27 research outputs found

    Angiogenesis: the VE-cadherin switch.

    Get PDF
    International audienceBecause angiogenesis is a key step in a number of pathologic processes, including tumor growth and atherosclerosis, many research studies have investigated the regulatory signals active at various stages of vascular invasion. The differential activities of the endothelial junction protein vascular endothelial (VE)-cadherin reflect the versatile behavior of endothelial cells between vascular quiescence and angiogenesis. VE-cadherin function and signaling are deeply modified in proliferating cells, and this conversion is accompanied by phosphorylation of the protein on tyrosine residues and enhanced transcription of its gene. Recent advances in the complex interplay between protein tyrosine kinases and phosphatases regulating VE-cadherin phosphorylation and function are discussed in this review

    The ATR Inhibitor AZD6738 Synergizes with Gemcitabine In Vitro and In Vivo to Induce Pancreatic Ductal Adenocarcinoma Regression.

    Get PDF
    Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest cancers, and overall survival rates have barely improved over the past five decades. The antimetabolite gemcitabine remains part of the standard of care but shows very limited antitumor efficacy. Ataxia telangiectasia and Rad3-related protein (ATR), the apical kinase of the intra-S-phase DNA damage response, plays a central role in safeguarding cells from replication stress and can therefore limit the efficacy of antimetabolite drug therapies. We investigated the ability of the ATR inhibitor, AZD6738, to prevent the gemcitabine-induced intra-S-phase checkpoint activation and evaluated the antitumor potential of this combination in vitro and in vivo In PDAC cell lines, AZD6738 inhibited gemcitabine-induced Chk1 activation, prevented cell-cycle arrest, and restrained RRM2 accumulation, leading to the strong induction of replication stress markers only with the combination. Moreover, synergistic growth inhibition was identified in a panel of 5 mouse and 7 human PDAC cell lines using both Bliss Independence and Loewe models. In clonogenic assays, the combination abrogated survival at concentrations for which single agents had minor effects. In vivo, AZD6738 in combination with gemcitabine was well tolerated and induced tumor regression in a subcutaneous allograft model of a KrasG12D; Trp53R172H; Pdx-Cre (KPC) mouse cancer cell line, significantly extending survival. Remarkably, the combination also induced regression of a subgroup of KPC autochthonous tumors, which generally do not respond well to conventional chemotherapy. Altogether, our data suggest that AZD6738 in combination with gemcitabine merits evaluation in a clinical trial in patients with PDAC. Mol Cancer Ther; 17(8); 1670-82. ©2018 AACR

    Quantifying cell cycle-dependent drug sensitivities in cancer using a high throughput synchronisation and screening approach.

    Get PDF
    BACKGROUND: Chemotherapy and targeted agent anti-cancer efficacy is largely dependent on the proliferative state of tumours, as exemplified by agents that target DNA synthesis/replication or mitosis. As a result, cell cycle specificities of a number of cancer drugs are well known. However, they are yet to be described in a quantifiable manner. METHODS: A scalable cell synchronisation protocol used to screen a library of 235 anti-cancer compounds exposed over six hours in G1 or S/G2 accumulated AsPC-1 cells to generate a cell cycle specificity (CCS) score. FINDINGS: The synchronisation method was associated with reduced method-related cytotoxicity compared to nocodazole, delivering sufficient cell cycle purity and cell numbers to run high-throughput drug library screens. Compounds were identified with G1 and S/G2-associated specificities that, overall, functionally matched with a compound's target/mechanism of action. This annotation was used to describe a synergistic schedule using the CDK4/6 inhibitor, palbociclib, prior to gemcitabine/AZD6738 as well as describe the correlation between the CCS score and published synergistic/antagonistic drug schedules. INTERPRETATION: This is the first highly quantitative description of cell cycle-dependent drug sensitivities that utilised a tractable and tolerated method with potential uses outside the present study. Drug treatments such as those shown to be G1 or S/G2 associated may benefit from scheduling considerations such as after CDK4/6 inhibitors and being first in drug sequences respectively. FUNDING: Cancer Research UK (CRUK) Institute core grants C14303/A17197 and C9545/A29580. The Li Ka Shing Centre where this work was performed was generously funded by CK Hutchison Holdings Limited, the University of Cambridge, CRUK, The Atlantic Philanthropies and others

    Mechanistic Distinctions between CHK1 and WEE1 Inhibition Guide the Scheduling of Triple Therapy with Gemcitabine.

