Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters








Publication year range
2.
Cell Rep Med ; 4(10): 101223, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37794584

ABSTRACT

Wet age-related macular degeneration (AMD), characterized by leaky neovessels emanating from the choroid, is a main cause of blindness. As current treatments for wet AMD require regular intravitreal injections of anti-vascular endothelial growth factor (VEGF) biologics, there is a need for the development of less invasive treatments. Here, we designed an allosteric inhibitor of end binding-3 (EB3) protein, termed EBIN, which reduces the effects of environmental stresses on endothelial cells by limiting pathological calcium signaling. Delivery of EBIN via eye drops in mouse and non-human primate (NHP) models of wet AMD prevents both neovascular leakage and choroidal neovascularization. EBIN reverses the epigenetic changes induced by environmental stresses, allowing an activation of a regenerative program within metabolic-active endothelial cells comprising choroidal neovascularization (CNV) lesions. These results suggest the therapeutic potential of EBIN in preventing the degenerative processes underlying wet AMD.


Subject(s)
Choroidal Neovascularization , Wet Macular Degeneration , Mice , Animals , Endothelial Cells/metabolism , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Wet Macular Degeneration/drug therapy , Wet Macular Degeneration/metabolism
3.
iScience ; 26(5): 106661, 2023 May 19.
Article in English | MEDLINE | ID: mdl-37168565

ABSTRACT

Endothelial cells (ECs) continuously sense and adapt to changes in shear stress generated by blood flow. Here, we show that the activation of the mechanosensitive channel Piezo1 by defined shear forces induces Ca2+ entry into the endoplasmic reticulum (ER) via the ER Ca2+ ATPase pump. This entry is followed by inositol trisphosphate receptor 2 (IP3R2)-elicited ER Ca2+ release into the cytosol. The mechanism of ER Ca2+ release involves the generation of cAMP by soluble adenylyl cyclase (sAC), leading to IP3R2-evoked Ca2+ gating. Depleting sAC or IP3R2 prevents ER Ca2+ release and blocks EC alignment in the direction of flow. Overexpression of constitutively active Akt1 restores the shear-induced alignment of ECs lacking Piezo1 or IP3R2, as well as the flow-induced vasodilation in endothelial restricted Piezo1 knockout mice. These studies describe an unknown Piezo1-cAMP-IP3R2 circuit as an essential mechanism activating Akt signaling and inducing adaptive changes in ECs to laminar flow.

4.
Sci Signal ; 14(679)2021 04 20.
Article in English | MEDLINE | ID: mdl-33879602

ABSTRACT

Chloride intracellular channels 1 (CLIC1) and 4 (CLIC4) are expressed in endothelial cells and regulate angiogenic behaviors in vitro, and the expression of Clic4 is important for vascular development and function in mice. Here, we found that CLIC1 and CLIC4 in endothelial cells regulate critical G protein-coupled receptor (GPCR) pathways associated with vascular development and disease. In cultured endothelial cells, we found that CLIC1 and CLIC4 transiently translocated to the plasma membrane in response to sphingosine 1-phosphate (S1P). Both CLIC1 and CLIC4 were essential for mediating S1P-induced activation of the small guanosine triphosphatase (GTPase) Rac1 downstream of S1P receptor 1 (S1PR1). In contrast, only CLIC1 was essential for S1P-induced activation of the small GTPase RhoA downstream of S1PR2 and S1PR3. Neither were required for other S1P-S1PR signaling outputs. Rescue experiments revealed that CLIC1 and CLIC4 were not functionally interchangeable, suggesting distinct and specific functions for CLICs in transducing GPCR signaling. These CLIC-mediated mechanisms were critical for S1P-induced stimulation of the barrier function in endothelial cell monolayers. Our results define CLICs as previously unknown players in the pathways linking GPCRs to small GTPases and vascular endothelial function.


