Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 49
Filtrer
1.
Blood ; 140(20): 2154-2169, 2022 11 17.
Article de Anglais | MEDLINE | ID: mdl-35981497

RÉSUMÉ

Cerebral cavernous malformation (CCM) is a neurovascular disease that results in various neurological symptoms. Thrombi have been reported in surgically resected CCM patient biopsies, but the molecular signatures of these thrombi remain elusive. Here, we investigated the kinetics of thrombi formation in CCM and how thrombi affect the vasculature and contribute to cerebral hypoxia. We used RNA sequencing to investigate the transcriptome of mouse brain endothelial cells with an inducible endothelial-specific Ccm3 knock-out (Ccm3-iECKO). We found that Ccm3-deficient brain endothelial cells had a higher expression of genes related to the coagulation cascade and hypoxia when compared with wild-type brain endothelial cells. Immunofluorescent assays identified key molecular signatures of thrombi such as fibrin, von Willebrand factor, and activated platelets in Ccm3-iECKO mice and human CCM biopsies. Notably, we identified polyhedrocytes in Ccm3-iECKO mice and human CCM biopsies and report it for the first time. We also found that the parenchyma surrounding CCM lesions is hypoxic and that more thrombi correlate with higher levels of hypoxia. We created an in vitro model to study CCM pathology and found that human brain endothelial cells deficient for CCM3 expressed elevated levels of plasminogen activator inhibitor-1 and had a redistribution of von Willebrand factor. With transcriptomics, comprehensive imaging, and an in vitro CCM preclinical model, this study provides experimental evidence that genes and proteins related to the coagulation cascade affect the brain vasculature and promote neurological side effects such as hypoxia in CCMs. This study supports the concept that antithrombotic therapy may be beneficial for patients with CCM.


Sujet(s)
Hémangiome caverneux du système nerveux central , Humains , Animaux , Souris , Hémangiome caverneux du système nerveux central/génétique , Hémangiome caverneux du système nerveux central/métabolisme , Cellules endothéliales/métabolisme , Protéines régulatrices de l'apoptose/génétique , Thrombo-inflammation , Facteur de von Willebrand/métabolisme , Hypoxie/métabolisme
3.
Cell Mol Life Sci ; 79(4): 206, 2022 Mar 25.
Article de Anglais | MEDLINE | ID: mdl-35333979

RÉSUMÉ

Cerebral Cavernous Malformation (CCM) is a brain vascular disease with various neurological symptoms. In this study, we describe the inflammatory profile in CCM and show for the first time the formation of neutrophil extracellular traps (NETs) in rodents and humans with CCM. Through RNA-seq analysis of cerebellum endothelial cells from wild-type mice and mice with an endothelial cell-specific ablation of the Ccm3 gene (Ccm3iECKO), we show that endothelial cells from Ccm3iECKO mice have an increased expression of inflammation-related genes. These genes encode proinflammatory cytokines and chemokines, as well as adhesion molecules, which promote recruitment of inflammatory and immune cells. Similarly, immunoassays showed elevated levels of these cytokines and chemokines in the cerebellum of the Ccm3iECKO mice. Consistently, both flow cytometry and immunofluorescence analysis showed infiltration of different subsets of leukocytes into the CCM lesions. Neutrophils, which are known to fight against infection through different strategies, including the formation of NETs, represented the leukocyte subset within the most pronounced increase in CCM. Here, we detected elevated levels of NETs in the blood and the deposition of NETs in the cerebral cavernomas of Ccm3iECKO mice. Degradation of NETs by DNase I treatment improved the vascular barrier. The deposition of NETs in the cavernomas  of patients with CCM confirms the clinical relevance of NETs in CCM.


Sujet(s)
Pièges extracellulaires , Hémangiome caverneux du système nerveux central , Animaux , Protéines régulatrices de l'apoptose/génétique , Cellules endothéliales/métabolisme , Pièges extracellulaires/métabolisme , Hémangiome caverneux du système nerveux central/génétique , Hémangiome caverneux du système nerveux central/métabolisme , Hémangiome caverneux du système nerveux central/anatomopathologie , Humains , Inflammation/anatomopathologie , Protéines membranaires/métabolisme , Souris
5.
Elife ; 92020 11 03.
Article de Anglais | MEDLINE | ID: mdl-33138917

RÉSUMÉ

Cerebral cavernous malformation (CCM) is a rare neurovascular disease that is characterized by enlarged and irregular blood vessels that often lead to cerebral hemorrhage. Loss-of-function mutations to any of three genes results in CCM lesion formation; namely, KRIT1, CCM2, and PDCD10 (CCM3). Here, we report for the first time in-depth single-cell RNA sequencing, combined with spatial transcriptomics and immunohistochemistry, to comprehensively characterize subclasses of brain endothelial cells (ECs) under both normal conditions and after deletion of Pdcd10 (Ccm3) in a mouse model of CCM. Integrated single-cell analysis identifies arterial ECs as refractory to CCM transformation. Conversely, a subset of angiogenic venous capillary ECs and respective resident endothelial progenitors appear to be at the origin of CCM lesions. These data are relevant for the understanding of the plasticity of the brain vascular system and provide novel insights into the molecular basis of CCM disease at the single cell level.


