Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Shock ; 58(1): 68-77, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35670456

ABSTRACT

ABSTRACT: With-No lysine Kinases (WNKs) have been newly implicated in alveolar fluid clearance (AFC). Epithelial sodium channels (ENaCs) serve a vital role in AFC. The potential protective effect of WNK4 in acute respiratory distress syndrome (ARDS), mediated by ENaC-associated AFC was investigated in the study. A model of lipopolysaccharide (LPS)-induced ARDS was established in C57BL/6 mice. WNK4, Sterile 20-related proline-alanine-rich kinase (SPAK), small interfering RNA (siRNA)-WNK4 or siRNA-SPAK were transfected into mouse lung or primary alveolar epithelial type II (ATII) cells. AFC, bronchoalveolar lavage fluid and lung histomorphology were determined. The expression of ENaC was determined to investigate the regulation of AFC by WNK4-SPAK signaling pathway. Activation of WNK4-SPAK signaling improved lung injury and survival rate, with enhanced AFC and reduced pulmonary edema via the upregulation of ENaC in ARDS. In primary rat ATII cells, gene-silencing by siRNA transfection reduced ENaC expression and the level of WNK4-associated SPAK phosphorylation. Immunoprecipitation revealed that the level of neural precursor cell-expressed developmentally downregulated gene 4 (Nedd4-2) binding to ENaC was decreased as a result of WNK4-SPAK signaling. The present study demonstrated that the WNK4/SPAK pathway improved AFC during LPS-induced ARDS, which is mainly dependent on the upregulation of ENaC with Nedd4-2-mediated ubiquitination.


Subject(s)
Epithelial Sodium Channels , Protein Serine-Threonine Kinases , Respiratory Distress Syndrome , Animals , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Rats , Respiratory Distress Syndrome/chemically induced , Signal Transduction , Up-Regulation
2.
Oxid Med Cell Longev ; 2022: 5475832, 2022.
Article in English | MEDLINE | ID: mdl-35571250

ABSTRACT

The "obesity paradox in acute respiratory distress syndrome" (ARDS) refers to the phenomenon in which obesity is associated with higher morbidity but lower mortality in patients with ARDS. Endothelial-to-mesenchymal transition (EndMT) represents a key link in the interaction between endothelial disruption and mesenchymal fibrosis under inflammatory and oxidative conditions, which represent the intersectional pathophysiology of ARDS. Adipose tissue is considered to constitute the major source of circulating exosomal microRNAs (miRNAs), which act as genetic forms of adipokines for cell-cell crosstalk. We aimed to demonstrate the regulation and mechanism of adipose-derived exosomes in the obesity paradox in ARDS. High-fat-induced obese mice and lean control mice were subjected to ARDS insult to investigate the effects of obesity on ARDS and microarray analysis was performed to screen for differences in circulating miRNAs. In addition, mice and pulmonary endothelial cells were administered with adipose-derived exosomal miR-122-5p to investigate the underlying molecular mechanisms. We found high-fat diet-induced obesity protected against ARDS in mice by reinforcing endothelial barrier and attenuating fibroproliferation. Circulating exosomes produced in the obese state mediated these protective effects by inhibiting EndMT and oxidative stress. Mechanistically, adipose-derived exosomal miR-122-5p promoted the integrity and function of pulmonary endothelial barrier and alleviated fibrogenesis by suppressing EndMT and oxidative stress through down-regulation of the transforming growth factor ß1 (TGF-ß1)/TGF-ß receptor 1 (TGF-ßR1)/Smad2 pathway in vivo and in vitro. In conclusion, adipose-derived circulating exosomal miR-122-5p protects against ARDS by reinforcing pulmonary endothelial barrier through inhibition of EndMT and oxidative stress via down-regulation of the TGF-ß pathway, which propose a potential explanation for the obesity paradox in ARDS and indicate promising prospects for adipose-derived exosomes in cell-free therapies for ARDS.


