Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Am J Respir Cell Mol Biol ; 70(1): 11-25, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37725486

RESUMEN

The generation of bioactive truncated oxidized phospholipids (Tr-OxPLs) from oxidation of cell-membrane or circulating lipoproteins is a common feature of various pathological states. Scavenger receptor CD36 is involved in lipid transport and acts as a receptor for Tr-OxPLs. Interestingly, Tr-OxPLs and CD36 are involved in endothelial dysfunction-derived acute lung injury, but the precise mechanistic connections remain unexplored. In the present study, we investigated the role of CD36 in mediating pulmonary endothelial cell (EC) dysfunction caused by Tr-OxPLs. Our results demonstrated that the Tr-OxPLs KOdia-PC, Paz-PC, PGPC, PON-PC, POV-PC, and lysophosphocholine caused an acute EC barrier disruption as revealed by measurements of transendothelial electrical resistance and VE-cadherin immunostaining. More importantly, a synthetic amphipathic helical peptide, L37pA, targeting human CD36 strongly attenuated Tr-OxPL-induced EC permeability. L37pA also suppressed Tr-OxPL-induced endothelial inflammatory activation monitored by mRNA expression of inflammatory cytokines/chemokines and adhesion molecules. In addition, L37pA blocked Tr-OxPL-induced NF-κB activation and tyrosine phosphorylation of Src kinase and VE-cadherin. The Src inhibitor SU6656 attenuated KOdia-PC-induced EC permeability and inflammation, but inhibition of the Toll-like receptors (TLRs) TLR1, TLR2, TLR4, and TLR6 had no such protective effects. CD36-knockout mice were more resistant to Tr-OxPL-induced lung injury. Treatment with L37pA was equally effective in ameliorating Tr-OxPL-induced vascular leak and lung inflammation as determined by an Evans blue extravasation assay and total cell and protein content in BAL fluid. Altogether, these results demonstrate an essential role of CD36 in mediating Tr-OxPL-induced EC dysfunction and suggest a strong therapeutic potential of CD36 inhibitory peptides in mitigating lung injury and inflammation.


Asunto(s)
Lesión Pulmonar Aguda , Fosfolípidos , Animales , Ratones , Humanos , Fosfolípidos/metabolismo , Lesión Pulmonar Aguda/patología , Inflamación , Péptidos , Pulmón/patología
2.
FASEB J ; 36(9): e22470, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35969180

RESUMEN

Extracellular DNA-binding proteins such as histones are danger-associated molecular pattern released by the injured tissues in trauma and sepsis settings, which trigger host immune response and vascular dysfunction. Molecular events leading to histone-induced endothelial cell (EC) dysfunction remain poorly understood. This study performed comparative analysis of H1, H2A, H2B, H3, and H4 histone subunits effects on human pulmonary EC permeability and inflammatory response. Analysis of transendothelial electrical resistance and EC monolayer permeability for macromolecues revealed that H3 and H4, but not H1, H2A, or H2B caused dose-dependent EC permeability accompanied by disassembly of adherens junctions. At higher doses, H3 and H4 activated nuclear factor kappa B inflammatory cascade leading to upregulation EC adhesion molecules ICAM1, VCAM1, E-selectin, and release of inflammatory cytokines. Inhibitory receptor analysis showed that toll-like receptor (TLR) 4 but not TLR1/2 or receptor for advanced glycation end inhibition significantly attenuated deleterious effects of H3 and H4 histones. Inhibitor of Rho-kinase was without effect, while inhibition of Src kinase caused partial preservation of cell-cell junctions, H3/H4-induced permeability and inflammation. Deleterious effects of H3/H4 were blocked by heparin. Activation of Epac-Rap1 signaling restored EC barrier properties after histone challenge. Intravenous injection of histones in mice caused elevation of inflammatory markers and increased vascular leak. Post-treatment with pharmacological Epac/Rap1 activator suppressed injurious effects of histones in vitro and in vivo. These results identify H3 and H4 as key histone subunits exhibiting deleterious effects on pulmonary vascular endothelium via TLR4-dependent mechanism. In conclusion, elevation of circulating histones may represent a serious risk of exacerbated acute lung injury (ALI) and multiple organ injury during severe trauma and infection.


Asunto(s)
Histonas , Inflamación , Animales , Endotelio Vascular/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Histonas/metabolismo , Humanos , Inflamación/metabolismo , Ratones , Permeabilidad
3.
Am J Respir Cell Mol Biol ; 67(3): 375-388, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35679261

RESUMEN

TLR7 (Toll-like receptor 7), the sensor for single-stranded RNA, contributes to systemic inflammation and mortality in murine polymicrobial sepsis. Recent studies show that extracellular miR-146a-5p serves as a TLR7 ligand and plays an important role in regulating host innate immunity. However, the role of miR-146a-5p and TLR7 signaling in pulmonary inflammation, endothelial activation, and sepsis-associated acute respiratory distress syndrome remains unclear. Here, we show that intratracheal administration of exogenous miR-146a-5p in mice evokes lung inflammation, activates endothelium, and increases endothelial permeability via TLR7-dependent mechanisms. TLR7 deficiency attenuates pulmonary barrier dysfunction and reduces lung inflammatory response in a murine sepsis model. Moreover, the impact of miR-146a-5p-TLR7 signaling on endothelial activation appears to be a secondary effect because TLR7 is undetectable in the human pulmonary artery and microvascular endothelial cells (ECs), which show no response to direct miR-146a-5p treatment in vitro. Both conditioned media of miR-146a-5p-treated macrophages (Mϕ) and septic sera of wild-type mice induce a marked EC barrier disruption in vitro, whereas Mϕ conditioned media or septic sera of TLR7-/- mice do not exhibit such effect. Cytokine array and pathway enrichment analysis of the Mϕ conditioned media and septic sera identify TNFα (tumor necrosis factor α) as the main downstream effector of miR-146a-5p-TLR7 signaling responsible for the EC barrier dysfunction, which is further supported by neutralizing anti-TNFα antibody intervention. Together, these data demonstrate that TLR7 activation elicits pulmonary inflammation and endothelial barrier disruption by sensing extracellular miR-146a-5p and contributes to sepsis-associated acute respiratory distress syndrome.


Asunto(s)
Glicoproteínas de Membrana , MicroARNs , Síndrome de Dificultad Respiratoria , Sepsis , Receptor Toll-Like 7 , Animales , Medios de Cultivo Condicionados , Células Endoteliales/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Síndrome de Dificultad Respiratoria/inmunología , Sepsis/complicaciones , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
4.
J Biol Chem ; 296: 100239, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33372035

RESUMEN

Proinflammatory cytokines such as IL-6 induce endothelial cell (EC) barrier disruption and trigger an inflammatory response in part by activating the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway. The protein suppressor of cytokine signaling-3 (SOCS3) is a negative regulator of JAK-STAT, but its role in modulation of lung EC barrier dysfunction caused by bacterial pathogens has not been investigated. Using human lung ECs and EC-specific SOCS3 knockout mice, we tested the hypothesis that SOCS3 confers microtubule (MT)-mediated protection against endothelial dysfunction. SOCS3 knockdown in cultured ECs or EC-specific SOCS3 knockout in mice resulted in exacerbated lung injury characterized by increased permeability and inflammation in response to IL-6 or heat-killed Staphylococcus aureus (HKSA). Ectopic expression of SOCS3 attenuated HKSA-induced EC dysfunction, and this effect required assembled MTs. SOCS3 was enriched in the MT fractions, and treatment with HKSA disrupted SOCS3-MT association. We discovered that-in addition to its known partners gp130 and JAK2-SOCS3 interacts with MT plus-end binding proteins CLIP-170 and CLASP2 via its N-terminal domain. The resulting SOCS3-CLIP-170/CLASP2 complex was essential for maximal SOCS3 anti-inflammatory effects. Both IL-6 and HKSA promoted MT disassembly and disrupted SOCS3 interaction with CLIP-170 and CLASP2. Moreover, knockdown of CLIP-170 or CLASP2 impaired SOCS3-JAK2 interaction and abolished the anti-inflammatory effects of SOCS3. Together, these findings demonstrate for the first time an interaction between SOCS3 and CLIP-170/CLASP2 and reveal that this interaction is essential to the protective effects of SOCS3 in lung endothelium.


Asunto(s)
Inflamación/genética , Lesión Pulmonar/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Proteína 3 Supresora de la Señalización de Citocinas/genética , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Animales , Citoesqueleto/genética , Células Endoteliales , Endotelio Vascular/metabolismo , Endotelio Vascular/microbiología , Endotelio Vascular/patología , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Uniones Intercelulares/genética , Interleucina-6/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/microbiología , Lesión Pulmonar/patología , Ratones , Ratones Noqueados , Permeabilidad , Staphylococcus aureus/patogenicidad
5.
Am J Physiol Lung Cell Mol Physiol ; 323(3): L223-L239, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35852995

RESUMEN

Extracellular histones released into the circulation following trauma, sepsis, and ARDS may act as potent damage-associated molecular pattern signals leading to multiple organ failure. Endothelial cell (EC) dysfunction caused by extracellular histones has been demonstrated in vitro and in vivo; however, precise mechanistic details of histone-induced EC dysfunction and exacerbation of ongoing inflammation remain poorly understood. This study investigated the role of extracellular histones in exacerbating preexisting endothelial dysfunction and acute lung injury. Histone subunits H3 and H4, but not H1, H2A, or H2B, induced permeability in human pulmonary EC. H3 and H4 at concentrations above 30 µg/mL caused EC inflammation reflected by activation of the NF-κB pathway, transcriptional activation, and release of cytokines and chemokines including IL-6 and IL-8, and increased mRNA and protein expression of EC adhesion molecules VCAM-1 and ICAM-1. Pharmacological inhibitors targeting Toll-like receptor TLR4 but not TLR2/6, blocked histone-induced EC dysfunction. H3 and H4 also strongly augmented EC permeability and inflammation caused by Gram-negative and Gram-positive bacterial particles, endotoxin, and TNFα. Heparin blocked histone-induced augmentation of EC inflammation caused by endotoxin and TNFα. Injection of histone in mouse models of lung injury caused by bacterial wall lipopolysaccharide (LPS) and heat-killed Staphylococcus aureus (HKSA) augmented ALI parameters: increased protein content, cell count, and inflammatory cytokine secretion in bronchoalveolar lavage fluid. Important clinical significance of these findings is in the demonstration that even a modest increase in extracellular histone levels can act as a severe exacerbating factor in conjunction with other EC barrier disruptive or proinflammatory agents.


Asunto(s)
Lesión Pulmonar Aguda , Histonas , Lesión Pulmonar Aguda/metabolismo , Animales , Humanos , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Ratones , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
FASEB J ; 35(4): e21388, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33724556

RESUMEN

Suppressors of cytokine signaling (SOCS) provide negative regulation of inflammatory reaction. The role and precise cellular mechanisms of SOCS1 in control of endothelial dysfunction and barrier compromise associated with acute lung injury remain unexplored. Our results show that siRNA-mediated SOCS1 knockdown augmented lipopolysaccharide (LPS)-induced pulmonary endothelial cell (EC) permeability and enhanced inflammatory response. Consistent with in vitro data, EC-specific SOCS1 knockout mice developed more severe lung vascular leak and accumulation of inflammatory cells in bronchoalveolar lavage fluid. SOCS1 overexpression exhibited protective effects against LPS-induced endothelial permeability and inflammation, which were dependent on microtubule (MT) integrity. Biochemical and image analysis of unstimulated EC showed SOCS1 association with the MT, while challenge with LPS or MT depolymerizing agent colchicine impaired this association. SOCS1 directly interacted with N2 domains of MT-associated proteins CLIP-170 and CLASP2. Furthermore, N-terminal region of SOCS1 was indispensable for these interactions and SOCS1-ΔN mutant lacking N-terminal 59 amino acids failed to rescue LPS-induced endothelial dysfunction. Depletion of endogenous CLIP-170 or CLASP2 abolished SOCS1 interaction with Toll-like receptor-4 and Janus kinase-2 leading to impairment of SOCS1 inhibitory effects on LPS-induced inflammation. Altogether, these findings suggest that endothelial barrier protective and anti-inflammatory effects of SOCS1 are critically dependent on its targeting to the MT.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Lipopolisacáridos/toxicidad , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Animales , Línea Celular , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Inflamación/inducido químicamente , Ratones , Ratones Noqueados , Proteína 1 Supresora de la Señalización de Citocinas/genética
7.
Infect Immun ; 89(10): e0030121, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34097506

RESUMEN

Recent studies suggest an anti-inflammatory protective role for class B scavenger receptor BI (SR-BI) in endotoxin-induced inflammation and sepsis. Other data, including ours, provide evidence for an alternative role of SR-BI, facilitating bacterial and endotoxin uptake and contributing to inflammation and bacterial infection. Enhanced endotoxin susceptibility of SR-BI-deficient mice due to their anti-inflammatory glucocorticoid deficiency complicates the understanding of SR-BI's role in endotoxemia/sepsis, calling for the use of alternative models. In this study, using human SR-BI (hSR-BI) and hSR-BII transgenic mice, we found that SR-BI and, to a lesser extent, its splicing variant SR-BII protect against LPS-induced lung damage. At 20 h after intratracheal LPS instillation, the extent of pulmonary inflammation and vascular leakage was significantly lower in hSR-BI and hSR-BII transgenic mice than in wild-type mice. Higher bronchoalveolar lavage fluid (BALF) inflammatory cell count and protein content and lung tissue neutrophil infiltration found in wild-type mice were associated with markedly (2 to 3 times) increased proinflammatory cytokine production compared to these parameters in transgenic mice following LPS administration. The markedly lower endotoxin levels detected in BALF of transgenic versus wild-type mice and the significantly increased BODIPY-LPS uptake observed in lungs of hSR-BI and hSR-BII mice 20 h after the i.t. LPS injection suggest that hSR-BI- and hSR-BII-mediated enhanced LPS clearance in the airways could represent the mechanism of their protective role against LPS-induced acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Receptores Depuradores/metabolismo , Receptores Depuradores de Clase B/metabolismo , Células A549 , Lesión Pulmonar Aguda/inducido químicamente , Animales , Líquido del Lavado Bronquioalveolar , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Endotoxemia/metabolismo , Humanos , Inflamación/inmunología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neutrófilos/metabolismo , Sepsis/metabolismo
8.
J Biol Chem ; 294(10): 3369-3384, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622143

RESUMEN

Staphylococcus aureus is a major etiological agent of sepsis and induces endothelial cell (EC) barrier dysfunction and inflammation, two major hallmarks of acute lung injury. However, the molecular mechanisms of bacterial pathogen-induced EC barrier disruption are incompletely understood. Here, we investigated the role of microtubules (MT) in the mechanisms of EC barrier compromise caused by heat-killed S. aureus (HKSA). Using a customized monolayer permeability assay in human pulmonary EC and MT fractionation, we observed that HKSA-induced barrier disruption is accompanied by MT destabilization and increased histone deacetylase-6 (HDAC6) activity resulting from elevated reactive oxygen species (ROS) production. Molecular or pharmacological HDAC6 inhibition rescued barrier function in HKSA-challenged vascular endothelium. The HKSA-induced EC permeability was associated with impaired MT-mediated delivery of cytoplasmic linker-associated protein 2 (CLASP2) to the cell periphery, limiting its interaction with adherens junction proteins. HKSA-induced EC barrier dysfunction was also associated with increased Rho GTPase activity via activation of MT-bound Rho-specific guanine nucleotide exchange factor-H1 (GEF-H1) and was abolished by HDAC6 down-regulation. HKSA activated the NF-κB proinflammatory pathway and increased the expression of intercellular and vascular cell adhesion molecules in EC, an effect that was also HDAC6-dependent and mediated, at least in part, by a GEF-H1/Rho-dependent mechanism. Of note, HDAC6 knockout mice or HDAC6 inhibitor-treated WT mice were partially protected from vascular leakage and inflammation caused by both HKSA or methicillin-resistant S. aureus (MRSA). Our results indicate that S. aureus-induced, ROS-dependent up-regulation of HDAC6 activity destabilizes MT and thereby activates the GEF-H1/Rho pathway, increasing both EC permeability and inflammation.


Asunto(s)
Células Endoteliales/metabolismo , Microtúbulos/metabolismo , Staphylococcus aureus/fisiología , Células Endoteliales/microbiología , Histona Desacetilasa 6/metabolismo , Calor , Humanos , Inflamación/microbiología , Viabilidad Microbiana , Oxidación-Reducción , Permeabilidad , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas de Unión al GTP rho/metabolismo
9.
Am J Physiol Lung Cell Mol Physiol ; 318(3): L533-L548, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31913681

RESUMEN

Transfusion of red blood cells (RBCs) is a common life-saving clinical practice in severely anemic or hemorrhagic patients; however, it may result in serious pathological complications such as transfusion-related acute lung injury. The factors mediating the deleterious effects of RBC transfusion remain unclear. In this study, we tested the effects of washed long-term (RBC-O; >28 days) versus short-term (RBC-F; <14 days) stored RBCs and their supernatants on lung endothelial (EC) permeability under control and inflammatory conditions. RBCs enhanced basal EC barrier function as evidenced by an increase in transendothelial electrical resistance and decrease in permeability for macromolecules. RBCs also attenuated EC hyperpermeability and suppressed secretion of EC adhesion molecule ICAM-1 and proinflammatory cytokine IL-8 in response to LPS or TNF-α. In both settings, RBC-F had slightly higher barrier protective effects as compared with RBC-O. In contrast, supernatants from both RBC-F and RBC-O disrupted the EC barrier. The early phase of EC permeability response caused by RBC supernatants was partially suppressed by antioxidant N-acetyl cysteine and inhibitor of Src kinase family PP2, while addition of heme blocker and inhibition of NOD-like receptor family pyrin domain containing protein 3 (NLRP3), stress MAP kinases, receptor for advanced glycation end-products (RAGE), or Toll-like receptor-4 (TLR4) signaling were without effect. Morphological analysis revealed that RBC supernatants increased LPS- and TNF-α-induced breakdown of intercellular junctions and formation of paracellular gaps. RBC supernatants augmented LPS- and TNF-α-induced EC inflammation reflected by increased production of IL-6, IL-8, and soluble ICAM-1. These findings demonstrate the deleterious effects of RBC supernatants on EC function, which may have a major impact in pathological consequences associated with RBC transfusion.


Asunto(s)
Conservación de la Sangre/efectos adversos , Permeabilidad de la Membrana Celular , Endotelio Vascular/patología , Eritrocitos/patología , Inflamación/patología , Pulmón/patología , Células Alogénicas , Eliminación de Componentes Sanguíneos/métodos , Endotelio Vascular/inmunología , Transfusión de Eritrocitos/efectos adversos , Humanos , Inflamación/etiología , Inflamación/inmunología , Pulmón/inmunología
10.
FASEB J ; 33(3): 3887-3900, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30521374

RESUMEN

As mechanisms controlling redox homeostasis become impaired with aging, exaggerated oxidant stress may cause disproportional oxidation of cell membranes and circulating phospholipids (PLs), leading to the formation of truncated oxidized PL products (Tr-OxPLs), which exhibit deleterious effects. This study investigated the role of elevated Tr-OxPLs as a factor exacerbating inflammation and lung barrier dysfunction in an animal model of aging. Mass spectrometry analysis of Tr-OxPL species in young (2-4 mo) and aging (18-24 mo) mice revealed elevated basal levels of several products [1-palmitoyl-2-(5-oxovaleroyl)- sn-glycero-phosphocholine (POVPC), 1-palmitoyl-2-glutaroyl- sn-glycero-phosphocholine, lysophosphocholine, 1-palmitoyl-2-(9-oxo-nonanoyl)- sn-glycero-3-phosphocholine, 1-palmitoyl-2-azelaoyl- sn-glycero-3-phosphocholine, O-1-O-palmitoyl-2-O-(5,8-dioxo-8-hydroxy-6-octenoyl)-l-glycero-3-phosphocholine, and others] in the aged lungs. An intratracheal (i.t.) injection of bacterial LPS caused increased generation of Tr-OxPLs in the lungs but not in the liver, with higher levels detected in the aged group. In addition, OxPLs clearance from the lung tissue after LPS challenge was delayed in the aged group. The impact of Tr-OxPLs on endothelial cell (EC) barrier compromise under inflammatory conditions was further evaluated in the 2-hit cell culture model of acute lung injury (ALI). EC barrier dysfunction caused by cell treatment with a cytokine mixture (CM) was augmented by cotreatment with low-dose Tr-OxPLs, which did not significantly affect endothelial function when added alone. Deleterious effects of Tr-OxPLs on inflamed ECs stimulated with CM were associated with further weakening of cell junctions and more robust EC hyperpermeability. Aged mice injected intratracheally with TNF-α exhibited a more pronounced elevation of cell counts and protein content in bronchoalveolar lavage (BAL) samples. Interestingly, intravenous administration of low POVPC doses-which did not affect BAL parameters alone in young mice exposed to i.t. TNF-α challenge-augmented lung injury to the levels observed in aged mice stimulated with TNF-α alone. Inhibition of Tr-OxPL generation by ectopic expression of PL-specific platelet-activating factor acetylhydrolase 2 (PAFAH2) markedly reduced EC dysfunction induced by CM, whereas PAFAH2 pharmacologic inhibition augmented deleterious effects of cytokines on EC barrier function. Moreover, exacerbating effects of PAFAH2 inhibition on TNF-α-induced lung injury were observed in vivo. These results demonstrate an age-dependent increase in Tr-OxPL production under basal conditions and augmented Tr-OxPL generation upon inflammatory stimulation, suggesting a major role for elevated Tr-OxPLs in more severe ALI and delayed resolution in aging lungs.-Ke, Y., Karki, P., Kim, J., Son, S., Berdyshev, E., Bochkov, V. N., Birukova, A. A., Birukov, K. G. Elevated truncated oxidized phospholipids as a factor exacerbating ALI in the aging lungs.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Envejecimiento/metabolismo , Células Epiteliales Alveolares/metabolismo , Pulmón/metabolismo , Fosfolípidos/metabolismo , Lesión Pulmonar Aguda/patología , Envejecimiento/patología , Células Epiteliales Alveolares/patología , Animales , Células Cultivadas , Femenino , Humanos , Pulmón/citología , Pulmón/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción
11.
Circ Res ; 121(3): 244-257, 2017 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-28522438

RESUMEN

RATIONALE: Oxidation of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC) generates a group of bioactive oxidized phospholipid products with a broad range of biological activities. Barrier-enhancing and anti-inflammatory effects of OxPAPC on pulmonary endothelial cells are critical for prevention of acute lung injury caused by bacterial pathogens or excessive mechanical ventilation. Anti-inflammatory properties of OxPAPC are associated with its antagonistic effects on Toll-like receptors and suppression of RhoA GTPase signaling. OBJECTIVE: Because OxPAPC exhibits long-lasting anti-inflammatory and lung-protective effects even after single administration in vivo, we tested the hypothesis that these effects may be mediated by additional mechanisms, such as OxPAPC-dependent production of anti-inflammatory and proresolving lipid mediator, lipoxin A4 (LXA4). METHODS AND RESULTS: Mass spectrometry and ELISA assays detected significant accumulation of LXA4 in the lungs of OxPAPC-treated mice and in conditioned medium of OxPAPC-exposed pulmonary endothelial cells. Administration of LXA4 reproduced anti-inflammatory effect of OxPAPC against tumor necrosis factor-α in vitro and in the animal model of lipopolysaccharide-induced lung injury. The potent barrier-protective and anti-inflammatory effects of OxPAPC against tumor necrosis factor-α and lipopolysaccharide challenge were suppressed in human pulmonary endothelial cells with small interfering RNA-induced knockdown of LXA4 formyl peptide receptor-2 (FPR2/ALX) and in mFPR2-/- (mouse formyl peptide receptor 2) mice lacking the mouse homolog of human FPR2/ALX. CONCLUSIONS: This is the first demonstration that inflammation- and injury-associated phospholipid oxidation triggers production of anti-inflammatory and proresolution molecules, such as LXA4. This lipid mediator switch represents a novel mechanism of OxPAPC-assisted recovery of inflamed lung endothelium.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Lipoxinas/metabolismo , Fosfatidilcolinas/uso terapéutico , Lesión Pulmonar Aguda/prevención & control , Animales , Antiinflamatorios no Esteroideos/farmacología , Células Cultivadas , Humanos , Lipoxinas/farmacología , Lipoxinas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilcolinas/farmacología , Resultado del Tratamiento
12.
FASEB J ; 31(9): 4187-4202, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28572443

RESUMEN

Unlike other agonists that cause transient endothelial cell (EC) response, the products of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (PAPC) oxidation that contain cyclopenthenone groups, which recapitulate prostaglandin-like structure, cause sustained enhancement of the pulmonary EC barrier. The mechanisms that drive the sustained effects by oxidized PAPC (OxPAPC) remain unexplored. On the basis of the structural similarity of isoprostanoid moieties that are present in full-length oxygenated PAPC species, we used an inhibitory approach to perform the screening of prostanoid receptors as potential candidates that mediate OxPAPC effects. Results show that only prostaglandin E receptor-4 (EP4) was involved and mediated the sustained phase of the barrier-enhancing effects of OxPAPC that are associated with the activation of Rac GTPase and its cytoskeletal targets. EC incubation with OxPAPC also induced EP4 mRNA expression in pulmonary ECs and lung tissue. EP4 knockdown using gene-specific small interfering RNA did not affect the rapid phase of OxPAPC-induced EC barrier enhancement or the protective effects against thrombin-induced EC permeability, but abolished the advanced barrier enhancement phase and suppressed the protective effects of OxPAPC against more sustained EC barrier dysfunction and cell inflammatory response caused by TNF-α. Endothelial-specific knockout of the EP4 receptor in mice attenuated the protective effect of intravenous OxPAPC administration in the model of acute lung injury caused by intratracheal injection of LPS. Taken together, these results demonstrate a novel role for prostaglandin receptor EP4 in the mediation of barrier-enhancing and anti-inflammatory effects caused by oxidized phospholipids.-Oskolkova, O., Gawlak, G., Tian, Y., Ke, Y., Sarich, N., Son, S., Andreasson, K., Bochkov, V. N., Birukova, A. A., Birukov, K. G. Prostaglandin E receptor-4 receptor mediates endothelial barrier-enhancing and anti-inflammatory effects of oxidized phospholipids.


Asunto(s)
Células Endoteliales/fisiología , Fosfatidilcolinas/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Uniones Adherentes/fisiología , Animales , Citoesqueleto , Impedancia Eléctrica , Humanos , Inflamación/metabolismo , Lesión Pulmonar , Ratones , Ratones Noqueados , Oxidación-Reducción , Fosfatidilcolinas/química , Fosfolípidos , Subtipo EP4 de Receptores de Prostaglandina E/genética , Trombina , Factor de Necrosis Tumoral alfa , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
13.
J Immunol ; 197(2): 611-9, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27316682

RESUMEN

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.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Antiinflamatorios/farmacología , Antígenos CD36/antagonistas & inhibidores , Inflamación/inmunología , Lesión Pulmonar Aguda/patología , Animales , Apolipoproteína A-I/inmunología , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inflamación/patología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos/farmacología
14.
J Biol Chem ; 291(19): 10032-45, 2016 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-26884340

RESUMEN

High tidal volume mechanical ventilation and the resultant excessive mechanical forces experienced by lung vascular endothelium are known to lead to increased vascular endothelial leak, but the underlying molecular mechanisms remain incompletely understood. One reported mechanotransduction pathway of increased endothelial cell (EC) permeability caused by high magnitude cyclic stretch (18% CS) involves CS-induced activation of the focal adhesion associated signalosome, which triggers Rho GTPase signaling. This study identified an alternative pathway of CS-induced EC permeability. We show here that high magnitude cyclic stretch (18% CS) rapidly activates VEGF receptor 2 (VEGFR2) signaling by dissociating VEGFR2 from VE-cadherin at the cell junctions. This results in VEGFR2 activation, Src-dependent VE-cadherin tyrosine phosphorylation, and internalization leading to increased endothelial permeability. This process is also accompanied by CS-induced phosphorylation and internalization of PECAM1. Importantly, CS-induced endothelial barrier disruption was attenuated by VEGFR2 inhibition. 18% CS-induced EC permeability was linked to dissociation of cell junction scaffold afadin from the adherens junctions. Forced expression of recombinant afadin in pulmonary endothelium attenuated CS-induced VEGFR2 and VE-cadherin phosphorylation, preserved adherens junction integrity and VEGFR2·VE-cadherin complex, and suppressed CS-induced EC permeability. This study shows for the first time a mechanism whereby VEGFR2 activation mediates EC permeability induced by pathologically relevant cyclic stretch. In this mechanism, CS induces dissociation of the VE-cadherin·VEGFR2 complex localized at the adherens juctions, causing activation of VEGFR2, VEGFR2-mediated Src-dependent phosphorylation of VE-cadherin, disassembly of adherens junctions, and EC barrier failure.


Asunto(s)
Permeabilidad Capilar/fisiología , Endotelio Vascular/metabolismo , Mecanotransducción Celular , Arteria Pulmonar/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Uniones Adherentes/fisiología , Western Blotting , Adhesión Celular/fisiología , Células Cultivadas , Endotelio Vascular/citología , Humanos , Inmunoprecipitación , Fosforilación , Arteria Pulmonar/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
J Biol Chem ; 291(45): 23681-23692, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27590342

RESUMEN

Agonist-induced activation of Rho GTPase signaling leads to endothelial cell (EC) permeability and may culminate in pulmonary edema, a devastating complication of acute lung injury. Cingulin is an adaptor protein first discovered in epithelium and is involved in the organization of the tight junctions. This study investigated the role of cingulin in control of agonist-induced lung EC permeability via interaction with RhoA-specific activator GEF-H1. The siRNA-induced cingulin knockdown augmented thrombin-induced EC permeability monitored by measurements of transendothelial electrical resistance and endothelial cell permeability for macromolecules. Increased thrombin-induced permeability in ECs with depleted cingulin was associated with increased activation of GEF-H1 and RhoA detected in pulldown activation assays. Increased GEF-H1 association with cingulin was essential for down-regulation of thrombin-induced RhoA barrier disruptive signaling. Using cingulin-truncated mutants, we determined that GEF-H1 interaction with the rod + tail domain of cingulin was required for inactivation of GEF-H1 and endothelial cell barrier preservation. The results demonstrate the role for association of GEF-H1 with cingulin as the mechanism of RhoA pathway inactivation and rescue of EC barrier after agonist challenge.


Asunto(s)
Permeabilidad Capilar , Células Endoteliales/metabolismo , Pulmón/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Trombina/metabolismo , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/fisiopatología , Línea Celular , Células Endoteliales/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Humanos , Pulmón/fisiopatología , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/genética , Mutación , Mapas de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal , Proteína de Unión al GTP rhoA/metabolismo
17.
Am J Physiol Lung Cell Mol Physiol ; 313(4): L710-L721, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28663336

RESUMEN

Prostaglandins (PG), the products of cyclooxygenase-mediated conversion of arachidonic acid, become upregulated in many situations including allergic response, inflammation, and injury, and exhibit a variety of biological activities. Previous studies described barrier-enhancing and anti-inflammatory effects of PGE2 and PGI2 on vascular endothelial cells (EC). Yet, the effects of other PG members on EC barrier and inflammatory activation have not been systematically analyzed. This study compared effects of PGE2, PGI2, PGF2α, PGA2, PGJ2, and PGD2 on human pulmonary EC. EC permeability was assessed by measurements of transendothelial electrical resistance and cell monolayer permeability for FITC-labeled tracer. Anti-inflammatory effects of PGs were evaluated by analysis of expression of adhesion molecule ICAM1 and secretion of soluble ICAM1 and cytokines by EC. PGE2, PGI2, and PGA2 exhibited the most potent barrier-enhancing effects and most efficient attenuation of thrombin-induced EC permeability and contractile response, whereas PGI2 effectively suppressed thrombin-induced permeability but was less efficient in the attenuation of prolonged EC hyperpermeability caused by interleukin-6 or bacterial wall lipopolysaccharide, LPS. PGD2 showed a modest protective effect on the EC inflammatory response, whereas PGF2α and PGJ2 were without effect on agonist-induced EC barrier dysfunction. In vivo, PGE2, PGI2, and PGA2 attenuated LPS-induced lung inflammation, whereas PGF2α and PGJ2 were without effect. Interestingly, PGD2 exhibited a protective effect in the in vivo model of LPS-induced lung injury. This study provides a comprehensive analysis of barrier-protective and anti-inflammatory effects of different prostaglandins on lung EC in vitro and in vivo and identifies PGE2, PGI2, and PGA2 as prostaglandins with the most potent protective properties.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Inflamación/tratamiento farmacológico , Lesión Pulmonar/tratamiento farmacológico , Prostaglandinas/farmacología , Animales , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Hemostáticos/efectos adversos , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/efectos adversos , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Ratones , Trombina/efectos adversos
18.
Am J Respir Cell Mol Biol ; 55(4): 476-486, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27115795

RESUMEN

Increased vascular endothelial cell (EC) permeability is a result of intercellular gap formation that may be induced by contraction-dependent and contraction-independent mechanisms. This study investigated a role of the adaptor protein vinculin in EC permeability induced by contractile (thrombin) and noncontractile (IL-6) agonists. Although thrombin and IL-6 caused a similar permeability increase in human pulmonary ECs and disrupted the association between vinculin and vascular endothelial-cadherin, they induced different patterns of focal adhesion (FA) arrangement. Thrombin, but not IL-6, caused formation of large, vinculin-positive FAs, phosphorylation of FA proteins, FA kinase and Crk-associated substrate, and increased vinculin-talin association. Thrombin-induced formation of talin-positive FA and intercellular gaps were suppressed in ECs with small interfering RNA-induced vinculin knockdown. Vinculin knockdown and inhibitors of Rho kinase and myosin-II motor activity also attenuated thrombin-induced EC permeability. Importantly, ectopic expression of the vinculin mutant lacking the F-actin-binding domain decreased thrombin-induced Rho pathway activation and EC permeability. In contrast, IL-6-induced EC permeability did not involve RhoA- or myosin-dependent mechanisms but engaged Janus kinase/signal transducer and activator of transcription-mediated phosphorylation and internalization of vascular endothelial-cadherin. This process was vinculin independent but Janus kinase/tyrosine kinase Src-dependent. These data suggest that vinculin participates in a contractile-dependent mechanism of permeability by integrating FA with stress fibers, leading to maximal RhoA activation and EC permeability response. Vinculin inhibition does not affect contractile-independent mechanisms of EC barrier failure. This study provides, for the first time, a comparative analysis of two alternative mechanisms of vascular endothelial barrier dysfunction and defines a specific role for vinculin in the contractile type of permeability response.

19.
J Biol Chem ; 290(7): 4097-109, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25492863

RESUMEN

Hepatocyte growth factor (HGF) attenuates agonist-induced endothelial cell (EC) permeability and increases pulmonary endothelial barrier function via Rac-dependent enhancement of the peripheral actin cytoskeleton. However, the precise mechanisms of HGF effects on the peripheral cytoskeleton are not well understood. This study evaluated a role for Rac/Cdc42-specific guanine nucleotide exchange factor Asef and the multifunctional Rac effector, IQGAP1, in the mechanism of HGF-induced EC barrier enhancement. HGF induced Asef and IQGAP1 co-localization at the cell cortical area and stimulated formation of an Asef-IQGAP1 functional protein complex. siRNA-induced knockdown of Asef or IQGAP1 attenuated HGF-induced EC barrier enhancement. Asef knockdown attenuated HGF-induced Rac activation and Rac association with IQGAP1, and it abolished both IQGAP1 accumulation at the cell cortical layer and IQGAP1 interaction with actin cytoskeletal regulators cortactin and Arp3. Asef activation state was essential for Asef interaction with IQGAP1 and protein complex accumulation at the cell periphery. In addition to the previously reported role of the IQGAP1 RasGAP-related domain in the Rac-dependent IQGAP1 activation and interaction with its targets, we show that the IQGAP1 C-terminal domain is essential for HGF-induced IQGAP1/Asef interaction and Asef-Rac-dependent activation leading to IQGAP1 interaction with Arp3 and cortactin as a positive feedback mechanism of IQGAP1 activation. These results demonstrate a novel feedback mechanism of HGF-induced endothelial barrier enhancement via Asef/IQGAP1 interactions, which regulate the level of HGF-induced Rac activation and promote cortical cytoskeletal remodeling via IQGAP1-Arp3/cortactin interactions.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Permeabilidad de la Membrana Celular , Endotelio Vascular/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Arteria Pulmonar/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Western Blotting , Células Cultivadas , Endotelio Vascular/citología , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Arteria Pulmonar/citología , ARN Interferente Pequeño/genética , Factores de Intercambio de Guanina Nucleótido Rho/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas Activadoras de ras GTPasa/antagonistas & inhibidores , Proteínas Activadoras de ras GTPasa/genética
20.
Biochim Biophys Acta ; 1852(1): 104-19, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25445540

RESUMEN

Apart from control of circulating fluid, atrial natriuretic peptide (ANP) exhibits anti-inflammatory effects in the lung. However, molecular mechanisms of ANP anti-inflammatory effects are not well-understood. Peripheral microtubule (MT) dynamics is essential for agonist-induced regulation of vascular endothelial permeability. Here we studied the role of MT-dependent signaling in ANP protective effects against endothelial cell (EC) barrier dysfunction and acute lung injury induced by Staphylococcus aureus-derived peptidoglican-G (PepG). PepG-induced vascular endothelial dysfunction was accompanied by MT destabilization and disruption of MT network. ANP attenuated PepG-induced MT disassembly, NFκB signaling and activity of MT-associated Rho activator GEF-H1 leading to attenuation of EC inflammatory activation reflected by expression of adhesion molecules ICAM1 and VCAM1. ANP-induced EC barrier preservation and MT stabilization were linked to phosphorylation and inactivation of MT-depolymerizing protein stathmin. Expression of stathmin phosphorylation-deficient mutant abolished ANP protective effects against PepG-induced inflammation and EC permeability. In contrast, siRNA-mediated stathmin knockdown prevented PepG-induced peripheral MT disassembly and endothelial barrier dysfunction. ANP protective effects in a murine model of PepG-induced lung injury were associated with increased phosphorylation of stathmin, while exacerbated lung injury in the ANP knockout mice was accompanied by decreased pool of stable MT. Stathmin knockdown in vivo reversed exacerbation of lung injury in the ANP knockout mice. These results show a novel MT-mediated mechanism of endothelial barrier protection by ANP in pulmonary EC and animal model of PepG-induced lung injury via stathmin-dependent control of MT assembly.


Asunto(s)
Factor Natriurético Atrial/fisiología , Endotelio Vascular/fisiopatología , Microtúbulos/fisiología , Peptidoglicano/metabolismo , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Noqueados , Fosforilación , Transducción de Señal , Estatmina/genética , Estatmina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA