Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Immunol ; 197(2): 611-9, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27316682

ABSTRACT

Synthetic amphipathic helical peptides (SAHPs) designed as apolipoprotein A-I mimetics are known to bind to class B scavenger receptors (SR-Bs), SR-BI, SR-BII, and CD36, receptors that mediate lipid transport and facilitate pathogen recognition. In this study, we evaluated SAHPs, selected for targeting human CD36, by their ability to attenuate LPS-induced inflammation, endothelial barrier dysfunction, and acute lung injury (ALI). L37pA, which targets CD36 and SR-BI equally, inhibited LPS-induced IL-8 secretion and barrier dysfunction in cultured endothelial cells while reducing lung neutrophil infiltration by 40% in a mouse model of LPS-induced ALI. A panel of 20 SAHPs was tested in HEK293 cell lines stably transfected with various SR-Bs to identify SAHPs with preferential selectivity toward CD36. Among several SAHPs targeting both SR-BI/BII and CD36 receptors, ELK-B acted predominantly through CD36. Compared with L37pA, 5A, and ELK SAHPs, ELK-B was most effective in reducing the pulmonary barrier dysfunction, neutrophil migration into the lung, and lung inflammation induced by LPS. We conclude that SAHPs with relative selectivity toward CD36 are more potent at inhibiting acute pulmonary inflammation and dysfunction. These data indicate that therapeutic strategies using SAHPs targeting CD36, but not necessarily mimicking all apolipoprotein A-I functions, may be considered a possible new treatment approach for inflammation-induced ALI and pulmonary edema.


Subject(s)
Acute Lung Injury/immunology , Anti-Inflammatory Agents/pharmacology , CD36 Antigens/antagonists & inhibitors , Inflammation/immunology , Acute Lung Injury/pathology , Animals , Apolipoprotein A-I/immunology , Disease Models, Animal , HEK293 Cells , Humans , Inflammation/pathology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Peptides/pharmacology
2.
PLoS One ; 9(4): e92670, 2014.
Article in English | MEDLINE | ID: mdl-24739883

ABSTRACT

Acute lung injury (ALI) is accompanied by decreased lung compliance. However, a role of tissue mechanics in modulation of inflammation remains unclear. We hypothesized that bacterial lipopolysacharide (LPS) stimulates extracellular matrix (ECM) production and vascular stiffening leading to stiffness-dependent exacerbation of endothelial cell (EC) inflammatory activation and lung barrier dysfunction. Expression of GEF-H1, ICAM-1, VCAM-1, ECM proteins fibronectin and collagen, lysyl oxidase (LOX) activity, interleukin-8 and activation of Rho signaling were analyzed in lung samples and pulmonary EC grown on soft (1.5 or 2.8 kPa) and stiff (40 kPa) substrates. LPS induced EC inflammatory activation accompanied by expression of ECM proteins, increase in LOX activity, and activation of Rho signaling. These effects were augmented in EC grown on stiff substrate. Stiffness-dependent enhancement of inflammation was associated with increased expression of Rho activator, GEF-H1. Inhibition of ECM crosslinking and stiffening by LOX suppression reduced EC inflammatory activation and GEF-H1 expression in response to LPS. In vivo, LOX inhibition attenuated LPS-induced expression of GEF-H1 and lung dysfunction. These findings present a novel mechanism of stiffness-dependent exacerbation of vascular inflammation and escalation of ALI via stimulation of GEF-H1-Rho pathway. This pathway represents a fundamental mechanism of positive feedback regulation of inflammation.


Subject(s)
Lipopolysaccharides/toxicity , Pneumonia/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Animals , Cells, Cultured , Collagen/metabolism , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Feedback, Physiological , Fibronectins/metabolism , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/metabolism , Mice, Inbred C57BL , Pneumonia/chemically induced , Pneumonia/pathology , Protein-Lysine 6-Oxidase/metabolism , Rho Factor/metabolism , Signal Transduction , Vascular Cell Adhesion Molecule-1/metabolism
3.
J Biol Chem ; 289(8): 5168-83, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24352660

ABSTRACT

Microtubule (MT) dynamics is involved in a variety of cell functions, including control of the endothelial cell (EC) barrier. Release of Rho-specific nucleotide exchange factor GEF-H1 from microtubules activates the Rho pathway of EC permeability. In turn, pathologic vascular leak can be prevented by treatment with atrial natriuretic peptide (ANP). This study investigated a novel mechanism of vascular barrier protection by ANP via modulation of GEF-H1 function. In pulmonary ECs, ANP suppressed thrombin-induced disassembly of peripheral MT and attenuated Rho signaling and cell retraction. ANP effects were mediated by the Rac1 GTPase effector PAK1. Activation of Rac1-PAK1 promoted PAK1 interaction with the Rho activator GEF-H1, inducing phosphorylation of total and MT-bound GEF-H1 and leading to attenuation of Rho-dependent actin remodeling. In vivo, ANP attenuated lung injury caused by excessive mechanical ventilation and TRAP peptide (TRAP/HTV), which was further exacerbated in ANP(-/-) mice. The protective effects of ANP against TRAP/HTV-induced lung injury were linked to the increased pool of stabilized MT and inactivation of Rho signaling via ANP-induced, PAK1-dependent inhibitory phosphorylation of GEF-H1. This study demonstrates a novel protective mechanism of ANP against pathologic hyperpermeability and suggests a novel pharmacological intervention for the prevention of increased vascular leak via PAK1-dependent modulation of GEF-H1 activity.