Subject(s)
Chloride Channels/metabolism , Mitochondrial Proteins/metabolism , Neuropeptides , Sphingosine-1-Phosphate Receptors , rac1 GTP-Binding Protein , rhoA GTP-Binding Protein , Animals , Cell Line , Cells, Cultured , Endothelial Cells , Lysophospholipids , Mice , Neuropeptides/metabolism , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine , Sphingosine-1-Phosphate Receptors/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
5.
PLoS One ; 15(5): e0232338, 2020.
Article in English | MEDLINE | ID: mdl-32421702

ABSTRACT

End-binding proteins (EBs) associate with the growing microtubule plus ends to regulate microtubule dynamics as well as the interaction with intracellular structures. EB3 contributes to pathological vascular leakage through interacting with the inositol 1,4,5-trisphosphate receptor 3 (IP3R3), a calcium channel located at the endoplasmic reticulum membrane. The C-terminal domain of EB3 (residues 200-281) is functionally important for this interaction because it contains the effector binding sites, a prerequisite for EB3 activity and specificity. Structural data for this domain is limited. Here, we report the backbone chemical shift assignments for the human EB3 C-terminal domain and computationally explore its EB3 conformations. Backbone assignments, along with computational models, will allow future investigation of EB3 structural dynamics, interactions with effectors, and will facilitate the development of novel EB3 inhibitors.


Subject(s)
Microtubule-Associated Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Humans , Models, Molecular , Protein Domains , Protein Structure, Secondary
6.
Am J Respir Cell Mol Biol ; 62(2): 168-177, 2020 02.
Article in English | MEDLINE | ID: mdl-31409093

ABSTRACT

Disruption of alveolar-capillary barriers is a major complication of high-volume mechanical ventilation referred to as "ventilator-induced lung injury." The stretching force in alveoli is transmitted to endothelial cells, increasing the tension on underlying endothelial plasma membrane. The mechanosensor Piezo1, a plasma membrane cation channel, was inducibly deleted in endothelial cells of mice (Piezo1iEC-/-), which allowed us to study its role in regulating the endothelial barrier response to alveolar stretch. We observed significant increase in lung vascular permeability in Piezo1iEC-/- mice as compared with control Piezo1fl/fl mice in response to high-volume mechanical ventilation. We also observed that human lung endothelial monolayers depleted of Piezo1 and exposed to cyclic stretch had increased permeability. We identified the calcium-dependent cysteine protease calpain as a downstream target of Piezo1. Furthermore, we showed that calpain maintained stability of the endothelial barrier in response to mechanical stretch by cleaving Src kinase, which was responsible for disassembling endothelial adherens junctions. Pharmacological activation of calpain caused Src cleavage and thereby its inactivation, and it restored the disrupted lung endothelial barrier seen in Piezo1iEC-/- mice undergoing high-volume mechanical ventilation. Our data demonstrate that downregulation of Piezo1 signaling in endothelium is a critical factor in the pathogenesis of ventilator-induced lung injury, and thus augmenting Piezo1 expression or pharmacologically activating Piezo1 signaling may be an effective therapeutic strategy.


Subject(s)
Adherens Junctions/metabolism , Endothelial Cells/metabolism , Ion Channels/metabolism , Lung/metabolism , Animals , Capillary Permeability/drug effects , Cell Membrane/metabolism , Endothelium, Vascular/metabolism , Mice , Pulmonary Alveoli/metabolism , Ventilator-Induced Lung Injury/metabolism
7.
Am J Physiol Lung Cell Mol Physiol ; 317(3): L392-L401, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31313617

ABSTRACT

Here we describe a novel method for studying the protein "interactome" in primary human cells and apply this method to investigate the effect of posttranslational protein modifications (PTMs) on the protein's functions. We created a novel "biomimetic microsystem platform" (Bio-MSP) to isolate the protein complexes in primary cells by covalently attaching purified His-tagged proteins to a solid microscale support. Using this Bio-MSP, we have analyzed the interactomes of unphosphorylated and phosphomimetic end-binding protein-3 (EB3) in endothelial cells. Pathway analysis of these interactomes demonstrated the novel role of EB3 phosphorylation at serine 162 in regulating the protein's function. We showed that phosphorylation "switches" the EB3 biological network to modulate cellular processes such as cell-to-cell adhesion whereas dephosphorylation of this site promotes cell proliferation. This novel technique provides a useful tool to study the role of PTMs or single point mutations in activating distinct signal transduction networks and thereby the biological function of the protein in health and disease.


