGFR-3 versus 107 receptors, enzymes, ion channels, and 65 kinases. However, it was moderately active on VEGFR-2 with a VEGFR-3/VEGFR-2 ratio of about 10. SAR131675 had no antiproliferative activity on a panel of 30 tumors and primary cells, further showing its high specificity and indicating that SAR131675 is not a cytotoxic or cytostatic agent. SAR131675 was very well tolerated in mice and showed a potent antitumoral effect in several orthotopic and syngenic models, including mammary 4T1 carcinoma and RIP1.Tag2 tumors. Interestingly, it significantly reduced lymph node invasion and lung metastasis, showing its antilymphangiogenic activity in vivo. Moreover, treatment of mice before resection of 4T1 primary tumors was sufficient to prevent metastasis. Tumor-associated macrophages (TAM) play an important role in tumor growth and metastasis. The expression of VEGFR-3 on TAMs has been recently described. F4/80 immunostaining clearly showed that SAR131675 significantly reduced TAM infiltration and aggregation in 4T1 tumors. Taken together, SAR131675 is the first highly specific VEGFR-3-TK inhibitor described to date, displaying significant antitumoral and antimetastatic activities in vivo through inhibition of lymphangiogenesis and TAM invasion.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Naphthyridines/pharmacology , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Angiogenesis Inhibitors/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Cell Movement/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Enzyme Activation/drug effects , Female , Humans , Lymphangiogenesis/drug effects , Lymphocytes/drug effects , Lymphocytes/metabolism , Macrophages/drug effects , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Naphthyridines/administration & dosage , Neoplasm Metastasis , Neovascularization, Physiologic/drug effects , Protein Kinase Inhibitors/administration & dosage , Tumor Burden/drug effects , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Xenograft Model Antitumor Assays , Zebrafish
12.
J Reprod Immunol ; 76(1-2): 17-22, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17467060

ABSTRACT

In this review, we summarize the results of a number of our recent in vitro and in vivo experiments demonstrating that, in addition to the immunoregulatory functions, soluble HLA-G molecules also affect endothelial cell activity. We have found that soluble HLA-G1 (also designated HLA-G5) inhibits endothelial cell proliferation, migration and tubule formation, and this occurred through binding to the CD160 receptor and via an apoptotic pathway. Moreover, we have demonstrated that soluble HLA-G1 blocks in vivo rabbit corneal neoangiogenesis. Although it cannot be excluded that other soluble HLA class I molecules may have similar effects, as soluble forms of HLA-G are being produced by trophoblast cells at the maternal-fetal interface during early gestation, we discuss how such anti-angiogenic properties of soluble HLA-G1 may locally influence uterine vascular remodeling.


Subject(s)
Antigens, CD/metabolism , HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Neovascularization, Physiologic , Receptors, Immunologic/metabolism , Angiogenesis Modulating Agents/metabolism , Animals , Endothelial Cells/physiology , Female , GPI-Linked Proteins , HLA-G Antigens , Humans , Placenta/blood supply , Placenta/immunology , Pregnancy , Trophoblasts/immunology , Uterus/blood supply
13.
Blood ; 108(8): 2608-15, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16809620

ABSTRACT

HLA-G is a major histocompatibility complex class Ib molecule whose constitutive tissue distribution is restricted mainly to trophoblast cells at the maternal-fetal interface during pregnancy. In this study, we demonstrated the ability of the soluble HLA-G1 (sHLA-G1) isoform to inhibit fibroblast growth factor-2 (FGF2)-induced capillary-like tubule formation. Using a rabbit corneal neovascularization model, we further showed that sHLA-G1 inhibits FGF2-induced angiogenesis in vivo. We also demonstrated that sHLA-G1 induces endothelial cell apoptosis through binding to BY55/CD160, a glycosylphosphatidylinositolanchored receptor expressed by endothelial cells. Furthermore, we showed that the specific CL1-R2 anti-CD160 monoclonal antibody mimics sHLA-G1-mediated inhibition of endothelial cell tube formation and induction of apoptosis. Thus, the engagement of CD160 in endothelial cells may be essential for the inhibition of angiogenesis. sHLA-G1/CD160-mediated antiangiogenic property may participate in the vascular remodeling of maternal spiral arteries during pregnancy, and, given that we found that CD160 is strongly expressed in the vasculature of a murine tumor, it offers an attractive therapeutic target for preventing pathologic neovascularization.


