Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Brain ; 139(Pt 6): 1762-82, 2016 06.
Article in English | MEDLINE | ID: mdl-27084575

ABSTRACT

The p75 neurotrophin receptor is important in multiple physiological actions including neuronal survival and neurite outgrowth during development, and after central nervous system injury. We have discovered a novel piperazine-derived compound, EVT901, which interferes with p75 neurotrophin receptor oligomerization through direct interaction with the first cysteine-rich domain of the extracellular region. Using ligand binding assays with cysteine-rich domains-fused p75 neurotrophin receptor, we confirmed that EVT901 interferes with oligomerization of full-length p75 neurotrophin receptor in a dose-dependent manner. Here we report that EVT901 reduces binding of pro-nerve growth factor to p75 neurotrophin receptor, blocks pro-nerve growth factor induced apoptosis in cells expressing p75 neurotrophin receptor, and enhances neurite outgrowth in vitro Furthermore, we demonstrate that EVT901 abrogates p75 neurotrophin receptor signalling by other ligands, such as prion peptide and amyloid-ß. To test the efficacy of EVT901 in vivo, we evaluated the outcome in two models of traumatic brain injury. We generated controlled cortical impacts in adult rats. Using unbiased stereological analysis, we found that EVT901 delivered intravenously daily for 1 week after injury, reduced lesion size, protected cortical neurons and oligodendrocytes, and had a positive effect on neurological function. After lateral fluid percussion injury in adult rats, oral treatment with EVT901 reduced neuronal death in the hippocampus and thalamus, reduced long-term cognitive deficits, and reduced the occurrence of post-traumatic seizure activity. Together, these studies provide a new reagent for altering p75 neurotrophin receptor actions after injury and suggest that EVT901 may be useful in treatment of central nervous system trauma and other neurological disorders where p75 neurotrophin receptor signalling is affected.


Subject(s)
Oligodendroglia/drug effects , Piperazines/pharmacology , Receptor, Nerve Growth Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cell Count , Cell Proliferation/drug effects , Cells, Cultured , Demyelinating Diseases/pathology , Dose-Response Relationship, Drug , Humans , Male , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligodendroglia/metabolism , Phosphorylation/drug effects , Primary Cell Culture , Radioligand Assay , Rats , Receptor, Nerve Growth Factor/biosynthesis , Receptor, trkA/metabolism , Recovery of Function
2.
Angiogenesis ; 19(3): 389-406, 2016 07.
Article in English | MEDLINE | ID: mdl-27234973

ABSTRACT

Anti-vascular endothelial growth factor (VEGF) therapies have improved clinical outcomes for patients with cancers and retinal vascular diseases. Three anti-VEGF agents, pegaptanib, ranibizumab, and aflibercept, are approved for ophthalmic indications, while bevacizumab is approved to treat colorectal, lung, and renal cancers, but is also used off-label to treat ocular vascular diseases. The efficacy of bevacizumab relative to ranibizumab in treating neovascular age-related macular degeneration has been assessed in several trials. However, questions persist regarding its safety, as bevacizumab can form large complexes with dimeric VEGF165, resulting in multimerization of the Fc domain and platelet activation. Here, we compare binding stoichiometry, Fcγ receptor affinity, platelet activation, and binding to epithelial and endothelial cells in vitro for bevacizumab and aflibercept, in the absence or presence of VEGF. In contrast to bevacizumab, aflibercept forms a homogenous 1:1 complex with each VEGF dimer. Unlike multimeric bevacizumab:VEGF complexes, the monomeric aflibercept:VEGF complex does not exhibit increased affinity for low-affinity Fcγ receptors, does not activate platelets, nor does it bind to the surface of epithelial or endothelial cells to a greater degree than unbound aflibercept or control Fc. The latter finding reflects the fact that aflibercept binds VEGF in a unique manner, distinct from antibodies not only blocking the amino acids necessary for VEGFR1/R2 binding but also occluding the heparin-binding site on VEGF165.