Subject(s)
Biomimetics , Endothelial Cells/metabolism , Endothelium/metabolism , Protein Processing, Post-Translational/physiology , Biomimetics/methods , Computational Biology/methods , Humans , Phosphorylation , Proteins/metabolism , Proteomics/methods , Signal Transduction/physiology
8.
Proc Natl Acad Sci U S A ; 116(26): 12980-12985, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31186359

ABSTRACT

Increased pulmonary microvessel pressure experienced in left heart failure, head trauma, or high altitude can lead to endothelial barrier disruption referred to as capillary "stress failure" that causes leakage of protein-rich plasma and pulmonary edema. However, little is known about vascular endothelial sensing and transduction of mechanical stimuli inducing endothelial barrier disruption. Piezo1, a mechanosensing ion channel expressed in endothelial cells (ECs), is activated by elevated pressure and other mechanical stimuli. Here, we demonstrate the involvement of Piezo1 in sensing increased lung microvessel pressure and mediating endothelial barrier disruption. Studies were made in mice in which Piezo1 was deleted conditionally in ECs (Piezo1iΔEC ), and lung microvessel pressure was increased either by raising left atrial pressure or by aortic constriction. We observed that lung endothelial barrier leakiness and edema induced by raising pulmonary microvessel pressure were abrogated in Piezo1iΔEC mice. Piezo1 signaled lung vascular hyperpermeability by promoting the internalization and degradation of the endothelial adherens junction (AJ) protein VE-cadherin. Breakdown of AJs was the result of activation of the calcium-dependent protease calpain and degradation of the AJ proteins VE-cadherin, ß-catenin, and p120-catenin. Deletion of Piezo1 in ECs or inhibition of calpain similarly prevented reduction in the AJ proteins. Thus, Piezo1 activation in ECs induced by elevated lung microvessel pressure mediates capillary stress failure and edema formation secondary to calpain-induced disruption of VE-cadherin adhesion. Inhibiting Piezo1 signaling may be a useful strategy to limit lung capillary stress failure injury in response to elevated vascular pressures.


Subject(s)
Endothelium, Vascular/pathology , Ion Channels/metabolism , Microvessels/pathology , Pulmonary Edema/pathology , Respiratory Insufficiency/pathology , Adherens Junctions/pathology , Adherens Junctions/ultrastructure , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Arterial Pressure/physiology , Blood Pressure/physiology , Cadherins/genetics , Cadherins/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Endothelium, Vascular/cytology , Endothelium, Vascular/ultrastructure , Female , Gene Knock-In Techniques , Humans , Hydrostatic Pressure/adverse effects , Intercellular Signaling Peptides and Proteins/pharmacology , Ion Channels/antagonists & inhibitors , Ion Channels/genetics , Lung/blood supply , Male , Mechanotransduction, Cellular , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Microvessels/cytology , Microvessels/drug effects , Primary Cell Culture , Pulmonary Edema/etiology , Pulmonary Edema/physiopathology , Respiratory Insufficiency/etiology , Respiratory Insufficiency/prevention & control , Spider Venoms/pharmacology
9.
J Cell Biol ; 218(1): 299-316, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30463880

ABSTRACT

Vascular endothelial (VE)-cadherin forms homotypic adherens junctions (AJs) in the endothelium, whereas N-cadherin forms heterotypic adhesion between endothelial cells and surrounding vascular smooth muscle cells and pericytes. Here we addressed the question whether both cadherin adhesion complexes communicate through intracellular signaling and contribute to the integrity of the endothelial barrier. We demonstrated that deletion of N-cadherin (Cdh2) in either endothelial cells or pericytes increases junctional endothelial permeability in lung and brain secondary to reduced accumulation of VE-cadherin at AJs. N-cadherin functions by increasing the rate of VE-cadherin recruitment to AJs and induces the assembly of VE-cadherin junctions. We identified the dual Rac1/RhoA Rho guanine nucleotide exchange factor (GEF) Trio as a critical component of the N-cadherin adhesion complex, which activates both Rac1 and RhoA signaling pathways at AJs. Trio GEF1-mediated Rac1 activation induces the recruitment of VE-cadherin to AJs, whereas Trio GEF2-mediated RhoA activation increases intracellular tension and reinforces Rac1 activation to promote assembly of VE-cadherin junctions and thereby establish the characteristic restrictive endothelial barrier.