Subject(s)
Antigens, CD/metabolism , Endothelial Cells/drug effects , HLA Antigens/metabolism , HLA Antigens/pharmacology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/pharmacology , Neovascularization, Physiologic/drug effects , Receptors, Immunologic/metabolism , Amino Acid Sequence , Animals , Antigens, CD/genetics , Apoptosis/drug effects , Apoptosis/immunology , Apoptosis/physiology , Cells, Cultured , Cornea/blood supply , Cornea/drug effects , Endothelial Cells/cytology , Endothelial Cells/immunology , Fibroblast Growth Factor 2/metabolism , GPI-Linked Proteins , HLA-G Antigens , Humans , In Vitro Techniques , Mice , Molecular Sequence Data , Rabbits , Receptors, Immunologic/genetics , Solubility
14.
Blood ; 108(4): 1243-50, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16621967

ABSTRACT

Neuropilin 2 (NRP2) is a receptor for the vascular endothelial growth factor (VEGF) and the semaphorin (SEMA) families, 2 unrelated ligand families involved in angiogenesis and neuronal guidance. NRP2 specifically binds VEGF-A and VEGF-C, although the biological relevance of these interactions in human endothelial cells is poorly understood. In this study, we show that both VEGF-A and VEGF-C induce the interaction of NRP2 with VEGFR-2. This interaction correlated with an enhancement of the VEGFR-2 phosphorylation threshold. Overexpression of NRP2 in primary human endothelial cells promoted cell survival induced by VEGF-A and VEGF-C. In contrast, SEMA3F, another ligand for NRP2, was able to inhibit human endothelial cell survival and migration induced by VEGF-A and VEGF-C. Moreover, a siRNA targeting specifically NRP2 was a potent inhibitor of human endothelial cell migration induced by VEGF-A and VEGF-C. Thus, our data indicate that NRP2 acts as a coreceptor that enhances human endothelial cell biological responses induced by VEGF-A and VEGF-C.


Subject(s)
Cell Movement/physiology , Endothelial Cells/physiology , Neuropilin-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Gene Expression , Humans , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Neuropilin-2/antagonists & inhibitors , Protein Binding/drug effects , Protein Binding/physiology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/pharmacology
15.
Blood ; 105(1): 139-44, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15388575

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is a complication of heparin therapy caused by antibodies against a complex of platelet factor 4 and heparin. Fondaparinux (Arixtra) is a new synthetic selective factor Xa inhibitor. We performed a serologic study to determine the cross-reactivity of HIT sera with fondaparinux. Using a prospective, blinded study design, 39 clinically and serologically confirmed sera from patients with HIT and 15 control sera were sent to 3 different laboratories, each of which specialized in a particular HIT assay. These include the serotonin release assay, heparin-induced platelet agglutination assay, and platelet aggregation assay. Two of 82 assays (2.4%) performed in the presence of control sera were positive, both with unfractionated heparin. In the presence of HIT sera, 75 of 94 (79.8%) evaluable assays were positive with unfractionated heparin; fondaparinux was significantly (P < .001) less reactive than unfractionated heparin, only 3 of 91 evaluable assays (3.3%) being positive. Using flow cytometry, unlike unfractionated heparin, fondaparinux did not induce the binding of PAC1 and anti-CD62 monoclonal antibodies or of annexin V to platelets with HIT sera. Together, these results suggest that fondaparinux is nonreactive to HIT sera and raise the possibility that the drug may be used for prophylaxis and treatment of thrombosis in patients with a history of HIT.


Subject(s)
Autoantibodies/immunology , Heparin/immunology , Heparin/pharmacology , Platelet Activation/drug effects , Polysaccharides/pharmacology , Flow Cytometry , Fondaparinux , Humans , Serotonin/blood , Single-Blind Method , Thrombocytosis/blood , Thrombocytosis/chemically induced , Thrombocytosis/complications , Thrombocytosis/drug therapy
16.
Thromb Haemost ; 92(5): 929-39, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15543318