Subject(s)
Atrial Natriuretic Factor/metabolism , Capillary Permeability , Rho Guanine Nucleotide Exchange Factors/metabolism , Actins/metabolism , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Animals , Atrial Natriuretic Factor/pharmacology , Capillary Permeability/drug effects , Cytoprotection/drug effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Genes, Dominant , HEK293 Cells , Humans , Lung/blood supply , Lung/pathology , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Microtubules/metabolism , Models, Biological , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Pneumonia/enzymology , Pneumonia/pathology , Protein Binding/drug effects , Thrombin/pharmacology , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
4.
Transl Res ; 162(1): 45-55, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23571093

ABSTRACT

Lung inflammation and alterations in endothelial cell (EC) micro- and macrovascular permeability are key events to development of acute lung injury. Using ECs derived from human pulmonary artery and lung microvasculature, we investigated the interplay between p38 stress mitogen-activated protein kinase (MAPK) and Rho guanosine triphosphatase signaling in inflammatory and hyperpermeability responses. Both cell types were treated with Staphylococcus aureus-derived peptidoglycan (PepG) and lipoteichoic acid (LTA) with or without pretreatment with p38 MAPK or Rho kinase inhibitors. LTA and PepG increased permeability markedly in both pulmonary macrovascular and microvascular ECs. Agonist-induced hyperpermeability was accompanied by cytoskeletal remodeling, disruption of cell-cell contacts, formation of paracellular gaps, and activation of p38 MAPK, nuclear factor kappa-B (NFκB), and Rho/Rho kinase signaling. In macrovascular ECs, pharmacologic inhibition of Rho kinase with Y27632 suppressed p38 MAP kinase cascade activation significantly, whereas inhibition of p38 MAPK with SB203580 had no effect on Rho activation. In contrast, inhibition of p38 MAPK in microvascular ECs suppressed LTA/PepG-induced activation of Rho, whereas the Rho inhibitor suppressed activation of p38 MAPK. Inhibition of either p38 MAPK or Rho kinase attenuated activation of NFκB signaling substantially. These results demonstrate cell-type-specific differences in signaling induced by Staphylococcus aureus-derived pathogens in pulmonary endothelium. Thus, although Gram-positive bacterial compounds caused barrier dysfunction in both EC types, it was induced by a different pattern of crosstalk between Rho, p38 MAPK, and NFκB signaling. These observations may have important implications in defining microvasculature-specific therapeutic strategies aimed at the treatment of sepsis and acute lung injury induced by Gram-positive bacterial pathogens.


Subject(s)
Endothelium, Vascular/pathology , Lung/pathology , Receptor Cross-Talk/physiology , Staphylococcal Infections/physiopathology , Staphylococcus aureus/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , rho-Associated Kinases/metabolism , Cells, Cultured , Endothelial Cells/enzymology , Endothelial Cells/pathology , Humans , Inflammation , Lipopolysaccharides/metabolism , Lung Injury/physiopathology , Peptidoglycan/metabolism , Signal Transduction , Staphylococcal Infections/enzymology , Teichoic Acids/metabolism
5.
Eur Respir J ; 41(1): 165-76, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22790920

ABSTRACT

The protective effects of prostacyclin and its stable analogue iloprost are mediated by elevation of intracellular cyclic AMP (cAMP) leading to enhancement of the peripheral actin cytoskeleton and cell-cell adhesive structures. This study tested the hypothesis that iloprost may exhibit protective effects against lung injury and endothelial barrier dysfunction induced by bacterial wall lipopolysaccharide (LPS). Endothelial barrier dysfunction was assessed by measurements of transendothelial permeability, morphologically and by analysis of LPS-activated inflammatory signalling. In vivo, C57BL/6J mice were challenged with LPS with or without iloprost or 8-bromoadenosine-3',5'-cyclic monophosphate (Br-cAMP) treatment. Lung injury was monitored by measurements of bronchoalveolar lavage protein content, cell count and Evans blue extravasation. Iloprost and Br-cAMP attenuated the disruption of the endothelial monolayer, and suppressed the activation of p38 mitogen-activated protein kinase (MAPK), the nuclear factor (NF)-κB pathway, Rho signalling, intercellular adhesion molecular (ICAM)-1 expression and neutrophil migration after LPS challenge. In vivo, iloprost was effective against LPS-induced protein and neutrophil accumulation in bronchoalveolar lavage fluid, and reduced myeloperoxidase activation, ICAM-1 expression and Evans blue extravasation in the lungs. Inhibition of Rac activity abolished the barrier-protective and anti-inflammatory effects of iloprost and Br-cAMP. Iloprost-induced elevation of intracellular cAMP triggers Rac signalling, which attenuates LPS-induced NF-κB and p38 MAPK inflammatory pathways and the Rho-dependent mechanism of endothelial permeability.