Subject(s)
Adherens Junctions/metabolism , Cadherins/genetics , Endothelial Cells/metabolism , Guanine Nucleotide Exchange Factors/genetics , Pericytes/metabolism , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Adherens Junctions/ultrastructure , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Aorta/cytology , Aorta/metabolism , Brain/cytology , Brain/metabolism , Cadherins/deficiency , Cadherins/metabolism , Endothelial Cells/ultrastructure , Female , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/metabolism , Humans , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/metabolism , Pericytes/ultrastructure , Permeability , Phosphoproteins/metabolism , Primary Cell Culture , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
10.
Cell Mol Life Sci ; 74(22): 4189-4207, 2017 11.
Article in English | MEDLINE | ID: mdl-28803370

ABSTRACT

The endothelium, a monolayer of endothelial cells lining vessel walls, maintains tissue-fluid homeostasis by restricting the passage of the plasma proteins and blood cells into the interstitium. The ion Ca2+, a ubiquitous secondary messenger, initiates signal transduction events in endothelial cells that is critical to control of vascular tone and endothelial permeability. The ion Ca2+ is stored inside the intracellular organelles and released into the cytosol in response to environmental cues. The inositol 1,4,5-trisphosphate (IP3) messenger facilitates Ca2+ release through IP3 receptors which are Ca2+-selective intracellular channels located within the membrane of the endoplasmic reticulum. Binding of IP3 to the IP3Rs initiates assembly of IP3R clusters, a key event responsible for amplification of Ca2+ signals in endothelial cells. This review discusses emerging concepts related to architecture and dynamics of IP3R clusters, and their specific role in propagation of Ca2+ signals in endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Animals , Calcium/metabolism , Cytoskeleton/metabolism , Humans , Inositol 1,4,5-Trisphosphate/chemistry , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Kinase C-alpha/metabolism , Signal Transduction
11.
Circ Res ; 120(1): 179-206, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-28057793

ABSTRACT

The monolayer of endothelial cells lining the vessel wall forms a semipermeable barrier (in all tissue except the relatively impermeable blood-brain and inner retinal barriers) that regulates tissue-fluid homeostasis, transport of nutrients, and migration of blood cells across the barrier. Permeability of the endothelial barrier is primarily regulated by a protein complex called adherens junctions. Adherens junctions are not static structures; they are continuously remodeled in response to mechanical and chemical cues in both physiological and pathological settings. Here, we discuss recent insights into the post-translational modifications of junctional proteins and signaling pathways regulating plasticity of adherens junctions and endothelial permeability. We also discuss in the context of what is already known and newly defined signaling pathways that mediate endothelial barrier leakiness (hyperpermeability) that are important in the pathogenesis of cardiovascular and lung diseases and vascular inflammation.


Subject(s)
Adherens Junctions/metabolism , Capillary Permeability/physiology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Signal Transduction/physiology , Animals , Gap Junctions/metabolism , Humans , Protein Binding/physiology
13.
Cell Rep ; 12(1): 79-89, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26119739

ABSTRACT

The mechanisms by which the microtubule cytoskeleton regulates the permeability of endothelial barrier are not well understood. Here, we demonstrate that microtubule-associated end-binding protein 3 (EB3), a core component of the microtubule plus-end protein complex, binds to inositol 1,4,5-trisphosphate receptors (IP3Rs) through an S/TxIP EB-binding motif. In endothelial cells, α-thrombin, a pro-inflammatory mediator that stimulates phospholipase Cß, increases the cytosolic Ca(2+) concentration and elicits clustering of IP3R3s. These responses, and the resulting Ca(2+)-dependent phosphorylation of myosin light chain, are prevented by depletion of either EB3 or mutation of the TxIP motif of IP3R3 responsible for mediating its binding to EB3. We also show that selective EB3 gene deletion in endothelial cells of mice abrogates α-thrombin-induced increase in endothelial permeability. We conclude that the EB3-mediated interaction of IP3Rs with microtubules controls the assembly of IP3Rs into effective Ca(2+) signaling clusters, which thereby regulate microtubule-dependent endothelial permeability.