Subject(s)
Bevacizumab/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombinant Fusion Proteins/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/metabolism , Bevacizumab/adverse effects , Bevacizumab/therapeutic use , Cell Line , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Macular Degeneration/immunology , Macular Degeneration/metabolism , Macular Degeneration/therapy , Mice , Mice, Transgenic , Platelet Activation , Protein Binding , Protein Multimerization , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/therapeutic use , Thrombocytopenia/etiology , Thrombosis/etiology , Vascular Endothelial Growth Factor A/immunology
3.
Angiogenesis ; 15(3): 349-60, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22437266

ABSTRACT

Communication between endothelial cells and cardiomyocytes is critical for cardiac development and regeneration. However the mechanisms involved in these endothelial-cardiomyocyte interactions remain poorly understood. Nucleotides are released within the heart, especially under ischemia or pressure overload. The function of P2Y nucleotide receptors in cardiac development has never been investigated. Here we show that adult P2Y(4)-null mice display microcardia. P2Y(4) nucleotide receptor is expressed in cardiac endothelial cells but not in cardiomyocytes. Loss of P2Y(4) in cardiac endothelial cells strongly inhibits their growth, migration and PDGF-B secretion in response to UTP. Proliferation of microvessels and cardiomyocytes is reduced in P2Y(4)-null hearts early after birth, resulting in reduced heart growth. Our study uncovers mouse P2Y(4) receptor as an essential regulator of cardiac endothelial cell function, and illustrates the involvement of endothelial-cardiomyocyte interactions in post-natal heart development. We also detected P2Y(4) expression in human cardiac microvessels. P2Y(4) receptor could constitute a therapeutic target to regulate cardiac remodelling and post-ischemic revascularisation.


Subject(s)
Heart/growth & development , Receptors, Purinergic P2/physiology , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Knockout , Neovascularization, Physiologic , Reverse Transcriptase Polymerase Chain Reaction
4.
J Biol Chem ; 285(34): 26628-40, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20547770

ABSTRACT

Fibroblast growth factor (FGF) signaling regulates mammalian development and metabolism, and its dysregulation is implicated in many inherited and acquired diseases, including cancer. Heparan sulfate glycosaminoglycans (HSGAGs) are essential for FGF signaling as they promote FGF.FGF receptor (FGFR) binding and dimerization. Using novel organic synthesis protocols to prepare homogeneously sulfated heparin mimetics (HM), including hexasaccharide (HM(6)), octasaccharide (HM(8)), and decasaccharide (HM(10)), we tested the ability of these HM to support FGF1 and FGF2 signaling through FGFR4. Biological assays show that both HM(8) and HM(10) are significantly more potent than HM(6) in promoting FGF2-mediated FGFR4 signaling. In contrast, all three HM have comparable activity in promoting FGF1.FGFR4 signaling. To understand the molecular basis for these differential activities in FGF1/2.FGFR4 signaling, we used NMR spectroscopy, isothermal titration calorimetry, and size-exclusion chromatography to characterize binding interactions of FGF1/2 with the isolated Ig-domain 2 (D2) of FGFR4 in the presence of HM, and binary interactions of FGFs and D2 with HM. Our data confirm the existence of both a secondary FGF1.FGFR4 interaction site and a direct FGFR4.FGFR4 interaction site thus supporting the formation of the symmetric mode of FGF.FGFR dimerization in solution. Moreover, our results show that the observed higher activity of HM(8) relative to HM(6) in stimulating FGF2.FGFR4 signaling correlates with the higher affinity of HM(8) to bind and dimerize FGF2. Notably FGF2.HM(8) exhibits pronounced positive binding cooperativity. Based on our findings we propose a refined symmetric FGF.FGFR dimerization model, which incorporates the differential ability of HM to dimerize FGFs.


Subject(s)
Fibroblast Growth Factors/metabolism , Heparin/analogs & derivatives , Oligosaccharides/pharmacology , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Animals , Binding Sites , Cell Line , Humans , Mice , Multiprotein Complexes/biosynthesis , Oligosaccharides/chemistry , Protein Binding , Protein Multimerization , Structure-Activity Relationship
5.
J Clin Invest ; 115(2): 237-46, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15650770

ABSTRACT

Mechanisms regulating thrombus stabilization remain largely unknown. Here, we report that loss of any 1 of the Gas6 receptors (Gas6-Rs), i.e., Tyro3, Axl, or Mer, or delivery of a soluble extracellular domain of Axl that traps Gas6 protects mice against life-threatening thrombosis. Loss of a Gas6-R does not prevent initial platelet aggregation but impairs subsequent stabilization of platelet aggregates, at least in part by reducing "outside-in" signaling and platelet granule secretion. Gas6, through its receptors, activates PI3K and Akt and stimulates tyrosine phosphorylation of the beta3 integrin, thereby amplifying outside-in signaling via alphaIIbbeta3. Blocking the Gas6-R-alphaIIbbeta3 integrin cross-talk might be a novel approach to the reduction of thrombosis.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Platelet Aggregation/physiology , Signal Transduction/physiology , Thrombosis/metabolism , Animals , Integrin beta3/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor Protein-Tyrosine Kinases/administration & dosage , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/genetics , Thrombosis/drug therapy , Thrombosis/genetics , Thrombosis/pathology
6.
J Clin Invest ; 115(1): 118-27, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15630451

