Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Sci ; 133(9)2020 05 14.
Article in English | MEDLINE | ID: mdl-32198280

ABSTRACT

Endothelial barrier dysfunction leads to edema and vascular leak, causing high morbidity and mortality. Previously, Abl kinase inhibition has been shown to protect against vascular leak. Using the distinct inhibitory profiles of clinically available Abl kinase inhibitors, we aimed to provide a mechanistic basis for novel treatment strategies against vascular leakage syndromes. We found that the inhibitor bosutinib most potently protected against inflammation-induced endothelial barrier disruption. In vivo, bosutinib prevented lipopolysaccharide (LPS)-induced alveolar protein extravasation in an acute lung injury mice model. Mechanistically, mitogen-activated protein 4 kinase 4 (MAP4K4) was identified as important novel mediator of endothelial permeability, which signaled via ezrin, radixin and moesin proteins to increase turnover of integrin-based focal adhesions. The combined inhibition of MAP4K4 and Abl-related gene (Arg, also known as ABL2) by bosutinib preserved adherens junction integrity and reduced turnover of focal adhesions, which synergistically act to stabilize the endothelial barrier during inflammation. We conclude that MAP4K4 is an important regulator of endothelial barrier integrity, increasing focal adhesion turnover and disruption of cell-cell junctions during inflammation. Because it inhibits both Arg and MAP4K4, use of the clinically available drug bosutinib might form a viable strategy against vascular leakage syndromes.


Subject(s)
Focal Adhesions , Pharmaceutical Preparations , Adherens Junctions , Aniline Compounds , Animals , Capillary Permeability , Mice , Nitriles , Quinolines
2.
Sci Rep ; 9(1): 8131, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31148579

ABSTRACT

RhoGTPases regulate cytoskeletal dynamics, migration and cell-cell adhesion in endothelial cells. Besides regulation at the level of guanine nucleotide binding, they also undergo post-translational modifications, for example ubiquitination. RhoGTPases are ubiquitinated by Cullin RING ligases which are in turn regulated by neddylation. Previously we showed that inhibition of Cullin RING ligase activity by the neddylation inhibitor MLN4924 is detrimental for endothelial barrier function, due to accumulation of RhoB and the consequent induction of contractility. Here we analyzed the effect of pharmacological activation of Cullin RING ligases on endothelial barrier integrity in vitro and in vivo. CSN5i-3 induced endothelial barrier disruption and increased macromolecule leakage in vitro and in vivo. Mechanistically, CSN5i-3 strongly induced the expression and activation of RhoB and to lesser extent of RhoA in endothelial cells, which enhanced cell contraction. Elevated expression of RhoGTPases was a consequence of activation of the NF-κB pathway. In line with this notion, CSN5i-3 treatment decreased IκBα expression and increased NF-κB-mediated ICAM-1 expression and consequent adhesion of neutrophils to endothelial cells. This study shows that sustained neddylation of Cullin RING-ligases leads to activation the NF-κB pathway in endothelial cells, elevated expression of RhoGTPases, Rho/ROCK-dependent activation of MLC and disruption of the endothelial barrier.


Subject(s)
COP9 Signalosome Complex/metabolism , Endothelium, Vascular/metabolism , Inflammation , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Hydrolases/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , rhoB GTP-Binding Protein/metabolism , Animals , Cyclopentanes/pharmacology , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Adhesion Molecule-1/metabolism , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B p50 Subunit/metabolism , Neutrophils/metabolism , Pyrimidines/pharmacology , Ubiquitin/chemistry , Up-Regulation , Zebrafish
3.
Mol Biol Cell ; 30(5): 607-621, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30601691

ABSTRACT

Rho GTPases control both the actin cytoskeleton and adherens junction stability and are recognized as essential regulators of endothelial barrier function. They act as molecular switches and are primarily regulated by the exchange of GDP and GTP. However, posttranslational modifications such as phosphorylation, prenylation, and ubiquitination can additionally alter their localization, stability, and activity. F-box proteins are involved in the recognition of substrate proteins predestined for ubiquitination and subsequent degradation. Given the importance of ubiquitination, we studied the effect of the loss of 62 members of the F-box protein family on endothelial barrier function in human umbilical vein endothelial cells. Endothelial barrier function was quantified by electrical cell impedance sensing and macromolecule passage assay. Our RNA interference-based screen identified FBXW7 as a key regulator of endothelial barrier function. Mechanistically, loss of FBXW7 induced the accumulation of the RhoB GTPase in endothelial cells, resulting in their increased contractility and permeability. FBXW7 knockdown induced activation of the cholesterol biosynthesis pathway and changed the prenylation of RhoB. This effect was reversed by farnesyl transferase inhibitors and by the addition of geranylgeranyl pyrophosphate. In summary, this study identifies FBXW7 as a novel regulator of endothelial barrier function in vitro. Loss of FBXW7 indirectly modulates RhoB activity via alteration of the cholesterol biosynthesis pathway and, consequently, of the prenylation status and activity of RhoB, resulting in increased contractility and disruption of the endothelial barrier.


Subject(s)
Biosynthetic Pathways , Cholesterol/metabolism , F-Box-WD Repeat-Containing Protein 7/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Prenylation , rhoB GTP-Binding Protein/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/metabolism , Biosynthetic Pathways/drug effects , Cell Membrane Permeability/drug effects , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Models, Biological , RNA, Small Interfering/metabolism , Thrombin/pharmacology
4.
Small GTPases ; 10(6): 466-484, 2019 11.
Article in English | MEDLINE | ID: mdl-28949796

ABSTRACT

RhoGTPases are known regulators of intracellular actin dynamics that are important for maintaining endothelial barrier function. RhoA is most extensively studied as a key regulator of endothelial barrier function, however the function of the 2 highly homologous family-members (> 88%) RhoB and RhoC in endothelial barrier function is still poorly understood. This study aimed to determine whether RhoA, RhoB and RhoC have overlapping or distinct roles in barrier function and permeability in resting and activated endothelium. By using primary endothelial cells in combination with siRNA transfection to establish individual, double or triple knockdown of the RhoA/B/C RhoGTPases, we found that RhoB, but not RhoA or RhoC, is in resting endothelium a negative regulator of permeability. Loss of RhoB accounted for an accumulation of VE-cadherin at cell-cell contacts. Thrombin-induced loss of endothelial integrity is mediated primarily by RhoA and RhoB. Combined loss of RhoA/B showed decreased phosphorylation of Myosin Light Chain and increased expression of VE-cadherin at cell-cell contacts after thrombin stimulation. RhoC contributes to the Rac1-dependent restoration of endothelial barrier function. In summary, this study shows that these highly homologous RhoGTPases differentially control the dynamics of endothelial barrier function.


Subject(s)
Human Umbilical Vein Endothelial Cells/physiology , rhoA GTP-Binding Protein/physiology , rhoB GTP-Binding Protein/physiology , rhoC GTP-Binding Protein/physiology , Actins/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Communication , Cells, Cultured , Humans , RNA, Small Interfering , Thrombin/pharmacology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , rhoB GTP-Binding Protein/genetics , rhoC GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...