Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biomedicines ; 11(6)2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37371733

ABSTRACT

Vascular barrier dysfunction is characterized by increased permeability and inflammation of endothelial cells (ECs), which are prominent features of acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and sepsis, and a major complication of the SARS-CoV-2 infection and COVID-19. Functional impairment of the EC barrier and accompanying inflammation arises due to microbial toxins and from white blood cells of the lung as part of a defensive action against pathogens, ischemia-reperfusion or blood product transfusions, and aspiration syndromes-based injury. A loss of barrier function results in the excessive movement of fluid and macromolecules from the vasculature into the interstitium and alveolae resulting in pulmonary edema and collapse of the architecture and function of the lungs, and eventually culminates in respiratory failure. Therefore, EC barrier integrity, which is heavily dependent on cytoskeletal elements (mainly actin filaments, microtubules (MTs), cell-matrix focal adhesions, and intercellular junctions) to maintain cellular contacts, is a critical requirement for the preservation of lung function. EC cytoskeletal remodeling is regulated, at least in part, by Ser/Thr phosphorylation/dephosphorylation of key cytoskeletal proteins. While a large body of literature describes the role of phosphorylation of cytoskeletal proteins on Ser/Thr residues in the context of EC barrier regulation, the role of Ser/Thr dephosphorylation catalyzed by Ser/Thr protein phosphatases (PPases) in EC barrier regulation is less documented. Ser/Thr PPases have been proposed to act as a counter-regulatory mechanism that preserves the EC barrier and opposes EC contraction. Despite the importance of PPases, our knowledge of the catalytic and regulatory subunits involved, as well as their cellular targets, is limited and under-appreciated. Therefore, the goal of this review is to discuss the role of Ser/Thr PPases in the regulation of lung EC cytoskeleton and permeability with special emphasis on the role of protein phosphatase 1 (PP1) and protein phosphatase 2A (PP2A) as major mammalian Ser/Thr PPases. Importantly, we integrate the role of PPases with the structural dynamics of the cytoskeleton and signaling cascades that regulate endothelial cell permeability and inflammation.

2.
Sci Rep ; 6: 39018, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27976727

ABSTRACT

The goal of this study was to investigate the role of MLC phosphatase (MLCP) in a LPS model of acute lung injury (ALI). We demonstrate that ectopic expression of a constitutively-active (C/A) MLCP regulatory subunit (MYPT1) attenuates the ability of LPS to increase endothelial (EC) permeability. Down-regulation of MYPT1 exacerbates LPS-induced expression of ICAM1 suggesting an anti-inflammatory role of MLCP. To determine whether MLCP contributes to LPS-induced ALI in vivo, we utilized a nanoparticle DNA delivery method to specifically target lung EC. Expression of a C/A MYPT1 reduced LPS-induced lung inflammation and vascular permeability. Further, increased expression of the CS1ß (MLCP catalytic subunit) also reduced LPS-induced lung inflammation, whereas the inactive CS1ß mutant increased vascular leak. We next examined the role of the cytoskeletal targets of MLCP, the ERM proteins (Ezrin/Radixin/Moesin), in mediating barrier dysfunction. LPS-induced increase in EC permeability was accompanied by PKC-mediated increase in ERM phosphorylation, which was more prominent in CS1ß-depleted cells. Depletion of Moesin and Ezrin, but not Radixin attenuated LPS-induced increases in permeability. Further, delivery of a Moesin phospho-null mutant into murine lung endothelium attenuated LPS-induced lung inflammation and vascular leak suggesting that MLCP opposes LPS-induced ALI by mediating the dephosphorylation of Moesin and Ezrin.


Subject(s)
Acute Lung Injury , Cytoskeletal Proteins/metabolism , Endothelium, Vascular/metabolism , Lipopolysaccharides/toxicity , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Capillary Permeability/drug effects , Endothelium, Vascular/pathology , Humans , Mice , Phosphorylation
3.
Am J Respir Cell Mol Biol ; 50(3): 614-25, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24134589