ABSTRACT

The angiogenic mechanism and therapeutic potential of PDGF-CC, a recently discovered member of the VEGF/PDGF superfamily, remain incompletely characterized. Here we report that PDGF-CC mobilized endothelial progenitor cells in ischemic conditions; induced differentiation of bone marrow cells into ECs; and stimulated migration of ECs. Furthermore, PDGF-CC induced the differentiation of bone marrow cells into smooth muscle cells and stimulated their growth during vessel sprouting. Moreover, delivery of PDGF-CC enhanced postischemic revascularization of the heart and limb. Modulating the activity of PDGF-CC may provide novel opportunities for treating ischemic diseases.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/drug effects , Ischemia/drug therapy , Ischemia/pathology , Neovascularization, Physiologic/drug effects , Platelet-Derived Growth Factor/pharmacology , Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Hindlimb/blood supply , Hindlimb/drug effects , Humans , Ischemia/chemically induced , Ischemia/metabolism , Lymphokines , Mice , Microcirculation/drug effects , Myocardium/metabolism , Myocardium/pathology , Phosphotyrosine/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Stem Cells/cytology
7.
Curr Opin Investig Drugs ; 6(3): 275-82, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15816504

ABSTRACT

The dual role of P2Y1 and P2Y12 receptors in platelet aggregation by ADP has been firmly established, based on the action of selective inhibitors, gene targeting in mice and human genetic evidence. Both of these receptor subtypes constitute targets for antithrombotic agents, and compounds with a dual action might also be of interest. However, the agents currently on the market (ticlopidine and clopidogrel), or known to be in development (cangrelor, AZD-6140 and prasugrel), all target the P2Y12 receptor. The thienopyridines (ticlopidine, clopidogrel and prasugrel) irreversibly inactivate the P2Y12 receptor via the covalent binding of an active metabolite generated in the liver, while the other compounds are competitive antagonists. Cangrelor, an ATP derivative, is suitable for intravenous perfusion, whereas AZD-6140 is in clinical development as an orally active agent.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Purinergic P2 Receptor Antagonists , Thrombosis/drug therapy , Ticlopidine/analogs & derivatives , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/pharmacology , Animals , Clopidogrel , Humans , Mice , Ticlopidine/chemistry , Ticlopidine/pharmacology
8.
Thromb Haemost ; 90(5): 839-43, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14597979

ABSTRACT

N-acetyl L-cysteine (NAC) is widely used to treat obstructive bronchopulmonary diseases. It has thiol reactive properties, accounting for its mucolytic activity. Clopidogrel is a potent antithrombotic compound, metabolised by the liver which generates an active metabolite containing a thiol reactive group, responsible for an irreversible interaction with the platelet P2Y(12) ADP receptor. The aim of this study was to determine if NAC interferes with the antiaggregating activity of clopidogrel. For this purpose, NAC (100 micro M) was incubated with platelets from rats treated or not with clopidogrel (5 mg/kg, PO, -2 h). Clopidogrel treatment strongly inhibited aggregation but this effect was not modified by NAC. In another experiment, a low concentration of the active metabolite of clopidogrel (0.3 micro g/ml) was incubated with platelets from men or rats, in the absence or presence of NAC (100 micro M). When stimulated by ADP (2.5 micro M), platelet aggregation was inhibited by the active metabolite when incubated alone. In the presence of NAC, the inhibition by the active metabolite was not modified, therefore clearly indicating that NAC cannot reduce the thiol reactive part of the active metabolite of clopidogrel and does not interfere with its anti-aggregating activity. Moreover, in rats treated for 5 days with NAC (150 mg/kg), the activity of clopidogrel (5 or 10 mg/kg) against ADP-induced platelet aggregation was neither inhibited nor increased. This demonstrates that the generation of the active metabolite of clopidogrel is not affected by NAC. In conclusion, we have found that NAC does not restore the "normal" properties of P2Y(12) on platelets from clopidogrel-treated animals, it does not interfere with the antiaggregating activity of the active metabolite of clopidogrel, and does not interfere with the generation of the active metabolite.