ABSTRACT

The abilities of three synthetic oligosaccharides to accelerate antithrombin inhibition of ten clotting or fibrinolytic proteinases were compared with those of unfractionated, fractionated high-affinity and low-molecular-weight heparins. The results show that the anticoagulant effects of the latter three heparins under conditions approximating physiologic are exerted almost exclusively by acceleration of the inactivation of thrombin, factor Xa and factor IXa to near diffusion-controlled rate constants of approximately 10(6) - 10(7) M(-1).s(-1). All other proteinases are inhibited with at least 20-fold lower rate constants. The anti-coagulant ability of the synthetic regular (fondaparinux) and high-affinity (idraparinux) pentasaccharides is due to a common mechanism, involving acceleration of only factor Xa inhibition to rate constants of approximately 10(6) M(-1).s(-1) . A synthetic hexadecasaccharide, containing both the pentasaccharide sequence and a proteinase binding site, exerts its anticoagulant effect by accelerating antithrombin inactivation of both thrombin and factor Xa to rate constants of approximately 10(6) - 10(7) M(-1).s(-1), although thrombin appears to be the more important target. In contrast, factor IXa inhibition is appreciably less stimulated. The conformational change of antithrombin induced both by the pentasaccharides and longer heparins contributes substantially, approximately 150-500-fold, to accelerating the inactivation of factors Xa, IXa and VIIa and moderately, approximately 50-fold, to that of factor XIIa and tissue plasminogen activator inhibition. The bridging effect due to binding of antithrombin and proteinase to the same, long heparin chain is dominating, approximately 1000-3000-fold, for thrombin inhibition and is appreciably smaller, although up to approximately 250-350-fold, for the inactivation of factors IXa and XIa. These results establish the proteinase targets of heparin derivatives currently used in or considered for thrombosis therapy and give new insights into the mechanism of heparin acceleration of antithrombin inhibition of proteinases.


Subject(s)
Antithrombin III/drug effects , Hemostasis/drug effects , Heparin/pharmacology , Oligosaccharides/pharmacology , Serine Endopeptidases/drug effects , Antithrombin III/isolation & purification , Antithrombin III/metabolism , Blood Coagulation/drug effects , Blood Coagulation Factors/drug effects , Blood Coagulation Factors/isolation & purification , Fibrinolysis/drug effects , Fondaparinux , Heparin/chemistry , Humans , Kinetics , Oligosaccharides/chemistry , Polysaccharides/chemistry , Polysaccharides/pharmacology , Protein Binding , Serine Endopeptidases/isolation & purification , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology
17.
Biochem Biophys Res Commun ; 324(2): 909-15, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15474514

ABSTRACT

VEGFR-3 is essential for vascular development and maintenance of lymphatic vessel's integrity. Little is known about its cooperative effect with other receptors of the same family. Contrary to VEGFR-2, stimulation of VEGFR-3 by VEGF-C and -D failed to enhance its phosphorylation either in HEK293T or in PAE cells. These ligands were unable to induce angiogenesis of PAEC expressing VEGFR-3 alone. In the presence of VEGFR-2, VEGF-C and -D induced heterodimerization of VEGFR-3 with VEGFR-2. This heterodimerization was associated with enhanced VEGFR-3 phosphorylation and subsequent cellular responses as evidenced by the formation of capillary-like structures in PAE cells and proliferation of primary human endothelial cells expressing both receptors. Taken together, these results show for the first time that VEGFR-3 needs to be associated to VEGFR-2 to induce ligand-dependent cellular responses.


Subject(s)
Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-3/chemistry , Blotting, Western , Carbazoles/pharmacology , Cell Line , Cell Proliferation , Cells, Cultured , Dimerization , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Humans , Immunoprecipitation , Ligands , Neovascularization, Pathologic , Peptides/chemistry , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Vascular Endothelial Growth Factor C/chemistry , Vascular Endothelial Growth Factor D/chemistry
18.
J Cell Physiol ; 200(3): 351-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15254962

ABSTRACT

Tumor growth and metastasis require the generation of new blood vessels, a process known as neo-angiogenesis. Recent studies have indicated that early tumor vascularization is characterized by the differentiation and mobilization of human bone marrow cells. Vascular endothelial growth factor-A (VEGF-A) is one of the growth factors, which enhances their differentiation into endothelial cells, but little is known about the implication of the VEGF-receptor tyrosine kinases and about the implication of the VEGF-R co-receptor, neuropilin-1, in this process. In this context, the identification of the molecular pathways that support the proliferation and differentiation of vascular stem and progenitor cells was investigated in order to define the pharmaceutical targets involved in tissue vascularization associated with this process. For this purpose, an in vitro model of differentiation of human bone marrow AC133+ (BM-AC133+) cells into vascular precursors was used. In this work, we have demonstrated for the first time that the effect of VEGF-A on BM-AC133+ cells relies on an early action of VEGF-A on the expression of its tyrosine kinase receptors followed by an activation of a VEGF-R2/neuropilin-1-dependent signaling pathway. This signaling promotes the differentiation of BM-AC133+ cells into endothelial precursor cells, followed by the proliferation of these differentiated cells. Altogether, these results strongly suggest that VEGF inhibitors, acting at the level of VEGF-R2 and/or neuropilin-1, by inhibiting differentiation and proliferation of these cells, could be potentially active compounds to prevent progenitor cells to be involved in tumor angiogenesis leading to tumor growth.