Subject(s)
Iloprost/therapeutic use , Lung Injury/drug therapy , Lung/drug effects , Lung/physiopathology , Animals , Cells, Cultured , Endothelium/drug effects , Endothelium/physiology , Lipopolysaccharides/administration & dosage , Lung Injury/chemically induced , Mice , Mice, Inbred C57BL , Neuropeptides/drug effects , Neuropeptides/physiology , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/physiology , rac1 GTP-Binding Protein
6.
Am J Respir Cell Mol Biol ; 47(5): 688-97, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22842495

ABSTRACT

Oxidative stress is an important part of host innate immune response to foreign pathogens, such as bacterial LPS, but excessive activation of redox signaling may lead to pathologic endothelial cell (EC) activation and barrier dysfunction. Microtubules (MTs) play an important role in agonist-induced regulation of vascular endothelial permeability, but their impact in modulation of inflammation and EC barrier has not been yet investigated. This study examined the effects of LPS-induced oxidative stress on MT dynamics and the involvement of MTs in the LPS-induced mechanisms of Rho activation, EC permeability, and lung injury. LPS treatment of pulmonary vascular EC induced elevation of reactive oxygen species (ROS) and caused oxidative stress associated with EC hyperpermeability, cytoskeletal remodeling, and formation of paracellular gaps, as well as activation of Rho, p38 stress kinase, and NF-κB signaling, the hallmarks of endothelial barrier dysfunction. LPS also triggered ROS-dependent disassembly of the MT network, leading to activation of MT-dependent signaling. Stabilization of MTs with epothilone B, or inhibition of MT-associated guanine nucleotide exchange factor (GEF)-H1 activity by silencing RNA-mediated knockdown, suppressed LPS-induced EC barrier dysfunction in vitro, and attenuated vascular leak and lung inflammation in vivo. LPS disruptive effects were linked to activation of Rho signaling caused by LPS-induced MT disassembly and release of Rho-specific GEF-H1 from MTs. These studies demonstrate, for the first time, the mechanism of ROS-induced Rho activation via destabilization of MTs and GEF-H1-dependent activation of Rho signaling, leading to pulmonary EC barrier dysfunction and exacerbation of LPS-induced inflammation.


Subject(s)
Acute Lung Injury/metabolism , Capillary Permeability , Microtubules/metabolism , Oxidative Stress , Acetylcysteine/pharmacology , Acute Lung Injury/immunology , Animals , Antioxidants/pharmacology , Cells, Cultured , Electric Impedance , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Epothilones/pharmacology , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/physiology , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Protein Stability , Pulmonary Artery/immunology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , RNA Interference , Rho Guanine Nucleotide Exchange Factors , Tubulin Modulators/pharmacology
7.
FASEB J ; 26(9): 3862-74, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22700873

ABSTRACT

Microtubule (MT) dynamics in vascular endothelium are modulated by vasoactive mediators and are critically involved in the control of endothelial cell (EC) permeability via Rho GTPase-dependent crosstalk with the actin cytoskeleton. However, the role of regulators in MT stability in these mechanisms remains unclear. This study investigated the involvement of the MT-associated protein stathmin in the mediation of agonist-induced permeability in EC cultures and vascular leak in vivo. Thrombin treatment of human pulmonary ECs induced rapid dephosphorylation and activation of stathmin. Inhibition of stathmin activity by small interfering RNA-based knockdown or cAMP-mediated phosphorylation abrogated thrombin-induced F-actin remodeling and Rho-dependent EC hyperpermeability, while expression of a phosphorylation-deficient stathmin mutant exacerbated thrombin-induced EC barrier disruption. Stathmin suppression preserved the MT network against thrombin-induced MT disassembly and release of Rho-specific guanine nucleotide exchange factor, GEF-H1. The protective effects of stathmin knockdown were observed in vivo in the mouse 2-hit model of ventilator-induced lung injury and were linked to MT stabilization and down-regulation of Rho signaling in the lung. These results demonstrate the mechanism of stathmin-dependent control of MT dynamics, Rho signaling, and permeability and suggest novel potential pharmacological interventions in the prevention of increased vascular leak via modulation of stathmin activity.


Subject(s)
Cell Membrane Permeability , Lung/cytology , Microtubules/physiology , Stathmin/physiology , Animals , Endothelium/cytology , Endothelium/drug effects , Fluorescent Antibody Technique , Gene Knockdown Techniques , Lung/drug effects , Male , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Small Interfering/genetics , Respiration, Artificial/adverse effects , Stathmin/genetics , Stathmin/metabolism , Thrombin/pharmacology
8.
PLoS One ; 7(1): e30957, 2012.
Article in English | MEDLINE | ID: mdl-22303475

ABSTRACT

INTRODUCTION: Oxidation products of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphatidylcholine (OxPAPC) differentially modulate endothelial cell (EC) barrier function in a dose-dependent fashion. Vascular endothelial growth factor receptor-2 (VEGFR2) is involved in the OxPAPC-induced EC inflammatory activation. This study examined a role of VEGFR2 in barrier dysfunction caused by high concentrations of OxPAPC and evaluated downstream signaling mechanisms resulting from the effect of OxPAPC in EC from pulmonary and systemic circulation. METHODS: EC monolayer permeability in human pulmonary artery endothelial cells (HPAEC) and human aortic endothelial cells (HAEC) was monitored by changes in transendothelial electrical resistance (TER) across EC monolayers. Actin cytoskeleton was examined by immunostaining with Texas Red labeled phalloidin. Phosphorylation of myosin light chains (MLC) and VE-Cadherin was examined by Western blot and immunofluorescence techniques. The role of VEGFR2 in OxPAPC-induced permeability and cytoskeletal arrangement were determined using siRNA-induced VEGFR2 knockdown. RESULTS: Low OxPAPC concentrations (5-20 µg/ml) induced a barrier protective response in both HPAEC and HAEC, while high OxPAPC concentrations (50-100 µg/ml) caused a rapid increase in permeability; actin stress fiber formation and increased MLC phosphorylation were observed as early as 30 min after treatment. VEGFR2 knockdown dramatically decreased the amount of MLC phosphorylation and stress fiber formation caused by high OxPAPC concentrations with modest effects on the amount of VE-cadherin phosphorylation at Y(731). We present evidence that activation of Rho is involved in the OxPAPC/VEGFR2 mechanism of EC permeability induced by high OxPAPC concentrations. Knockdown of VEGFR2 did not rescue the early drop in TER but prevented further development of OxPAPC-induced barrier dysfunction. CONCLUSIONS: This study shows that VEGFR2 is involved in the delayed phase of EC barrier dysfunction caused by high OxPAPC concentrations and contributes to stress fiber formation and increased MLC phosphorylation.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/enzymology , Phosphatidylcholines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Antigens, CD/metabolism , Aorta/cytology , Cadherins/metabolism , Cell Membrane Permeability/drug effects , Electric Impedance , Enzyme Activation/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Myosin Light Chains/metabolism , Protein Transport/drug effects , Pulmonary Artery/cytology , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism
9.
Am J Physiol Lung Cell Mol Physiol ; 302(9): L965-75, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22345573

ABSTRACT

Most patients with acute lung injury (ALI) and acute respiratory distress syndrome of septic and nonseptic nature require assisted ventilation with positive pressure, which at suboptimal range may further exacerbate lung dysfunction. Previous studies described enhancement of agonist-induced Rho GTPase signaling and endothelial cell (EC) permeability in EC cultures exposed to pathologically relevant cyclic stretch (CS) magnitudes. This study examined a role of pathologic CS in modulation of pulmonary EC permeability caused by IL-6, a cytokine increased in sepsis and acting in a Rho-independent manner. IL-6 increased EC permeability, which was associated with activation of Jak/signal transducers and activators of transcription, p38 MAP kinase, and NF-κB signaling and was augmented by EC exposure to 18% CS. Rho kinase inhibitor Y-27632 suppressed the synergistic effect of 18% CS on IL-6-induced EC monolayer disruption but did not alter the IL-6 effects on static EC culture. 18% CS also increased IL-6-induced ICAM-1 expression by pulmonary EC and neutrophil adhesion, which was attenuated by Y-27632. Intratracheal IL-6 administration in C57BL/6J mice increased protein content and cell count in bronchoalveolar lavage fluid. These changes were augmented by high tidal volume mechanical ventilation (HTV; 30 ml/kg, 4 h). Intravenous injection of Y-27632 suppressed IL6/HTV-induced lung injury. In conclusion, this study proposes a novel mechanism contributing to two-hit model of ALI: in addition to synergistic effects on Rho-dependent endothelial hyper-permeability triggered by thrombin, TNFα, LPS, or other agonists, ventilator-induced lung injury-relevant CS may also exacerbate Rho-independent mechanisms of EC permeability induced by other inflammatory mediators such as IL-6 via mechanisms involving Rho activity.


Subject(s)
Acute Lung Injury/metabolism , Interleukin-6/physiology , Stress, Physiological , rho GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Acute Lung Injury/physiopathology , Amides/pharmacology , Animals , Cells, Cultured , Cytoskeletal Proteins/metabolism , Electric Impedance , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung/blood supply , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Permeability , Proteolysis , Pulmonary Artery/cytology , Pyridines/pharmacology , Random Allocation , Respiration, Artificial/adverse effects , Signal Transduction , Stress, Mechanical , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
10.
J Cell Physiol ; 227(5): 1883-90, 2012 May.
Article in English | MEDLINE | ID: mdl-21732359

ABSTRACT

Afadin is a novel regulator of epithelial cell junctions assembly. However, its role in the formation of endothelial cell junctions and the regulation of vascular permeability remains obscure. We previously described protective effects of oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC) in the in vitro and in vivo models of lung endothelial barrier dysfunction and acute lung injury, which were mediated by Rac GTPase. This study examined a role of afadin in the OxPAPC-induced enhancement of interactions between adherens junctions and tight junctions as a novel mechanism of endothelial cell (EC) barrier preservation. OxPAPC induced Rap1-dependent afadin accumulation at the cell periphery and Rap1-dependent afadin interaction with adherens junction and tight junction proteins p120-catenin and ZO-1, respectively. Afadin knockdown using siRNA or ectopic expression of afadin mutant lacking Rap1 GTPase binding domain suppressed OxPAPC-induced EC barrier enhancement and abolished barrier protective effects of OxPAPC against thrombin-induced EC permeability. Afadin knockdown also abolished protective effects of OxPAPC against ventilator-induced lung injury in vivo. These results demonstrate for the first time a critical role of afadin in the regulation of vascular barrier function in vitro and in vivo via coordination of adherens junction-tight junction interactions.


Subject(s)
Catenins/metabolism , Endothelium/metabolism , Lung/anatomy & histology , Lung/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Phospholipids/metabolism , Phosphoproteins/metabolism , Adherens Junctions/metabolism , Animals , Cell Adhesion/physiology , Cell Line , Cells, Cultured , Cytoskeleton/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Phospholipids/chemistry , Tight Junctions/metabolism , Zonula Occludens-1 Protein , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism , Delta Catenin
11.
Am J Respir Cell Mol Biol ; 46(3): 331-41, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21997484

ABSTRACT

The generation of phospholipid oxidation products in atherosclerosis, sepsis, and lung pathologies affects endothelial barrier function, which exerts significant consequences on disease outcomes in general. Our group previously showed that oxidized 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphocholine (OxPAPC) at low concentrations increases endothelial cell (EC) barrier function, but decreases it at higher concentrations. In this study, we determined the mechanisms responsible for the pulmonary endothelial cell barrier dysfunction induced by high OxPAPC concentrations. OxPAPC at a range of 5-20 µg/ml enhanced EC barriers, as indicated by increased transendothelial electrical resistance. In contrast, higher OxPAPC concentrations (50-100 µg/ml) rapidly increased EC permeability, which was accompanied by increased total cell protein tyrosine (Tyr) phosphorylation, phosphorylation at Tyr-418, the activation of Src kinase, and the phosphorylation of adherens junction (AJ) protein vascular endothelial cadherin (VE-cadherin) at Tyr-731 and Tyr-658, which was not observed in ECs stimulated with low OxPAPC doses. The early tyrosine phosphorylation of VE-cadherin was linked to the dissociation of VE-cadherin-p120-catenin/ß-catenin complexes and VE-cadherin internalization, whereas low OxPAPC doses promoted the formation of VE-cadherin-p120-catenin/ß-catenin complexes. High but not low doses of OxPAPC increased the production of reactive oxygen species (ROS) and protein oxidation. The inhibition of Src by PP2 and ROS production by N-acetyl cysteine inhibited the disassembly of VE-cadherin-p120-catenin complexes, and attenuated high OxPAPC-induced EC barrier disruption. These results show the differential effects of OxPAPC doses on VE-cadherin-p120-catenin complex assembly and EC barrier function. These data suggest that the rapid tyrosine phosphorylation of VE-cadherin and other potential targets mediated by Src and ROS-dependent mechanisms plays a key role in the dissociation of AJ complexes and EC barrier dysfunction induced by high OxPAPC doses.


Subject(s)
Capillary Permeability/drug effects , Endothelial Cells/drug effects , Phosphatidylcholines/pharmacology , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Catenins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Electric Impedance , Endothelial Cells/metabolism , Humans , Phosphorylation , Reactive Oxygen Species/metabolism , Time Factors , Tyrosine , beta Catenin/metabolism , src-Family Kinases/metabolism , Delta Catenin
12.
Exp Cell Res ; 317(6): 859-72, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21111731