ABSTRACT

Acute lung injury (ALI) is a severe hypoxemic respiratory insufficiency associated with lung leak, diffuse alveolar damage, inflammation, and loss of lung function. Decreased dimethylaminohydrolase (DDAH) activity and increases in asymmetric dimethylarginine (ADMA), together with exaggerated oxidative/nitrative stress, contributes to the development of ALI in mice exposed to LPS. Whether restoring DDAH function and suppressing ADMA levels can effectively ameliorate vascular hyperpermeability and lung injury in ALI is unknown, and was the focus of this study. In human lung microvascular endothelial cells, DDAH II overexpression prevented the LPS-dependent increase in ADMA, superoxide, peroxynitrite, and protein nitration. DDAH II also attenuated the endothelial barrier disruption associated with LPS exposure. Similarly, in vivo, we demonstrated that the targeted overexpression of DDAH II in the pulmonary vasculature significantly inhibited the accumulation of ADMA and the subsequent increase in oxidative/nitrative stress in the lungs of mice exposed to LPS. In addition, augmenting pulmonary DDAH II activity before LPS exposure reduced lung vascular leak and lung injury and restored lung function when DDAH activity was increased after injury. Together, these data suggest that enhancing DDAH II activity may prove a useful adjuvant therapy to treat patients with ALI.


Subject(s)
Acute Lung Injury/prevention & control , Amidohydrolases/metabolism , Endothelial Cells/enzymology , Genetic Therapy , Lipopolysaccharides , Lung/blood supply , Microvessels/enzymology , Pulmonary Edema/prevention & control , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Acute Lung Injury/genetics , Amidohydrolases/genetics , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Capillary Permeability , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Humans , Lung/enzymology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Microvessels/pathology , Oxidative Stress , Peroxynitrous Acid/metabolism , Pulmonary Edema/chemically induced , Pulmonary Edema/enzymology , Pulmonary Edema/genetics , Superoxides/metabolism , Time Factors , Transfection , Up-Regulation
4.
Am J Respir Cell Mol Biol ; 47(4): 445-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22582175

ABSTRACT

Antibiotics-induced release of the pore-forming virulence factor pneumolysin (PLY) in patients with pneumococcal pneumonia results in its presence days after lungs are sterile and is a major factor responsible for the induction of permeability edema. Here we sought to identify major mechanisms mediating PLY-induced endothelial dysfunction. We evaluated PLY-induced endothelial hyperpermeability in human lung microvascular endothelial cells (HL-MVECs) and human lung pulmonary artery endothelial cells in vitro and in mice instilled intratracheally with PLY. PLY increases permeability in endothelial monolayers by reducing stable and dynamic microtubule content and modulating VE-cadherin expression. These events, dependent upon an increased calcium influx, are preceded by protein kinase C (PKC)-α activation, perturbation of the RhoA/Rac1 balance, and an increase in myosin light chain phosphorylation. At later time points, PLY treatment increases the expression and activity of arginase in HL-MVECs. Arginase inhibition abrogates and suppresses PLY-induced endothelial barrier dysfunction by restoring NO generation. Consequently, a specific PKC-α inhibitor and the TNF-derived tonoplast intrinsic protein peptide, which blunts PLY-induced PKC-α activation, are able to prevent activation of arginase in HL-MVECs and to reduce PLY-induced endothelial hyperpermeability in mice. Arginase I (AI)(+/-)/arginase II (AII)(-/-) C57BL/6 mice, displaying a significantly reduced arginase I expression in the lungs, are significantly less sensitive to PLY-induced capillary leak than their wild-type or AI(+/+)/AII(-/-) counterparts, indicating an important role for arginase I in PLY-induced endothelial hyperpermeability. These results identify PKC-α and arginase I as potential upstream and downstream therapeutic targets in PLY-induced pulmonary endothelial dysfunction.


Subject(s)
Arginase/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Lung/pathology , Protein Kinase C-alpha/metabolism , Streptolysins/pharmacology , Animals , Antigens, CD/metabolism , Arginase/antagonists & inhibitors , Bacterial Proteins/pharmacology , Cadherins/metabolism , Calcium Signaling , Cells, Cultured , Endothelial Cells/enzymology , Enzyme Inhibitors/pharmacology , Humans , Lung/blood supply , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Microvessels/pathology , Pneumonia/enzymology , Pneumonia/immunology , Pneumonia/pathology , Protein Kinase C-alpha/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
5.
Vascul Pharmacol ; 56(1-2): 56-63, 2012.
Article in English | MEDLINE | ID: mdl-22074808

ABSTRACT

We have previously shown that the anti-cancer agent 2-methoxyestradiol (2ME) induces hyperpermeability across endothelial monolayers. Here, we show that both microtubule disruptor, 2ME, and microtubule stabilizer, paclitaxel (taxol), increase vascular lung permeability in vitro and in vivo. Simultaneous application of 2ME and taxol alleviates 2ME-induced endothelial barrier dysfunction, which is evident by the decreased Evans Blue Dye accumulation in lung tissue and increased transendothelial resistance across monolayers. 2ME significantly increases the level of p38 and MLC phosphorylation in both endothelial monolayers and murine lungs; this increase is suppressed in the presence of taxol. Taxol treatment leads to an immediate and sustained increase in tubulin acetylation in human pulmonary artery endothelial cells (HPAEC). Surprisingly, 2ME treatment also increases tubulin acetylation; however, the onset of this process is delayed and coincides with the stage of a partial barrier restoration in HPAEC monolayer. Inhibition of histone deacetylase 6 (HDAC6) with tubacin increases tubulin acetylation level, suppresses 2ME-induced HSP27 and MLC phosphorylation, and decreases 2ME-induced barrier dysfunction, suggesting barrier-protective and/or barrier-restorative role for tubulin acetylation in vascular endothelium.


Subject(s)
Capillary Permeability/drug effects , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Estradiol/analogs & derivatives , Paclitaxel/pharmacology , 2-Methoxyestradiol , Acetylation/drug effects , Anilides/pharmacology , Animals , Cells, Cultured , Drug Interactions , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Estradiol/pharmacology , HSP27 Heat-Shock Proteins/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Pulmonary Artery/metabolism , Tubulin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Respir Physiol Neurobiol ; 179(2-3): 334-7, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21907835

ABSTRACT

TIMAP is a regulatory subunit of protein phosphatase 1, whose role remains largely unknown. Our recent data suggested that TIMAP is involved in the regulation of barrier function in cultured pulmonary endothelial monolayers [Csortos et al., 2008. Am. J. Physiol. Lung Cell. Mol. Physiol. 295, L440-L450]. Here we showed that TIMAP depletion exacerbates lipopolysaccharide (LPS)-induced vascular leakage in murine lung, suggesting that TIMAP has a barrier-protective role in vivo. Real-Time RT PCR analysis revealed that treatment with LPS significantly suppressed Timap mRNA level. This suppression was not achieved via the down-regulation of Timap promoter activity, suggesting that LPS decreased Timap mRNA stability. Pretreatment with protein kinase A (PKA) inhibitor H-89 reduced TIMAP mRNA level, whereas pretreatment with PKA activator, bnz-cAMP, increased this level and attenuated LPS-induced decrease in TIMAP mRNA. Altogether, these data confirmed the barrier-protective role of TIMAP and suggested that barrier-disruptive and barrier-protective agents may employ modulation of TIMAP expression as a mechanism affecting barrier permeability.


Subject(s)
Acute Lung Injury/metabolism , Capillary Permeability/physiology , Endothelium, Vascular/metabolism , Lung/metabolism , Membrane Proteins/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Animals , Capillary Permeability/drug effects , Cells, Cultured , Down-Regulation , Humans , Lipopolysaccharides/toxicity , Lung/blood supply , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
7.
Am J Respir Cell Mol Biol ; 45(6): 1185-94, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21659656

ABSTRACT

We showed previously that microtubule disruptor 2-methoxyestradiol (2ME) induces hyperpermeability of the endothelial monolayer via mechanisms that include the activation of p38 and Rho kinase (ROCK) and rearrangement of the actin cytoskeleton. Using the protein kinase C (PKC) inhibitors Ro-31-7549 and Ro-32-0432, we show in vitro and in vivo that 2ME-induced barrier dysfunction is also PKC-dependent. The known PKC substrates ezrin, radixin, and moesin (ERM) were recently implicated in the regulation of endothelial permeability. This study tested the hypotheses that ERM proteins are phosphorylated in response to 2ME, and that this phosphorylation is involved in 2ME-induced barrier dysfunction. We show that the application of 2ME leads to a dramatic increase in the level of ERM phosphorylation. This increase is attenuated in cells pretreated with the microtubule stabilizer taxol. In human pulmonary artery endothelial cells (HPAECs), the phosphorylation of ERM occurs in a p38-dependent and PKC-dependent manner. The activation of p38 appears to occur upstream from the activation of PKC, in response to 2ME. Phosphorylated ERM are localized at the cell periphery during the early phase of response to 2ME (15 minutes), and colocalize with F-actin branching points during the later phase of response (60 minutes). Using the short interfering RNA approach, we also showed that individual ERM depletion significantly attenuates 2ME-induced hyperpermeability. HPAEC monolayers, depleted of ERM proteins and monolayers, overexpressing phosphorylation-deficient ERM mutants, exhibit less attenuation of 2ME-induced barrier disruption in response to the PKC inhibitor Ro-31-7549. These results suggest a critical role of PKC activation in response to microtubule-disrupting agents, and implicate the phosphorylation of ERM in the barrier dysfunction induced by 2ME.


Subject(s)
Capillary Permeability/drug effects , Cytoskeletal Proteins/metabolism , Endothelium, Vascular/metabolism , Estradiol/analogs & derivatives , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Tubulin Modulators/pharmacology , 2-Methoxyestradiol , Animals , Capillary Permeability/genetics , Cells, Cultured , Cytoskeletal Proteins/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Humans , Indoles/pharmacology , Male , Maleimides/pharmacology , Membrane Proteins/genetics , Mice , Microfilament Proteins/genetics , Paclitaxel/pharmacology , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Pyrroles/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
8.
J Biomed Biotechnol ; 2010: 671536, 2010.
Article in English | MEDLINE | ID: mdl-20445745

ABSTRACT

To understand how microtubules contribute to the dynamic reorganization of the endothelial cell (EC) cytoskeleton, we established an EC model expressing EB3-GFP, a protein that marks microtubule plus-ends. Using this model, we were able to measure microtubule growth rate at the centrosome region and near the cell periphery of a single human EC and in the EC monolayer. We demonstrate that the majority of microtubules in EC are dynamic, the growth rate of their plus-ends is highest in the internal cytoplasm, in the region of the centrosome. Growth rate of microtubule plus-ends decreases from the cell center toward the periphery. Our data suggest the existing mechanism(s) of local regulation of microtubule plus-ends growth in EC. Microtubule growth rate in the internal cytoplasm of EC in the monolayer is lower than that of single EC suggesting the regulatory effect of cell-cell contacts. Centrosomal microtubule growth rate distribution in single EC indicated the presence of two subpopulations of microtubules with "normal" (similar to those in monolayer EC) and "fast" (three times as much) growth rates. Our results indicate functional interactions between cell-cell contacts and microtubules.


Subject(s)
Endothelial Cells/physiology , Microtubules/physiology , Models, Biological , Cell Line , Centrosome/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Fluorescence , Microscopy, Video , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
J Cell Physiol ; 223(1): 215-23, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20054824

ABSTRACT

Extracellular beta-NAD is known to elevate intracellular levels of calcium ions, inositol 1,4,5-trisphate and cAMP. Recently, beta-NAD was identified as an agonist for P2Y1 and P2Y11 purinergic receptors. Since beta-NAD can be released extracellularly from endothelial cells (EC), we have proposed its involvement in the regulation of EC permeability. Here we show, for the first time, that endothelial integrity can be enhanced in EC endogenously expressing beta-NAD-activated purinergic receptors upon beta-NAD stimulation. Our data demonstrate that extracellular beta-NAD increases the transendothelial electrical resistance (TER) of human pulmonary artery EC (HPAEC) monolayers in a concentration-dependent manner indicating endothelial barrier enhancement. Importantly, beta-NAD significantly attenuated thrombin-induced EC permeability as well as the barrier-compromising effects of Gram-negative and Gram-positive bacterial toxins representing the barrier-protective function of beta-NAD. Immunofluorescence microscopy reveals more pronounced staining of cell-cell junctional protein VE-cadherin at the cellular periphery signifying increased tightness of the cell-cell contacts after beta-NAD stimulation. Interestingly, inhibitory analysis (pharmacological antagonists and receptor sequence specific siRNAs) indicates the participation of both P2Y1 and P2Y11 receptors in beta-NAD-induced TER increase. beta-NAD-treatment attenuates the lipopolysaccharide (LPS)-induced phosphorylation of myosin light chain (MLC) indicating its involvement in barrier protection. Our studies also show the involvement of cAMP-dependent protein kinase A and EPAC1 pathways as well as small GTPase Rac1 in beta-NAD-induced EC barrier enhancement. With these results, we conclude that beta-NAD regulates the pulmonary EC barrier integrity via small GTPase Rac1- and MLCP- dependent signaling pathways.


Subject(s)
Actins/metabolism , Capillary Permeability , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeleton/metabolism , Endothelial Cells/enzymology , Guanine Nucleotide Exchange Factors/metabolism , NAD/metabolism , Pulmonary Artery/enzymology , rac1 GTP-Binding Protein/metabolism , Antigens, CD/metabolism , Bacterial Proteins/pharmacology , Cadherins/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Electric Impedance , Endothelial Cells/drug effects , Guanine Nucleotide Exchange Factors/genetics , Humans , Intercellular Junctions/metabolism , Lipopolysaccharides/pharmacology , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Phosphorylation , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , RNA Interference , RNA, Messenger/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y1 , Signal Transduction , Streptolysins/pharmacology , Thrombin/metabolism , Time Factors , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...