Subject(s)
Bone Marrow Cells/physiology , Cell Differentiation , Hematopoietic Stem Cells/physiology , Neuropilin-1/metabolism , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Cell Adhesion , Cell Division , Cell Movement , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Neovascularization, Pathologic/prevention & control , Platelet Endothelial Cell Adhesion Molecule-1 , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Time Factors
19.
Bioorg Med Chem ; 12(7): 1713-30, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15028263

ABSTRACT

SSR182289A 1 is the result of a rational optimisation process leading to an orally active thrombin inhibitor. The structure incorporates an original 2-(acetylamino)-[1,1'-biphenyl]-3-sulfonyl N-terminal motif, a central l-Arg surrogate carrying a weakly basic 3-amino-pyridine, and an unusual 4-difluoropiperidine at the C-terminus. Its synthesis is convergent and palladium catalysis has been employed for the construction of the key C-C bonds: Suzuki coupling for the bis-aryl fragment and Sonogashira reaction for the delta- bond of the central amino-acid chain. The compound is a potent inhibitor of thrombin's activities in vitro and demonstrates potent oral anti-thrombotic potencies in three rat models of thrombosis. The observed in vitro potency could be rationalized through the examination of the interactions within the SSR182289A 1 - thrombin crystal structure. SSR182289A 1, has been therefore selected for further development.


Subject(s)
Aminopyridines/pharmacology , Sulfonamides/pharmacology , Thrombin/antagonists & inhibitors , Administration, Oral , Aminopyridines/chemical synthesis , Animals , Azetidines/pharmacology , Benzylamines , Blood Coagulation/drug effects , Crystallography, X-Ray , Disease Models, Animal , Humans , Hydrogen Bonding , Male , Models, Molecular , Molecular Structure , Platelet Aggregation/drug effects , Rats , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Thrombin/physiology , Thrombosis/drug therapy , Thrombosis/physiopathology , Thrombosis/prevention & control
20.
J Pharmacol Exp Ther ; 309(1): 235-40, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14718579

ABSTRACT

SanOrg123781A is a synthetic hexadecasaccharide that displays antithrombin-dependent inhibition of factor Xa and thrombin and potent antithrombotic effects. The antithrombotic activity of SanOrg123781A has been studied in a new mouse model of arterial thrombosis, where thrombus formation was induced by the application of an electrical current to the adventitial surface of a carotid artery. In this model, antiplatelet agents such as the ADP-receptor antagonist clopidogrel (30 mg/kg, p.o. 2 h before stimulation) and the GpIIb/IIIa antagonist SR121566A [3-(N-[4-(4-[amino(imino)methyl]phenyl)-1,3-thiazol-2-yl]-N-[1-(carboxymethyl)piperidin-4-yl]amino)propionic acid, trihydrochloride] (0.3 mg/kg, i.v. 5 min before stimulation) strongly prolonged the time to occlusion (TTO) (761 and 473% increases, respectively), whereas aspirin was devoid of antithrombotic activity. Standard heparin (2 mg/kg, i.v.), the low molecular weight heparin enoxaparin (20 mg/kg, i.v.), and the synthetic, antithrombin-dependent inhibitor of factor Xa fondaparinux (10 mg/kg, i.v.) were also active in this model (742, 707, and 602% TTO increases, respectively). Interestingly, SanOrg123781A was active at much lower doses than the other oligosaccharides (554% increase in TTO at 0.3 mg/kg, i.v. 5 min before stimulation). Low doses of SanOrg123781A administered in combination with low doses of clopidogrel led to a marked increase in TTO, which was statistically more important than the additive effects of the two compounds given alone. These results indicate that SanOrg123781A exerts a potent antithrombotic activity in a mouse model of arterial thrombosis when compared with reference compounds and show that the combination of SanOrg123781A with clopidogrel leads to a marked synergistic antithrombotic effect.


Subject(s)
Carotid Artery Injuries/drug therapy , Carotid Artery Thrombosis/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Polysaccharides/therapeutic use , Ticlopidine/analogs & derivatives , Ticlopidine/therapeutic use , Animals , Clopidogrel , Drug Synergism , Electric Injuries , Male , Mice , Mice, Inbred BALB C , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Polysaccharides/pharmacology , Thrombosis/metabolism , Ticlopidine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...