Subject(s)
Calcium Signaling , Endothelial Cells/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Microtubule-Associated Proteins/metabolism , Animals , CHO Cells , Capillary Permeability , Cricetinae , Cricetulus , HEK293 Cells , Humans , Mice , Microtubule-Associated Proteins/genetics , Protein Binding
14.
Stem Cell Reports ; 5(1): 10-21, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26120059

ABSTRACT

Understanding epigenetic mechanisms regulating embryonic stem cell (ESC) differentiation to endothelial cells may lead to increased efficiency of generation of vessel wall endothelial cells needed for vascular engineering. Here we demonstrated that the histone demethylases KDM4A and KDM4C played an indispensable but independent role in mediating the expression of fetal liver kinase (Flk)1 and VE-cadherin, respectively, and thereby the transition of mouse ESCs (mESCs) to endothelial cells. KDM4A was shown to bind to histones associated with the Flk1 promoter and KDM4C to bind to histones associated with the VE-cadherin promoter. KDM4A and KDM4C were also both required for capillary tube formation and vasculogenesis in mice. We observed in zebrafish that KDM4A depletion induced more severe vasculogenesis defects than KDM4C depletion, reflecting the early involvement of KDM4A in specifying endothelial cell fate. These findings together demonstrate the essential role of KDM4A and KDM4C in orchestrating mESC differentiation to endothelial cells through the activation of Flk1 and VE-cadherin promoters, respectively.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cell Differentiation/genetics , Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Antigens, CD/genetics , Cadherins/genetics , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Histones/genetics , Histones/metabolism , Mice , Promoter Regions, Genetic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish
15.
Appl Opt ; 54(8): 2113-7, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25968391

ABSTRACT

A femtosecond pulse train with THz repetition rate generated by the interference of two phase-modulated pulses has been recorded experimentally. Pulse repetition rates and their duration have been measured. It has been shown that at the 50-fs time delay between phase-modulated pulses the repetition rate is 3.1 THz with a pulse width of 200 fs, while at the 120-fs time delay the repetition rate is 7.1 THz with a pulse width of 67 fs.

18.
J Cell Biol ; 208(1): 23-32, 2015 01 05.
Article in English | MEDLINE | ID: mdl-25559184

ABSTRACT

The role of the RhoGTPase Rac1 in stabilizing mature endothelial adherens junctions (AJs) is not well understood. In this paper, using a photoactivatable probe to control Rac1 activity at AJs, we addressed the relationship between Rac1 and the dynamics of vascular endothelial cadherin (VE-cadherin). We demonstrated that Rac1 activation reduced the rate of VE-cadherin dissociation, leading to increased density of VE-cadherin at AJs. This response was coupled to a reduction in actomyosin-dependent tension across VE-cadherin adhesion sites. We observed that inhibiting myosin II directly or through photo-release of the caged Rho kinase inhibitor also reduced the rate of VE-cadherin dissociation. Thus, Rac1 functions by stabilizing VE-cadherin trans-dimers in mature AJs by counteracting the actomyosin tension. The results suggest a new model of VE-cadherin adhesive interaction mediated by Rac1-induced reduction of mechanical tension at AJs, resulting in the stabilization of VE-cadherin adhesions.