ABSTRACT

Products resulting from oxidation of cell membrane phospholipid 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC) exhibit potent protective effects against lung endothelial cell (EC) barrier dysfunction caused by pathologically relevant mechanical forces and inflammatory agents. These effects were linked to enhancement of peripheral cytoskeleton and cell adhesion interactions mediated by small GTPase Rac and inhibition of Rho-mediated barrier-disruptive signaling. However, the mechanism of OxPAPC-induced, Rac-dependent Rho downregulation critical for vascular barrier protection remains unclear. This study tested the hypothesis that Rho negative regulator p190RhoGAP is essential for OxPAPC-induced lung barrier protection against ventilator-induced lung injury (VILI), and investigated potential mechanism of p190RhoGAP targeting to adherens junctions (AJ) via p120-catenin. OxPAPC induced peripheral translocation of p190RhoGAP, which was abolished by knockdown of Rac-specific guanine nucleotide exchange factors Tiam1 and Vav2. OxPAPC also induced Rac-dependent tyrosine phosphorylation and association of p190RhoGAP with AJ protein p120-catenin. siRNA-induced knockdown of p190RhoGAP attenuated protective effects of OxPAPC against EC barrier compromise induced by thrombin and pathologically relevant cyclic stretch (18% CS). In vivo, p190RhoGAP knockdown significantly attenuated protective effects of OxPAPC against ventilator-induced lung vascular leak, as detected by increased cell count and protein content in the bronchoalveolar lavage fluid, and tissue neutrophil accumulation in the lung. These results demonstrate for the first time a key role of p190RhoGAP for the vascular endothelial barrier protection in VILI.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Phospholipids/metabolism , Repressor Proteins/metabolism , Ventilator-Induced Lung Injury/physiopathology , Animals , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/physiology , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/genetics , Humans , Mice , Oxidation-Reduction , Phosphatidylcholines/pharmacology , Repressor Proteins/genetics
13.
J Neurochem ; 114(6): 1734-44, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20598021

ABSTRACT

Cholesteryl ester rich apolipoprotein B100 (apoB100) lipoproteins accumulate in Bruch's membrane before the development of age-related macular degeneration. It is not known if these lipoproteins come from the circulation or local ocular tissue. Emerging, but incomplete evidence suggests that the retinal pigmented epithelium (RPE) can secrete lipoproteins. The purpose of this investigation was to determine (i) whether human RPE cells synthesize and secrete apoB100, and (ii) whether this secretion is driven by cellular cholesterol, and if so, (iii) whether statins inhibit this response. The established, human derived ARPE-19 cells challenged with 0-0.8 mM oleic acid accumulated cellular cholesterol, but not triglycerides. Oleic acid increased the amount of apoB100 protein recovered from the medium by both western blot analysis and (35) S-radiolabeled immunoprecipitation while negative stain electron microscopy showed lipoprotein-like particles. Of nine statins evaluated, lipophilic statins induced HMG-CoA reductase mRNA expression the most. The lipophilic Cerivastatin (5 µM) reduced cellular cholesterol by 39% and abrogated apoB100 secretion by 3-fold. In contrast, the hydrophilic statin Pravastatin had minimal effect on apoB100 secretion. These data suggest that ARPE-19 cells synthesize and secrete apoB100 lipoproteins, that this secretion is driven by cellular cholesterol, and that statins can inhibit apoB100 secretion by reducing cellular cholesterol.


Subject(s)
Apolipoprotein B-100/metabolism , Cholesterol/metabolism , Antioxidants/pharmacology , Cell Line , Cell Survival/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Oleic Acid/pharmacology , Pyridines/pharmacology , Triglycerides/metabolism
15.
Lab Invest ; 90(6): 906-14, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19434059

ABSTRACT

To maintain normal retinal function, retinal pigment epithelial (RPE) cells engulf photoreceptor outer segments (ROS) enriched in free fatty acids (FFAs). We have previously demonstrated fatty acid-binding protein 5 (FABP5) downregulation in the RPE/choroidal complex in a mouse model of aging and early age-related macular degeneration. FABPs are involved in intracellular transport of FFAs and their targeting to specific metabolic pathways. To elucidate the role of FABP5 in lipid metabolism, the production of the FABP5 protein in a human RPE cell line was inhibited using RNA interference technology. As a result, the levels of cholesterol and cholesterol ester were decreased by about 40%, whereas FFAs and triglycerides were increased by 18 and 67% after siRNA treatment, respectively. Some species of phospholipids were decreased in siRNA-treated cells. Cellular lipid droplets were evident and apoB secretion was decreased by 76% in these cells. Additionally, we discovered that ARPE-19 cells could synthesize and secrete Apolipoprotein B100 (apoB100), which may serve as a backbone structure for the formation of lipoprotein particles in these cells. Our results indicate that FABP5 mRNA knockdown results in the accumulation of cellular triglycerides, decreased cholesterol levels, and reduced secretion of apoB100 protein and lipoprotein-like particles. These observations indicated that FABP5 plays a critical role in lipid metabolism in RPE cells, suggesting that FABP5 downregulation in the RPE/choroid complex in vivo might contribute to aging and early age-related macular degeneration.


Subject(s)
Cholesterol/metabolism , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , RNA, Messenger/genetics , Retinal Pigment Epithelium/physiology , Triglycerides/metabolism , Animals , Apolipoprotein B-100/metabolism , Cell Line , Cholesterol Esters/metabolism , DNA Primers , Fatty Acids, Nonesterified/metabolism , Humans , Lipoproteins/metabolism , Macular Degeneration/genetics , Macular Degeneration/therapy , Mice , Phospholipids/metabolism , RNA, Small Interfering/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Transfection
16.
J Neurochem ; 105(4): 1187-97, 2008 May.
Article in English | MEDLINE | ID: mdl-18182060

ABSTRACT

The accumulation of apolipoprotein B100 lipoproteins in Bruch membrane is an early event thought to promote age-related macular degeneration (AMD). Immunohistochemistry using an anti-oxidized low density lipoprotein antibody on 10 AMD specimens showed staining in Bruch membrane including basal deposits, a marker of AMD. To determine whether retinal pigmented epithelial cells develop a pathologic phenotype after interaction with lipoproteins, ARPE-19 cells were exposed to low density lipoproteins (LDL) or oxidized LDLs (oxLDL). Analysis using the Affymetrix U133 Plus 2.0 (Affymetrix, Inc., Santa Clara, CA, USA) gene chip showed physiological and pathological transcriptional responses after LDL and oxLDL treatment, respectively. LDL induced a down-regulation of cholesterol biosynthesis genes while oxLDL induced transcriptional alterations in genes related to lipid metabolism, oxidative stress, inflammation and apoptosis. Electrospray mass spectrometry showed that oxLDL, but not LDL induced large cellular increases of sphingomyelin, ceramides, and cholesteryl esters. With TUNEL labeling, oxLDL caused 14.6% apoptosis compared to <1% after LDL. Addition of an inhibitor of sphingomyelin synthase inhibited this apoptosis by 41%. These data support the hypothesis that oxidized lipoproteins are one trigger for initiating early events in the pathogenesis of AMD.


Subject(s)
Lipoproteins, LDL/physiology , Pigment Epithelium of Eye/pathology , Adult , Aged , Aged, 80 and over , Apoptosis/physiology , Cell Line, Transformed , Female , Humans , Lipid Peroxidation/physiology , Lipoproteins, LDL/metabolism , Macular Degeneration/etiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Middle Aged , Pigment Epithelium of Eye/metabolism
17.
Vaccine ; 23(44): 5177-85, 2005 Oct 25.
Article in English | MEDLINE | ID: mdl-16039021

ABSTRACT

Non-typeable Haemophilus influenzae (NTHi) outer membrane protein P6 was used as a new protein carrier for NTHi detoxified lipooligosaccharide (dLOS) conjugates due to its conservation and potential to elicit bactericidal antibodies. P6 was covalently conjugated to dLOS of strain 9274 through adipic acid dihydrazide with different ratios of dLOS to P6, which resulted in two conjugate formulations with weight ratios of dLOS to P6 of 3.7 for dLOS-P6 (I) and 1.6 for dLOS-P6 (II). Binding activity of the conjugates was examined by an enzyme-linked immunosorbent assay with mouse monoclonal antibodies specific to LOS and P6 and a rabbit anti-P6 serum. The results showed that the conjugates bound not only to the LOS antibody but also to both P6 antibodies, suggesting that the conjugates retained epitopes of both LOS and P6 antigens. Animal studies revealed that dLOS-P6 (II) induced high levels of anti-LOS and anti-P6 IgGs in mice and rabbits. However, dLOS-P6 (I) induced lower levels of anti-LOS IgGs in mice and rabbits and anti-P6 IgGs in rabbits with no anti-P6 IgGs in mice. In addition, all rabbit, but not mouse, antisera elicited by the conjugates showed bactericidal activity against the homologous strain, and two of them elicited by each conjugate plus Ribi adjuvant showed cross-bactericidal activity against three of five major serotype stains. These data indicate that P6 could serve as an effective carrier for dLOS or other carbohydrate conjugates and that the ratio of carbohydrate to P6 might contribute to immune responses in vivo.


Subject(s)
Bacterial Outer Membrane Proteins/administration & dosage , Drug Delivery Systems , Haemophilus Vaccines/administration & dosage , Lipopolysaccharides/administration & dosage , Vaccines, Conjugate/administration & dosage , Animals , Antibodies, Bacterial/biosynthesis , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins , Haemophilus Vaccines/immunology , Immune Sera/immunology , Immunization , Lipopolysaccharides/immunology , Mice , Rabbits , Vaccines, Conjugate/immunology
18.
Invest Ophthalmol Vis Sci ; 46(4): 1117-23, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15790868

ABSTRACT

PURPOSE: Although light-induced oxidative stress in the retina has been extensively reported, little information regarding light-induced oxidative stress in choroidal endothelial cells (CECs) is available. In the current study, light-induced DNA oxidation and the activation of nuclear factor-kappaB (NF-kappaB), a major oxidative responsive transcription factor, were investigated in mouse CECs. METHODS: Mice were exposed to green light. Light-induced DNA oxidation in CECs was detected by in situ 8-hydroxy-2-deoxyguanosine (8-oxo-dG) immunolabeling. CECs were isolated from the retinal pigment epithelium (RPE)/choroid by using immunomagnetic beads. The isolated CECs were immunochemically characterized by the expression of endothelial markers, CD31, and P1H12. The quality of total RNA from CECs was assessed by a bioanalyzer and RT-PCR. NF-kappaB activation in situ and in isolated CECs was investigated. RESULTS: After a 3-hour exposure to light, the immunoreactivity to anti-8-oxo-dG antibody or anti-NF-kappaB p65 antibody in CECs in situ was significantly increased when compared with unexposed mice. Isolated CECs expressed CD31 and P1H12. The 28S/18S rRNA ratio of RNA isolated from CECs was 1.5:1. CD31 and von Willebrand Factor (vWF) transcripts were predominantly expressed in the RNA from isolated CECs. IkappaBalpha was more heavily phosphorylated in light-exposed than untreated CECs. IkappaBalpha expression levels were increased fivefold in isolated CECs after exposure to light compared to unexposed control subjects. CONCLUSIONS: Exposure to light induces oxidative stress in CECs in vivo. A method for CEC isolation from the mouse RPE/choroid with preservation of RNA quality has been developed. The results of this study may facilitate the ability to identify CEC-specific genes and gene products that respond to photo-oxidative stress.


Subject(s)
Choroid/blood supply , Deoxyguanosine/analogs & derivatives , Endothelium, Vascular/radiation effects , Light , Oxidative Stress/radiation effects , 8-Hydroxy-2'-Deoxyguanosine , Animals , DNA/metabolism , Dark Adaptation , Deoxyguanosine/metabolism , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique, Indirect , I-kappa B Proteins/metabolism , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
J Comp Neurol ; 478(2): 149-63, 2004 Oct 11.
Article in English | MEDLINE | ID: mdl-15349976

ABSTRACT

Several mutations in the opsin gene have been associated with congenital stationary night blindness, considered to be a relatively nonprogressive disorder. In the present study, we examined the structural and functional changes induced by one of these mutations, i.e., substitution of aspartic acid for glycine at position 90 (G90D). Transgenic mice were created in which the ratio of transgenic opsin transcript to endogenous was 0.5:1, 1.7:1, or 2.5:1 and were studied via light and electron microscopy, immunocytochemistry, electroretinography (ERG), and spectrophotometry. Retinas with transgenic opsin levels equivalent to one endogenous allele (G0.5) appeared normal for a period of about 3-4 months, but at later ages there were disorganized, shortened rod outer segments (ROS), and a loss of photoreceptor nuclei. Higher levels of G90D opsin expression produced earlier signs of retinal degeneration and more severe disruption of photoreceptor morphology. Despite these adverse effects, the mutation had a positive effect on the retinas of rhodopsin knockout (R-/-) mice, whose visual cells fail to form ROS and rapidly degenerate. Incorporation of the transgene in the null background (G+/-/R-/- or G+/+/R-/-) led to the development of ROS containing G90D opsin and prolonged survival of photoreceptors. Absorbance spectra measured both in vitro and in situ showed a significant reduction of more than 90% in the amount of light-sensitive pigment in the retinas of G+/+/R-/- mice, and ERG recordings revealed a >1 log unit loss in sensitivity. However, the histological appearances of the retinas of these mice show no significant loss of photoreceptors and little change in the lengths of their outer segments. These findings suggest that much of the ERG sensitivity loss derives from the reduced quantal absorption that results from a failure of G90D opsin to bind to its chromophore and form a normal complement of light-sensitive visual pigment.


Subject(s)
Nerve Degeneration/pathology , Night Blindness/genetics , Retina/pathology , Rhodopsin/metabolism , Rod Opsins/genetics , Animals , Blotting, Northern , Blotting, Western , Disease Models, Animal , Electroretinography , Immunohistochemistry , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron , Nerve Degeneration/metabolism , Point Mutation , Retina/ultrastructure , Rhodopsin/genetics , Spectrophotometry , Transgenes
20.
Biochim Biophys Acta ; 1647(1-2): 315-20, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12686151

ABSTRACT

Dimeric mitochondrial aspartate aminotransferase (mAAT) contains a molecule of pyridoxal 5'-phosphate (PLP) tightly attached to each of its two identical active sites. The presence of this natural reporter allows us to study separately local perturbations in the architecture of this critical region of the molecule during unfolding. Upon unfolding of the enzyme with guanidine hydrochloride (GdnHCl), the coenzyme is completely released from the active site. The transition midpoint for the dissociation of PLP is 1.4+/-0.02 M when determined by size-exclusion chromatography (SEC) and 1.6+/-0.02 M when the protein-bound PLP is estimated by electrospray mass spectrometry (ESI-MS). In both cases the transition midpoint is higher than that of inactivation (1.3+/-0.01 M). On the other hand, the midpoint of the unfolding transition obtained by monitoring changes in ellipticity at 356 nm, which reflects the asymmetric environment of the PLP cofactor at the active site, is 1.19+/-0.011 M guanidine. These results indicate that the unfolding of mAAT is a multi-step process which includes an intermediate containing bound PLP but lacking catalytic activity.


Subject(s)
Aspartate Aminotransferase, Mitochondrial/chemistry , Protein Folding , Pyridoxal Phosphate/chemistry , Animals , Binding Sites , Circular Dichroism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...