Subject(s)
Exosomes , MicroRNAs , Respiratory Distress Syndrome , Adipose Tissue/metabolism , Animals , Down-Regulation , Endothelial Cells/metabolism , Exosomes/metabolism , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/complications , Obesity/metabolism , Oxidative Stress , Signal Transduction , Transforming Growth Factor beta/metabolism
3.
Int Immunopharmacol ; 109: 108805, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35504205

ABSTRACT

Pulmonary vascular endothelial dysfunction is a key pathogenic mechanism in acute respiratory distress syndrome (ARDS), resulting in fibrosis in lung tissues, including in the context of COVID-19. Pirfenidone (PFD) has become a novel therapeutic agent for treating idiopathic pulmonary fibrosis (IPF) and can improve lung function, inhibit fibrosis and inhibit inflammation. Recently, endothelial-to-mesenchymal transition (EndMT) was shown to play a crucial role in various respiratory diseases. However, the role of PFD in the course of EndMT in LPS-induced ARDS remains poorly understood. The purpose of this study was to explore the anti-EndMT effects of PFD on pulmonary fibrosis after LPS-induced ARDS. First, we determined that PFD significantly reduced LPS-induced ARDS, as shown by significant pathological alterations, and alleviated the oxidative stress and inflammatory response in vitro and in vivo. Furthermore, PFD decreased pulmonary fibrosis in LPS-induced ARDS by inhibiting EndMT and reduced the expression levels of Hedgehog (HH) pathway target genes, such as Gli1 and α-SMA, after LPS induction. In summary, this study confirmed that inhibiting the HH pathway by PFD could decrease pulmonary fibrosis by downregulating EndMT in LPS-induced ARDS. In conclusion, we demonstrate that PFD is a promising agent to attenuate pulmonary fibrosis following ARDS in the future.


Subject(s)
Hedgehog Proteins , Pulmonary Fibrosis , Pyridones , Respiratory Distress Syndrome , Animals , Hedgehog Proteins/metabolism , Lipopolysaccharides , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pyridones/pharmacology , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/drug therapy , Signal Transduction
4.
Pulm Pharmacol Ther ; 73-74: 102123, 2022 06.
Article in English | MEDLINE | ID: mdl-35306165

ABSTRACT

PURPOSE: The "obesity paradox" phenomenon occurs in critically ill patients who receive mechanical ventilation. Our previous studies found that the adipose-derived exosomes secreted by obese mice have a protective effect on the pulmonary microvascular endothelial barrier. However, the extraction of exosomes is cumbersome, their yield is low, and their storage is difficult. After further research, we discovered a new type of adipose-derived bioactive material called: lipoaspirate nanoparticles (Lipo-NPs). METHODS: Lipo-NPs were extracted and identified using a tangential flow filtration system. The Lipo-NPs were used as an intervention in ventilator-induced lung injury (VILI) models in vivo and in vitro to investigate whether they have a protective effect on lung tissue damage (haematoxylin and eosin staining), lung barrier function (lung wet/dry [W/D] weight ratio, protein concentration in bronchoalveolar lavage fluid (BALF), and Vascular endothelial (VE)-expression), as well as their related mechanisms. RESULTS: In both in vivo and in vitro studies, Lipo-NPs can attenuate lung injury, reduce lung W/D ratio and protein concentration in BALF, and augment the expression of the adhesion link-protein VE-cadherin, thus playing a protective role in lung barrier function. This protective effect involves the activation of the transient receptor potential vanilloid 4 (TRPV4)/Rho-associated kinase1 (ROCK1) signalling pathway. We further verified the role of this signalling pathway via activation and inhibition of TRPV4 and ROCK1. Moreover, phosphorylation of myosin light chain 2 (MLC2) regulates F-actin and is a target of the ROCK pathway. CONCLUSION: Lipo-NPs can enhance the expression of VE-cadherin by inhibiting the TRPV4/ROCK1/pMLC2 signalling pathway in the mechanical ventilation model, thereby exerting a protective effect on the VILI pulmonary microvascular endothelial barrier.


Subject(s)
Nanoparticles , TRPV Cation Channels , Ventilator-Induced Lung Injury , rho-Associated Kinases , Animals , Humans , Lung/metabolism , Mice , Mice, Inbred C57BL , Respiration, Artificial , TRPV Cation Channels/metabolism , Ventilator-Induced Lung Injury/prevention & control , rho-Associated Kinases/metabolism
5.
Mol Med Rep ; 24(4)2021 Oct.
Article in English | MEDLINE | ID: mdl-34396442

ABSTRACT

Chronic alcohol abuse increases the risk of mortality and poor outcomes in patients with acute respiratory distress syndrome. However, the underlying mechanisms remain to be elucidated. The present study aimed to investigate the effects of chronic alcohol consumption on lung injury and clarify the signaling pathways involved in the inhibition of alveolar fluid clearance (AFC). In order to produce rodent models with chronic alcohol consumption, wild­type C57BL/6 mice were treated with alcohol. A2a adenosine receptor (AR) small interfering (si)RNA or A2bAR siRNA were transfected into the lung tissue of mice and primary rat alveolar type II (ATII) cells. The rate of AFC in lung tissue was measured during exposure to lipopolysaccharide (LPS). Epithelial sodium channel (ENaC) expression was determined to investigate the mechanisms underlying alcohol­induced regulation of AFC. In the present study, exposure to alcohol reduced AFC, exacerbated pulmonary edema and worsened LPS­induced lung injury. Alcohol caused a decrease in cyclic adenosine monophosphate (cAMP) levels and inhibited α­ENaC, ß­ENaC and γ­ENaC expression levels in the lung tissue of mice and ATII cells. Furthermore, alcohol decreased α­ENaC, ß­ENaC and γ­ENaC expression levels via the A2aAR or A2bAR­cAMP signaling pathways in vitro. In conclusion, the results of the present study demonstrated that chronic alcohol consumption worsened lung injury by aggravating pulmonary edema and impairing AFC. An alcohol­induced decrease of α­ENaC, ß­ENaC and γ­ENaC expression levels by the A2AR­mediated cAMP pathway may be responsible for the exacerbated effects of chronic alcohol consumption in lung injury.


Subject(s)
Acute Lung Injury/metabolism , Alveolar Epithelial Cells/metabolism , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/metabolism , Ethanol/pharmacology , Receptors, Adenosine A2/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Alveolar Epithelial Cells/pathology , Animals , Cyclic AMP/metabolism , Cytokines , Lipopolysaccharides/adverse effects , Lung/metabolism , Lung Injury/chemically induced , Lung Injury/metabolism , Lung Injury/pathology , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/metabolism , Pulmonary Edema/chemically induced , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Rats , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Signal Transduction
6.
Int Immunopharmacol ; 88: 106951, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32892076

ABSTRACT

Inflammatory storms and endothelial barrier dysfunction are the central pathophysiological features of acute respiratory distress syndrome (ARDS). Intermedin (IMD), a member of the calcitonin gene-related peptide (CGRP) family, has been reported to alleviate inflammation and protect endothelial cell (EC) integrity. However, the effects of IMD on ARDS have not been clearly elucidated. In the present study, clinical ARDS data were used to explore the relationship between serum IMD levels and disease severity and prognosis, and we then established a model to predict the possibility of hospital survival. Mouse models of ARDS and LPS-challenged endothelial cells were used to analyze the protective effect and underlying mechanism of IMD. We found that in patients with ARDS, increased serum IMD levels were associated with reduced disease severity and increased rates of hospital survival. IMD alleviated the LPS-induced inflammatory response by decreasing proinflammatory cytokines, NF-κB p65 expression and NF-κB p65 nuclear translocation. In addition, IMD stabilized the endothelial barrier by repairing adherens junctions (AJs), cytoskeleton and capillary leakage. IMD exerted protective effects against ARDS on pulmonary endothelial cells, at least partly, through PI3K/Akt/eNOS signaling, while IMD's anti-inflammation effect was mediated through an eNOS-independent mechanism. Our study may provide new therapeutic insight for ARDS treatment.


Subject(s)
Peptide Hormones/blood , Respiratory Distress Syndrome/blood , Animals , Endothelial Cells/metabolism , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides , Lung/metabolism , Lung/pathology , Male , Mice, Inbred C57BL , Neuropeptides/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Shock ; 54(1): 110-118, 2020 07.
Article in English | MEDLINE | ID: mdl-32530844

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a critical clinical syndrome with high mortality rate, and few effective therapies have been found in the past 50 years, indicating that the pathogenesis of ARDS remains unclear. Exosomes, a novel cross-communication mechanism, are involved in critical diseases. However, the role of circulating exosomes in the development of ARDS remains poorly understood. METHODS: In the present study, naive mice were treated with circulating exosomes from lipopolysaccharide (LPS)-induced ARDS mice or exosome-depleted serum. Histological lung damage, bronchoalveolar lavage fluid (BALF), and endoplasmic reticulum (ER) stress were measured. RESULTS: Increased tumor necrosis factor (TNF)-α, interleukin (IL)-6, total cell counts, polymorphonuclear (PMN) leukocyte proportions and myeloperoxidase (MPO) activity in BALF, and increased wet/dry weight ratios and protein concentrations in BALF were found in mice after exosome injection but not in mice treated with exosome-depleted serum. Furthermore, western blot analysis showed that circulating exosomes from ARDS mice upregulated glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP) expression and downregulated ß-Catenin and VE-cadherin expression in lung tissues. CONCLUSIONS: Collectively, these data demonstrate that circulating exosomes from LPS-induced ARDS mice trigger ER stress in lung tissue, facilitating the development of ARDS, at least partly by promoting endothelial dysfunction.


Subject(s)
Endoplasmic Reticulum Stress , Exosomes/metabolism , Lung/pathology , Respiratory Distress Syndrome/pathology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Interleukin-6/analysis , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Neutrophils , Peroxidase/metabolism , Tumor Necrosis Factor-alpha/analysis
8.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L723-L741, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32073873

ABSTRACT

Mechanical ventilation (MV) is the main supportive treatment of acute respiratory distress syndrome (ARDS), but it may lead to ventilator-induced lung injury (VILI). Large epidemiological studies have found that obesity was associated with lower mortality in mechanically ventilated patients with acute lung injury, which is known as "obesity paradox." However, the effects of obesity on VILI are unknown. In the present study, wild-type mice were fed a high-fat diet (HFD) and ventilated with high tidal volume to investigate the effects of obesity on VILI in vivo, and pulmonary microvascular endothelial cells (PMVECs) were subjected to 18% cyclic stretching (CS) to further investigate its underlying mechanism in vitro. We found that HFD protects mice from VILI by alleviating the pulmonary endothelial barrier injury and inflammatory responses in mice. Adipose-derived exosomes can regulate distant tissues as novel adipokines, providing a new mechanism for cell-cell interactions. We extracted three adipose-derived exosomes, including HFD mouse serum exosome (S-Exo), adipose tissue exosome (AT-Exo), and adipose-derived stem cell exosome (ADSC-Exo), and further explored their effects on MV or 18% CS-induced VILI in vivo and in vitro. Administration of three exosomes protected against VILI by suppressing pulmonary endothelial barrier hyperpermeability, repairing the expression of adherens junctions, and alleviating inflammatory response in vivo and in vitro, accompanied by transient receptor potential vanilloid 4 (TRPV4)/Ca2+ pathway inhibition. Collectively, these data indicated that HFD-induced obesity plays a protective role in VILI by alleviating the pulmonary endothelial barrier injury and inflammatory response via adipose-derived exosomes, at least partially, through inhibiting the TRPV4/Ca2+ pathway.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Endothelial Cells/metabolism , Exosomes/metabolism , Respiratory Distress Syndrome/metabolism , TRPV Cation Channels/metabolism , Ventilator-Induced Lung Injury/metabolism , Adherens Junctions/metabolism , Animals , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Respiration, Artificial/adverse effects , Signal Transduction/physiology , Tidal Volume/physiology
9.
Cell Mol Biol Lett ; 24: 35, 2019.
Article in English | MEDLINE | ID: mdl-31160894

ABSTRACT

BACKGROUND: Pulmonary edema is one of the pathological characteristics of acute respiratory distress syndrome (ARDS). The epithelial sodium channel (ENaC) is thought to be the rate-limiting factor for alveolar fluid clearance (AFC) during pulmonary edema. The peroxisome proliferator-activated receptor γ (PPARγ) agonist rosiglitazone was shown to stimulate ENaC-mediated salt absorption in the kidney. However, its role in the lung remains unclear. Here, we investigated the role of the PPARγ agonist in the lung to find out whether it can regulate AFC during acute lung injury (ALI). We also attempted to elucidate the mechanism for this. METHODS: Our ALI model was established through intratracheal instillation of lipopolysaccharide (LPS) in C57BL/6 J mice. The mice were randomly divided into 4 groups of 10. The control group underwent a sham operation and received an equal quantity of saline. The three experimental groups underwent intratracheal instillation of 5 mg/kg LPS, followed by intraperitoneal injection of 4 mg/kg rosiglitazone, 4 mg/kg rosiglitazone plus 1 mg/kg GW9662, or only equal quantity of saline. The histological morphology of the lung, the levels of TNF-α and IL-1ß in the bronchoalveolar lavage fluid (BALF), the level of AFC, and the expressions of αENaC and serum and glucocorticoid-induced kinase-1 (SGK1) were determined. Type 2 alveolar (AT II) cells were incubated with rosiglitazone (15 µM) with or without GW9662 (10 µM). The expressions of αENaC and SGK1 were determined 24 h later. RESULTS: A mouse model of ALI was successfully established. Rosiglitazone significantly ameliorated the lung injury, decreasing the TNF-α and IL-1ß levels in the BALF, enhancing AFC, and promoting the expressions of αENaC and SGK1 in ALI mice, which were abolished by the specific PPARγ blocker GW9662. In vitro, rosiglitazone increased the expressions of αENaC and SGK1. This increase was prevented by GW9662. CONCLUSIONS: Rosiglitazone ameliorated the lung injury and promoted ENaC-mediated AFC via a PPARγ/SGK1-dependent signaling pathway, alleviating pulmonary edema in a mouse model of ALI.


Subject(s)
Acute Lung Injury/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Epithelial Sodium Channels/metabolism , PPAR gamma/metabolism , Protein Serine-Threonine Kinases/metabolism , Rosiglitazone/pharmacology , Signal Transduction , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Disease Models, Animal , Inflammation Mediators/metabolism , Lipopolysaccharides , Lung/metabolism , Lung/pathology , Male , Mice, Inbred C57BL , Phosphorylation/drug effects , Signal Transduction/drug effects
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(3): 283-288, 2018 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-29643033

ABSTRACT

OBJECTIVE: To investigate the effects of Vaspin on lipopolysaccharide (LPS)-induced acute respiratory distress syndrome (ARDS) in mice and explore the possible mechanism. METHODS: Forty male C57B/L6 mice were randomized equally into control group, LPS group, Vaspin group and wortmannin group with corresponding treatments. The pathological changes of the lung tissues were evaluated by HE staining, and the severity of pulmonary edema was measured according to the wet/dry ratio (W/D) of the lung tissue. The lung permeability was evaluated by detecting total protein concentrations in the bronchoalveolar lavage fluid (BALF) using bicinchoninic acid (BCA) assay. Myeloperoxidase (MPO) activity in the lung tissue was detected using a MPO assay kit, and the levels of interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) in the lungs were measured using ELISA. Immunohistochemical staining was performed to detect the expression of vascular cell adhesion molecule-1 (VCAM-1) and Western blotting was used to detect the protein expressions of cleaved caspase-3 and p-Akt in the lung tissues. RESULTS: Compared with the control group, the mice in LPS group displayed typical ARDS pathological changes in the lungs with significantly increased W/D, total protein concentrations in BALF, lung MPO activity, levels of IL-1ß and TNF-α, and pulmonary expressions of VCAM-1 and cleaved caspase-3 (P<0.05) but decreased expression of p-Akt (P<0.05). These changes induced by LPS were significantly alleviated by the administration of Vaspin (P<0.05). The protective effects of Vaspin against ARDS were obviously attenuated by the PI3K inhibitor wortmannin (P<0.05). CONCLUSION: Vaspin protects against LPS-induced ARDS in mice possibly by inhibiting inflammation and protecting vascular endothelium through upregulation of the PI3K/Akt signal pathway.


Subject(s)
Adipokines/metabolism , Endothelium, Vascular/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Respiratory Distress Syndrome/metabolism , Serpins/metabolism , Signal Transduction , Animals , Bronchoalveolar Lavage Fluid , Caspase 3/metabolism , Inflammation , Interleukin-1beta/metabolism , Lipopolysaccharides , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Random Allocation , Respiratory Distress Syndrome/chemically induced , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Wortmannin/pharmacology
11.
Int J Mol Med ; 40(6): 1803-1817, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039444

ABSTRACT

Acute respiratory distress syndrome (ARDS) is characterized by uncontrolled extravasation of protein­rich fluids, which is caused by disruption and dysfunction of the barrier of pulmonary endothelial cells (ECs). Visceral adipose tissue­derived serine protease inhibitor (vaspin) is a novel adipokine with pleiotropic properties, which has been reported to exert beneficial effects against obesity­associated systemic vascular diseases; however, its effects on ARDS remain unknown. In the present study, mice were subjected to systemic administration of adenoviral vector expressing vaspin (Ad­vaspin) to examine its effects on lipopolysaccharide (LPS)­induced ARDS in vivo. Histological analysis was then conducted, and cytokine [tumor necrosis factor (TNF)­α, interleukin (IL)­6 and IL­10] levels, and intercellular cell adhesion molecule­1 (ICAM­1) and adherens junctions (AJs) expression were detected. In addition, human pulmonary microvascular ECs (HPMECs) were treated with recombinant human (rh)­vaspin to further investigate its molecular basis and underlying mechanism. The mRNA expression levels of inflammatory cytokines (TNF­α and IL­6) and endothelial­specific adhesion markers [vascular cell adhesion molecule­1 and E­selectin], activation of nuclear factor­κB, and cell viability and apoptosis were then examined. Furthermore, the expression of AJs and organization of the cytoskeleton, as well as expression and activity of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and generation of reactive oxygen species (ROS) were determined. The results indicated that Ad­vaspin protected against LPS­induced ARDS by alleviating the pulmonary inflammatory response and pulmonary EC barrier dysfunction in mice, which was accompanied by activation of the protein kinase B (Akt)/glycogen synthase kinase (GSK)­3ß pathway. In addition, pretreatment of HPMECs with rh­vaspin attenuated inflammation, apoptosis and ROS generation without alterations in AJs and cytoskeletal organization following LPS insult, which was accompanied by activation of the Akt/GSK3ß pathway. In conclusion, the present study demonstrated that vaspin protects against LPS­induced ARDS by reversing EC barrier dysfunction via the suppression of inflammation, apoptosis and ROS production in pulmonary ECs, at least partially via activation of the Akt/GSK3ß pathway. These findings provide evidence of a causal link between vaspin and EC dysfunction in ARDS, and suggest a potential therapeutic intervention for patients with ARDS.


Subject(s)
Apoptosis/drug effects , Inflammation/metabolism , Reactive Oxygen Species/metabolism , Respiratory Distress Syndrome/metabolism , Serpins/pharmacology , Signal Transduction/physiology , Animals , Cell Line , Cell Survival/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Lipopolysaccharides/adverse effects , Lung , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/analysis , Recombinant Proteins/pharmacology , Respiratory Distress Syndrome/chemically induced , Signal Transduction/drug effects
12.
Cell Death Dis ; 7(9): e2360, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27607575

ABSTRACT

Acute respiratory distress syndrome (ARDS) is characterized by increased pulmonary inflammation and endothelial barrier permeability. Omentin has been shown to benefit obesity-related systemic vascular diseases; however, its effects on ARDS are unknown. In the present study, the level of circulating omentin in patients with ARDS was assessed to appraise its clinical significance in ARDS. Mice were subjected to systemic administration of adenoviral vector expressing omentin (Ad-omentin) and one-shot treatment of recombinant human omentin (rh-omentin) to examine omentin's effects on lipopolysaccharide (LPS)-induced ARDS. Pulmonary endothelial cells (ECs) were treated with rh-omentin to further investigate its underlying mechanism. We found that a decreased level of circulating omentin negatively correlated with white blood cells and procalcitonin in patients with ARDS. Ad-omentin protected against LPS-induced ARDS by alleviating the pulmonary inflammatory response and endothelial barrier injury in mice, accompanied by Akt/eNOS pathway activation. Treatment of pulmonary ECs with rh-omentin attenuated inflammatory response and restored adherens junctions (AJs), and cytoskeleton organization promoted endothelial barrier after LPS insult. Moreover, the omentin-mediated enhancement of EC survival and differentiation was blocked by the Akt/eNOS pathway inactivation. Therapeutic rh-omentin treatment also effectively protected against LPS-induced ARDS via the Akt/eNOS pathway. Collectively, these data indicated that omentin protects against LPS-induced ARDS by suppressing inflammation and promoting the pulmonary endothelial barrier, at least partially, through an Akt/eNOS-dependent mechanism. Therapeutic strategies aiming to restore omentin levels may be valuable for the prevention or treatment of ARDS.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cytokines/pharmacology , Endothelial Cells/drug effects , Lectins/pharmacology , Nitric Oxide Synthase Type III/immunology , Pneumonia/drug therapy , Proto-Oncogene Proteins c-akt/immunology , Respiratory Distress Syndrome/drug therapy , Adenoviridae/genetics , Adenoviridae/metabolism , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Adherens Junctions/ultrastructure , Animals , Anti-Inflammatory Agents, Non-Steroidal/immunology , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Calcitonin/genetics , Calcitonin/immunology , Cell Survival/drug effects , Cytokines/genetics , Cytokines/immunology , Endothelial Cells/cytology , Endothelial Cells/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , GPI-Linked Proteins/pharmacology , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Lectins/genetics , Lectins/immunology , Lipopolysaccharides , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Nitric Oxide Synthase Type III/genetics , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/mortality , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/mortality , Signal Transduction , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...