Sujet(s)
Cellules endothéliales/cytologie , Hémangiome caverneux du système nerveux central/physiopathologie , Animaux , Protéines régulatrices de l'apoptose/métabolisme , Artères/anatomopathologie , Encéphale/vascularisation , Encéphale/anatomopathologie , Différenciation cellulaire , Modèles animaux de maladie humaine , Délétion de gène , Immunohistochimie , Souris , Souris de lignée C57BL , Microscopie confocale , Mitose , Néovascularisation pathologique , Phénotype , RNA-Seq , Analyse de séquence d'ARN , Transduction du signal/génétique , Analyse sur cellule unique , Tamoxifène/pharmacologie , Transcriptome
6.
Development ; 147(16)2020 08 24.
Article de Anglais | MEDLINE | ID: mdl-32747434

RÉSUMÉ

Central nervous system (CNS) blood vessels contain a functional blood-brain barrier (BBB) that is necessary for neuronal survival and activity. Although Wnt/ß-catenin signaling is essential for BBB development, its downstream targets within the neurovasculature remain poorly understood. To identify targets of Wnt/ß-catenin signaling underlying BBB maturation, we performed a microarray analysis that identified Fgfbp1 as a novel Wnt/ß-catenin-regulated gene in mouse brain endothelial cells (mBECs). Fgfbp1 is expressed in the CNS endothelium and secreted into the vascular basement membrane during BBB formation. Endothelial genetic ablation of Fgfbp1 results in transient hypervascularization but delays BBB maturation in specific CNS regions, as evidenced by both upregulation of Plvap and increased tracer leakage across the neurovasculature due to reduced Wnt/ß-catenin activity. In addition, collagen IV deposition in the vascular basement membrane is reduced in mutant mice, leading to defective endothelial cell-pericyte interactions. Fgfbp1 is required cell-autonomously in mBECs to concentrate Wnt ligands near cell junctions and promote maturation of their barrier properties in vitro Thus, Fgfbp1 is a crucial extracellular matrix protein during BBB maturation that regulates cell-cell interactions and Wnt/ß-catenin activity.


Sujet(s)
Barrière hémato-encéphalique/embryologie , Collagène de type IV/métabolisme , Protéines et peptides de signalisation intercellulaire/métabolisme , Voie de signalisation Wnt , bêta-Caténine/métabolisme , Animaux , Collagène de type IV/génétique , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Protéines et peptides de signalisation intercellulaire/génétique , Souris , Souris transgéniques , Péricytes/cytologie , Péricytes/métabolisme , bêta-Caténine/génétique
7.
Arterioscler Thromb Vasc Biol ; 39(11): 2273-2288, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31533473

RÉSUMÉ

OBJECTIVE: Activation of endothelial ß-catenin signaling by neural cell-derived Norrin or Wnt ligands is vital for the vascularization of the retina and brain. Mutations in members of the Norrin/ß-catenin pathway contribute to inherited blinding disorders because of defective vascular development and dysfunctional blood-retina barrier. Despite a vital role for endothelial ß-catenin signaling in central nervous system health and disease, its contribution to central nervous system angiogenesis and its interactions with downstream signaling cascades remains incompletely understood. Approach and Results: Here, using genetically modified mouse models, we show that impaired endothelial ß-catenin signaling caused hypovascularization of the postnatal retina and brain because of deficient endothelial cell proliferation and sprouting. Mosaic genetic analysis demonstrated that endothelial ß-catenin promotes but is not required for tip cell formation. In addition, pharmacological treatment revealed that angiogenesis under conditions of inhibited Notch signaling depends upon endothelial ß-catenin. Importantly, impaired endothelial ß-catenin signaling abrogated the expression of the VEGFR (vascular endothelial growth factor receptor)-2 and VEGFR3 in brain microvessels but not in the lung endothelium. CONCLUSIONS: Our study identifies molecular crosstalk between the Wnt/ß-catenin and the Notch and VEGF-A signaling pathways and strongly suggest that endothelial ß-catenin signaling supports central nervous system angiogenesis by promoting endothelial cell sprouting, tip cell formation, and VEGF-A/VEGFR2 signaling.


