17 research outputs found

    Effets cellulaires et voies de signalisation activés par le facteur anticoagulant, la protéine S, sur les cellules endothéliales (implication lors de l'angiogenèse)

    Get PDF
    L'angiogenèse est un processus physiologique qui correspond à la formation de nouveaux vaisseaux sanguins à partir d'un réseau vasculaire préexistant et est régulée par l'équilibre entre les facteurs endogènes pro- et anti-angiogéniques. La rupture de cet équilibre est associée à de nombreuses pathologies dont l'ischémie, la rétinopathie ou encore la progression tumorale. Etant donné que les cellules endothéliales, principal type cellulaire composant les vaisseaux sanguins expriment les récepteurs à activité tyrosine kinase du facteur anticoagulant, la protéine S, Tyro3, Axl et Mer et produisent de la protéine S, l'objectif de ce travail est d'étudier le rôle, de la protéine S dans l'angiogenèse. Dans la première partie de ce travail, nous avons montré in vivo que la protéine S inhibe l'angiogenèse induite par les facteurs pro-angiogéniques (VEGFA et FGF2). Parallèlement, nous avons observé in vitro une inhibition par la protéine S de la prolifération et de la migration des cellules endothéliales induites par le VEGFA. Cet effet est corrélé à l'inhibition par la protéine S des voies de signalisation des MAP-Kinases et de la phosphatidylinositol 3-kinase (PIK3) induites par le VEGFA. Nous avons ensuite mis en évidence, par l'utilisation d'inhibiteurs pharmacologiques et de petits ARNs interférents, que la protéine S inhibe, via l'activation du récepteur Mer et le recrutement de la protéine phosphatase SHP2, l'activation du VEGFR2, le principal récepteur du VEGFA. Dans la deuxième partie, nous avons montré de manière intéressante que le rôle joué par la protéine S lors de l'angiogenèse est plus complexe, puisqu'elle est capable d'activer directement la voie de signalisatioAngiogenesis is a physiological process that leads to new blood vessel formation and is regulated by a balance between pro-and anti-angiogenic endogenous factors. Disruption of this balance leads to many pathologies such as ischemia, retinopathies or tumor growth. Because endothelial cells, the main cellular type composing blood vessels, produce the anticoagulant factor, protein S and express its tyrosine kinase receptors Tyro3, Axl and Mer, we investigated the implication of protein S in angiogenesis. In the first part of this work, we demonstrated that protein S inhibits pro-angiogenic factors (VEGFA and FGF2)-induced angiogenesis in vivo. We also observed an inhibition of VEGFA-dependent endothelial cell proliferation and migration induced by protein S. These effects were correlated with protein S induced inhibition of VEGFA-dependent MAP-Kinases (Erk1, Erk2) and phosphatidylinositol 3-kinase (PIK3) activation. Furthermore, we demonstrated, using pharmacological inhibitors and small interfering RNAs, that protein S inhibits VEGFA-induced VEGFR-2 activation through Mer receptor activation and SHP2 protein phosphatase recruitment. In the second part, we demonstrated that, protein S on its own, is able to induce MAP-kinases pathway activation and endothelial cells proliferation. These cellular and molecular effects involved Mer receptor and SHP2 protein activation and required protein kinase SRC recruitment. Our results describe for the first time that protein S is an endogenous regulator for angiogenesis both in vitro and in vivo and may form the framework for the use of protein S as part of an anti-angiogenic treatment.POITIERS-SCD-Bib. électronique (861949901) / SudocSudocFranceF

    Lymphatic and Immune Cell Cross-Talk Regulates Cardiac Recovery After Experimental Myocardial Infarction

    Get PDF
    Objective: Lymphatics play an essential pathophysiological role in promoting fluid and immune cell tissue clearance. Conversely, immune cells may influence lymphatic function and remodeling. Recently, cardiac lymphangiogenesis has been proposed as a therapeutic target to prevent heart failure after myocardial infarction (MI). We investigated the effects of gene therapy to modulate cardiac lymphangiogenesis post-MI in rodents. Second, we determined the impact of cardiac-infiltrating T cells on lymphatic remodeling in the heart. Approach and Results: Comparing adenoviral versus adeno-associated viral gene delivery in mice, we found that only sustained VEGF (vascular endothelial growth factor)-C(C156S)therapy, achieved by adeno-associated viral vectors, increased cardiac lymphangiogenesis, and led to reduced cardiac inflammation and dysfunction by 3 weeks post-MI. Conversely, inhibition of VEGF-C/-D signaling, through adeno-associated viral delivery of soluble VEGFR3 (vascular endothelial growth factor receptor 3), limited infarct lymphangiogenesis. Unexpectedly, this treatment improved cardiac function post-MI in both mice and rats, linked to reduced infarct thinning due to acute suppression of T-cell infiltration. Finally, using pharmacological, genetic, and antibody-mediated prevention of cardiac T-cell recruitment in mice, we discovered that both CD4(+)and CD8(+)T cells potently suppress, in part through interferon-gamma, cardiac lymphangiogenesis post-MI. Conclusions: We show that resolution of cardiac inflammation after MI may be accelerated by therapeutic lymphangiogenesis based on adeno-associated viral gene delivery of VEGF-C-C156S. Conversely, our work uncovers a major negative role of cardiac-recruited T cells on lymphatic remodeling. Our results give new insight into the interconnection between immune cells and lymphatics in orchestration of cardiac repair after injury.Peer reviewe

    Contribution à l'étude écotoxicologique des eaux de rejets agricoles en milieu marin : tests larvaires sur l'huître Crassostrea gigas

    No full text
    L'étude menée dans ce rapport concerne la toxicité des eaux de rejets agricoles à proximité des exploitations conchylicoles. Cette étude s'est tenue au cours du mois de mai 2004, dans le marais de Moëze-Brouage en Charente Maritime qui représente de façon significative le problème agro-conchylicole puisque le marais se situe entre une zone de production agricole intensive et la première région productrice d' huître en France (le bassin Marennes-Oléron). Le test utilisé pour quantifier la toxicité des eaux de rejets agricoles est le test larves d' huîtres basé sur le pourcentage d'anomalies larvaires obtenues suite à l'exposition des embryons de Crassostrea gigas (obtenus par fécondation et sélection des géniteurs) avec un échantillon d'eau de mer à analyser. Les résultats obtenus au cours de cette étude montrent les limites de ce test par rapport à la dépendance des facteurs environnementaux

    Cellular effects and signaling pathways activated by the anticoagulant factor, protein S, on endothelial cells in the angiogenic process

    No full text
    L'angiogenèse est un processus physiologique qui correspond à la formation de nouveaux vaisseaux sanguins à partir d'un réseau vasculaire préexistant et est régulée par l'équilibre entre les facteurs endogènes pro- et anti-angiogéniques. La rupture de cet équilibre est associée à de nombreuses pathologies dont l'ischémie, la rétinopathie ou encore la progression tumorale. Etant donné que les cellules endothéliales, principal type cellulaire composant les vaisseaux sanguins expriment les récepteurs à activité tyrosine kinase du facteur anticoagulant, la protéine S, Tyro3, Axl et Mer et produisent de la protéine S, l'objectif de ce travail est d'étudier le rôle, de la protéine S dans l'angiogenèse. Dans la première partie de ce travail, nous avons montré in vivo que la protéine S inhibe l'angiogenèse induite par les facteurs pro-angiogéniques (VEGFA et FGF2). Parallèlement, nous avons observé in vitro une inhibition par la protéine S de la prolifération et de la migration des cellules endothéliales induites par le VEGFA. Cet effet est corrélé à l'inhibition par la protéine S des voies de signalisation des MAP-Kinases et de la phosphatidylinositol 3-kinase (PIK3) induites par le VEGFA. Nous avons ensuite mis en évidence, par l'utilisation d'inhibiteurs pharmacologiques et de petits ARNs interférents, que la protéine S inhibe, via l'activation du récepteur Mer et le recrutement de la protéine phosphatase SHP2, l'activation du VEGFR2, le principal récepteur du VEGFA. Dans la deuxième partie, nous avons montré de manière intéressante que le rôle joué par la protéine S lors de l'angiogenèse est plus complexe, puisqu'elle est capable d'activer directement la voie de signalisatioAngiogenesis is a physiological process that leads to new blood vessel formation and is regulated by a balance between pro-and anti-angiogenic endogenous factors. Disruption of this balance leads to many pathologies such as ischemia, retinopathies or tumor growth. Because endothelial cells, the main cellular type composing blood vessels, produce the anticoagulant factor, protein S and express its tyrosine kinase receptors Tyro3, Axl and Mer, we investigated the implication of protein S in angiogenesis. In the first part of this work, we demonstrated that protein S inhibits pro-angiogenic factors (VEGFA and FGF2)-induced angiogenesis in vivo. We also observed an inhibition of VEGFA-dependent endothelial cell proliferation and migration induced by protein S. These effects were correlated with protein S induced inhibition of VEGFA-dependent MAP-Kinases (Erk1, Erk2) and phosphatidylinositol 3-kinase (PIK3) activation. Furthermore, we demonstrated, using pharmacological inhibitors and small interfering RNAs, that protein S inhibits VEGFA-induced VEGFR-2 activation through Mer receptor activation and SHP2 protein phosphatase recruitment. In the second part, we demonstrated that, protein S on its own, is able to induce MAP-kinases pathway activation and endothelial cells proliferation. These cellular and molecular effects involved Mer receptor and SHP2 protein activation and required protein kinase SRC recruitment. Our results describe for the first time that protein S is an endogenous regulator for angiogenesis both in vitro and in vivo and may form the framework for the use of protein S as part of an anti-angiogenic treatment

    Epigenetic regulation of endothelial-cell-mediated vascular repair

    No full text
    International audienceMaintenance of vascular integrity is essential for the prevention of vascular disease and for recovery following cardiovascular, cerebrovascular and peripheral vascular events including limb ischemia, heart attack and stroke. Endothelial stem/progenitor cells have recently gained considerable interest due to their potential use in stem cell therapies to mediate revascularization after ischemic injury. Therefore, there is an urgent need to understand fundamental mechanisms regulating vascular repair in specific cell types to develop new beneficial therapeutic interventions. In this review, we highlight recent studies demonstrating that epigenetic mechanisms (including post-translational modifications of DNA and histones as well as non-coding RNA-mediated processes) play essential roles in the regulation of endo-thelial stem/progenitor cell functions through modifying chromatin structure. Furthermore, we discuss the potential of using small molecules that modulate the activities of epigenetic enzymes to enhance the vascular repair function of endothelial cells and offer insight on potential strategies that may accelerate clinical applications

    The vitamin K–dependent anticoagulant factor, protein S, inhibits multiple VEGF-A–induced angiogenesis events in a Mer- and SHP2-dependent manner

    No full text
    International audienceProtein S is a vitamin K-dependent glyco-protein, which, besides its anticoagulant function, acts as an agonist for the ty-rosine kinase receptors Tyro3, Axl, and Mer. The endothelium expresses Tyro3, Axl, and Mer and produces protein S. The interaction of protein S with endothelial cells and particularly its effects on angio-genesis have not yet been analyzed. Here we show that human protein S, at circulating concentrations, inhibited vascular en-dothelial growth factor (VEGF) receptor 2-dependent vascularization of Matrigel plugs in vivo and the capacity of endo-thelial cells to form capillary-like networks in vitro as well as VEGF-A-induced endothelial migration and proliferation. Furthermore, protein S inhibited VEGF-A-induced endothelial VEGFR2 phosphorylation and activation of mitogen-activated kinase-Erk1/2 and Akt. Protein S activated the tyrosine phos-phatase SHP2, and the SHP2 inhibitor NSC 87877 reversed the observed inhibition of VEGF-A-induced endothelial proliferation. Using siRNA directed against Tyro3, Axl, and Mer, we demonstrate that protein S-mediated SHP2 activation and inhibition of VEGF-A-stimulated proliferation were mediated by Mer. Our report provides the first evidence for the existence of a protein S/Mer/SHP2 axis, which inhibits VEGFR2 signaling, regulates en-dothelial function, and points to a role for protein S as an endogenous angiogen-esis inhibitor. (Blood. 2012;120(25): 5073-5083

    Gene Expression of Protein Tyrosine Phosphatase 1B and Endoplasmic Reticulum Stress During Septic Shock

    No full text
    International audienceIntroduction: Protein Tyrosine Phosphatase 1B (PTP1B) and endoplasmic reticulum stress (ERS) are involved in the septic inflammatory response. Their inhibition is associated with improved survival in murine models of sepsis. The objective was to describe PTP1B and ERS expression during septic shock in human. Material and Methods: Prospective study including patients admitted to intensive care unit (ICU) for septic shock. Blood samples were collected on days 1 (D1), 3 and 5 (D5). Quantitative PCR (performed from whole blood) evaluated the expression of genes coding for PTP1B (PTPN1) and key elements of ERS (GRP78, ATF6, CHOP) or for endothelial dysfunction-related markers (ICAM1 and ET1). We analyzed gene variation between D5 and D1, collected glycemic parameters, insulin resistance and organ failure was evaluated by Sequential Organ Failure Assessment (SOFA) score. Results: We included 44 patients with a mean SAPS II 50 ± 16 and a mortality rate of 13.6%. Between D1 and D5, there was a significant decrease of PTPN1 (p < 0.001) and ATF6 (p < 0.001) expressions. Their variations of expression were correlated with SOFA variation (PTPN1, r = 0.35, CI 95% [0.05; 0.54], p = 0.03 and ATF6, r = 0.45 CI 95% [0.20; 0.65], p < 0.001). We did not find any correlation between PTPN1 expression and insulin resistance or glycemic parameters. Between D1 and D5, ATF6 and PTPN1 expressions were correlated with that of ET1. Conclusions: Our study has evaluated for the first time the expression of PTP1B and ERS in patients with septic shock, revealing that gene expression variation of PTPN1 and ATF6 are partly correlated with the evolution of septic organ failure and with endothelial dysfunction markers expression

    Adipogenic Mesenchymal Stromal Cells from Bone Marrow and Their Hematopoietic Supportive Role: Towards Understanding the Permissive Marrow Microenvironment in Acute Myeloid Leukemia

    No full text
    International audiencePurpose The role of bone marrow-derived mesenchymal stem/stromal cells (MSCs) in creating a permissive microen-vironment that supports the emergence and progression of acute myeloid leukemia (AML) is not well established. We investigated the extent to which adipogenic differentiation in normal MSCs alters hematopoietic supportive capacity and we undertook an in-depth comparative study of human bone marrow MSCs derived from newly diagnosed AML patients and healthy donors, including an assessment of adipogenic differentiation capacity. Findings MSCs from healthy controls with partial induction of adipogenic differentiation, in comparison to MSCs undergoing partial osteogenic differentiation, expressed increased levels of hematopoietic factors and induced greater proliferation , decreased quiescence and reduced in vitro hematopoietic colony forming capacity of CD34 + hematopoietic stem and progenitor cells (HSPCs). Moreover, we observed that AML-derived MSCs had markedly increased adipogenic potential and delayed osteogenic differentiation, while maintaining normal morphology and viability. AML-derived MSCs, however, possessed reduced proliferative capacity and decreased frequency of subendothelial quiescent MSCs compared to controls. Conclusion Our results support the notion of a bone marrow microenvironment characterized by increased propensity toward adipogenesis in AML, which may negatively impact normal hematopoiesis. Larger confirmatory studies are needed to understand the impact of various clinical factors. Novel leukemia treatments aimed at normalizing bone marrow niches may enhance the competitive advantage of normal he-matopoietic progenitors over leukemia cells

    #1409 Enhancer of zeste homolog 2 (EZH2) as a new therapeutic target to prevent indoxyl-sulfate-induced aortic valve calcification

    No full text
    International audienceBackground and Aims Calcific aortic valve disease (CAVD) is highly prevalent in chronic kidney disease (CKD) patients and is associated with a poor prognosis. To date, there is no pharmacological treatment to slow down this process. Our group recently reported that indoxyl-sulfate (IS) -induced IL-6 secretion in human interstitial valvular cells (hVICs) promotes their osteogenic transition and calcification in an autocrine manner. Preliminary data suggest that IS also influences macrophage secretion of IL-6. Whether this phenomenon affects the hVICs mineralization remains to be elucidated. In this context, the epigenetic enzyme Enhancer of Zeste Homolog 2 (EZH2), a key modulator of macrophages’ inflammation, may represent an interesting therapeutic target. Thus, this work aimed to verify whether IS-induced macrophages’ secretion of IL-6 modulates hVICs mineralization, and to determine whether EZH2 is involved in this inflammatory process. Method THP1-derived macrophages were exposed to increasing concentrations of IS (IS normal [Isn]: 0.5 μg/mL, IS uremic [Isu]: 37 μg/mL, IS intermediate [int]: 100 μg/mL or IS maximum [Ismax]: 233 μg/mL) in the presence or absence of an EZH2 inhibitor called GSK-343 (5µM). EZH2 expression and activity (evidenced by the trimethylation of the lysine 27 of the histones H3 (H3K27me3)) were assessed by western blot. Macrophages’ inflammatory potential was checked by RT-qPCR, western blot and Elisa. The impact of macrophages exposed or not to IS and/or GSK-343 on hVICs osteogenic transition was evaluated using conditioned media. HVICs osteogenic transition was assessed following runx2 expression by qRT-PCR and western blot. Mineralization was quantified by the o-cresolphthalein method. Results IS induced macrophages secretion of IL-1β, IL-6, CCL2 and TNF-α. Conditioned media from IS-polarized macrophages promoted a 6-fold increase in hVICs calcification and favored their osteogenic transition as evidenced by increased runx2 expression. IL-6 was the only cytokine able to promote runx2 expression in hVICs, confirming the importance of this specific cytokine in the process. Exposure to IS upregulated macrophages’ expression EZH2, an effect blocked by GSK-343. If GSK-343 did not affect IS-induced macrophages’ secretion of IL-1β, CCL2 and TNF-α, it efficiently blocked the secretion of IL-6 and subsequent induction of runx2 in hVICs. In this model, neither IS, nor GSK-343 modulated macrophages’ H3K27me3, suggesting that EZH2 may act as a transcriptional factor for IL-6 through its non-canonical pathway. In this latter, EZH2 is thought to promote inflammation by binding to p65, rather than through its trimethylation activity. In line with this hypothesis, exposure to IS promoted EZH2 and p65 nuclear translocation in the meantime as well as their co-immunoprecipitation, two phenomena blocked by GSK343. Conclusion This work demonstrates that IS-induced macrophages’ secretion of IL-6 promotes hVICs osteogenic transition and mineralization. Our data suggest that IL-6 secretion in response to IS depends on the EZH2-p65 pathway. The fact that GSK-343 can block this mechanism sheds light on EZH2 as a new therapeutic target to prevent CAVD in CKD patients

    A galactosidase-responsive doxorubicin-folate conjugate for selective targeting of acute myelogenous leukemia blasts

    No full text
    International audienceCytarabine combined with an anthracycline or an anthracenedione represents the usual intensive induction therapy for the treatment of AML. However, this protocol induces severe side effects and treatment-related mortality due to the lack of selectivity of these cytotoxic agents. In this paper, we present the study of the first galactosidase-responsive molecular "Trojan Horse" programmed for the delivery of doxorubicin exclusively inside AML blasts over-expressing the folate receptor (FR). This targeting system allows the selective killing of AML blasts without affecting normal endothelial, cardiac or hematologic cells from healthy donors suggesting that FDC could reduce adverse events usually recorded with anthracyclines
    corecore