Subject(s)
Acetylcysteine/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology , Adenosine Diphosphate , Animals , Blood Platelets/drug effects , Cells, Cultured , Clopidogrel , Drug Interactions , Female , Humans , Male , Membrane Proteins/metabolism , Platelet Aggregation/drug effects , Rats , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y12 , Ticlopidine/metabolism
9.
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.

10.
PLoS One ; 9(11): e113215, 2014.
Article in English | MEDLINE | ID: mdl-25405900

ABSTRACT

RGS18 is a myeloerythroid lineage-specific regulator of G-protein signaling, highly expressed in megakaryocytes (MKs) and platelets. In the present study, we describe the first generation of a RGS18 knockout mouse model (RGS18-/-). Interesting phenotypic differences between RGS18-/- and wild-type (WT) mice were identified, and show that RGS18 plays a significant role in both platelet generation and function. RGS18 deficiency produced a gain of function phenotype in platelets. In resting platelets, the level of CD62P expression was increased in RGS18-/- mice. This increase correlated with a higher level of plasmatic serotonin concentration. RGS18-/- platelets displayed a higher sensitivity to activation in vitro. RGS18 deficiency markedly increased thrombus formation in vivo. In addition, RGS18-/- mice presented a mild thrombocytopenia, accompanied with a marked deficit in MK number in the bone marrow. Analysis of MK maturation in vitro and in vivo revealed a defective megakaryopoiesis in RGS18-/- mice, with a lower bone marrow content of only the most committed MK precursors. Finally, RGS18 deficiency was correlated to a defect of platelet recovery in vivo under acute conditions of thrombocytopenia. Thus, we highlight a role for RGS18 in platelet generation and function, and provide additional insights into the physiology of RGS18.


Subject(s)
Megakaryocytes/metabolism , Platelet Activation/physiology , RGS Proteins/genetics , RGS Proteins/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Blood Cell Count , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Phylogeny , Platelet Activation/genetics , Promoter Regions, Genetic/genetics , Serotonin/blood , Signal Transduction/genetics , Thrombosis/metabolism
11.
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
12.
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
13.
Mol Biol Cell ; 22(18): 3366-78, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21775632

ABSTRACT

Endothelial cells (ECs) are the primary sensors of variations in blood oxygen concentrations. They use the hypoxia-sensitive stabilization of the hypoxia-inducible factor-1α (HIF-1α) transcription factor to engage specific transcriptional programs in response to oxygen changes. The regulation of HIF-1α expression is well documented at the protein level, but much less is known about the control of its mRNA stability. Using small interfering RNA knockdown experiments, reporter gene analyses, ribonucleoprotein immunoprecipitations, and mRNA half-life determinations, we report a new regulatory mechanism of HIF-1α expression in ECs. We demonstrate that 1) sustained hypoxia progressively decreases HIF-1α mRNA while HIF-1α protein levels rapidly peak after 3 h and then slowly decay; 2) silencing the mRNA-destabilizing protein tristetraprolin (TTP) in ECs reverses hypoxia-induced down-regulation of HIF-1α mRNA; 3) the decrease in the half-life of Luciferase-HIF-1α-3'UTR reporter transcript that is observed after prolonged hypoxia is mediated by TTP; 4) TTP binds specifically to HIF-1α 3'UTR; and 5) the most distal AU-rich elements present in HIF-1α 3'UTR (composed of two hexamers) are sufficient for TTP-mediated repression. Finally, we bring evidence that silencing TTP expression enhances hypoxia-induced increase in HIF-1α protein levels with a concomitant increase in the levels of the carbonic anhydrase enzyme CA IX, thus suggesting that TTP physiologically controls the expression of a panel of HIF-1α target genes. Altogether, these data reveal a new role for TTP in the control of gene expression during the response of endothelial cell to hypoxia.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , RNA Stability , RNA, Messenger/metabolism , Tristetraprolin/metabolism , 3' Untranslated Regions , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Base Sequence , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Cell Hypoxia/genetics , Cell Line , Endothelium, Vascular/metabolism , Gene Expression Regulation , Gene Silencing , Genes, Reporter , Half-Life , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Tristetraprolin/genetics
14.
Thromb Haemost ; 105(4): 696-705, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21301779