    Get PDF
    Combination of cytotoxic therapy with emerging DNA damage response inhibitors (DDRi) has been limited by tolerability issues. However, the goal of most combination trials has been to administer DDRi with standard-of-care doses of chemotherapy. We hypothesized that mechanism-guided treatment scheduling could reduce the incidence of dose-limiting toxicities and enable tolerable multitherapeutic regimens. Integrative analyses of mathematical modeling and single-cell assays distinguished the synergy kinetics of WEE1 inhibitor (WEE1i) from CHEK1 inhibitor (CHK1i) by potency, spatiotemporal perturbation, and mitotic effects when combined with gemcitabine. These divergent properties collectively supported a triple-agent strategy, whereby a pulse of gemcitabine and CHK1i followed by WEE1i durably suppressed tumor cell growth. In xenografts, CHK1i exaggerated replication stress without mitotic CDK hyperactivation, enriching a geminin-positive subpopulation and intratumoral gemcitabine metabolite. Without overt toxicity, addition of WEE1i to low-dose gemcitabine and CHK1i was most effective in tumor control compared with single and double agents. Overall, our work provides quantitative insights into the mechanisms of DDRi chemosensitization, leading to the rational development of a tolerable multitherapeutic regimen.Significance: Multiple lines of mechanistic insight regarding DNA damage response inhibitors rationally guide the preclinical development of a tolerable multitherapeutic regimen.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/11/3054/F1.large.jpg Cancer Res; 78(11); 3054-66. ©2018 AACR

    Endothelial adherens and tight junctions in vascular homeostasis, inflammation and angiogenesis.

    Get PDF
    International audienceEndothelial cells lining the vessel wall are connected by adherens, tight and gap junctions. These junctional complexes are related to those found at epithelial junctions but with notable changes in terms of specific molecules and organization. Endothelial junctional proteins play important roles in tissue integrity but also in vascular permeability, leukocyte extravasation and angiogenesis. In this review, we will focus on specific mechanisms of endothelial tight and adherens junctions

    Phosphorylation sur tyrosine de la VE-cadhérine en réponse au VEGF : mécanismes moléculaires et implications physiopathologiques

    Get PDF
    Tyrosine phosphorylation of VE-cadherin in response to VEGF : molecular mechanisms and physiopathologic implications. Vascular integrity deeply depends on endothelial cell-cell junction stability. VE- (Vascular Endothelial)-cadherin is a key component of this junctions, and its tyrosine phosphorylation is thought to be a potent mechanism that controls vascular permeability. In this report, we demonstrated the existence of a tyrosine phosphorylated form of VE-cadherin in vivo. Remarkably, VE-cadherin tyrosine phosphorylation was strongly increased in ovary and uterus during hormone-induced angiogenesis, but not in other adult quiescent tissues. For the first time, we demonstrated that Src had a permanent association with VE-cadherin, in vivo and in vitro. Moreover, in agreement with other in vitro data, we found a transient association between VEGFR-2 and VE-cadherin in vivo in angiogenic tissues. Using several approaches, we demonstrated that VE-cadherin is a direct substrate for Src kinase in VEGF signaling pathway, and that tyrosine 685 is the unique phosphorylated site. In vitro wound healing test suggested that phosphorylation of Y685 in response to VEGF may be a key event in the intracellular signaling involved in migration process of endothelial cells. Finally, studying human breast carcinoma we also demonstrated the existence of a tyrosine phosphorylated form of VE-cadherin in tumor angiogenesis and its association with a PI3K and Src family kinase activity. Overall, this data concluded that Src kinase directly phosphorylates VE-cadherin on tyrosine 685 in response to VEGF, and this phosphorylation may be in vivo a sign of an angiogenic endothelium.L'intégrité de l'endothélium vasculaire est dépendante de l'organisation des jonctions adhérentes entre les cellules endothéliales. La VE (Vascular Endothelial)-cadhérine un constituant essentiel de ces jonctions et sa phosphorylation sur tyrosine pourrait être un moyen de régulation de la perméabilité endothéliale. Ce travail de thèse a permis de valider l'existence in vivo de la phosphorylation sur tyrosine de la VE-cadhérine. Nous montrons en effet la très forte induction de cette phosphorylation au cours de l'angiogenèse physiologique dans l'ovaire et l'utérus, par rapport aux tissus quiescents. Pour la première fois, nous rapportons l'association constante de la tyrosine kinase Src à la VE-cadhérine, à la fois in vivo et in vitro. De plus, en accord avec les données de la littérature in vitro, nous confirmons in vivo l'association transitoire du VEGFR2 à la VE-cadhérine dans un contexte angiogénique. Par différentes approches, nous apportons la preuve que la VE-cadhérine est un substrat direct de Src, et que la tyrosine 685 du domaine cytoplasmique est la cible unique de la kinase en réponse au VEGF. Grâce à un test de blessure d'HUVECs, nous montrons l'importance de la phosphorylation par Src de la tyrosine 685 dans la migration des cellules endothéliales induite par le VEGF. Enfin, l'étude de carcinomes mammaires nous as permis de démontrer l'existence d'une forme phosphorylée sur tyrosine de la VE-cadhérine également dans l'angiogenèse tumorale, et son association avec une activité kinase de type Src et PI3K. L'ensemble de ce travail permet de conclure à la phosphorylation par Src de la VEcadhérine sur la Y685 en réponse au VEGF, et suggère que cette phosphorylation représente in vivo la signature d'un endothélium activé

    Phosphorylation sur tyrosine de la VE-cadhérine en réponse au VEGF (mécanismes moléculaires et implications physiopathologiques )

    No full text
    L'intégrité de l'endothélium vasculaire est dépendante de l'organisation des jonctions adhérentes entre les cellules endothéliales. La VE (Vascular Endothelial)-cadhérine un constituant essentiel de ces jonctions et sa phosphorylation sur tyrosine pourrait être un moyen de régulation de la perméabilité endothéliale. Ce travail a permis de valider l'existence in vivo de la phosphorylation sur tyrosine de la VE-cadhérine. Nous montrons une forte induction de cette phosphorylation au cours de l'angiogenèse physiologique dans l'ovaire et l'utérus, par rapport aux tissus quiescents. Pour la première fois, nous rapportons l'association constante de la kinase Src à la VE-cadhérine, à la fois in vivo et in vitro. De plus, en accord avec les données de la littérature in vitro, nous confirmons in vivo l'association transitoire du VEGF-R2 à la VE-cadhérine dans un contexte angiogénique. Nous apportons la preuve que la VE-cadhérine est un substrat de Src, et que la tyrosine 685 du domaine cytoplasmique est la cible unique de la kinase en réponse au VEGF. Grâce à un test de blessure d'HUVECs, nous montrons l'importance de la phosphorylation par Src de la tyrosine 685 dans la migration des cellules endothéliales induite par le VEGF. Enfin, l'étude de carcinomes mammaires nous as permis de démontrer l'existence d'une forme phosphorylée sur tyrosine de la VE-cadhérine également dans l'angiogenèse tumorale, et son association avec une activité kinase de type Src et PI3K. L'ensemble de ce travail conclut à la phosphorylation par Src de la VE-cadhérine sur la Y685 en réponse au VEGF, et suggère que cette phosphorylation pourrait représenter in vivo la signature d'un endothélium activé.Vascular integrity deeply depends on endothelial cell-cell junction stability. VE- (Vascular Endothelial)-cadherin is a key component ofthis junctions, and its tyrosine phosphorylation is thought to be a potent mechanism that controls vascular permeability. ln this report, we demonstrated the existence of a tyrosine phosphorylated form ofVE-cadherin in vivo. Remarkably, VE-cadherin tyrosine phosphorylation was strongly increased in ovary and uterus during hormone-induced angiogenesis, but not in other adult quiescent tissues. For the first time, we demonstrated that Src had a permanent association with VE-cadherin, in vivo and in vitro. Moreover, in agreement with other in vitro data, we found a transient association between VEGFR-2 and VE-cadherin in vivo in angiogenic tissues. Using several approaches, we demonstrated that VE-cadherin is a direct substrate for Src kinase in VEGF signaling pathway, and that tyrosine 685 is the unique phosphorylated site. ln vitro wound healing test suggested that phosphorylation of Y685 in response to VEGF may be a key event in the intracellular signaling involved in migration process of endothelial cells. Finally, studying human breast carcinoma we also demonstrated the existence ofa tyrosine phosphorylated form ofVE-cadherin in tumor angiogenesis and its association with a PI3K and Src family kinase activity.GRENOBLE1-BU Sciences (384212103) / SudocSudocFranceF
    corecore