Subject(s)
Adherens Junctions/enzymology , Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/enzymology , rac1 GTP-Binding Protein/metabolism , Actomyosin/metabolism , Adherens Junctions/drug effects , Antigens, CD/genetics , Cadherins/genetics , Cell Adhesion , Cells, Cultured , Endothelial Cells/drug effects , Enzyme Activation , Humans , Kinetics , Microscopy, Fluorescence , Microscopy, Video , Models, Biological , Myosin Type II/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Protein Stability , Time-Lapse Imaging , Transfection , rac1 GTP-Binding Protein/genetics , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
19.
J Clin Invest ; 125(2): 652-64, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25574837

ABSTRACT

Vascular endothelial barrier dysfunction underlies diseases such as acute respiratory distress syndrome (ARDS), characterized by edema and inflammatory cell infiltration. The transcription factor HIF2α is highly expressed in vascular endothelial cells (ECs) and may regulate endothelial barrier function. Here, we analyzed promoter sequences of genes encoding proteins that regulate adherens junction (AJ) integrity and determined that vascular endothelial protein tyrosine phosphatase (VE-PTP) is a HIF2α target. HIF2α-induced VE-PTP expression enhanced dephosphorylation of VE-cadherin, which reduced VE-cadherin endocytosis and thereby augmented AJ integrity and endothelial barrier function. Mice harboring an EC-specific deletion of Hif2a exhibited decreased VE-PTP expression and increased VE-cadherin phosphorylation, resulting in defective AJs. Mice lacking HIF2α in ECs had increased lung vascular permeability and water content, both of which were further exacerbated by endotoxin-mediated injury. Treatment of these mice with Fg4497, a prolyl hydroxylase domain 2 (PHD2) inhibitor, activated HIF2α-mediated transcription in a hypoxia-independent manner. HIF2α activation increased VE-PTP expression, decreased VE-cadherin phosphorylation, promoted AJ integrity, and prevented the loss of endothelial barrier function. These findings demonstrate that HIF2α enhances endothelial barrier integrity, in part through VE-PTP expression and the resultant VE-cadherin dephosphorylation-mediated assembly of AJs. Moreover, activation of HIF2α/VE-PTP signaling via PHD2 inhibition has the potential to prevent the formation of leaky vessels and edema in inflammatory diseases such as ARDS.


Subject(s)
Acute Lung Injury/metabolism , Adherens Junctions/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Endothelial Cells/metabolism , Respiratory Distress Syndrome/metabolism , Signal Transduction , Acute Lung Injury/pathology , Adherens Junctions/genetics , Adherens Junctions/pathology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cadherins/genetics , Cadherins/metabolism , Cell Line , Endothelial Cells/pathology , Gene Expression Regulation, Enzymologic/genetics , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mice , Mice, Knockout , Phosphorylation/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/biosynthesis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/pathology
20.
Mol Cell ; 48(6): 914-25, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23159740

ABSTRACT

Vascular endothelial (VE)-cadherin homophilic adhesion controls endothelial barrier permeability through assembly of adherens junctions (AJs). We observed that loss of VE-cadherin-mediated adhesion induced the activation of Src and phospholipase C (PLC)γ2, which mediated Ca(2+) release from endoplasmic reticulum (ER) stores, resulting in activation of calcineurin (CaN), a Ca(2+)-dependent phosphatase. Downregulation of CaN activity induced phosphorylation of serine 162 in end binding (EB) protein 3. This phospho-switch was required to destabilize the EB3 dimer, suppress microtubule (MT) growth, and assemble AJs. The phospho-defective S162A EB3 mutant, in contrast, induced MT growth in confluent endothelial monolayers and disassembled AJs. Thus, VE-cadherin outside-in signaling regulates cytosolic Ca(2+) homeostasis and EB3 phosphorylation, which are required for assembly of AJs. These results identify a pivotal function of VE-cadherin homophilic interaction in modulating endothelial barrier through the tuning of MT dynamics.


Subject(s)
Adherens Junctions/metabolism , Antigens, CD/physiology , Cadherins/physiology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Processing, Post-Translational , Antigens, CD/metabolism , Cadherins/metabolism , Calcineurin/metabolism , Calcium/metabolism , Calcium Signaling , Calmodulin/metabolism , Cell Adhesion , Cells, Cultured , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Enzyme Activation , Homeostasis , Humans , Kinetics , Microscopy, Confocal , Phospholipase C gamma/metabolism , Phosphorylation , Protein Binding , Time-Lapse Imaging , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL