Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Signal ; 28(6): 541-51, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26923917

ABSTRACT

Endothelial cell (EC) barrier disruption induced by edemagenic agonists such as thrombin is a result of increased actomyosin contraction and enforcement of focal adhesions (FA) anchoring contracting stress fibers, which leads to cell retraction and force-induced disruption of cell junctions. In turn, EC barrier enhancement by oxidized phospholipids (OxPAPC) and other agonists is a result of increased tethering forces due to enforcement of the peripheral actin rim and enhancement of cell-cell adherens junction (AJ) complexes promoting EC barrier integrity. This study tested participation of the mechanosensitive adaptor, vinculin, which couples FA and AJ to actin cytoskeleton, in control of the EC permeability response to barrier disruptive (thrombin) and barrier enhancing (OxPAPC) stimulation. OxPAPC and thrombin induced different patterns of FA remodeling. Knockdown of vinculin attenuated both, OxPAPC-induced decrease and thrombin-induced increase in EC permeability. Thrombin stimulated the vinculin association with FA protein talin and suppressed the interaction with AJ protein, VE-cadherin. In contrast, OxPAPC stimulated the vinculin association with VE-cadherin. Thrombin and OxPAPC induced different levels of myosin light chain (MLC) phosphorylation and caused different patterns of intracellular phospho-MLC distribution. Thrombin-induced talin-vinculin and OxPAPC-induced VE-cadherin-vinculin association were abolished by myosin inhibitor blebbistatin. Expression of the vinculin mutant unable to interact with actin attenuated EC permeability changes and MLC phosphorylation caused by both, thrombin and OxPAPC. These data suggest that the specific vinculin interaction with FA or AJ in different contexts of agonist stimulation is defined by development of regional actyomyosin-based tension and participates in both, the barrier-disruptive and barrier-enhancing endothelial responses.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Vinculin/physiology , Cadherins/metabolism , Cell Adhesion , Cell Membrane Permeability/drug effects , Cells, Cultured , Cytoskeleton/metabolism , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Focal Adhesions/metabolism , Humans , Myosin Light Chains/metabolism , Phospholipids/pharmacology , Talin/metabolism , Thrombin/pharmacology , Vinculin/metabolism
2.
Am J Physiol Lung Cell Mol Physiol ; 308(5): L452-63, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25539852

ABSTRACT

Increased vascular endothelial permeability and inflammation are major pathological mechanisms of pulmonary edema and its life-threatening complication, the acute respiratory distress syndrome (ARDS). We have previously described potent protective effects of hepatocyte growth factor (HGF) against thrombin-induced hyperpermeability and identified the Rac pathway as a key mechanism of HGF-mediated endothelial barrier protection. However, anti-inflammatory effects of HGF are less understood. This study examined effects of HGF on the pulmonary endothelial cell (EC) inflammatory activation and barrier dysfunction caused by the gram-negative bacterial pathogen lipopolysaccharide (LPS). We tested involvement of the novel Rac-specific guanine nucleotide exchange factor Asef in the HGF anti-inflammatory effects. HGF protected the pulmonary EC monolayer against LPS-induced hyperpermeability, disruption of monolayer integrity, activation of NF-kB signaling, expression of adhesion molecules intercellular adhesion molecule-1 and vascular cell adhesion molecule-1, and production of IL-8. These effects were critically dependent on Asef. Small-interfering RNA-induced downregulation of Asef attenuated HGF protective effects against LPS-induced EC barrier failure. Protective effects of HGF against LPS-induced lung inflammation and vascular leak were also diminished in Asef knockout mice. Taken together, these results demonstrate potent anti-inflammatory effects by HGF and delineate a key role of Asef in the mediation of the HGF barrier protective and anti-inflammatory effects. Modulation of Asef activity may have important implications in therapeutic strategies aimed at the treatment of sepsis and acute lung injury/ARDS-induced gram-negative bacterial pathogens.


Subject(s)
Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Guanine Nucleotide Exchange Factors/metabolism , Hepatocyte Growth Factor/pharmacology , Lung Injury/pathology , Lung Injury/physiopathology , Cell Adhesion/drug effects , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Humans , Inflammation/pathology , Lipopolysaccharides , Lung/drug effects , Lung/pathology , Lung/physiopathology , Neutrophils/cytology , Neutrophils/drug effects , Protective Agents/pharmacology , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/drug effects
3.
PLoS One ; 9(9): e105912, 2014.
Article in English | MEDLINE | ID: mdl-25198505

ABSTRACT

Microtubules (MT) play a vital role in many cellular functions, but their role in peripheral actin cytoskeletal dynamics which is essential for control of endothelial barrier and monolayer integrity is less understood. We have previously described the enhancement of lung endothelial cell (EC) barrier by hepatocyte growth factor (HGF) which was associated with Rac1-mediated remodeling of actin cytoskeleton. This study investigated involvement of MT-dependent mechanisms in the HGF-induced enhancement of EC barrier. HGF-induced Rac1 activation was accompanied by phosphorylation of stathmin, a regulator of MT dynamics. HGF also stimulated MT peripheral growth monitored by time lapse imaging and tracking analysis of EB-1-decorated MT growing tips, and increased the pool of acetylated tubulin. These effects were abolished by EC pretreatment with HGF receptor inhibitor, downregulation of Rac1 pathway, or by expression of a stathmin-S63A phosphorylation deficient mutant. Expression of stathmin-S63A abolished the HGF protective effects against thrombin-induced activation of RhoA cascade, permeability increase, and EC barrier dysfunction. These results demonstrate a novel MT-dependent mechanism of HGF-induced EC barrier regulation via Rac1/PAK1/stathmin-dependent control of MT dynamics.


Subject(s)
Endothelium, Vascular/physiology , Hepatocyte Growth Factor/physiology , Lung/physiology , Microtubules/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Lung/metabolism , Phosphorylation , Stathmin/metabolism , Subcellular Fractions/metabolism , rac1 GTP-Binding Protein/physiology
4.
Cell Signal ; 26(11): 2306-16, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25101856

ABSTRACT

Previous reports described an important role of hepatocyte growth factor (HGF) in mitigation of pulmonary endothelial barrier dysfunction and cell injury induced by pathologic agonists and mechanical forces. HGF protective effects have been associated with Rac-GTPase signaling pathway activated by Rac-specific guanine nucleotide exchange factor Tiam1 and leading to enhancement of intercellular adherens junctions. This study tested involvement of a novel Rac-specific activator, Asef, in endothelial barrier enhancement by HGF and investigated a mechanism of HGF-induced Asef activation. Si-RNA-based knockdown of Tiam1 and Asef had an additive effect on attenuation of HGF-induced Rac activation and endothelial cell (EC) barrier enhancement. Tiam1 and Asef activation was abolished by pharmacologic inhibitors of HGF receptor and PI3-kinase. In contrast to Tiam1, Asef interacted with APC and associated with microtubule fraction upon HGF stimulation. EC treatment by low dose nocodazole to inhibit peripheral microtubule dynamics partially attenuated HGF-induced Asef peripheral translocation, but had negligible effect on Tiam1 translocation. These effects were associated with attenuation of HGF-induced barrier enhancement in EC pretreated with low ND dose and activation of Rac and its cytoskeletal effectors PAK1 and cortactin. These data demonstrate, that in addition to microtubule-independent Tiam1 activation, HGF engages additional microtubule- and APC-dependent pathway of Asef activation. These mechanisms may complement each other to provide the fine tuning of Rac signaling and endothelial barrier enhancement in response to various agonists.


Subject(s)
Endothelium, Vascular/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Hepatocyte Growth Factor/metabolism , Signal Transduction/physiology , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Cell Line , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Guanine Nucleotide Exchange Factors/genetics , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/genetics , Humans , Microtubules/genetics , Microtubules/metabolism , Nocodazole/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/drug effects , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Tubulin Modulators , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
5.
PLoS One ; 9(6): e100169, 2014.
Article in English | MEDLINE | ID: mdl-24940746

ABSTRACT

RATIONALE: Growth arrest DNA damage inducible alpha (GADD45a) is a stress-induced gene we have shown to participate in the pathophysiology of ventilator-induced lung injury (VILI) via regulation of mechanical stress-induced Akt ubiquitination and phosphorylation. The regulation of GADD45a expression by mechanical stress and its relationship with acute lung injury (ALI) susceptibility and severity, however, remains unknown. OBJECTIVES: We examined mechanical stress-dependent regulatory elements (MSRE) in the GADD45a promoter and the contribution of promoter polymorphisms in GADD45a expression and ALI susceptibility. METHODS AND RESULTS: Initial studies in GADD45a knockout and heterozygous mice confirmed the relationship of GADD45a gene dose to VILI severity. Human lung endothelial cells (EC) transfected with a luciferase vector containing the full length GADD45a promoter sequence (-771 to +223) demonstrated a >4 fold increase in GADD45a expression in response to 18% cyclic stretch (CS, 4 h) compared to static controls while specific promoter regions harboring CS-dependent MSRE were identified using vectors containing serial deletion constructs of the GADD45a promoter. In silico analyses of GADD45a promoter region (-371 to -133) revealed a potential binding site for specificity protein 1 (SP1), a finding supported by confirmed SP1 binding with the GADD45a promoter and by the significant attenuation of CS-dependent GADD45a promoter activity in response to SP1 silencing. Separately, case-control association studies revealed a significant association of a GADD45a promoter SNP at -589 (rs581000, G>C) with reduced ALI susceptibility. Subsequently, we found allelic variation of this SNP is associated with both differential GADD45a expression in mechanically stressed EC (18% CS, 4 h) and differential binding site of interferon regulatory factor 7 (IRF7) at this site. CONCLUSION: These results strongly support a functional role for GADD45a in ALI/VILI and identify a specific gene variant that confers risk for ALI.