Sujet(s)
Encéphale/vascularisation , Endothélium vasculaire/métabolisme , Néovascularisation physiologique , Rétine/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Voie de signalisation Wnt , bêta-Caténine/métabolisme , Animaux , Axine/métabolisme , Barrière hémato-encéphalique/métabolisme , Prolifération cellulaire , Cellules endothéliales/métabolisme , Souris transgéniques , Microcirculation , Interactions entre récepteurs , Récepteur Notch1/génétique , Récepteur Notch1/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique
8.
Nat Commun ; 10(1): 2761, 2019 06 24.
Article de Anglais | MEDLINE | ID: mdl-31235698

RÉSUMÉ

Cerebral cavernous malformation (CCM) is a neurovascular familial or sporadic disease that is characterised by capillary-venous cavernomas, and is due to loss-of-function mutations to any one of three CCM genes. Familial CCM follows a two-hit mechanism similar to that of tumour suppressor genes, while in sporadic cavernomas only a small fraction of endothelial cells shows mutated CCM genes. We reported that in mouse models and in human patients, endothelial cells lining the lesions have different features from the surrounding endothelium, as they express mesenchymal/stem-cell markers. Here we show that cavernomas originate from clonal expansion of few Ccm3-null endothelial cells that express mesenchymal/stem-cell markers. These cells then attract surrounding wild-type endothelial cells, inducing them to express mesenchymal/stem-cell markers and to contribute to cavernoma growth. These characteristics of Ccm3-null cells are reminiscent of the tumour-initiating cells that are responsible for tumour growth. Our data support the concept that CCM has benign tumour characteristics.


Sujet(s)
Protéines régulatrices de l'apoptose/génétique , Tumeurs du système nerveux central/anatomopathologie , Cellules endothéliales/anatomopathologie , Hémangiome caverneux du système nerveux central/anatomopathologie , Protéines et peptides de signalisation intracellulaire/génétique , Protéines membranaires/génétique , Protéines proto-oncogènes/génétique , Animaux , Protéines régulatrices de l'apoptose/métabolisme , Marqueurs biologiques/métabolisme , Encéphale/vascularisation , Encéphale/cytologie , Encéphale/anatomopathologie , Différenciation cellulaire/génétique , Lignée cellulaire , Tumeurs du système nerveux central/génétique , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Endothélium vasculaire/cytologie , Endothélium vasculaire/anatomopathologie , Femelle , Techniques de knock-out de gènes , Hémangiome caverneux du système nerveux central/génétique , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Mutation perte de fonction , Protéines membranaires/métabolisme , Cellules souches mésenchymateuses/métabolisme , Souris , Souris knockout , Protéines proto-oncogènes/métabolisme
9.
Circ Res ; 124(4): 511-525, 2019 02 15.
Article de Anglais | MEDLINE | ID: mdl-30591003

RÉSUMÉ

RATIONALE: The microvasculature of the central nervous system includes the blood-brain barrier (BBB), which regulates the permeability to nutrients and restricts the passage of toxic agents and inflammatory cells. Canonical Wnt/ß-catenin signaling is responsible for the early phases of brain vascularization and BBB differentiation. However, this signal declines after birth, and other signaling pathways able to maintain barrier integrity at postnatal stage are still unknown. OBJECTIVE: Sox17 (SRY [sex-determining region Y]-box 17) constitutes a major downstream target of Wnt/ß-catenin in endothelial cells and regulates arterial differentiation. In the present article, we asked whether Sox17 may act downstream of Wnt/ß-catenin in inducing BBB differentiation and maintenance. METHODS AND RESULTS: Using reporter mice and nuclear staining of Sox17 and ß-catenin, we report that although ß-catenin signaling declines after birth, Sox17 activation increases and remains high in the adult. Endothelial-specific inactivation of Sox17 leads to increase of permeability of the brain microcirculation. The severity of this effect depends on the degree of BBB maturation: it is strong in the embryo and progressively declines after birth. In search of Sox17 mechanism of action, RNA sequencing analysis of gene expression of brain endothelial cells has identified members of the Wnt/ß-catenin signaling pathway as downstream targets of Sox17. Consistently, we found that Sox17 is a positive inducer of Wnt/ß-catenin signaling, and it acts in concert with this pathway to induce and maintain BBB properties. In vivo, inhibition of the ß-catenin destruction complex or expression of a degradation-resistant ß-catenin mutant, prevent the increase in permeability and retina vascular malformations observed in the absence of Sox17. CONCLUSIONS: Our data highlight a novel role for Sox17 in the induction and maintenance of the BBB, and they underline the strict reciprocal tuning of this transcription factor and Wnt/ß-catenin pathway. Modulation of Sox17 activity may be relevant to control BBB permeability in pathological conditions.


