Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Arterioscler Thromb Vasc Biol ; 43(7): 1199-1218, 2023 07.
Article in English | MEDLINE | ID: mdl-37199159

ABSTRACT

BACKGROUND: Endothelial cells (ECs) are sensitive to physical forces created by blood flow, especially to laminar shear stress. Among the cell responses to laminar flow, EC polarization against the flow direction emerges as a key event, particularly during the development and remodeling of the vascular network. EC adopt an elongated planar cell shape with an asymmetrical distribution of intracellular organelles along the axis of blood flow. This study aimed to investigate the involvement of planar cell polarity via the receptor ROR2 (receptor tyrosine kinase-like orphan receptor 2) in endothelial responses to laminar shear stress. METHODS: We generated a genetic mouse model with EC-specific deletion of Ror2, in combination with in vitro approaches involving loss- and gain-of-function experiments. RESULTS: During the first 2 weeks of life, the endothelium of the mouse aorta undergoes a rapid remodeling associated with a loss of EC polarization against the flow direction. Notably, we found a correlation between ROR2 expression and endothelial polarization levels. Our findings demonstrate that deletion of Ror2 in murine ECs impaired their polarization during the postnatal development of the aorta. In vitro experiments further validated the essential role of ROR2 in both EC collective polarization and directed migration under laminar flow conditions. Exposure to laminar shear stress triggered the relocalization of ROR2 to cell-cell junctions where it formed a complex with VE-Cadherin and ß-catenin, thereby regulating adherens junctions remodeling at the rear and front poles of ECs. Finally, we showed that adherens junctions remodeling and cell polarity induced by ROR2 were dependent on the activation of the small GTPase Cdc42. CONCLUSIONS: This study identified ROR2/planar cell polarity pathway as a new mechanism controlling and coordinating collective polarity patterns of EC during shear stress response.


Subject(s)
Endothelial Cells , Receptor Tyrosine Kinase-like Orphan Receptors , Mice , Animals , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Cell Polarity/physiology , Endothelium, Vascular/metabolism , Intercellular Junctions , Stress, Mechanical
2.
Arterioscler Thromb Vasc Biol ; 42(6): 745-763, 2022 06.
Article in English | MEDLINE | ID: mdl-35510550

ABSTRACT

BACKGROUND: While endothelial dysfunction is suggested to contribute to heart failure with preserved ejection fraction pathophysiology, understanding the importance of the endothelium alone, in the pathogenesis of diastolic abnormalities has not yet been fully elucidated. Here, we investigated the consequences of specific endothelial dysfunction on cardiac function, independently of any comorbidity or risk factor (diabetes or obesity) and their potential effect on cardiomyocyte. METHODS: The ubiquitine ligase Pdzrn3, expressed in endothelial cells (ECs), was shown to destabilize tight junction. A genetic mouse model in which Pdzrn3 is overexpressed in EC (iEC-Pdzrn3) in adults was developed. RESULTS: EC-specific Pdzrn3 expression increased cardiac leakage of IgG and fibrinogen blood-born molecules. The induced edema demonstrated features of diastolic dysfunction, with increased end-diastolic pressure, alteration of dP/dt min, increased natriuretic peptides, in addition to limited exercise capacity, without major signs of cardiac fibrosis and inflammation. Electron microscopic images showed edema with disrupted EC-cardiomyocyte interactions. RNA sequencing analysis of gene expression in cardiac EC demonstrated a decrease in genes coding for endothelial extracellular matrix proteins, which could be related to the fragile blood vessel phenotype. Irregularly shaped capillaries with hemorrhages were found in heart sections of iEC-Pdzrn3 mice. We also found that a high-fat diet was not sufficient to provoke diastolic dysfunction; high-fat diet aggravated cardiac inflammation, associated with an altered cardiac metabolic signature in EC-Pdzrn3 mice, reminiscent of heart failure with preserved ejection fraction features. CONCLUSIONS: An increase of endothelial permeability is responsible for mediating diastolic dysfunction pathophysiology and for aggravating detrimental effects of a high-fat diet on cardiac inflammation and metabolism.


Subject(s)
Cardiomyopathies , Heart Failure , Animals , Capillary Permeability , Endothelial Cells/metabolism , Fibrosis , Heart Failure/genetics , Heart Failure/metabolism , Inflammation/metabolism , Mice , Myocytes, Cardiac/metabolism , Stroke Volume/physiology , Ubiquitin-Protein Ligases
3.
Arterioscler Thromb Vasc Biol ; 42(5): 597-609, 2022 05.
Article in English | MEDLINE | ID: mdl-35387477

ABSTRACT

BACKGROUND: Genome-wide association studies have revealed robust associations of common genetic polymorphisms in an intron of the PHACTR-1 (phosphatase and actin regulator 1) gene (chr6p24), with cervical artery dissection, spontaneous coronary artery dissection, and fibromuscular dysplasia. The aim was to assess its role in the pathogenesis of cervical artery dissection or fibromuscular dysplasia. METHODS: Using various tissue-specific Cre-driver mouse lines, Phactr1 was deleted either in endothelial cells using 2 tissue-specific Cre-driver (PDGFB [platelet-derived growth factor B]-CreERT2 mice and Tie2 [tyrosine kinase with immunoglobulin and EGF homology domains]-Cre) and smooth muscle cells (smooth muscle actin-CreERT2) with a third tissue-specific Cre-driver. RESULTS: To test the efficacy of the Phactr1 deletion after cre-induction, we confirmed first, a decrease in Phactr1 transcription and Phactr1 expression in endothelial cell and smooth muscle cell isolated from Phactr1iPDGFB and Phactr1iSMA mice. Irrespective to the tissue or the duration of the deletion, mice did not spontaneously display pathological phenotype or vascular impairment: mouse survival, growth, blood pressure, large vessel morphology, or actin organization were not different in knockout mice than their comparatives littermates. Challenging vascular function and repair either by angiotensin II-induced hypertension or limb ischemia did not lead to vascular morphology or function impairment in Phactr1-deleted mice. Similarly, there were no more consequences of Phactr1 deletion during embryogenesis in endothelial cells. CONCLUSIONS: Loss of PHACTR-1 function in the cells involved in vascular physiology does not appear to induce a pathological vascular phenotype. The in vivo effect of the intronic variation described in genome-wide association studies is unlikely to involve downregulation in PHACTR-1 expression.


Subject(s)
Actins , Arterial Occlusive Diseases/metabolism , Fibromuscular Dysplasia , Microfilament Proteins/metabolism , Actins/metabolism , Animals , Endothelial Cells/metabolism , Fibromuscular Dysplasia/genetics , Genome-Wide Association Study , Mice , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Myocytes, Smooth Muscle/metabolism , Phosphoric Monoester Hydrolases/metabolism
4.
Article in English | MEDLINE | ID: mdl-35074794

ABSTRACT

The Wnt/frizzled signaling pathway is one of the major regulators of endothelial biology, controlling key cellular activities. Many secreted Wnt ligands have been identified and can initiate diverse signaling via binding to a complex set of Frizzled (Fzd) transmembrane receptors and coreceptors. Roughly, Wnt signaling is subdivided into two pathways: the canonical Wnt/ß-catenin signaling pathway whose main downstream effector is the transcriptional coactivator ß-catenin, and the noncanonical Wnt signaling pathway, which is subdivided into the Wnt/Ca2+ pathway and the planar cell polarity pathway. Here, we will focus on its cross talk with other angiogenic pathways and on its role in blood-retinal- and blood-brain-barrier formation and its maintenance in a differentiated state. We will unravel how retinal vascular pathologies and neurovascular degenerative diseases result from disruption of the Wnt pathway related to vascular instability, and highlight current research into therapeutic options.


Subject(s)
Blood-Brain Barrier , Wnt Signaling Pathway , Endothelium , Frizzled Receptors , Humans , Ligands
5.
Sci Rep ; 12(1): 8, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34996942

ABSTRACT

Heart failure is the final common stage of most cardiopathies. Cardiomyocytes (CM) connect with others via their extremities by intercalated disk protein complexes. This planar and directional organization of myocytes is crucial for mechanical coupling and anisotropic conduction of the electric signal in the heart. One of the hallmarks of heart failure is alterations in the contact sites between CM. Yet no factor on its own is known to coordinate CM polarized organization. We have previously shown that PDZRN3, an ubiquitine ligase E3 expressed in various tissues including the heart, mediates a branch of the Planar cell polarity (PCP) signaling involved in tissue patterning, instructing cell polarity and cell polar organization within a tissue. PDZRN3 is expressed in the embryonic mouse heart then its expression dropped significantly postnatally corresponding with heart maturation and CM polarized elongation. A moderate CM overexpression of Pdzrn3 (Pdzrn3 OE) during the first week of life, induced a severe eccentric hypertrophic phenotype with heart failure. In models of pressure-overload stress heart failure, CM-specific Pdzrn3 knockout showed complete protection against degradation of heart function. We reported that Pdzrn3 signaling induced PKC ζ expression, c-Jun nuclear translocation and a reduced nuclear ß catenin level, consistent markers of the planar non-canonical Wnt signaling in CM. We then show that subcellular localization (intercalated disk) of junction proteins as Cx43, ZO1 and Desmoglein 2 was altered in Pdzrn3 OE mice, which provides a molecular explanation for impaired CM polarization in these mice. Our results reveal a novel signaling pathway that controls a genetic program essential for heart maturation and maintenance of overall geometry, as well as the contractile function of CM, and implicates PDZRN3 as a potential therapeutic target for the prevention of human heart failure.


Subject(s)
Heart Failure/enzymology , Heart Failure/prevention & control , Heart/growth & development , Ubiquitin-Protein Ligases/metabolism , Animals , Heart Failure/genetics , Heart Failure/physiopathology , Humans , Male , Mice , Mice, Knockout , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/genetics , beta Catenin/genetics , beta Catenin/metabolism
6.
J Cereb Blood Flow Metab ; 42(4): 613-629, 2022 04.
Article in English | MEDLINE | ID: mdl-34644209

ABSTRACT

Blood brain barrier (BBB) disruption is a critical component of the pathophysiology of cognitive impairment of vascular etiology (VCI) and associated with Alzheimer's disease (AD). The Wnt pathway plays a crucial role in BBB maintenance, but there is limited data on its role in cognitive pathologies. The E3 ubiquitin ligase PDZRN3 is a regulator of the Wnt pathway. In a murine model of VCI, overexpressing Pdzrn3 in endothelial cell (EC) exacerbated BBB hyperpermeability and accelerated cognitive decline. We extended these observations, in both VCI and AD models, showing that EC-specific depletion of Pdzrn3, reinforced the BBB, with a decrease in vascular permeability and a subsequent spare in cognitive decline. We found that in cerebral vessels, Pdzrn3 depletion protects against AD-induced Wnt target gene alterations and enhances endothelial tight junctional proteins. Our results provide evidence that Wnt signaling could be a molecular link regulating BBB integrity and cognitive decline under VCI and AD pathologies.


Subject(s)
Alzheimer Disease , Blood-Brain Barrier , Ubiquitin-Protein Ligases , Alzheimer Disease/metabolism , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Homeostasis , Mice , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Development ; 147(24)2020 12 21.
Article in English | MEDLINE | ID: mdl-33144399

ABSTRACT

Sense organs acquire their distinctive shapes concomitantly with the differentiation of sensory cells and neurons necessary for their function. Although our understanding of the mechanisms controlling morphogenesis and neurogenesis in these structures has grown, how these processes are coordinated remains largely unexplored. Neurogenesis in the zebrafish olfactory epithelium requires the bHLH proneural transcription factor Neurogenin 1 (Neurog1). To address whether Neurog1 also controls morphogenesis, we analysed the migratory behaviour of early olfactory neural progenitors in neurog1 mutant embryos. Our results indicate that the oriented movements of these progenitors are disrupted in this context. Morphogenesis is similarly affected by mutations in the chemokine receptor gene, cxcr4b, suggesting it is a potential Neurog1 target gene. We find that Neurog1 directly regulates cxcr4b through an E-box cluster located just upstream of the cxcr4b transcription start site. Our results suggest that proneural transcription factors, such as Neurog1, directly couple distinct aspects of nervous system development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Morphogenesis/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Olfactory Mucosa/growth & development , Receptors, CXCR4/genetics , Zebrafish Proteins/genetics , Animals , E-Box Elements/genetics , Embryo, Nonmammalian , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Mutation/genetics , Neurons/metabolism , Transcription Initiation Site , Zebrafish/genetics , Zebrafish/growth & development
8.
Front Physiol ; 11: 861, 2020.
Article in English | MEDLINE | ID: mdl-32848833

ABSTRACT

Blood flow produces mechanical frictional forces, parallel to the blood flow exerted on the endothelial wall of the vessel, the so-called wall shear stress (WSS). WSS sensing is associated with several vascular pathologies, but it is first a physiological phenomenon. Endothelial cell sensitivity to WSS is involved in several developmental and physiological vascular processes such as angiogenesis and vascular morphogenesis, vascular remodeling, and vascular tone. Local conditions of blood flow determine the characteristics of WSS, i.e., intensity, direction, pulsatility, sensed by the endothelial cells that, through their effect of the vascular network, impact WSS. All these processes generate a local-global retroactive loop that determines the ability of the vascular system to ensure the perfusion of the tissues. In order to account for the physiological role of WSS, the so-called shear stress set point theory has been proposed, according to which WSS sensing acts locally on vessel remodeling so that WSS is maintained close to a set point value, with local and distant effects of vascular blood flow. The aim of this article is (1) to review the existing literature on WSS sensing involvement on the behavior of endothelial cells and its short-term (vasoreactivity) and long-term (vascular morphogenesis and remodeling) effects on vascular functioning in physiological condition; (2) to present the various hypotheses about WSS sensors and analyze the conceptual background of these representations, in particular the concept of tensional prestress or biotensegrity; and (3) to analyze the relevance, explanatory value, and limitations of the WSS set point theory, that should be viewed as dynamical, and not algorithmic, processes, acting in a self-organized way. We conclude that this dynamic set point theory and the biotensegrity concept provide a relevant explanatory framework to analyze the physiological mechanisms of WSS sensing and their possible shift toward pathological situations.

9.
FASEB J ; 34(1): 1288-1303, 2020 01.
Article in English | MEDLINE | ID: mdl-31914666

ABSTRACT

Retinopathies remain major causes of visual impairment in diabetic patients and premature infants. Introduction of anti-angiogenic drugs targeting vascular endothelial growth factor (VEGF) has transformed therapy for these proliferative retinopathies. However, limitations associated with anti-VEGF medications require to unravel new pathways of vessel growth to identify potential drug targets. Here, we investigated the role of Wnt/Frizzled-7 (Fzd7) pathway in a mouse model of oxygen-induced retinopathy (OIR). Using transgenic mice, which enabled endothelium-specific and time-specific Fzd7 deletion, we demonstrated that Fzd7 controls both vaso-obliteration and neovascular phases (NV). Deletion of Fzd7 at P12, after the ischemic phase of OIR, prevented formation of aberrant neovessels into the vitreous by suppressing proliferation of endothelial cells (EC) in tufts. Next we validated in vitro two Frd7 blocking strategies: a monoclonal antibody (mAbFzd7) against Fzd7 and a soluble Fzd7 receptor (CRD). In vivo a single intravitreal microinjection of mAbFzd7 or CRD significantly attenuated retinal neovascularization (NV) in mice with OIR. Molecular analysis revealed that Fzd7 may act through the activation of Wnt/ß-catenin and Jagged1 expression to control EC proliferation in extra-retinal neovessels. We identified Fzd7/ß-catenin signaling as new regulator of pathological retinal NV. Fzd7 appears to be a potent pharmacological target to prevent or treat aberrant angiogenesis of ischemic retinopathies.


Subject(s)
Diabetic Retinopathy/metabolism , Ischemia/metabolism , Repressor Proteins/metabolism , Retinal Neovascularization/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Gene Deletion , Ischemia/genetics , Ischemia/pathology , Jagged-1 Protein/biosynthesis , Jagged-1 Protein/genetics , Mice , Mice, Mutant Strains , Repressor Proteins/genetics , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , beta Catenin/genetics
10.
Elife ; 72018 06 28.
Article in English | MEDLINE | ID: mdl-29952749

ABSTRACT

Pitx2c, a homeodomain transcription factor, is classically known for its left-right patterning role. However, an early wave of pitx2 expression occurs at the onset of gastrulation in several species, indicating a possible earlier role that remains relatively unexplored. Here we show that in zebrafish, maternal-zygotic (MZ) pitx2c mutants exhibit a shortened body axis indicative of convergence and extension (CE) defects. Live imaging reveals that MZpitx2c mutants display less persistent mesendodermal migration during late stages of gastrulation. Transplant data indicate that Pitx2c functions cell non-autonomously to regulate this cell behavior by modulating cell shape and protrusive activity. Using transcriptomic analyses and candidate gene approaches, we identify transcriptional changes in components of the chemokine-ECM-integrin dependent mesendodermal migration network. Together, our results define pathways downstream of Pitx2c that are required during early embryogenesis and reveal novel functions for Pitx2c as a regulator of morphogenesis.


Subject(s)
Cell Movement/genetics , Embryonic Development/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Shape , Chemokines/genetics , Chemokines/metabolism , Embryo, Nonmammalian , Endoderm/cytology , Endoderm/metabolism , Epithelial Cells/cytology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gastrulation/genetics , Integrins/genetics , Integrins/metabolism , Mutation , Notochord/cytology , Notochord/metabolism , Time-Lapse Imaging , Transcription Factors/metabolism , Transcriptome , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Zygote/cytology , Zygote/growth & development , Zygote/metabolism
11.
Elife ; 72018 01 02.
Article in English | MEDLINE | ID: mdl-29292696

ABSTRACT

The zebrafish olfactory epithelium comprises a variety of neuronal populations, which are thought to have distinct embryonic origins. For instance, while ciliated sensory neurons arise from preplacodal ectoderm (PPE), previous lineage tracing studies suggest that both Gonadotropin releasing hormone 3 (Gnrh3) and microvillous sensory neurons derive from cranial neural crest (CNC). We find that the expression of Islet1/2 is restricted to Gnrh3 neurons associated with the olfactory epithelium. Unexpectedly, however, we find no change in Islet1/2+ cell numbers in sox10 mutant embryos, calling into question their CNC origin. Lineage reconstruction based on backtracking in time-lapse confocal datasets, and confirmed by photoconversion experiments, reveals that Gnrh3 neurons derive from the anterior PPE. Similarly, all of the microvillous sensory neurons we have traced arise from preplacodal progenitors. Our results suggest that rather than originating from separate ectodermal populations, cell-type heterogeneity is generated from overlapping pools of progenitors within the preplacodal ectoderm.


Subject(s)
Cell Lineage , Ectoderm/embryology , Neurons/physiology , Olfactory Mucosa/embryology , Zebrafish/embryology , Animals , Microscopy, Confocal , Time-Lapse Imaging
12.
Sci Signal ; 10(464)2017 01 31.
Article in English | MEDLINE | ID: mdl-28143902

ABSTRACT

Endothelial cells serve as a barrier between blood and tissues. Maintenance of the endothelial cell barrier depends on the integrity of intercellular junctions, which is regulated by a polarity complex that includes the ζ isoform of atypical protein kinase C (PKCζ) and partitioning defective 3 (PAR3). We revealed that the E3 ubiquitin ligase PDZ domain-containing ring finger 3 (PDZRN3) regulated endothelial intercellular junction integrity. Endothelial cell-specific overexpression of Pdzrn3 led to early embryonic lethality with severe hemorrhaging and altered organization of endothelial intercellular junctions. Conversely, endothelial-specific loss of Pdzrn3 prevented vascular leakage in a mouse model of transient ischemic stroke, an effect that was mimicked by pharmacological inhibition of PKCζ. PDZRN3 regulated Wnt signaling and associated with a complex containing PAR3, PKCζ, and the multi-PDZ domain protein MUPP1 (Discs Lost-multi-PDZ domain protein 1) and targeted MUPP1 for proteasomal degradation in transfected cells. Transient ischemic stroke increased the ubiquitination of MUPP1, and deficiency of MUPP1 in endothelial cells was associated with decreased localization of PKCζ and PAR3 at intercellular junctions. In endothelial cells, Pdzrn3 overexpression increased permeability through a PKCζ-dependent pathway. In contrast, Pdzrn3 depletion enhanced PKCζ accumulation at cell-cell contacts and reinforced the cortical actin cytoskeleton under stress conditions. These findings reveal how PDZRN3 regulates vascular permeability through a PKCζ-containing complex.


Subject(s)
Capillary Permeability , Endothelial Cells/metabolism , Intercellular Junctions , Protein Kinase C/metabolism , Ubiquitin-Protein Ligases/metabolism , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing , Animals , Brain/blood supply , Brain/embryology , Brain/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian/blood supply , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Endothelial Cells/cytology , Humans , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Membrane Proteins , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase C/genetics , Stroke/etiology , Stroke/genetics , Stroke/metabolism , Ubiquitin-Protein Ligases/genetics , Wnt Signaling Pathway/genetics
13.
Arterioscler Thromb Vasc Biol ; 36(12): 2369-2380, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27758766

ABSTRACT

OBJECTIVE: Vessel formation requires precise orchestration of a series of morphometric and molecular events controlled by a multitude of angiogenic factors and morphogens. Wnt/frizzled signaling is required for proper vascular formation. In this study, we investigated the role of the Fzd7 (frizzled-7) receptor in retinal vascular development and its relationship with the Wnt/ß-catenin canonical pathway and Notch signaling. APPROACH AND RESULTS: Using transgenic mice, we demonstrated that Fzd7 is required for postnatal vascular formation. Endothelial cell (EC) deletion of fzd7 (fzd7ECKO) delayed retinal plexus formation because of an impairment in tip cell phenotype and a decrease in stalk cell proliferation. Dvl (dishevelled) proteins are a main component of Wnt signaling and play a functionally redundant role. We found that Dvl3 depletion in dvl1-/- mice mimicked the fzd7ECKO vascular phenotype and demonstrated that Fzd7 acted via ß-catenin activation by showing that LiCl treatment rescued impairment in tip and stalk cell phenotypes induced in fzd7 mutants. Deletion of fzd7 or Dvl1/3 induced a strong decrease in Wnt canonical genes and Notch partners' expression. Genetic and pharmacological rescue strategies demonstrated that Fzd7 acted via ß-catenin activation, upstream of Notch signaling to control Dll4 and Jagged1 EC expression. CONCLUSIONS: Fzd7 expressed by EC drives postnatal angiogenesis via activation of Dvl/ß-catenin signaling and can control the integrative interaction of Wnt and Notch signaling during postnatal angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic , Receptors, G-Protein-Coupled/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Calcium-Binding Proteins , Cell Proliferation , Cells, Cultured , Dishevelled Proteins/deficiency , Dishevelled Proteins/genetics , Endothelial Cells/drug effects , Frizzled Receptors , Genotype , Intracellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/metabolism , Lithium Chloride/pharmacology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/drug effects , Phenotype , RNA Interference , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, Notch/metabolism , Retinal Neovascularization/genetics , Retinal Neovascularization/physiopathology , Retinal Vessels/drug effects , Transfection , Wnt Signaling Pathway/drug effects
14.
Mol Biol Cell ; 27(6): 941-53, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26792835

ABSTRACT

Angiogenesis involves the coordinated growth and migration of endothelial cells (ECs) toward a proangiogenic signal. The Wnt planar cell polarity (PCP) pathway, through the recruitment of Dishevelled (Dvl) and Dvl-associated activator of morphogenesis (Daam1), has been proposed to regulate cell actin cytoskeleton and microtubule (MT) reorganization for oriented cell migration. Here we report that Kif26b--a kinesin--and Daam1 cooperatively regulate initiation of EC sprouting and directional migration via MT reorganization. First, we find that Kif26b is recruited within the Dvl3/Daam1 complex. Using a three-dimensional in vitro angiogenesis assay, we show that Kif26b and Daam1 depletion impairs tip cell polarization and destabilizes extended vascular processes. Kif26b depletion specifically alters EC directional migration and mislocalized MT organizing center (MTOC)/Golgi and myosin IIB cell rear enrichment. Therefore the cell fails to establish a proper front-rear polarity. Of interest, Kif26b ectopic expression rescues the siDaam1 polarization defect phenotype. Finally, we show that Kif26b functions in MT stabilization, which is indispensable for asymmetrical cell structure reorganization. These data demonstrate that Kif26b, together with Dvl3/Daam1, initiates cell polarity through the control of PCP signaling pathway-dependent activation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Polarity , Dishevelled Proteins/metabolism , Endothelial Cells/metabolism , Kinesins/metabolism , Wnt Signaling Pathway , Animals , Cell Movement , Endothelial Cells/physiology , Humans , Mice , Microfilament Proteins , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Neovascularization, Physiologic , rho GTP-Binding Proteins
15.
Annu Rev Genet ; 49: 647-72, 2015.
Article in English | MEDLINE | ID: mdl-26442849

ABSTRACT

Although the left and right hemispheres of our brains develop with a high degree of symmetry at both the anatomical and functional levels, it has become clear that subtle structural differences exist between the two sides and that each is dominant in processing specific cognitive tasks. As the result of evolutionary conservation or convergence, lateralization of the brain is found in both vertebrates and invertebrates, suggesting that it provides significant fitness for animal life. This widespread feature of hemispheric specialization has allowed the emergence of model systems to study its development and, in some cases, to link anatomical asymmetries to brain function and behavior. Here, we present some of what is known about brain asymmetry in humans and model organisms as well as what is known about the impact of environmental and genetic factors on brain asymmetry development. We specifically highlight the progress made in understanding the development of epithalamic asymmetries in zebrafish and how this model provides an exciting opportunity to address brain asymmetry at different levels of complexity.


Subject(s)
Brain/anatomy & histology , Brain/physiology , Functional Laterality/physiology , Animals , Brain/embryology , Brain/growth & development , Epithalamus/anatomy & histology , Epithalamus/physiology , Functional Laterality/genetics , Hormones/metabolism , Humans , Language , Posture , Zebrafish/physiology
16.
Nat Commun ; 5: 4832, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25198863

ABSTRACT

Development and stabilization of a vascular plexus requires the coordination of multiple signalling processes. Wnt planar cell polarity (PCP) signalling is critical in vertebrates for diverse morphogenesis events, which coordinate cell orientation within a tissue-specific plane. However, its functional role in vascular morphogenesis is not well understood. Here we identify PDZRN3, an ubiquitin ligase, and report that Pdzrn3 deficiency impairs embryonic angiogenic remodelling and postnatal retinal vascular patterning, with a loss of two-dimensional polarized orientation of the intermediate retinal plexus. Using in vitro and ex vivo Pdzrn3 loss-of-function and gain-of-function experiments, we demonstrate a key role of PDZRN3 in endothelial cell directional and coordinated extension. PDZRN3 ubiquitinates Dishevelled 3 (Dvl3), to promote endocytosis of the Frizzled/Dvl3 complex, for PCP signal transduction. These results highlight the role of PDZRN3 to direct Wnt PCP signalling, and broadly implicate this pathway in the planar orientation and highly branched organization of vascular plexuses.


Subject(s)
Blood Vessels/embryology , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Morphogenesis/genetics , Neovascularization, Physiologic/genetics , Ubiquitin-Protein Ligases/genetics , Wnt Signaling Pathway/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blood Vessels/metabolism , Cell Polarity/genetics , Dishevelled Proteins , Endocytosis , Frizzled Receptors/metabolism , Mice , Mice, Knockout , Phosphoproteins/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Wnt Proteins/metabolism
17.
Cardiovasc Res ; 103(2): 291-303, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24866384

ABSTRACT

AIMS: Vascular permeability is essential for the health of normal tissues and is an important characteristic of many disease states. The role of the Wnt/frizzled pathway in vascular biology has recently been reported. The objectives of this study are to analyse the role of Frizzled7 (Fzd7) receptor in the control of vascular integrity. METHODS AND RESULTS: Fzd7 is expressed in endothelial cells and accumulates at the points of cell-cell contact in association with VE-cadherin and ß-catenin, two major adherens junction molecules. To selectively delete fzd7 in the vasculature, we developed gene targeting approaches using CreLox strategy in mice. Genetic fzd7 inhibition in the endothelium increases vascular permeability in basal and factor-induced conditions. On the cellular level, fzd7 knockdown or depletion leads to an increase in paracellular permeability with a loss of adherens junction organization. These impairments are associated with a decrease in both VE-Cadherin and ß-catenin expression, a decrease in their association and an increase of tyrosine phosphorylation of VE-cadherin/ß-catenin. Fzd7 transduces a Wnt/ß-catenin signalling cascade that is required to regulate ß-catenin and canonical target gene expression. Finally, LiCl, a GSK3 inhibitor, and ß-catenin overexpression rescued endothelial integrity and adherens junction organization, induced by fzd7 deletion. CONCLUSION: These findings establish that Fzd7 is a new partner of adherens junctional complex and represents a novel molecular switch for the control of vascular permeability via activation of the Wnt-canonical pathway.


Subject(s)
Cadherins/metabolism , Capillary Permeability/physiology , Endothelial Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Wnt Signaling Pathway/physiology , Animals , Cell Communication , Endothelium, Vascular/metabolism , Frizzled Receptors , Glycogen Synthase Kinase 3/metabolism , Intercellular Junctions/metabolism , Mice , Mice, Transgenic , Signal Transduction/physiology , beta Catenin/metabolism
18.
Development ; 141(7): 1572-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24598158

ABSTRACT

Left-right (L/R) asymmetries in the brain are thought to underlie lateralised cognitive functions. Understanding how neuroanatomical asymmetries are established has been achieved through the study of the zebrafish epithalamus. Morphological symmetry in the epithalamus is broken by leftward migration of the parapineal, which is required for the subsequent elaboration of left habenular identity; the habenular nuclei flank the midline and show L/R asymmetries in marker expression and connectivity. The Nodal target pitx2c is expressed in the left epithalamus, but nothing is known about its role during the establishment of asymmetry in the brain. We show that abrogating Pitx2c function leads to the right habenula adopting aspects of left character, and to an increase in parapineal cell numbers. Parapineal ablation in Pitx2c loss of function results in right habenular isomerism, indicating that the parapineal is required for the left character detected in the right habenula in this context. Partial parapineal ablation in the absence of Pitx2c, however, reduces the number of parapineal cells to wild-type levels and restores habenular asymmetry. We provide evidence suggesting that antagonism between Nodal and Pitx2c activities sets an upper limit on parapineal cell numbers. We conclude that restricting parapineal cell number is crucial for the correct elaboration of epithalamic asymmetry.


Subject(s)
Body Patterning/genetics , Habenula/embryology , Pineal Gland/embryology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Cell Count , Embryo, Nonmammalian , Epithalamus/cytology , Epithalamus/embryology , Habenula/cytology , Nodal Protein/physiology , Organ Size/genetics , Pineal Gland/cytology , Signal Transduction/physiology , Transcription Factors/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
19.
PLoS One ; 7(7): e42131, 2012.
Article in English | MEDLINE | ID: mdl-22860064

ABSTRACT

Thrombin receptor, F2R or PAR1 is a G-protein coupled receptor, located in the membrane of endothelial cells. It has been initially found to transduce signals in hemostasis, but recently also known to act in cancer and in vascular development. Mouse embryos lacking PAR1 function die from hemorrhages with varying frequency at midgestation. We have performed a survey of potential PAR1 homologs in the zebrafish genome and identified a teleost ortholog of mammalian PAR1. Knockdown of par1 function in zebrafish embryos demonstrates a requirement for Par1 in cardio-vascular development. Furthermore, we show that function of Par1 requires the presence of a phylogenetically conserved proteolytic cleavage site and a second intracellular domain. Altogether our results demonstrate a high degree of conservation of PAR1 proteins in the vertebrate lineage in respect to amino acid sequence as well as protein function.


Subject(s)
Cardiovascular System/embryology , Receptor, PAR-1/physiology , Zebrafish/embryology , Animals , Evolution, Molecular , Gene Knockdown Techniques , Heart Rate , Receptor, PAR-1/genetics , Regional Blood Flow
20.
Circ Res ; 110(1): 47-58, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22076635

ABSTRACT

RATIONALE: A growing body of evidence supports the hypothesis that the Wnt/planar cell polarity (PCP) pathway regulates endothelial cell proliferation and angiogenesis, but the components that mediate this regulation remain elusive. OBJECTIVE: We investigated the involvement of one of the receptors, Frizzled4 (Fzd4), in this process because its role has been implicated in retinal vascular development. METHODS AND RESULTS: We found that loss of fzd4 function in mice results in a striking reduction and impairment of the distal small artery network in the heart and kidney. We report that loss of fzd4 decreases vascular cell proliferation and migration and decreases the ability of the endothelial cells to form tubes. We show that fzd4 deletion induces defects in the expression level of stable acetylated tubulin and in Golgi organization during migration. Deletion of fzd4 favors Wnt noncanonical AP1-dependent signaling, indicating that Fzd4 plays a pivotal role favoring PCP signaling. Our data further demonstrate that Fzd4 is predominantly localized on the top of the plasma membrane, where it preferentially induces Dvl3 relocalization to promote its activation and α-tubulin recruitment during migration. In a pathological mouse angiogenic model, deletion of fzd4 impairs the angiogenic response and leads to the formation of a disorganized arterial network. CONCLUSIONS: These results suggest that Fzd4 is a major receptor involved in arterial formation and organization through a Wnt/PCP pathway.


Subject(s)
Arteries/cytology , Cell Polarity/physiology , Cell Proliferation , Frizzled Receptors/physiology , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Arteries/physiology , Arterioles/cytology , Arterioles/physiology , Cell Movement/physiology , Dishevelled Proteins , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Frizzled Receptors/genetics , Gene Expression Regulation, Developmental/physiology , Gene Knock-In Techniques , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Microtubules/physiology , Models, Animal , Phosphoproteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...