Subject(s)
Acute Lung Injury/genetics , Cell Cycle Proteins/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic , Ventilator-Induced Lung Injury/genetics , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Adult , Aged , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Dosage , Gene Expression Regulation , Genes, Reporter , Heterozygote , Humans , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Luciferases/genetics , Luciferases/metabolism , Lung/metabolism , Lung/pathology , Male , Mechanotransduction, Cellular , Mice , Mice, Knockout , Middle Aged , Nuclear Proteins/metabolism , Polymorphism, Single Nucleotide , Protein Binding , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Stress, Mechanical , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology
6.
Am J Respir Cell Mol Biol ; 50(2): 409-18, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24053186

ABSTRACT

Increased lung vascular permeability and alveolar edema are cardinal features of inflammatory conditions such as acute respiratory distress syndrome (ARDS) and ventilator-induced lung injury (VILI). We previously demonstrated that pre-B-cell colony-enhancing factor (PBEF)/NAMPT, the proinflammatory cytokine encoded by NAMPT, participates in ARDS and VILI inflammatory syndromes. The present study evaluated posttranscriptional regulation of PBEF/NAMPT gene expression in human lung endothelium via 3'-untranslated region (UTR) microRNA (miRNA) binding. In silico analysis identified hsa-miR-374a and hsa-miR-568 as potential miRNA candidates. Increased PBEF/NAMPT transcription (by RT-PCR) and expression (by Western blotting) induced by 18% cyclic stretch (CS) (2 h: 3.4 ± 0.06 mRNA fold increase (FI); 10 h: 1.5 ± 0.06 protein FI) and by LPS (4 h: 3.8 ± 0.2 mRNA FI; 48 h: 2.6 ± 0.2 protein FI) were significantly attenuated by transfection with mimics of hsa-miR-374a or hsa-miR-568 (40-60% reductions each). LPS and 18% CS increased the activity of a PBEF/NAMPT 3'-UTR luciferase reporter (2.4-3.25 FI) with induction reduced by mimics of each miRNA (44-60% reduction). Specific miRNA inhibitors (antagomirs) for each PBEF/NAMPT miRNA significantly increased the endogenous PBEF/NAMPT mRNA (1.4-3.4 ± 0.1 FI) and protein levels (1.2-1.4 ± 0.1 FI) and 3'-UTR luciferase activity (1.4-1.7 ± 0.1 FI) compared with negative antagomir controls. Collectively, these data demonstrate that increased PBEF/NAMPT expression induced by bioactive agonists (i.e., excessive mechanical stress, LPS) involves epigenetic regulation with hsa-miR-374a and hsa-miR-568, representing novel therapeutic strategies to reduce inflammatory lung injury.


Subject(s)
Cytokines/metabolism , Endothelium/metabolism , Epigenesis, Genetic , Lung/metabolism , MicroRNAs/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , RNA, Messenger/genetics , Stress, Mechanical , Ventilator-Induced Lung Injury/genetics , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Lipopolysaccharides/pharmacology , MicroRNAs/drug effects , MicroRNAs/genetics , RNA, Messenger/metabolism , Respiratory Distress Syndrome/genetics , Ventilator-Induced Lung Injury/metabolism
7.
Chem Phys Lipids ; 175-176: 9-19, 2013.
Article in English | MEDLINE | ID: mdl-23911706

ABSTRACT

The lipid membrane not only provides a rich interface with an array of receptor signaling complexes with which a cell communicates, but it also serves as a source of lipid derived bioactive molecules. In pathologic conditions of acute lung injury (ALI) associated with activation of oxidative stress, unsaturated phosphatidyl cholines overlooking a luminal space undergo oxidation leading to generation of fragmented phospholipids such as 1-palmitoyl-2-hydroxy-sn-glycero-3-phosphocholine (lysoPC), or 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (PAPC) full length oxygenation products (oxPAPC). Using Langmuir monolayers as models of the lipid bilayer, we evaluated the propensity of these phospholipids to solubilize from the cell membrane. The results suggest that lysoPC is rapidly released as it is produced, while oxPAPC has a longer membrane bound lifetime. After being released from cell membranes, these oxidized phospholipids exhibit potent agonist-like effects on neighboring cells. Therefore, we correlate the presence of the two phospholipid groups with the onset and resolution of increased vascular leakiness associated with ALI through testing their effect on vascular endothelial barrier integrity. Our work shows that cells respond differently to these two groups of products of phosphatidyl choline oxidation. LysoPC disrupts cell-cell junctions and increases endothelial permeability while oxPAPC enhances endothelial barrier. These data suggest a model whereby rapid release of lysoPC results in onset of ALI associated vascular leak, and the release of a reserve of oxPAPC as oxidative stress subsides restores the vascular barrier properties.


Subject(s)
Acute Lung Injury/metabolism , Endothelium, Vascular/pathology , Lysophosphatidylcholines/metabolism , Phosphatidylcholines/metabolism , Pulmonary Artery/pathology , Acute Lung Injury/pathology , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Kinetics , Oxidation-Reduction , Permeability , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Thermodynamics
8.
Am J Physiol Lung Cell Mol Physiol ; 305(3): L240-55, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23729486

ABSTRACT

Endothelial cell (EC) barrier disruption induced by inflammatory agonists such as thrombin leads to potentially lethal physiological dysfunction such as alveolar flooding, hypoxemia, and pulmonary edema. Thrombin stimulates paracellular gap and F-actin stress fiber formation, triggers actomyosin contraction, and alters EC permeability through multiple mechanisms that include protein kinase C (PKC) activation. We previously have shown that the ezrin, radixin, and moesin (ERM) actin-binding proteins differentially participate in sphingosine-1 phosphate-induced EC barrier enhancement. Phosphorylation of a conserved threonine residue in the COOH-terminus of ERM proteins causes conformational changes in ERM to unmask binding sites and is considered a hallmark of ERM activation. In the present study we test the hypothesis that ERM proteins are phosphorylated on this critical threonine residue by thrombin-induced signaling events and explore the role of the ERM family in modulating thrombin-induced cytoskeletal rearrangement and EC barrier function. Thrombin promotes ERM phosphorylation at this threonine residue (ezrin Thr567, radixin Thr564, moesin Thr558) in a PKC-dependent fashion and induces translocation of phosphorylated ERM to the EC periphery. Thrombin-induced ERM threonine phosphorylation is likely synergistically mediated by protease-activated receptors PAR1 and PAR2. Using the siRNA approach, depletion of either moesin alone or of all three ERM proteins significantly attenuates thrombin-induced increase in EC barrier permeability (transendothelial electrical resistance), cytoskeletal rearrangements, paracellular gap formation, and accumulation of phospho-myosin light chain. In contrast, radixin depletion exerts opposing effects on these indexes. These data suggest that ERM proteins play important differential roles in the thrombin-induced modulation of EC permeability, with moesin promoting barrier dysfunction and radixin opposing it.


Subject(s)
Cytoskeletal Proteins/metabolism , Endothelial Cells/physiology , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Thrombin/metabolism , Capillary Permeability , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , Electric Impedance , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Inflammation , Membrane Proteins/genetics , Microfilament Proteins/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering , Signal Transduction
9.
Am J Respir Cell Mol Biol ; 49(1): 58-66, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23492194

ABSTRACT

Increased lung vascular permeability, the consequence of endothelial cell (EC) barrier dysfunction, is a cardinal feature of inflammatory conditions such as acute lung injury and sepsis and leads to lethal physiological dysfunction characterized by alveolar flooding, hypoxemia, and pulmonary edema. We previously demonstrated that the nonmuscle myosin light chain kinase isoform (nmMLCK) plays a key role in agonist-induced pulmonary EC barrier regulation. The present study evaluated posttranscriptional regulation of MYLK expression, the gene encoding nmMLCK, via 3' untranslated region (UTR) binding by microRNAs (miRNAs) with in silico analysis identifying hsa-miR-374a, hsa-miR-374b, hsa-miR-520c-3p, and hsa-miR-1290 as miRNA candidates. We identified increased MYLK gene transcription induced by TNF-α (24 h; 4.7 ± 0.45 fold increase [FI]), LPS (4 h; 2.85 ± 0.15 [FI]), and 18% cyclic stretch (24 h; 4.6 ± 0.24 FI) that was attenuated by transfection of human lung ECs with mimics of hsa-miR-374a, hsa-miR-374b, hsa-miR-520c-3p, or hsa-miR-1290 (20-80% reductions by each miRNA). TNF-α, LPS, and 18% cyclic stretch each increased the activity of a MYLK 3'UTR luciferase reporter (2.5-7.0 FI) with induction reduced by mimics of each miRNA (30-60% reduction). MiRNA inhibitors (antagomirs) for each MYLK miRNA significantly increased 3'UTR luciferase activity (1.2-2.3 FI) and rescued the decreased MLCK-3'UTR reporter activity produced by miRNA mimics (70-110% increases for each miRNA; P < 0.05). These data demonstrate that increased human lung EC expression of MYLK by bioactive agonists (excessive mechanical stress, LPS, TNF-α) is regulated in part by specific miRNAs (hsa-miR-374a, hsa-miR-374b, hsa-miR-520c-3p, and hsa-miR-1290), representing a novel therapeutic strategy for reducing inflammatory lung injury.


Subject(s)
Calcium-Binding Proteins/metabolism , Endothelium/enzymology , MicroRNAs/metabolism , Myosin-Light-Chain Kinase/metabolism , 3' Untranslated Regions , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biomimetics , Calcium-Binding Proteins/genetics , Capillary Permeability , Cell Line , Endothelium/pathology , Gene Expression Regulation , Genes, Reporter , Humans , Lipopolysaccharides/pharmacology , Luciferases/metabolism , Lung/enzymology , Lung/pathology , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myosin-Light-Chain Kinase/genetics , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Stress, Mechanical , Transcription, Genetic , Transfection , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
10.
Cell Signal ; 23(12): 2086-96, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21864676

ABSTRACT

Endothelial cell (EC) barrier dysfunction induced by inflammatory agonists is a frequent pathophysiologic event in multiple diseases. The platelet-derived phospholipid sphingosine-1 phosphate (S1P) reverses this dysfunction by potently enhancing the EC barrier through a process involving Rac GTPase-dependent cortical actin rearrangement as an integral step. In this study we explored the role of the ezrin, radixin, and moesin (ERM) family of actin-binding linker protein in modulating S1P-induced human pulmonary EC barrier enhancement. S1P induces ERM translocation to the EC periphery and promotes ERM phosphorylation on a critical threonine residue (Ezrin-567, Radixin-564, Moesin-558). This phosphorylation is dependent on activation of PKC isoforms and Rac1. The majority of ERM phosphorylation on these critical threonine residues after S1P occurs in moesin and ezrin. Baseline radixin phosphorylation is higher than in the other two ERM proteins but does not increase after S1P. S1P-induced moesin and ezrin threonine phosphorylation is not mediated by the barrier enhancing receptor S1PR1 because siRNA downregulation of S1PR1 fails to inhibit these phosphorylation events, while stimulation of EC with the S1PR1-specific agonist SEW2871 fails to induce these phosphorylation events. Silencing of either all ERM proteins or radixin alone (but not moesin alone) reduced S1P-induced Rac1 activation and phosphorylation of the downstream Rac1 effector PAK1. Radixin siRNA alone, or combined siRNA for all three ERM proteins, dramatically attenuates S1P-induced EC barrier enhancement (measured by transendothelial electrical resistance (TER), peripheral accumulation of di-phospho-MLC, and cortical cytoskeletal rearrangement. In contrast, moesin depletion has the opposite effects on these parameters. Ezrin silencing partially attenuates S1P-induced EC barrier enhancement and cytoskeletal changes. Thus, despite structural similarities and reported functional redundancy, the ERM proteins differentially modulate S1P-induced alterations in lung EC cytoskeleton and permeability. These results suggest that ERM activation is an important regulatory event in EC barrier responses to S1P.


Subject(s)
Cytoskeletal Proteins/metabolism , Endothelial Cells/physiology , Lysophospholipids/physiology , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Pulmonary Artery/cytology , Sphingosine/analogs & derivatives , Actins/metabolism , Amides/pharmacology , Antigens, CD/metabolism , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Cadherins/metabolism , Cell Membrane/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electric Impedance , Endothelial Cells/metabolism , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Lysophospholipids/pharmacology , Membrane Proteins/genetics , Microfilament Proteins/genetics , Oxadiazoles/pharmacology , Permeability , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C/metabolism , Pulmonary Artery/physiology , Pyridines/pharmacology , RNA Interference , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/pharmacology , Sphingosine/physiology , Thiophenes/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
11.
J Appl Physiol (1985) ; 110(1): 213-24, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21051573

ABSTRACT

Lung inflammation and alterations in endothelial cell (EC) permeability are key events to development of acute lung injury (ALI). Protective effects of atrial natriuretic peptide (ANP) have been shown against inflammatory signaling and endothelial barrier dysfunction induced by gram-negative bacterial wall liposaccharide. We hypothesized that ANP may possess more general protective effects and attenuate lung inflammation and EC barrier dysfunction by suppressing inflammatory cascades and barrier-disruptive mechanisms shared by gram-negative and gram-positive pathogens. C57BL/6J wild-type or ANP knockout mice (Nppa-/-) were treated with gram-positive bacterial cell wall compounds, Staphylococcus aureus-derived peptidoglycan (PepG) and/or lipoteichoic acid (LTA) (intratracheal, 2.5 mg/kg each), with or without ANP (intravenous, 2 µg/kg). In vitro, human pulmonary EC barrier properties were assessed by morphological analysis of gap formation and measurements of transendothelial electrical resistance. LTA and PepG markedly increased pulmonary EC permeability and activated p38 and ERK1/2 MAP kinases, NF-κB, and Rho/Rho kinase signaling. EC barrier dysfunction was further elevated upon combined LTA and PepG treatment, but abolished by ANP pretreatment. In vivo, LTA and PepG-induced accumulation of protein and cells in the bronchoalveolar lavage fluid, tissue neutrophil infiltration, and increased Evans blue extravasation in the lungs was significantly attenuated by intravenous injection of ANP. Accumulation of bronchoalveolar lavage markers of LTA/PepG-induced lung inflammation and barrier dysfunction was further augmented in ANP-/- mice and attenuated by exogenous ANP injection. These results strongly suggest a protective role of ANP in the in vitro and in vivo models of ALI associated with gram-positive infection. Thus ANP may have important implications in therapeutic strategies aimed at the treatment of sepsis and ALI-induced gram-positive bacterial pathogens.


Subject(s)
Acute Lung Injury/microbiology , Acute Lung Injury/prevention & control , Atrial Natriuretic Factor/administration & dosage , Endothelium, Vascular/physiopathology , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Acute Lung Injury/physiopathology , Animals , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Staphylococcal Infections/physiopathology , Treatment Outcome
12.
Microvasc Res ; 79(2): 128-38, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19962392

ABSTRACT

Elevation in intracellular cAMP level has been associated with increased endothelial barrier integrity and linked to the activation of protein kinase A (PKA). Recent studies have shown a novel mechanism of cAMP-mediated endothelial barrier regulation via cAMP-dependent nucleotide exchange factor Epac1 and Rap1 GTPase. This study examined a contribution of PKA-dependent and PKA-independent pathways in the human pulmonary endothelial (EC) barrier protection by cAMP. Synthetic cAMP analog, 8-bromoadenosine-3',5'-cyclic monophosphate (Br-cAMP), induced dose-dependent increase in EC transendothelial electrical resistance which was associated with activation of PKA, Epac/Rap1, and Tiam/Vav/Rac cascades and significantly attenuated thrombin-induced EC barrier disruption. Both specific Epac/Rap1 activator 8CPT-2Me-cAMP (8CPT) and specific PKA activator N(6)-benzoyl-adenosine-3',5'-cyclic monophosphate (6Bnz) enhanced EC barrier, suppressed thrombin-induced EC permeability, and independently activated small GTPase Rac. SiRNA-induced Rac knockdown suppressed barrier protective effects of both PKA and Epac signaling in pulmonary EC. Intravenous administration of either 6Bnz, or 8CPT, significantly reduced lung vascular leak in the murine model of lung injury induced by high tidal volume mechanical ventilation (HTV, 30 ml/kg, 4 h), whereas combined treatment with 6Bnz and 8CPT showed no further additive effects. This study dissected for the first time PKA and Epac pathways of lung EC barrier protection caused by cAMP elevation and identified Rac GTPase as a hub for PKA and Epac signaling leading to enhancement of lung vascular barrier.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/physiology , Endothelium, Vascular/metabolism , Guanine Nucleotide Exchange Factors/metabolism , rac GTP-Binding Proteins/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cells, Cultured , Cyclic AMP/analogs & derivatives , Electric Impedance , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Gene Knockdown Techniques , Gene Silencing , Guanine Nucleotide Exchange Factors/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Pulmonary Artery/cytology , RNA, Small Interfering/genetics , Signal Transduction , rac GTP-Binding Proteins/genetics
13.
Plast Reconstr Surg ; 123(5): 1533-1543, 2009 May.
Article in English | MEDLINE | ID: mdl-19407626

ABSTRACT

BACKGROUND: To overcome the donor-site morbidity associated with radial forearm flaps, many modifications of this technique have been reported, including the development of the perforator flap method. In this article, the authors analyze the efficacy of their shape-modified radial artery perforator flap method. METHODS: The authors analyzed all of their 112 cases of reconstruction with the shape-modified method with regard to the cause of injury, recipient site, whether the flap was free or pedicled, flap size, number of components that were divided by perforators, flap survival, and quality of the outcome. Donor-site morbidity, including the development of scars and dorsal hand numbness, was also evaluated. RESULTS: The reconstructed areas were the head and neck (27 cases, 24.1 percent), upper extremity (58 cases, 51.8 percent), and lower extremity (27 cases, 24.1 percent). Free flaps were used in 75 cases (67.0 percent). In total, 109 flaps (97.3 percent) survived completely, but three free flaps exhibited venous thrombosis. In terms of aesthetics, 48 cases had excellent outcomes (42.9 percent), 61 cases had good outcomes (54.5 percent), and three cases had poor outcomes (2.7 percent). Impaired blood circulation and numbness of the donor hand were never observed, but one patient complained about a donor-site cosmetic problem. CONCLUSIONS: The authors' flap can be considered as a kind of propeller flap. It also resembles the flap-in-flap method. It is suitable for a wide range of surgical indications and is useful and safe in not only adults but also young and elderly patients.


Subject(s)
Radial Artery , Wounds and Injuries/surgery , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Forearm/surgery , Graft Survival , Humans , Male , Middle Aged , Surgical Flaps/blood supply , Young Adult
14.
Nanomedicine ; 5(1): 30-41, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18824415

ABSTRACT

Actomyosin contraction directly regulates endothelial cell (EC) permeability, but intracellular redistribution of cytoskeletal tension associated with EC permeability is poorly understood. We used atomic force microscopy (AFM), EC permeability assays, and fluorescence microscopy to link barrier regulation, cell remodeling, and cytoskeletal mechanical properties in EC treated with barrier-protective as well as barrier-disruptive agonists. Thrombin, vascular endothelial growth factor, and hydrogen peroxide increased EC permeability, disrupted cell junctions, and induced stress fiber formation. Oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine, hepatocyte growth factor, and iloprost tightened EC barriers, enhanced peripheral actin cytoskeleton and adherens junctions, and abolished thrombin-induced permeability and EC remodeling. AFM force mapping and imaging showed differential distribution of cell stiffness: barrier-disruptive agonists increased stiffness in the central region, and barrier-protective agents decreased stiffness in the center and increased it at the periphery. Attenuation of thrombin-induced permeability correlates well with stiffness changes from the cell center to periphery. These results directly link for the first time the patterns of cell stiffness with specific EC permeability responses.


Subject(s)
Cytoskeleton/metabolism , Endothelium, Vascular/physiology , Lung/cytology , Microscopy, Atomic Force/methods , Cell Line , Cytoskeleton/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique , Humans , Hydrogen Peroxide/pharmacology , Lung/blood supply , Microscopy, Fluorescence , Permeability/drug effects , Phosphatidylcholines/pharmacology , Thrombin/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
15.
Am J Respir Cell Mol Biol ; 40(1): 99-107, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18664639

ABSTRACT

Circulating levels of hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF) are increased during acute lung injury; however, combined effects of HGF and VEGF on pulmonary endothelial cell (EC) permeability remain to be elucidated. We have previously shown differential remodeling of focal adhesions (FA) caused by barrier-protective and barrier-disruptive mechanical and chemical stimuli. This study examined a role of FA protein paxillin in the pulmonary EC barrier responses induced by HGF and VEGF. VEGF increased, but HGF decreased, pulmonary EC permeability. These effects were accompanied by differential patterns of site-specific phosphorylation of focal adhesion kinase (FAK) and paxillin and FA redistribution. HGF antagonized random FA formation caused by VEGF challenge and promoted FA accumulation at the cell periphery. HGF attenuated VEGF-induced paxillin redistribution, FA remodeling, and endothelial permeability. SiRNA-based paxillin knockdown attenuated VEGF-induced EC permeability, myosin light chain phosphorylation, and stress fiber and paracellular gap formation. Paxillin knockdown also decreased HGF-induced EC barrier enhancement and suppressed activation of Rac and its effector PAK1. Expression of paxillin-S(273) deficient on PAK1 phosphorylation site prevented HGF-induced cytoskeletal remodeling. These data show a dual role of paxillin in the HGF- and VEGF-mediated endothelial barrier regulation and suggest essential paxillin role in the modulation of Rac-Rho crosstalk. Our results also support a model of pulmonary EC barrier recovery during resolution of ALI via switch from VEGF to HGF signaling.


Subject(s)
Endothelial Cells , Focal Adhesions/metabolism , Hepatocyte Growth Factor/metabolism , Paxillin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Electric Impedance , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Knockdown Techniques , Humans , Paxillin/genetics , Permeability , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
16.
Am J Physiol Lung Cell Mol Physiol ; 295(4): L612-23, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18689603

ABSTRACT

Mechanical ventilation at high tidal volumes compromises the blood-gas barrier and increases lung vascular permeability, which may lead to ventilator-induced lung injury and pulmonary edema. Using pulmonary endothelial cell (ECs) exposed to physiologically [5% cyclic stretch (CS)] and pathologically (18% CS) relevant magnitudes of CS, we evaluated the potential protective effects of hepatocyte growth factor (HGF) on EC barrier dysfunction induced by CS and vascular endothelial growth factor (VEGF). In static culture, HGF enhanced EC barrier function in a Rac-dependent manner and attenuated VEGF-induced EC permeability and paracellular gap formation. The protective effects of HGF were associated with the suppression of Rho-dependent signaling triggered by VEGF. Five percent CS promoted HGF-induced enhancement of the cortical F-actin rim and activation of Rac-dependent signaling, suggesting synergistic barrier-protective effects of physiological CS and HGF. In contrast, 18% CS further enhanced VEGF-induced EC permeability, activation of Rho signaling, and formation of actin stress fibers and paracellular gaps. These effects were attenuated by HGF pretreatment. EC preconditioning at 5% CS before HGF and VEGF further promoted EC barrier maintenance. Our data suggest synergistic effects of HGF and physiological CS in the Rac-mediated mechanisms of EC barrier protection. In turn, HGF reduced the barrier-disruptive effects of VEGF and pathological CS via downregulation of the Rho pathway. These results support the importance of HGF-VEGF balance in control of acute lung injury/acute respiratory distress syndrome severity via small GTPase-dependent regulation of lung endothelial permeability.


Subject(s)
Endothelium, Vascular/physiology , Hepatocyte Growth Factor/pharmacology , Mechanoreceptors/physiology , Pulmonary Artery/physiology , Vascular Endothelial Growth Factor A/pharmacology , Actins/metabolism , Animals , Cattle , Cell Communication/drug effects , Cell Communication/physiology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Endothelium, Vascular/drug effects , Enzyme Activation , GTP Phosphohydrolases/metabolism , Humans , Pulmonary Artery/drug effects , RNA, Small Interfering , rac1 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/genetics
17.
Am J Physiol Lung Cell Mol Physiol ; 292(2): L487-99, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17012370

ABSTRACT

2-Methoxyestradiol (2ME), a promising anti-tumor agent, is currently tested in phase I/II clinical trial to assess drug tolerance and clinical effects. 2ME is known to affect microtubule (MT) polymerization rather than act through estrogen receptors. We hypothesized that 2ME, similar to other MT inhibitors, disrupts endothelial barrier properties. We show that 2ME decreases transendothelial electrical resistance and increases FITC-dextran leakage across human pulmonary artery endothelial monolayer, which correlates with 2ME-induced MT depolymerization. Pretreatment of endothelium with MT stabilizer taxol significantly attenuates the decrease in transendothelial resistance. 2ME treatment results in the induction of F-actin stress fibers, accompanied by the increase in myosin light chain (MLC) phosphorylation. The experiments with Rho kinase (ROCK) and MLC kinase inhibitors and ROCK small interfering RNA (siRNA) revealed that increase in MLC phosphorylation is attributed to the ROCK activation rather than MLC kinase activation. 2ME induces significant ERK1/2, p38, and JNK phosphorylation and activation; however, only p38 activation is relevant to the 2ME-induced endothelial hyperpermeability. p38 activation is accompanied by a marked increase in MAPKAP2 and 27-kDa heat shock protein (HSP27) phosphorylation level. Taxol significantly decreases p38 phosphorylation and activation in response to 2ME stimulation. Vice versa, p38 inhibitor SB203580 attenuates MT rearrangement in 2ME-challenged cells. Together, these results indicate that 2ME-induced barrier disruption is governed by MT depolymerization and p38- and ROCK-dependent mechanisms. The fact that certain concentrations of 2ME induce endothelial hyperpermeability suggests that the issue of the maximum-tolerated dose of 2ME for cancer treatment should be addressed with caution.


Subject(s)
Blood-Air Barrier/enzymology , Blood-Air Barrier/physiopathology , Estradiol/analogs & derivatives , Intracellular Signaling Peptides and Proteins/metabolism , Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , 2-Methoxyestradiol , Actomyosin/metabolism , Amides/pharmacology , Blood-Air Barrier/drug effects , Cell Membrane Permeability/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Estradiol/pharmacology , Humans , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/deficiency , Microelectrodes , Models, Biological , Myosin-Light-Chain Kinase/metabolism , Paclitaxel/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/deficiency , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Pyridines/pharmacology , Tubulin/metabolism , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...