Sujet(s)
Barrière hémato-encéphalique/métabolisme , Perméabilité capillaire , Protéines HMGB/métabolisme , Facteurs de transcription SOX-F/métabolisme , Voie de signalisation Wnt , Animaux , Protéines HMGB/génétique , Souris , Souris de lignée C57BL , Facteurs de transcription SOX-F/génétique
10.
Circ Res ; 122(2): 231-245, 2018 01 19.
Article de Anglais | MEDLINE | ID: mdl-29233846

RÉSUMÉ

RATIONALE: The mechanistic foundation of vascular maturation is still largely unknown. Several human pathologies are characterized by deregulated angiogenesis and unstable blood vessels. Solid tumors, for instance, get their nourishment from newly formed structurally abnormal vessels which present wide and irregular interendothelial junctions. Expression and clustering of the main endothelial-specific adherens junction protein, VEC (vascular endothelial cadherin), upregulate genes with key roles in endothelial differentiation and stability. OBJECTIVE: We aim at understanding the molecular mechanisms through which VEC triggers the expression of a set of genes involved in endothelial differentiation and vascular stabilization. METHODS AND RESULTS: We compared a VEC-null cell line with the same line reconstituted with VEC wild-type cDNA. VEC expression and clustering upregulated endothelial-specific genes with key roles in vascular stabilization including claudin-5, vascular endothelial-protein tyrosine phosphatase (VE-PTP), and von Willebrand factor (vWf). Mechanistically, VEC exerts this effect by inhibiting polycomb protein activity on the specific gene promoters. This is achieved by preventing nuclear translocation of FoxO1 (Forkhead box protein O1) and ß-catenin, which contribute to PRC2 (polycomb repressive complex-2) binding to promoter regions of claudin-5, VE-PTP, and vWf. VEC/ß-catenin complex also sequesters a core subunit of PRC2 (Ezh2 [enhancer of zeste homolog 2]) at the cell membrane, preventing its nuclear translocation. Inhibition of Ezh2/VEC association increases Ezh2 recruitment to claudin-5, VE-PTP, and vWf promoters, causing gene downregulation. RNA sequencing comparison of VEC-null and VEC-positive cells suggested a more general role of VEC in activating endothelial genes and triggering a vascular stability-related gene expression program. In pathological angiogenesis of human ovarian carcinomas, reduced VEC expression paralleled decreased levels of claudin-5 and VE-PTP. CONCLUSIONS: These data extend the knowledge of polycomb-mediated regulation of gene expression to endothelial cell differentiation and vessel maturation. The identified mechanism opens novel therapeutic opportunities to modulate endothelial gene expression and induce vascular normalization through pharmacological inhibition of the polycomb-mediated repression system.


Sujet(s)
Antigènes CD/biosynthèse , Cadhérines/biosynthèse , Endothélium vasculaire/métabolisme , Épigenèse génétique/physiologie , Animaux , Antigènes CD/génétique , Cadhérines/génétique , Membrane cellulaire/génétique , Membrane cellulaire/métabolisme , Membrane cellulaire/ultrastructure , Endothélium vasculaire/ultrastructure , Expression des gènes , Cellules HEK293 , Humains , Souris , Protéines du groupe Polycomb/métabolisme , Liaison aux protéines/physiologie
12.
Am J Pathol ; 187(8): 1879-1892, 2017 Aug.
Article de Anglais | MEDLINE | ID: mdl-28728747

RÉSUMÉ

Primary myelofibrosis is characterized by the development of fibrosis in the bone marrow that contributes to ineffective hematopoiesis. Bone marrow fibrosis is the result of a complex and not yet fully understood interaction among megakaryocytes, myeloid cells, fibroblasts, and endothelial cells. Here, we report that >30% of the endothelial cells in the small vessels of the bone marrow and spleen of patients with primary myelofibrosis have a mesenchymal phenotype, which is suggestive of the process known as endothelial-to-mesenchymal transition (EndMT). EndMT can be reproduced in vitro by incubation of cultured endothelial progenitor cells or spleen-derived endothelial cells with inflammatory cytokines. Megakaryocytes appear to be implicated in this process, because EndMT mainly occurs in the microvessels close to these cells, and because megakaryocyte-derived supernatant fluid can reproduce the EndMT switch in vitro. Furthermore, EndMT is an early event in a JAK2-V617F knock-in mouse model of primary myelofibrosis. Overall, these data show for the first time that microvascular endothelial cells in the bone marrow and spleen of patients with primary myelofibrosis show functional and morphologic changes that are associated to the mesenchymal phenotype.


Sujet(s)
Moelle osseuse/anatomopathologie , Myélofibrose primitive/anatomopathologie , Rate/anatomopathologie , Animaux , Modèles animaux de maladie humaine , Humains , Mégacaryocytes/anatomopathologie , Souris
13.
Development ; 144(14): 2629-2639, 2017 07 15.
Article de Anglais | MEDLINE | ID: mdl-28619820

RÉSUMÉ

Arterial specification and differentiation are influenced by a number of regulatory pathways. While it is known that the Vegfa-Notch cascade plays a central role, the transcriptional hierarchy controlling arterial specification has not been fully delineated. To elucidate the direct transcriptional regulators of Notch receptor expression in arterial endothelial cells, we used histone signatures, DNaseI hypersensitivity and ChIP-seq data to identify enhancers for the human NOTCH1 and zebrafish notch1b genes. These enhancers were able to direct arterial endothelial cell-restricted expression in transgenic models. Genetic disruption of SoxF binding sites established a clear requirement for members of this group of transcription factors (SOX7, SOX17 and SOX18) to drive the activity of these enhancers in vivo Endogenous deletion of the notch1b enhancer led to a significant loss of arterial connections to the dorsal aorta in Notch pathway-deficient zebrafish. Loss of SoxF function revealed that these factors are necessary for NOTCH1 and notch1b enhancer activity and for correct endogenous transcription of these genes. These findings position SoxF transcription factors directly upstream of Notch receptor expression during the acquisition of arterial identity in vertebrates.


Sujet(s)
Artères/embryologie , Artères/métabolisme , Récepteur Notch1/génétique , Récepteur Notch1/métabolisme , Facteurs de transcription SOX-F/génétique , Facteurs de transcription SOX-F/métabolisme , Séquence d'acides aminés , Animaux , Animal génétiquement modifié , Malformations artérioveineuses/embryologie , Malformations artérioveineuses/génétique , Malformations artérioveineuses/métabolisme , Éléments activateurs (génétique) , Femelle , Régulation de l'expression des gènes au cours du développement , Cellules endothéliales de la veine ombilicale humaine , Humains , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Grossesse , Récepteur Notch1/déficit , Facteurs de transcription SOX-F/déficit , Similitude de séquences d'acides aminés , Transduction du signal , Danio zébré , Protéines de poisson-zèbre/déficit , Protéines de poisson-zèbre/génétique , Protéines de poisson-zèbre/métabolisme
14.
Stem Cells ; 35(5): 1328-1340, 2017 05.
Article de Anglais | MEDLINE | ID: mdl-28090691

RÉSUMÉ

Vascular associated endothelial cell (ECs) progenitors are still poorly studied and their role in the newly forming vasculature at embryonic or postnatal stage remains elusive. In the present work, we first defined a set of genes highly expressed during embryo development and strongly downregulated in the adult mouse. In this group, we then concentrated on the progenitor cell marker Peg3/PW1. By in vivo staining of the vasculature we found that only a subset of cells coexpressed endothelial markers and PW1. These cells were quite abundant in the embryo vasculature but declined in number at postnatal and adult stages. Using a reporter mouse for PW1 expression, we have been able to isolate PW1-positive (PW1posECs) and negative endothelial cells (PW1negECs). PW1-positive cells were highly proliferative in comparison to PW1negECs and were able to form colonies when seeded at clonal dilution. Furthermore, by RNAseq analysis, PW1posECs expressed endothelial cell markers together with mesenchymal and stem cell markers. When challenged by endothelial growth factors in vitro, PW1posECs were able to proliferate more than PW1negECs and to efficiently form new vessels in vivo. Taken together these data identify a subset of vessel associated endothelial cells with characteristics of progenitor cells. Considering their high proliferative potential these cells may be of particular importance to design therapies to improve the perfusion of ischemic tissues or to promote vascular repair. Stem Cells 2017;35:1328-1340.


Sujet(s)
Vaisseaux sanguins/cytologie , Progéniteurs endothéliaux/cytologie , Progéniteurs endothéliaux/métabolisme , Facteurs de transcription Krüppel-like/métabolisme , Animaux , Marqueurs biologiques/métabolisme , Vaisseaux sanguins/embryologie , Vaisseaux sanguins/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Séparation cellulaire , Développement embryonnaire/effets des médicaments et des substances chimiques , Progéniteurs endothéliaux/effets des médicaments et des substances chimiques , Analyse de profil d'expression de gènes , Protéines et peptides de signalisation intercellulaire/pharmacologie , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Souris de lignée C57BL , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Phénotype
15.
J Neurosci ; 36(34): 8921-35, 2016 08 24.
Article de Anglais | MEDLINE | ID: mdl-27559173

RÉSUMÉ

UNLABELLED: The canonical Wnt/ß-catenin signaling pathway is crucial for blood-brain barrier (BBB) formation in brain endothelial cells. Although glucose transporter 1, claudin-3, and plasmalemma vesicular-associated protein have been identified as Wnt/ß-catenin targets in brain endothelial cells, further downstream targets relevant to BBB formation and function are incompletely explored. By Affymetrix expression analysis, we show that the cytochrome P450 enzyme Cyp1b1 was significantly decreased in ß-catenin-deficient mouse endothelial cells, whereas its close homolog Cyp1a1 was upregulated in an aryl hydrocarbon receptor-dependent manner, hence indicating that ß-catenin is indispensable for Cyp1b1 but not for Cyp1a1 expression. Functionally, Cyp1b1 could generate retinoic acid from retinol leading to cell-autonomous induction of the barrier-related ATP-binding cassette transporter P-glycoprotein. Cyp1b1 could also generate 20-hydroxyeicosatetraenoic acid from arachidonic acid, decreasing endothelial barrier function in vitro In mice in vivo pharmacological inhibition of Cyp1b1 increased BBB permeability for small molecular tracers, and Cyp1b1 was downregulated in glioma vessels in which BBB function is lost. Hence, we propose Cyp1b1 as a target of ß-catenin indirectly influencing BBB properties via its metabolic activity, and as a potential target for modulating barrier function in endothelial cells. SIGNIFICANCE STATEMENT: Wnt/ß-catenin signaling is crucial for blood-brain barrier (BBB) development and maintenance; however, its role in regulating metabolic characteristics of endothelial cells is unclear. We provide evidence that ß-catenin influences endothelial metabolism by transcriptionally regulating the cytochrome P450 enzyme Cyp1b1 Furthermore, expression of its close homolog Cyp1a1 was inhibited by ß-catenin. Functionally, Cyp1b1 generated retinoic acid as well as 20-hydroxyeicosatetraenoic acid that regulated P-glycoprotein and junction proteins, respectively, thereby modulating BBB properties. Inhibition of Cyp1b1 in vivo increased BBB permeability being in line with its downregulation in glioma endothelia, potentially implicating Cyp1b1 in other brain pathologies. In conclusion, Wnt/ß-catenin signaling regulates endothelial metabolic barrier function through Cyp1b1 transcription.


Sujet(s)
Barrière hémato-encéphalique/métabolisme , Cytochrome P-450 CYP1B1/métabolisme , Cellules endothéliales/métabolisme , Régulation de l'expression des gènes/physiologie , Voie de signalisation Wnt/physiologie , bêta-Caténine/métabolisme , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Cadhérines/génétique , Cadhérines/métabolisme , Perméabilité capillaire/génétique , Immunoprécipitation de la chromatine , Cytochrome P-450 CYP1B1/génétique , Relation dose-effet des médicaments , Femelle , Régulation de l'expression des gènes/génétique , Gliome/métabolisme , Gliome/anatomopathologie , Histone/métabolisme , Acide hydroxyeïcosatétraénoïque/pharmacologie , Mâle , Souris , Souris nude , Modèles biologiques , Transplantation tumorale , ARN messager/métabolisme , Récepteurs à hydrocarbure aromatique/génétique , Récepteurs à hydrocarbure aromatique/métabolisme , Voie de signalisation Wnt/génétique , bêta-Caténine/génétique
16.
BMC Neurol ; 16(1): 144, 2016 Aug 18.
Article de Anglais | MEDLINE | ID: mdl-27538712

RÉSUMÉ

BACKGROUND: VE-cadherin is the chief constituent of endothelial adherens junctions. However, the role of VE-cadherin in the pathogenesis of cerebrovascular diseases including brain ischemia has not yet been investigated. METHODS: VE-cadherin heterozygous (VEC(+/-)) mice and wildtype controls were subjected to transient brain ischemia by 30 min filamentous middle cerebral artery occlusion (MCAo)/reperfusion. RESULTS: Acute lesion sizes as assessed by MR-imaging on day 3 did not differ between genotypes. Unexpectedly, however, partial loss of VE-cadherin resulted in long-term stroke protection measured histologically on day 28. Equally surprisingly, VEC(+/-) mice displayed no differences in post-stroke angiogenesis compared to littermate controls, but showed increased absolute regional cerebral blood flow in ischemic striatum at four weeks. The early induction of VE-cadherin mRNA transcription after stroke was reduced in VEC(+/-) mice. By contrast, N-cadherin and ß-catenin mRNA expression showed a delayed, but sustained, upregulation up to 28 days after MCAo, which was increased in VEC(+/-) mice. Furthermore, partial loss of VE-cadherin resulted in a pattern of elevated ischemia-triggered mRNA transcription of pericyte-related molecules α-smooth muscle actin (α-SMA), aminopeptidase N (CD13), and platelet-derived growth factor receptor ß (PDGFR-ß). CONCLUSIONS: Partial loss of VE-cadherin results in long term stroke protection. On the cellular and molecular level, this effect appears to be mediated by improved endothelial/pericyte interactions and the resultant increase in cerebral blood flow. Our study reinforces accumulating evidence that long-term stroke outcome depends critically on vascular mechanisms.


Sujet(s)
Antigènes CD/métabolisme , Cadhérines/métabolisme , Circulation cérébrovasculaire/physiologie , Endothélium vasculaire/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Accident ischémique transitoire/métabolisme , Péricytes/métabolisme , Animaux , Modèles animaux de maladie humaine , Infarctus du territoire de l'artère cérébrale moyenne/complications , Infarctus du territoire de l'artère cérébrale moyenne/physiopathologie , Accident ischémique transitoire/étiologie , Accident ischémique transitoire/physiopathologie , Mâle , Souris , Souris transgéniques
17.
Nat Commun ; 7: 12422, 2016 08 12.
Article de Anglais | MEDLINE | ID: mdl-27516371

RÉSUMÉ

Mural cells of the vessel wall, namely pericytes and vascular smooth muscle cells, are essential for vascular integrity. The developmental sources of these cells and molecular mechanisms controlling their progenitors in the heart are only partially understood. Here we show that endocardial endothelial cells are progenitors of pericytes and vascular smooth muscle cells in the murine embryonic heart. Endocardial cells undergo endothelial-mesenchymal transition and convert into primitive mesenchymal progenitors expressing the platelet-derived growth factor receptors, PDGFRα and PDGFRß. These progenitors migrate into the myocardium, differentiate and assemble the wall of coronary vessels, which requires canonical Wnt signalling involving Frizzled4, ß-catenin and endothelial cell-derived Wnt ligands. Our findings identify a novel and unexpected population of progenitors for coronary mural cells with potential relevance for heart function and disease conditions.


Sujet(s)
Différenciation cellulaire/physiologie , Cellules endothéliales/physiologie , Transition épithélio-mésenchymateuse/physiologie , Myocytes du muscle lisse/physiologie , Péricytes/physiologie , Animaux , Cellules cultivées , Endocarde/cytologie , Endocarde/physiologie , Femelle , Coeur/physiologie , Cellules souches mésenchymateuses/physiologie , Souris , Souris de lignée C57BL , Souris transgéniques , Modèles animaux , Muscles lisses vasculaires/cytologie , Muscles lisses vasculaires/physiologie , Myocarde/cytologie , Récepteur au PDGF alpha/métabolisme , Récepteur au PDGF bêta/génétique , Récepteur au PDGF bêta/métabolisme
18.
EMBO Mol Med ; 8(1): 6-24, 2016 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-26612856

RÉSUMÉ

Cerebral cavernous malformations (CCMs) are vascular malformations located within the central nervous system often resulting in cerebral hemorrhage. Pharmacological treatment is needed, since current therapy is limited to neurosurgery. Familial CCM is caused by loss-of-function mutations in any of Ccm1, Ccm2, and Ccm3 genes. CCM cavernomas are lined by endothelial cells (ECs) undergoing endothelial-to-mesenchymal transition (EndMT). This switch in phenotype is due to the activation of the transforming growth factor beta/bone morphogenetic protein (TGFß/BMP) signaling. However, the mechanism linking Ccm gene inactivation and TGFß/BMP-dependent EndMT remains undefined. Here, we report that Ccm1 ablation leads to the activation of a MEKK3-MEK5-ERK5-MEF2 signaling axis that induces a strong increase in Kruppel-like factor 4 (KLF4) in ECs in vivo. KLF4 transcriptional activity is responsible for the EndMT occurring in CCM1-null ECs. KLF4 promotes TGFß/BMP signaling through the production of BMP6. Importantly, in endothelial-specific Ccm1 and Klf4 double knockout mice, we observe a strong reduction in the development of CCM and mouse mortality. Our data unveil KLF4 as a therapeutic target for CCM.


Sujet(s)
Hémangiome caverneux du système nerveux central/génétique , Hémangiome caverneux du système nerveux central/anatomopathologie , Facteurs de transcription Krüppel-like/métabolisme , Animaux , Protéine morphogénétique osseuse de type 6/antagonistes et inhibiteurs , Protéine morphogénétique osseuse de type 6/génétique , Protéine morphogénétique osseuse de type 6/métabolisme , Prolifération cellulaire , Modèles animaux de maladie humaine , Évolution de la maladie , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Cellules HEK293 , Hémangiome caverneux du système nerveux central/métabolisme , Humains , Protéine KRIT1 , Facteur-4 de type Kruppel , Facteurs de transcription Krüppel-like/antagonistes et inhibiteurs , Facteurs de transcription Krüppel-like/génétique , Souris , Souris de lignée C57BL , Souris knockout , Protéines associées aux microtubules/antagonistes et inhibiteurs , Protéines associées aux microtubules/génétique , Protéines associées aux microtubules/métabolisme , Mitogen-Activated Protein Kinase 7/métabolisme , Mutation , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Interférence par ARN , Transduction du signal , Protéine Smad-1/métabolisme , Facteur de croissance transformant bêta/métabolisme
19.
J Cell Biol ; 211(6): 1177-92, 2015 Dec 21.
Article de Anglais | MEDLINE | ID: mdl-26668327

RÉSUMÉ

Vascular endothelial (VE)-cadherin transfers intracellular signals contributing to vascular hemostasis. Signaling through VE-cadherin requires association and activity of different intracellular partners. Yes-associated protein (YAP)/TAZ transcriptional cofactors are important regulators of cell growth and organ size. We show that EPS8, a signaling adapter regulating actin dynamics, is a novel partner of VE-cadherin and is able to modulate YAP activity. By biochemical and imaging approaches, we demonstrate that EPS8 associates with the VE-cadherin complex of remodeling junctions promoting YAP translocation to the nucleus and transcriptional activation. Conversely, in stabilized junctions, 14-3-3-YAP associates with the VE-cadherin complex, whereas Eps8 is excluded. Junctional association of YAP inhibits nuclear translocation and inactivates its transcriptional activity both in vitro and in vivo in Eps8-null mice. The absence of Eps8 also increases vascular permeability in vivo, but did not induce other major vascular defects. Collectively, we identified novel components of the adherens junction complex, and we introduce a novel molecular mechanism through which the VE-cadherin complex controls YAP transcriptional activity.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Antigènes CD/métabolisme , Cadhérines/métabolisme , Endothélium vasculaire/métabolisme , Phosphoprotéines/métabolisme , Transduction du signal , Protéines adaptatrices de la transduction du signal/déficit , Animaux , Sites de fixation , Protéines du cycle cellulaire , Lignée cellulaire , Souris , Souris de lignée C57BL , Souris knockout , Liaison aux protéines , Transport des protéines , Protéines de signalisation YAP
20.
Proc Natl Acad Sci U S A ; 112(27): 8421-6, 2015 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-26109568

RÉSUMÉ

Cerebral cavernous malformation (CCM) is a disease of the central nervous system causing hemorrhage-prone multiple lumen vascular malformations and very severe neurological consequences. At present, the only recommended treatment of CCM is surgical. Because surgery is often not applicable, pharmacological treatment would be highly desirable. We describe here a murine model of the disease that develops after endothelial-cell-selective ablation of the CCM3 gene. We report an early, cell-autonomous, Wnt-receptor-independent stimulation of ß-catenin transcription activity in CCM3-deficient endothelial cells both in vitro and in vivo and a triggering of a ß-catenin-driven transcription program that leads to endothelial-to-mesenchymal transition. TGF-ß/BMP signaling is then required for the progression of the disease. We also found that the anti-inflammatory drugs sulindac sulfide and sulindac sulfone, which attenuate ß-catenin transcription activity, reduce vascular malformations in endothelial CCM3-deficient mice. This study opens previously unidentified perspectives for an effective pharmacological therapy of intracranial vascular cavernomas.


Sujet(s)
Tumeurs du système nerveux central/traitement médicamenteux , Hémangiome caverneux du système nerveux central/traitement médicamenteux , Protéines et peptides de signalisation intracellulaire/déficit , Sulindac/analogues et dérivés , Animaux , Anti-inflammatoires non stéroïdiens/pharmacologie , Protéines régulatrices de l'apoptose , Tumeurs du système nerveux central/génétique , Tumeurs du système nerveux central/métabolisme , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Hémangiome caverneux du système nerveux central/génétique , Hémangiome caverneux du système nerveux central/métabolisme , Immunohistochimie , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Souris knockout , RT-PCR , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Sulindac/pharmacologie , Facteur de croissance transformant bêta/métabolisme , bêta-Caténine/génétique , bêta-Caténine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...