Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
bioRxiv ; 2023 Nov 06.
Article in English | MEDLINE | ID: mdl-37986982

ABSTRACT

Lung inflammation, caused by acute exposure to ozone (O3) - one of the six criteria air pollutants - is a significant source of morbidity in susceptible individuals. Alveolar macrophages (AMØs) are the most abundant immune cells in the normal lung and their number increases following O3 exposure. However, the role of AMØs in promoting or limiting O3-induced lung inflammation has not been clearly defined. Here, we used a mouse model of acute O3 exposure, lineage tracing, genetic knockouts, and data from O3-exposed human volunteers to define the role and ontogeny of AMØs during acute O3 exposure. Lineage tracing experiments showed that 12, 24, and 72 h after exposure to O3 (2 ppm) for 3h all AMØs were tissue-resident origin. Similarly, in humans exposed to FA and O3 (200 ppb) for 135 minutes, we did not observe ~21h post-exposure an increase in monocyte-derived AMØs by flow cytometry. Highlighting a role for tissue-resident AMØs, we demonstrate that depletion of tissue-resident AMØs with clodronate-loaded liposomes led to persistence of neutrophils in the alveolar space after O3 exposure, suggesting that impaired neutrophil clearance (i.e., efferocytosis) leads to prolonged lung inflammation. Moreover, depletion of tissue-resident AMØ demonstrated reduced clearance of intratracheally instilled apoptotic Jurkat cells, consistent with reduced efferocytosis. Genetic ablation of MerTK - a key receptor involved in efferocytosis - also resulted in impaired clearance of apoptotic neutrophils followed O3 exposure. Overall, these findings underscore the pivotal role of tissue-resident AMØs in resolving O3-induced inflammation via MerTK-mediated efferocytosis.

2.
Eur Respir J ; 33(3): 612-24, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19010997

ABSTRACT

Despite an encouraging outcome of antioxidant therapy in animal models of acute lung injury, effective antioxidant agents for clinical application remain to be developed. The present study investigated the effect of pre-treatment with amifostine, a thiol antioxidant compound, on lung endothelial barrier dysfunction induced by Gram-negative bacteria wall-lipopolysaccharide (LPS). Endothelial permeability was monitored by changes in transendothelial electrical resistance. Cytoskeletal remodelling and reactive oxygen species (ROS) production was examined by immunofluorescence. Cell signalling was assessed by Western blot. Measurements of Evans blue extravasation, cell count and protein content in bronchoalveolar lavage fluid were used as in vivo parameters of lung vascular permeability. Hydrogen peroxide, LPS and interleukin-6 caused cytoskeletal reorganisation and increased permeability in the pulmonary endothelial cells, reflecting endothelial barrier dysfunction. These disruptive effects were inhibited by pre-treatment with amifostine and linked to the amifostine-mediated abrogation of ROS production and redox-sensitive signalling cascades, including p38, extracellular signal regulated kinase 1/2, mitogen-activated protein kinases and the nuclear factor-kappaB pathway. In vivo, concurrent amifostine administration inhibited LPS-induced oxidative stress and p38 mitogen-activated protein kinase activation, which was associated with reduced vascular leak and neutrophil recruitment to the lungs. The present study demonstrates, for the first time, protective effects of amifostine against lipopolysaccharide-induced lung vascular leak in vitro and in animal models of lipopolysaccharide-induced acute lung injury.


Subject(s)
Amifostine/pharmacology , Capillary Permeability/drug effects , Lung/drug effects , Radiation-Protective Agents/pharmacology , Animals , Antioxidants/metabolism , Bronchoalveolar Lavage Fluid , Cytoskeleton/metabolism , Inflammation , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species , Signal Transduction
3.
Am J Physiol Lung Cell Mol Physiol ; 281(6): L1472-83, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11704544

ABSTRACT

NaF, a potent G protein activator and Ser/Thr phosphatase inhibitor, significantly increased albumin permeability and decreased transcellular electrical resistance (TER), indicating endothelial cell (EC) barrier impairment. EC barrier dysfunction induced by NaF was accompanied by the development of actin stress fibers, intercellular gap formation, and significant time-dependent increases in myosin light chain (MLC) phosphorylation. However, despite rapid, albeit transient, activation of Ca(2+)/calmodulin-dependent MLC kinase (MLCK), the specific MLCK inhibitor ML-7 failed to affect NaF-induced MLC phosphorylation, actin cytoskeletal rearrangement, and reductions in TER, suggesting a limited role of MLCK in NaF-induced EC activation. In contrast, strategies to reduce Rho (C3 exoenzyme or toxin B) or to inhibit Rho-associated kinase (Y-27632 or dominant/negative RhoK) dramatically reduced MLC phosphorylation and actin stress fiber formation and significantly attenuated NaF-induced EC barrier dysfunction. Consistent with this role for RhoK activity, NaF selectively inhibited myosin-specific phosphatase activity, whereas the total Ser/Thr phosphatase activity remained unchanged. These data strongly suggest that MLC phosphorylation, mediated primarily by RhoK, and not MLCK, participates in NaF-induced EC actin cytoskeletal changes and barrier dysfunction.


Subject(s)
Endothelium, Vascular/metabolism , Myosin Light Chains/metabolism , Pulmonary Artery/cytology , Sodium Fluoride/pharmacology , Actins/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cattle , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Cytoskeleton/metabolism , Electric Impedance , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Activation/drug effects , Intracellular Signaling Peptides and Proteins , Myosin-Light-Chain Phosphatase , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases
4.
Am J Physiol Lung Cell Mol Physiol ; 281(6): L1500-11, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11704547

ABSTRACT

The mechanisms responsible for the divergent physiological responses of endothelial cells to vascular endothelial growth factor (VEGF) are incompletely understood. We hypothesized that VEGF elicits increased endothelial permeability and cell migration via differential activation of intracellular signal transduction pathways. To test this hypothesis, we established a model of VEGF-induced endothelial barrier dysfunction and chemotaxis with bovine pulmonary endothelial cells. We compared the effects of VEGF on transendothelial electrical resistance (TER), actin cytoskeletal remodeling, and chemotaxis of lung endothelial cells and then evaluated the role of the mitogen-activated protein kinases (MAPKs) p38 and extracellular signal-regulated kinase (ERK)1/2 in VEGF-mediated endothelial responses. The dose response of pulmonary arterial and lung microvascular endothelial cells to VEGF differed when barrier regulation and chemotaxis were evaluated. Inhibition of tyrosine kinase, phosphoinositol 3-kinase, or p38 MAPK significantly attenuated VEGF-mediated TER, F-actin remodeling, and chemotaxis. VEGF-mediated decreased TER was also significantly attenuated by inhibition of ERK1/2 MAPK but not by inhibition of fetal liver kinase-1 (flk-1) or Src kinase. In contrast, VEGF-mediated endothelial migration was not attenuated by ERK1/2 inhibition but was abolished by inhibition of either flk-1 or Src kinase. These data suggest potential mechanisms by which VEGF may differentially mediate physiological responses in vivo.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Lymphokines/pharmacology , Pulmonary Artery/cytology , Actins/metabolism , Animals , Butadienes/pharmacology , Cattle , Cell Division/drug effects , Cell Membrane Permeability/drug effects , Cells, Cultured , Chemotaxis/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Endothelium, Vascular/cytology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , p38 Mitogen-Activated Protein Kinases , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
5.
J Clin Invest ; 108(5): 689-701, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11544274

ABSTRACT

Substances released by platelets during blood clotting are essential participants in events that link hemostasis and angiogenesis and ensure adequate wound healing and tissue injury repair. We assessed the participation of sphingosine 1-phosphate (Sph-1-P), a biologically active phosphorylated lipid growth factor released from activated platelets, in the regulation of endothelial monolayer barrier integrity, which is key to both angiogenesis and vascular homeostasis. Sph-1-P produced rapid, sustained, and dose-dependent increases in transmonolayer electrical resistance (TER) across both human and bovine pulmonary artery and lung microvascular endothelial cells. This substance also reversed barrier dysfunction elicited by the edemagenic agent thrombin. Sph-1-P-mediated barrier enhancement was dependent upon G(ialpha)-receptor coupling to specific members of the endothelial differentiation gene (Edg) family of receptors (Edg-1 and Edg-3), Rho kinase and tyrosine kinase-dependent activation, and actin filament rearrangement. Sph-1-P-enhanced TER occurred in conjunction with Rac GTPase- and p21-associated kinase-dependent endothelial cortical actin assembly with recruitment of the actin filament regulatory protein, cofilin. Platelet-released Sph-1-P, linked to Rac- and Rho-dependent cytoskeletal rearrangement, may act late in angiogenesis to stabilize newly formed vessels, which often display abnormally increased vascular permeability.


Subject(s)
Capillary Permeability , Cytoskeleton/ultrastructure , DNA-Binding Proteins/physiology , Endothelium, Vascular/physiology , I-kappa B Proteins , Immediate-Early Proteins/physiology , Lysophospholipids , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Sphingosine/pharmacology , Actin Depolymerizing Factors , Actins/metabolism , Animals , Capillary Permeability/drug effects , Cattle , Cells, Cultured , Cytoskeleton/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/ultrastructure , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , MAP Kinase Signaling System , Microfilament Proteins/metabolism , NF-KappaB Inhibitor alpha , Neovascularization, Physiologic , Protein Serine-Threonine Kinases/physiology , Receptors, Lysophospholipid , Sphingosine/analogs & derivatives , p21-Activated Kinases , rac GTP-Binding Proteins/physiology
6.
Am J Physiol Lung Cell Mol Physiol ; 281(3): L565-74, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11504682

ABSTRACT

Endothelial cell (EC) barrier regulation is critically dependent on cytoskeletal components (microfilaments and microtubules). Because several edemagenic agents induce actomyosin-driven EC contraction tightly linked to myosin light chain (MLC) phosphorylation and microfilament reorganization, we examined the role of microtubule components in bovine EC barrier regulation. Nocodazole or vinblastine, inhibitors of microtubule polymerization, significantly decreased transendothelial electrical resistance in a dose-dependent manner, whereas pretreatment with the microtubule stabilizer paclitaxel significantly attenuated this effect. Decreases in transendothelial electrical resistance induced by microtubule disruption correlated with increases in lung permeability in isolated ferret lung preparations as well as with increases in EC stress fiber content and MLC phosphorylation. The increases in MLC phosphorylation were attributed to decreases in myosin-specific phosphatase activity without significant increases in MLC kinase activity and were attenuated by paclitaxel or by several strategies (C3 exotoxin, toxin B, Rho kinase inhibition) to inhibit Rho GTPase. Together, these results suggest that microtubule disruption initiates specific signaling pathways that cross talk with microfilament networks, resulting in Rho-mediated EC contractility and barrier dysfunction.


Subject(s)
Capillary Permeability/physiology , Endothelium, Vascular/physiology , Microtubules/physiology , Myosin Light Chains/metabolism , Actin Cytoskeleton/physiology , Animals , Cattle , Cells, Cultured , Electric Impedance , Endothelium, Vascular/cytology , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Phosphatase , Phosphoprotein Phosphatases/metabolism , Phosphorylation , rho GTP-Binding Proteins/physiology
7.
Am J Physiol Lung Cell Mol Physiol ; 280(6): L1168-78, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11350795

ABSTRACT

Tumor necrosis factor (TNF)-alpha is released in acute inflammatory lung syndromes linked to the extensive vascular dysfunction associated with increased permeability and endothelial cell apoptosis. TNF-alpha induced significant decreases in transcellular electrical resistance across pulmonary endothelial cell monolayers, reflecting vascular barrier dysfunction (beginning at 4 h and persisting for 48 h). TNF-alpha also triggered endothelial cell apoptosis beginning at 4 h, which was attenuated by the caspase inhibitor Z-Val-Ala-Asp-fluoromethylketone. Exploring the involvement of the actomyosin cytoskeleton in these important endothelial cell responses, we determined that TNF-alpha significantly increased myosin light chain (MLC) phosphorylation, with prominent stress fiber and paracellular gap formation, which paralleled the onset of decreases in transcellular electrical resistance and enhanced apoptosis. Reductions in MLC phosphorylation by the inhibition of either MLC kinase (ML-7, cholera toxin) or Rho kinase (Y-27632) dramatically attenuated TNF-alpha-induced stress fiber formation, indexes of apoptosis, and caspase-8 activity but not TNF-alpha-induced barrier dysfunction. These studies indicate a central role for the endothelial cell cytoskeleton in TNF-alpha-mediated apoptosis, whereas TNF-alpha-induced vascular permeability appears to evolve independently of contractile tension generation.


Subject(s)
Apoptosis , Capillary Permeability/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Myosin-Light-Chain Kinase/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Actins/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Caspase Inhibitors , Cattle , Cells, Cultured , Cytoskeleton/metabolism , Electric Impedance , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Phosphorylation/drug effects , Pulmonary Artery/cytology , Respiratory Distress Syndrome/metabolism , Stress Fibers/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism
8.
Circ Res ; 81(6): 895-903, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9400369

ABSTRACT

The 42- and 44-kD mitogen-activated protein kinases, also referred to as extracellular signal-related kinase (ERK) 2 and 1, respectively, may be transiently activated by stretching vascular smooth muscle cells (VSMCs). Using an organ culture model of rabbit aorta, we studied short- and long-term ERK1/2 activation by intraluminal pressure (150 mm Hg). Activation of ERK1/2 was biphasic: it reached a maximum (217.5 +/- 8.4% of control) 5 minutes after pressurizing and decreased to 120.7 +/- 5.1% of control after 2 hours. Furthermore, after 24 hours of pressurizing, ERK1/2 activity was as high (241.8 +/- 14.7% of control) as in the acute phase. Long-term pressure-induced ERK1/2 activation correlated with stimulation of tyrosine phosphorylation of proteins in the 125- to 140-kD range. Neither protein kinase C inhibitors (1 mumol/L staurosporine or 50 mumol/L bisindolylmaleimide-I) nor tyrosine kinase inhibitors (50 mumol/L tyrphostin A48 or 50 mumol/L genistein) affected pressure-induced ERK1/2 activation. However, the Src-family tyrosine kinase inhibitor herbimycin A (500 nmol/L) did reduce both 5-minute (by 92 +/- 8%) and 24-hour (by 63 +/- 7%) pressure-induced ERK1/2 activation. Thus, our results demonstrate a sustained activation of ERK1/2 and tyrosine kinases by intraluminal pressure in the arterial wall. Pressure-induced ERK1/2 activation is PKC independent and Src-family tyrosine kinase dependent and possibly includes activation of extracellular matrix-associated tyrosine kinases.


Subject(s)
Aorta/enzymology , Blood Pressure , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinases , Protein Kinase C/physiology , Protein-Tyrosine Kinases/physiology , Quinones/pharmacology , Animals , Benzoquinones , Enzyme Activation , Lactams, Macrocyclic , Male , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Organ Culture Techniques , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Rabbits , Rifabutin/analogs & derivatives , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...