ABSTRACT

Pharmacokinetic analyses of clopidogrel are hampered by the existence of multiple active metabolite isomers (H1 to H4) and their instability in blood. We sought to retest the pharmacodynamic activities of the four individual active metabolite isomers in vitro, with the ultimate aim of determining the isomers responsible for clopidogrel activity in vivo. In vitro activity was evaluated by measuring binding of [³³P]-2-methylthio-ADP on P2Y12-expressing Chinese hamster ovary (CHO) cells and human platelets in platelet-rich plasma (PRP). A stereoselective method that used reverse-phase ultra high-performance liquid chromatography (UHPLC) and tandem mass spectrometry (MS) was developed to measure individual concentrations of the stable 3'-methoxyacetophenone (MP) derivatives of H1-H4. The new method was used to analyze plasma samples from clopidogrel-treated subjects enrolled in a phase I clinical trial. In vitro binding assays confirmed the previously observed biological activity of H4 (IC50: CHO-P2Y12: 0.12 µM; PRP: 0.97 µM) and inactivity of H3, and demonstrated that H1 was also inactive. Furthermore, H2 demonstrated approximately half of the biological activity in vitro compared with H4. Optimisation of UHPLC conditions and MS collision parameters allowed the resolution and detection of the four derivatised active metabolite isomers (MP-H1 to MP-H4). The stereoselective assay was extensively validated, and was accurate and precise over the concentration range 0.5-250 ng/ml. Only MP-H3 and MP-H4 were quantifiable in incurred clinical samples. Based on in vitro pharmacodynamic data and found concentrations, the active metabolite isomer H4 is the only diastereoisomer of clinical relevance for documenting the pharmacokinetic profile of the active metabolite of clopidogrel.


Subject(s)
Blood Platelets/metabolism , Plasma/cytology , Receptors, Purinergic P2Y12/metabolism , Ticlopidine/analogs & derivatives , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Animals , Blood Platelets/cytology , Blood Platelets/drug effects , CHO Cells , Chromatography, High Pressure Liquid , Clopidogrel , Cricetinae , Cricetulus , Humans , Mass Spectrometry , Phosphorus Isotopes/chemistry , Plasma/chemistry , Product Surveillance, Postmarketing/methods , Protein Binding/drug effects , Receptors, Purinergic P2Y12/genetics , Sensitivity and Specificity , Stereoisomerism , Thionucleotides/chemistry , Thionucleotides/metabolism , Ticlopidine/analysis , Ticlopidine/chemistry , Ticlopidine/pharmacology , Transgenes/genetics
16.
Proc Natl Acad Sci U S A ; 103(29): 11069-74, 2006 Jul 18.
Article in English | MEDLINE | ID: mdl-16835302

ABSTRACT

P2Y12, a G protein-coupled receptor that plays a central role in platelet activation has been recently identified as the receptor targeted by the antithrombotic drug, clopidogrel. In this study, we further deciphered the mechanism of action of clopidogrel and of its active metabolite (Act-Met) on P2Y12 receptors. Using biochemical approaches, we demonstrated the existence of homooligomeric complexes of P2Y12 receptors at the surface of mammalian cells and in freshly isolated platelets. In vitro treatment with Act-Met or in vivo oral administration to rats with clopidogrel induced the breakdown of these oligomers into dimeric and monomeric entities in P2Y12 expressing HEK293 and platelets respectively. In addition, we showed the predominant association of P2Y12 oligomers to cell membrane lipid rafts and the partitioning of P2Y12 out of rafts in response to clopidogrel and Act-Met. The raft-associated P2Y12 oligomers represented the functional form of the receptor, as demonstrated by binding and signal transduction studies. Finally, using a series of receptors individually mutated at each cysteine residue and a chimeric P2Y12/P2Y13 receptor, we pointed out the involvement of cysteine 97 within the first extracellular loop of P2Y12 in the mechanism of action of Act-Met.


Subject(s)
Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Receptors, Purinergic P2/metabolism , Ticlopidine/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Clopidogrel , Humans , Mutation/genetics , Protein Binding , Rats , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y12 , Ticlopidine/metabolism , Ticlopidine/pharmacology
17.
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
18.
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
19.
Semin Thromb Hemost ; 31(2): 174-83, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15852221

ABSTRACT

Ticlopidine and clopidogrel belong to the same chemical family of thienopyridine adenosine diphosphate (ADP)-receptor antagonists. They have shown their efficacy as platelet antiaggregant and antithrombotic agents in many animal models, both ex vivo and in vivo. Although ticlopidine was discovered more than 30 years ago, it was only recently that the mechanism of action of ADP-receptor antagonists was characterized in detail. Ticlopidine and clopidogrel both behave in vivo as specific antagonists of P2Y (12), one of the ADP receptors on platelets. Metabolic steps that involve cytochrome P450-dependent pathways are required to generate the active metabolite responsible for this in vivo activity. The active moiety is a reactive thiol derivative that targets P2Y (12) on platelets. The interaction is irreversible, accounting for the observation that platelets are definitely antiaggregated, even if no active metabolite is detectable in plasma. The interaction is specific for P2Y (12); other purinoceptors such as P2Y (1) and P2Y (13) are spared. This results in inhibition of the binding of the P2Y (12) agonist 2-methylthio-ADP and the ADP-induced downregulation of adenylyl cyclase. Platelet aggregation is affected not only when triggered by ADP but also by aggregation inducers when used at concentrations requiring released ADP as an amplifier. The efficacy and safety of clopidogrel has been established in several large, randomized, controlled trials. The clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE) trial demonstrated the superiority of clopidogrel over acetylsalicylic acid (ASA) in patients at risk of ischemic events, including ischemic stroke, myocardial infarction (MI), and peripheral arterial disease. The clopidogrel in unstable angina to prevent recurrent ischemic events (CURE) trial showed a sustained, incremental benefit when clopidogrel was added to standard therapy (including ASA) in patients with unstable angina and non-Q-wave MI. The clopidogrel for the reduction of events during observation (CREDO) trial demonstrated the benefit of continuing clopidogrel (plus ASA) for 12 months, as opposed to 1 month, after percutaneous coronary intervention. The proven efficacy of clopidogrel, coupled with its favorable safety and tolerability profile, has prompted its evaluation in an extensive, ongoing clinical trial program that will help to further characterize the benefit of clopidogrel in patients with a range of atherothrombotic profiles.


Subject(s)
Arteriosclerosis/complications , Fibrinolytic Agents/therapeutic use , Membrane Proteins/antagonists & inhibitors , Platelet Aggregation Inhibitors/therapeutic use , Purinergic P2 Receptor Antagonists , Thrombophilia/drug therapy , Thrombosis/prevention & control , Ticlopidine/analogs & derivatives , Ticlopidine/therapeutic use , Adenosine Diphosphate/pharmacology , Animals , Aspirin/pharmacology , Aspirin/therapeutic use , Clinical Trials as Topic , Clopidogrel , Cohort Studies , Double-Blind Method , Drug Resistance , Fibrinolytic Agents/pharmacokinetics , Fibrinolytic Agents/pharmacology , Forecasting , Humans , Meta-Analysis as Topic , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Pyridines/pharmacology , Randomized Controlled Trials as Topic , Rats , Receptors, Purinergic P2 , Receptors, Purinergic P2Y12 , Thrombophilia/etiology , Thrombosis/etiology , Ticlopidine/pharmacokinetics , Ticlopidine/pharmacology
20.
Expert Opin Pharmacother ; 6(5): 755-64, 2005 May.
Article in English | MEDLINE | ID: mdl-15934902

ABSTRACT

Patients suffering a transient ischaemic attack (TIA) or ischaemic stroke (IS) have a high risk of recurrence. The inhibition of platelet function is effective in the reduction of secondary vascular events in patients with TIA or stroke. This is true for acetylsalicylic acid (ASA), clopidogrel, ticlopidine and the combination of ASA plus slow-release dipyridamole. This overview analyses the results of recent trials and presents ongoing or future trials with clopidogrel as well as the combination of clopidogrel plus ASA. Clopidogrel is superior to ASA in the prevention of vascular events in patients with IS, myocardial infarction (MI) or peripheral arterial disease (PAD). The difference is highest for high-risk patients such as diabetics, patients who underwent coronary bypass surgery and patients with a remote prior history of ischaemic events. A prediction model is presented which allows the identification of patients in whom clopidogrel is superior to ASA for the secondary prevention of stroke. The combination of clopidogrel and ASA is better than ASA alone in patients undergoing coronary stent implantations and patients with unstable angina or non-Q-wave MI. In high-risk patients with TIA or stroke, the addition of ASA to clopidogrel is not superior to ASA monotherapy but results in a higher rate of bleeding complications. The long-term combination therapy is currently investigated in several large trials in > 30,000 patients, with a large number of stroke patients.


Subject(s)
Stroke/prevention & control , Ticlopidine/analogs & derivatives , Clinical Trials as Topic/statistics & numerical data , Clopidogrel , Humans , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/therapeutic use , Risk Factors , Stroke/blood , Ticlopidine/chemistry , Ticlopidine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL