Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Adv Res ; 2024 Jan 17.
Article de Anglais | MEDLINE | ID: mdl-38237770

RÉSUMÉ

INTRODUCTION: Acute respiratory distress syndrome (ARDS) is a pulmonary inflammatory process primarily caused by sepsis. The resolution of inflammation is an active process involving the endogenous biosynthesis of specialized pro-resolving mediators, including resolvin D1 (RvD1). Resident alveolar macrophages (RAMs) maintain pulmonary homeostasis and play a key role in the resolution phase. However, the role of RAMs in promoting the resolution of inflammation by RvD1 is unclear. OBJECTIVES: Here, we investigated the mechanisms of RvD1 on regulating RAMs to promote the resolution of ARDS. METHODS: Mice were administered lipopolysaccharide and/or Escherichia coli via aerosol inhalation to establish a self-limited ARDS model. Then, RvD1 was administered at the peak inflammatory response. RAMs self-renewal was measured by flow cytometry, RAM phagocytosis was measured by two-photon fluorescence imaging. In addition, plasma was collected from intensive care unit patients on days 0-2, 3-5, and 6-9 to measure RvD1 and S100A8/A9 levels using triple quadrupole/linear ion trap mass spectrometry. RESULTS: RAMs were found to play a pivotal role in resolving inflammation during ARDS, and RvD1 enhanced RAM proliferation and phagocytosis, which was abrogated by a lipoxin A4 receptor (ALX, RvD1 receptor) inhibitor. Both primary RAMs transfected with rS100A8/A9 and/or S100A8/A9 siRNA and S100A9-/- mice (also deficient in S100A8 function) showed higher turnover and phagocytic function, indicating that RvD1 exerted its effects on RAMs by inhibiting S100A8/A9 production in the resolution phase. RvD1 reduced S100A8/A9 and its upstream MAPK14 levels in vivo and in vitro. Finally, in the patients, RvD1 levels were lower, but S100A8/A9 levels were higher. CONCLUSIONS: We propose that RvD1 improved RAM self-renewal and phagocytosis via the ALX/MAPK14/S100A8/A9 signaling pathway. Plasma RvD1 and S100A8/A9 levels were negatively correlated, and associated with the outcome of sepsis-induced ARDS.

2.
Int Immunopharmacol ; 102: 108348, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-34920958

RÉSUMÉ

PURPOSE: Acute respiratory distress syndrome (ARDS) is characterized by uncontrollable inflammation. Cyclooxygenase-2(COX-2) and its metabolite prostaglandins are known to promote the inflammatory resolution of ARDS. Recently, a newly discovered endogenous lipid mediator, Protectin DX (PDX), was also shown to mediate the resolution of inflammation. However, the regulatory of PDX on the pro-resolving COX-2 in ARDS remains unknown. MATERIAL AND METHODS: PDX (5 µg/kg) was injected into rats intravenously 12 h after the lipopolysaccharide (LPS, 3 mg/kg) challenge. Primary rat lung fibroblasts were incubated with LPS (1 µg/ml) and/or PDX (100 nM). Lung pathological changes examined using H&E staining. Protein levels of COX-2, PGDS and PGES were evaluated using western blot. Inflammatory cytokines were tested by qPCR, and the concentration of prostaglandins measured by using ELISA. RESULTS: Our study revealed that, COX-2 and L-PGDS has biphasic activation characteristics that LPS could induce induced by LPS both in vivo and in vitro.. The secondary peak of COX-2, L-PGDS-PGD2 promoted the inflammatory resolution in ARDS model with the DP1 receptor being activated and PDX up-regulated the inflammatory resolutionvia enhancing the secondary peak of COX-2/L-PGDS-PGD2 and activating the DP1 receptor. CONCLUSION: PDX promoted the resolution of inflammation of ARDS model via enhancing the expression of secondary peak of COX-2/L-PGDS-PGD2 and activating the DP1 receptor. PDX shows promising therapeutic potential in the clinical management of ARDS.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Acide docosahexaénoïque/usage thérapeutique , /traitement médicamenteux , Animaux , Anti-inflammatoires/pharmacologie , Cellules cultivées , Cyclooxygenase 2/métabolisme , Acide docosahexaénoïque/pharmacologie , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Intramolecular oxidoreductases/métabolisme , Lipocalines/métabolisme , Lipopolysaccharides/pharmacologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Mâle , Prostaglandine D2/métabolisme , Rat Sprague-Dawley , Récepteur prostaglandine/métabolisme , /métabolisme
3.
Cell Death Discov ; 7(1): 339, 2021 Nov 08.
Article de Anglais | MEDLINE | ID: mdl-34750369

RÉSUMÉ

The uncontrolled inflammatory response caused by a disorder in inflammation resolution is one of the reasons for acute respiratory distress syndrome (ARDS). The macrophage pool markedly expands when inflammatory monocytes, known as recruited macrophages, migrate from the circulation to the lung. The persistent presence of recruited macrophages leads to chronic inflammation in the resolution phase of inflammation. On the contrary, elimination of the recruited macrophages at the injury site leads to the rapid resolution of inflammation. Resolvin D1 (RvD1) is an endogenous lipid mediator derived from docosahexaenoic acid. Mice were administered RvD1 via the tail vein 3 and 4 days after stimulation with lipopolysaccharide. RvD1 reduced the levels of the inflammatory factors in the lung tissue, promoted the anti-inflammatory M2 phenotype, and enhanced the phagocytic function of recruited macrophages to alleviate acute lung injury. We also found that the number of macrophages was decreased in BAL fluid after treatment with RvD1. RvD1 increased the apoptosis of recruited macrophages partly via the FasL-FasR/caspase-3 signaling pathway, and this effect could be blocked by Boc-2, an ALX/PRP2 inhibitor. Taken together, our findings reinforce the concept of therapeutic targeting leading to the apoptosis of recruited macrophages. Thus, RvD1 may provide a new therapy for the resolution of ARDS.

4.
J Pharmacol Exp Ther ; 379(2): 156-165, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34465632

RÉSUMÉ

Acute respiratory distress syndrome (ARDS), a common and fatal clinical condition, is characterized by the destruction of epithelium and augmented permeability of the alveolar-capillary barrier. Resolvin conjugates in tissue regeneration 1 (RCTR1) is an endogenous lipid mediator derived from docosahexaenoic acid , exerting proresolution effects in the process of inflammation. In our research, we evaluated the role of RCTR1 in alveolar fluid clearance (AFC) in lipopolysaccharide-induced ARDS/acute lung injury (ALI) rat model. Rats were injected with RCTR1 (5 µg/kg) via caudal veins 8 hours after lipopolysaccharide (LPS) (14 mg/kg) treatment, and then AFC was estimated after 1 hour of ventilation. Primary type II alveolar epithelial cells were incubated with LPS (1 ug/ml) with or without RCTR1 (10 nM) for 8 hours. Our results showed that RCTR1 significantly enhanced the survival rate, promoted the AFC, and alleviated LPS-induced ARDS/ALI in vivo. Furthermore, RCTR1 remarkably elevated the protein expression of sodium channels and Na, K-ATPase and the activity of Na, K-ATPase in vivo and in vitro. Additionally, RCTR1 also decreased neural precursor cell expressed developmentally downregulated 4-2 (Nedd4-2) level via upregulating Ser473-phosphorylated-Akt expression. Besides this, inhibitors of receptor for lipoxin A4 (ALX), cAMP, and phosphatidylinositol 3-kinase (PI3K) (BOC-2, KH-7, and LY294002) notably inhibited the effects of RCTR1 on AFC. In summary, RCTR1 enhances the protein levels of sodium channels and Na, K-ATPase and the Na, K-ATPase activity to improve AFC in ALI through ALX/cAMP/PI3K/Nedd4-2 pathway, suggesting that RCTR1 may become a therapeutic drug for ARDS/ALI. SIGNIFICANCE STATEMENT: RCTR1, an endogenous lipid mediator, enhanced the rate of AFC to accelerate the resolution of inflammation in the LPS-induced murine lung injury model. RCTR1 upregulates the expression of epithelial sodium channels (ENaCs) and Na, K-ATPase in vivo and in vitro to accelerate the AFC. The efficacy of RCTR1 on the ENaC and Na, K-ATPase level was in an ALX/cAMP/PI3K/Nedd4-2-dependent manner.


Sujet(s)
Lésion pulmonaire aigüe/métabolisme , Acide docosahexaénoïque/pharmacologie , Agonistes de canaux sodiques épithéliaux/pharmacologie , Canaux sodium épithéliaux/métabolisme , Alvéoles pulmonaires/métabolisme , Sodium-Potassium-Exchanging ATPase/métabolisme , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/traitement médicamenteux , Animaux , Acide docosahexaénoïque/analogues et dérivés , Acide docosahexaénoïque/usage thérapeutique , Lipopolysaccharides/toxicité , Mâle , Alvéoles pulmonaires/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley
5.
J Cell Mol Med ; 24(23): 14001-14012, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33098250

RÉSUMÉ

Acute respiratory distress syndrome/acute lung injury (ARDS/ALI) is histologically characterized by extensive alveolar barrier disruption and excessive fibroproliferation responses. Protectin DX (PDX) displays anti-inflammatory and potent inflammation pro-resolving actions. We sought to investigate whether PDX attenuates LPS (lipopolysaccharide)-induced lung injury via modulating epithelial cell injury repair, apoptosis and fibroblasts activation. In vivo, PDX was administered intraperitoneally (IP) with 200 ng/per mouse after intratracheal injection of LPS, which remarkedly stimulated proliferation of type II alveolar epithelial cells (AT II cells), reduced the apoptosis of AT II cells, which attenuated lung injury induced by LPS. Moreover, primary type II alveolar cells were isolated and cultured to assess the effects of PDX on wound repair, apoptosis, proliferation and transdifferentiation in vitro. We also investigated the effects of PDX on primary rat lung fibroblast proliferation and myofibroblast differentiation. Our result suggests PDX promotes primary AT II cells wound closure by inducing the proliferation of AT II cells and reducing the apoptosis of AT II cells induced by LPS, and promotes AT II cells transdifferentiation. Furthermore, PDX inhibits transforming growth factor-ß1 (TGF-ß1 ) induced fibroproliferation, fibroblast collagen production and myofibroblast transformation. Furthermore, the effects of PDX on epithelial wound healing and proliferation, fibroblast proliferation and activation partly via the ALX/ PI3K signalling pathway. These data present identify a new mechanism of PDX which targets the airway epithelial cell and fibroproliferation are potential for treatment of ARDS/ALI.


Sujet(s)
Pneumocytes/effets des médicaments et des substances chimiques , Pneumocytes/métabolisme , Kinase du lymphome anaplasique/métabolisme , Acide docosahexaénoïque/pharmacologie , Phosphatidylinositol 3-kinases/métabolisme , Lésion pulmonaire aigüe/étiologie , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/anatomopathologie , Angiotensine-II/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Modèles animaux de maladie humaine , Médiateurs de l'inflammation , Lipopolysaccharides/effets indésirables , Souris , Rats
6.
J Cell Mol Med ; 24(18): 10604-10614, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32735065

RÉSUMÉ

Inflammatory cell infiltration contributes to the pathogenesis of acute respiratory distress syndrome (ARDS). Protectin DX (PDX), an endogenous lipid mediator, shows anti-inflammatory and proresolution bioactions. In vivo, the mice were intraperitoneally injected with PDX (0.1 µg/mouse) after intratracheal (1 mg/kg) or intraperitoneal (10 mg/kg) LPS administration. Flow cytometry was used to measure inflammatory cell numbers. Clodronate liposomes were used to deplete resident macrophages. RT-PCR, and ELISA was used to measure MIP-2, MCP-1, TNF-α and MMP9 levels. In vitro, sorted neutrophils, resident and recruited macrophages (1 × 106 ) were cultured with 1 µg/mL LPS and/or 100 nmol/L PDX to assess the chemokine receptor expression. PDX attenuated LPS-induced lung injury via inhibiting recruited macrophage and neutrophil recruitment through repressing resident macrophage MCP-1, MIP-2 expression and release, respectively. Finally, PDX inhibition of neutrophil infiltration and transmembrane was associated with TNF-α/MIP-2/MMP9 signalling pathway. These data suggest that PDX attenuates LPS-stimulated lung injury via reduction of the inflammatory cell recruitment mediated via resident macrophages.


Sujet(s)
Lésion pulmonaire aigüe/anatomopathologie , Acide docosahexaénoïque/usage thérapeutique , Macrophages/effets des médicaments et des substances chimiques , Lésion pulmonaire aigüe/induit chimiquement , Administration par voie nasale , Animaux , Chimiokine CCL2/biosynthèse , Chimiokine CCL2/génétique , Chimiokine CXCL2/biosynthèse , Chimiokine CXCL2/génétique , Chimiokine CXCL2/physiologie , Chimiotaxie des leucocytes/effets des médicaments et des substances chimiques , Acide clodronique/administration et posologie , Acide clodronique/pharmacologie , Acide docosahexaénoïque/pharmacologie , Acide docosahexaénoïque/physiologie , Inflammation , Injections péritoneales , Lipopolysaccharides/administration et posologie , Lipopolysaccharides/toxicité , Liposomes , Macrophages/physiologie , Matrix metalloproteinase 9/physiologie , Souris , Souris de lignée C57BL , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Récepteurs CCR2/antagonistes et inhibiteurs , Récepteurs à l'interleukine-8B/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Migration transendothéliale et transépithéliale/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/physiologie
7.
J Cell Mol Med ; 24(17): 9646-9657, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32757380

RÉSUMÉ

Acute respiratory distress syndrome (ARDS) is a fatal disease characterized by excessive infiltration of inflammatory cells. MCTR1 is an endogenously pro-resolution lipid mediator. We tested the hypothesis that MCTR1 accelerates inflammation resolution through resident M2 alveolar macrophage polarization. The mice received MCTR1 via intraperitoneal administration 3 days after LPS stimulation, and then, the bronchoalveolar lavage (BAL) fluid was collected 24 hours later to measure the neutrophil numbers. Flow cytometry was used to sort the resident and recruited macrophages. Post-treatment with MCTR1 offered dramatic benefits in the resolution phase of LPS-induced lung injury, including decreased neutrophil numbers, reduced BAL fluid protein and albumin concentrations and reduced histological injury. In addition, the expression of the M2 markers Arg1, FIZZ1, Remlα, CD206 and Dectin-1 was increased on resident macrophages in the LPS + MCTR1 group. Resident macrophage depletion abrogated the therapeutic effects of MCTR1, and reinjection of the sorted resident macrophages into the lung decreased neutrophil numbers. Finally, treatment with MCTR1 increased STAT6 phosphorylation. The STAT6 inhibitor AS1517499 abolished the beneficial effects of MCTR1. In conclusion, MCTR1 promotes resident M2 alveolar macrophage polarization via the STAT6 pathway to accelerate resolution of LPS-induced lung injury.


Sujet(s)
Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/métabolisme , Polarité de la cellule/physiologie , Lipopolysaccharides/pharmacologie , Macrophages alvéolaires/métabolisme , Protéines oncogènes/métabolisme , Facteur de transcription STAT-6/métabolisme , Animaux , Liquide de lavage bronchoalvéolaire , Inflammation/métabolisme , Poumon/métabolisme , Activation des macrophages/physiologie , Souris , Souris de lignée C57BL , Granulocytes neutrophiles/métabolisme , /métabolisme , Transduction du signal/physiologie
8.
Br J Anaesth ; 125(1): e130-e139, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32223967

RÉSUMÉ

BACKGROUND: High ventilatory frequencies increase static lung strain and possibly lung stress by shortening expiratory time, increasing intrathoracic pressure, and causing dynamic hyperinflation. We hypothesised that high intraoperative ventilatory frequencies were associated with postoperative respiratory complications. METHODS: In this retrospective hospital registry study, we analysed data from adult non-cardiothoracic surgical cases performed under general anaesthesia with mechanical ventilation at a single centre between 2005 and 2017. We assessed the association between intraoperative ventilatory frequency (categorised into four groups) and postoperative respiratory complications, defined as composite of invasive mechanical ventilation within 7 days after surgery or peripheral oxygen desaturation after extubation, using multivariable logistic regression. In a subgroup, we adjusted analyses for arterial blood gas parameters. RESULTS: A total of 102 632 cases were analysed. Intraoperative ventilatory frequencies ranged from a median (inter-quartile range [IQR]) of 8 (8-9) breaths min-1 (Group 1) to 15 (14-18) breaths min-1 (Group 4). High ventilatory frequencies were associated with higher odds of postoperative respiratory complications (adjusted odds ratio=1.26; 95% confidence interval, 1.14-1.38; P<0.001), which was confirmed in a subgroup after adjusting for arterial partial pressure of carbon dioxide and the ratio of arterial oxygen partial pressure to fractional inspired oxygen. We identified considerable variability in the use of high ventilatory frequencies attributable to individual provider preference (ranging from 22% to 88%) and temporal change; however, the association with postoperative respiratory complications remained unaffected. CONCLUSIONS: High intraoperative ventilatory frequency was associated with increased risk of postoperative respiratory complications, and increased postoperative healthcare utilisation.


Sujet(s)
Soins peropératoires/effets indésirables , Soins peropératoires/méthodes , Complications postopératoires/physiopathologie , Troubles respiratoires/physiopathologie , Ventilation artificielle/effets indésirables , Ventilation artificielle/méthodes , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Hôpitaux , Humains , Mâle , Massachusetts/épidémiologie , Adulte d'âge moyen , Complications postopératoires/étiologie , Enregistrements , Troubles respiratoires/étiologie , Études rétrospectives , Volume courant , Temps , Jeune adulte
9.
Biochem Pharmacol ; 177: 113955, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32251673

RÉSUMÉ

Vitamin D regulates cell proliferation, inhibits cytokines release at sites of inflammation and reduces inflammatory responses. In this study, the aim was to investigate whether exogenous vitamin D attenuates LPS-induced lung injury via modulating epithelial cell proliferation, migration, apoptosis and epithelial mesenchymal transition (EMT). Murine and in vitro primary type II alveolar epithelial cell work were included in this study. In vivo, mice were mildly vitamin D deficient, 0.1, 1.5, 10 mg/kg 1,25(OH)2-vitamin D3 or 25(OH)-vitamin D3 was administrated by means of an intra-gastric injection for 14 days pre-intra-tracheal (IT) LPS, which remarkedly promoted alveolar epithelial type II cells proliferation, inhibited ATII cells apoptosis and inhibited EMT, with the outcome of attenuated LPS-induced lung injury. In vitro, vitamin D stimulated epithelial cell scratch wound repair, reduced primary ATII cells apoptosis as well. Vitamin D promoted primary human ATII cells proliferation through the PI3K/AKT signaling pathway and activation of vitamin D receptor (VDR). Moreover, vitamin D inhibited EMT in response to TGF-ß, which was vitamin D receptor dependent. In conclusion, vitamin D attenuates lung injury via stimulating ATII cells proliferation and migration, reducing epithelial cell apoptosis and inhibits TGF-ß induced EMT. Together, these results suggest that vitamin D has therapeutic potential for the resolution of ARDS.


Sujet(s)
Pneumocytes/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Lésion pulmonaire/prévention et contrôle , Facteur de croissance transformant bêta/pharmacologie , Vitamine D/pharmacologie , Pneumocytes/métabolisme , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Femelle , Humains , Lipopolysaccharides , Lésion pulmonaire/induit chimiquement , Lésion pulmonaire/métabolisme , Mâle , Souris de lignée C57BL , Adulte d'âge moyen , Phosphatidylinositol 3-kinases/métabolisme , Récepteur calcitriol/métabolisme , /métabolisme , /physiopathologie , /prévention et contrôle , Transduction du signal/effets des médicaments et des substances chimiques , Vitamine D/sang
10.
Acta Anaesthesiol Scand ; 64(8): 1187-1193, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32320051

RÉSUMÉ

BACKGROUND: Despite an increase in the rates of epidural labor analgesia, continuation of epidural labor analgesia in the second stage of labor (CEADSSOL) was interrupted by care providers due to fears of increased risk of operative delivery and adverse neonatal outcomes. Therefore, we evaluated the effect of CEADSSOL and the newer American College of Obstetricians and Gynecologists (ACOG) definition of arrest of labor on the length of secondary stage of labor, newborn outcomes, and mode of delivery. METHODS: This is a retrospective cohort study. Data collection began during March 2014 and ended in May 2015, 1 year after implementation of both interventions. The primary outcome was the length of secondary stage of labor, mode of delivery and neonatal outcome (Apgar < 7, at 5 minutes). The implementation of continuing epidural analgesia during the second stage of labor was performed with 0.08%-0.15% ropivacaine and 0.1-0.2 µg/mL sufentanil. RESULTS: There were a total 10 414 deliveries during the study period. The length of the second stage of labor has no significant differences among groups. The cesarean delivery rate decreased 4.1% (36% vs 40.1%, P = .0038). Moreover, no significant difference was found in neonatal Apgar scores less than 7 at 5 minutes between two phases. Maternal outcomes remained unchanged. Post-intervention neonatal parameters including NICU admissions (P < .001), incidences of antibiotics usage (P < .0001), intubation (P = .0003), and 7 days mortality (P = .0020) were remarkably reduced compared to pre-interventions. CONCLUSION: The important finding of this study was the improvement in neonatal outcomes by implementing two simultaneous interventions without a cost of increased operative delivery.


Sujet(s)
Analgésie péridurale/méthodes , Analgésie obstétricale/méthodes , Second stade du travail/effets des médicaments et des substances chimiques , Issue de la grossesse , Adulte , Score d'Apgar , Études de cohortes , Femelle , Humains , Nouveau-né , Mâle , Grossesse , Études rétrospectives
11.
Int Immunopharmacol ; 76: 105877, 2019 Nov.
Article de Anglais | MEDLINE | ID: mdl-31522017

RÉSUMÉ

Acute lung injury (ALI) and/or acute respiratory distress syndrome (ARDS) are life-threatening critical syndromes characterized by the infiltration of a large number of inflammatory cells that lead to an excessive inflammatory response. Resolvin D1 (RvD1), an endogenous lipid mediator, is believed to have anti-inflammatory and proresolving effects. In the present study, we examined the impact of RvD1 on the pulmonary inflammatory response, neutrophil influx, and lung damage in a murine model of lipopolysaccharide (LPS)-induced ALI. Treatment with RvD1 protected mice against LPS-induced ALI, and compared to untreated mice, RvD1-treated mice exhibited significantly ameliorated lung pathological changes, decreased tumor necrosis factor-α (TNF-α) concentrations and attenuated neutrophil infiltration. In addition, treatment with RvD1 attenuated LPS-induced neutrophil infiltration via the downregulation of CXCL2 expression on resident alveolar macrophages. Finally, BOC-2, which inhibits the RvD1 receptor lipoxin A4 receptor/formyl peptide receptor 2 (ALX/FPR2), reversed the protective effects of RvD1. These data demonstrate that RvD1 ameliorates LPS-induced ALI via the suppression of neutrophil infiltration by an ALX/FPR2-dependent reduction in CXCL2 expression on resident alveolar macrophages.


Sujet(s)
Lésion pulmonaire aigüe/traitement médicamenteux , Chimiokine CXCL2/antagonistes et inhibiteurs , Acide docosahexaénoïque/pharmacologie , Acide docosahexaénoïque/usage thérapeutique , Macrophages alvéolaires/effets des médicaments et des substances chimiques , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/immunologie , Lésion pulmonaire aigüe/anatomopathologie , Animaux , Chimiokine CXCL2/génétique , Chimiokine CXCL2/immunologie , Lipopolysaccharides , Poumon/effets des médicaments et des substances chimiques , Poumon/immunologie , Poumon/anatomopathologie , Macrophages alvéolaires/immunologie , Souris de lignée C57BL , Infiltration par les neutrophiles/effets des médicaments et des substances chimiques
12.
Respir Res ; 20(1): 192, 2019 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-31438948

RÉSUMÉ

BACKGROUND: Acute respiratory distress syndrome (ARDS) is characterized by alveolar epithelial disruption. Lipoxins (LXs), as so-called "braking signals" of inflammation, are the first mediators identified to have dual anti-inflammatory and inflammatory pro-resolving properties. METHODS: In vivo, lipoxinA4 was administrated intraperitoneally with 1 µg/per mouse after intra-tracheal LPS administration (10 mg/kg). Apoptosis, proliferation and epithelial-mesenchymal transition of AT II cells were measured by immunofluorescence. In vitro, primary human alveolar type II cells were used to model the effects of lipoxin A4 upon proliferation, apoptosis and epithelial-mesenchymal transition. RESULTS: In vivo, lipoxin A4 markedly promoted alveolar epithelial type II cells (AT II cells) proliferation, inhibited AT II cells apoptosis, reduced cleaved caspase-3 expression and epithelial-mesenchymal transition, with the outcome of attenuated LPS-induced lung injury. In vitro, lipoxin A4 increased primary human alveolar epithelial type II cells (AT II cells) proliferation and reduced LPS induced AT II cells apoptosis. LipoxinA4 also inhibited epithelial mesenchymal transition in response to TGF-ß1, which was lipoxin receptor dependent. In addition, Smad3 inhibitor (Sis3) and PI3K inhibitor (LY294002) treatment abolished the inhibitory effects of lipoxinA4 on the epithelial mesenchymal transition of primary human AT II cells. Lipoxin A4 significantly downregulated the expressions of p-AKT and p-Smad stimulated by TGF-ß1 in primary human AT II cells. CONCLUSION: LipoxinA4 attenuates lung injury via stimulating epithelial cell proliferation, reducing epithelial cell apoptosis and inhibits epithelial-mesenchymal transition.


Sujet(s)
Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/traitement médicamenteux , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules épithéliales/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Lipoxines/usage thérapeutique , /traitement médicamenteux , Lésion pulmonaire aigüe/métabolisme , Animaux , Cellules cultivées , Humains , Injections péritoneales , Lipopolysaccharides , Lipoxines/effets indésirables , Souris , Souris de lignée C57BL , Inhibiteurs de protéines kinases/usage thérapeutique , Alvéoles pulmonaires/cytologie , Alvéoles pulmonaires/effets des médicaments et des substances chimiques , /induit chimiquement
13.
Biochem Biophys Res Commun ; 517(1): 89-95, 2019 09 10.
Article de Anglais | MEDLINE | ID: mdl-31301770

RÉSUMÉ

Alveolar epithelial type II cells (ATII cells) are the main target cells being damaged and releasing the inflammatory mediators during acute respiratory distress syndrome (ARDS). Extensive apoptosis of epithelial cells leads to the breakdown of the alveolar-epithelial barrier in ARDS. Cyclooxygenase-2 (COX-2) plays an important role in pulmonary inflammatory response. Dexmedetomidine (DEX), a potent selective α2 adrenergic receptor (α2-AR) agonist, presents sedative, anxiolytic, and analgesic effects for anesthetic procedures. DEX has anti-apoptotic and anti-inflammatory properties. Our study demonstrated that DEX exerted anti-apoptotic effect on primary human epithelial cells with the inhibition of caspase activation, which was partly via the α2AR/PI3K/AKT pathway. Moreover, DEX significantly reduced the expression of COX-2 as well as prostaglandinE2 (PGE2) and tumor necrosis factor-α (TNF-α) production induced by lipopolysaccharide (LPS). Our next step is to determine whether DEX can regulate apoptosis in animal models. These results suggest DEX may be a promising therapy for preventing and treating ARDS as well as chronic diseases by directly targeting epithelial cell actions.


Sujet(s)
Agonistes des récepteurs alpha-2 adrénergiques/pharmacologie , Pneumocytes/effets des médicaments et des substances chimiques , Anti-inflammatoires/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Cyclooxygenase 2/immunologie , Dexmédétomidine/pharmacologie , Pneumocytes/immunologie , Cellules cultivées , Cyclooxygenase 2/analyse , Humains , Lipopolysaccharides/immunologie
14.
Arch Gynecol Obstet ; 298(1): 89-96, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-29777348

RÉSUMÉ

PURPOSE: To evaluate the impact of labor epidural analgesia on maternal-fetal safety outcomes in a signal Chinese academic medical center. METHODS: A single-intervention impact study was conducted at The Second Affiliated Hospital, Wenzhou Medical University. The study period was divided into three phases: (1) baseline phase: from January 1 and June 30, 2009 when no analgesic method was routinely employed during labor; (2) phase-in period: the epidural analgesia was implemented 8 a.m.-5 p.m. during weekdays; and (3) the post-No Pain Labor N'Delivery phase when the labor epidural was applied 24 h a day, 7 days a week, from June 1, 2010 and June 30, 2011. The maternal-fetal safety outcomes of delivery were compared between the different periods. RESULTS: There were 15,415 deliveries with 42.3% of nulliparous parturients in the 31-month study period. As the primary outcomes, the labor epidural analgesia rate increased from 0 to 57%, the vaginal delivery rate increased, and cesarean delivery rate decreased by 3.5% after full implementation. As the secondary outcomes, the rate of episiotomy and severe perineal injury after the implementation periods were significant decreased. The rate of postpartum oxytocin administration was decreased by 17.8%. No significant difference between the baseline and implementation periods was found in the rate of postpartum hemorrhage, Apgar scores less than 7 at both 1 and 5 min, 7-day mortality, and the overall neonatal intensive care unit admission rate. CONCLUSION: Implementation of labor epidural analgesia increased the vaginal delivery rate and use of labor epidural analgesia is safe to parturients and fetus.


Sujet(s)
Analgésie péridurale/effets indésirables , Analgésie obstétricale/effets indésirables , Travail obstétrical/effets des médicaments et des substances chimiques , Adulte , Analgésie péridurale/méthodes , Analgésie obstétricale/méthodes , Études de cohortes , Femelle , Humains , Nouveau-né , Grossesse , Études rétrospectives
15.
Lab Invest ; 98(1): 130-140, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-29083412

RÉSUMÉ

Acute and chronic inflammatory lung diseases are often associated with epithelial cell injury/loss and fibroproliferative responses. ResolvinD1 (RvD1) is biosynthesized during the resolution phase of inflammatory response and exerts potent anti-inflammatory and promotes resolution of inflammatory lung diseases. The aim of this study was to investigate whether RvD1 exerts protective effects on alveolar epithelial cell function/differentiation and protects against fibroproliferative stimuli. Primary human alveolar type II cells were used to model the effects of RvD1 in vitro upon wound repair, proliferation, apoptosis, transdifferentiation, and epithelial-mesenchymal transition (EMT). Effects of RvD1 upon primary human lung fibroblast proliferation, collagen production, and myofibroblast differentiation were also examined. RvD1 promoted alveolar type II (ATII) cell wound repair and proliferation. RvD1 protected ATII cells against sFas-ligand/TNF-α-induced apoptosis and inhibition on cell proliferation and viability. RvD1 promoted ATII cells transdifferentiation. Moreover, we demonstrate that RvD1 inhibited EMT in response to TGF-ß. Furthermore RvD1 inhibited human lung fibroblast proliferation, collagen production, and myofibroblast differentiation induced by both TGF-ß and bronchoalveolar lavage fluid from acute respiratory distress syndrome (ARDS) patients. The effects of RvD1 were PI3-kinase dependent and mediated via the resolvin receptor. RvD1 seems to promote alveolar epithelial repair by stimulating ATII cells wound repair, proliferation, reducing apoptosis, and inhibiting TGF-ß-induced EMT. While RvD1 reduced fibroproliferation, collagen production, and myofibroblast differentiation. Together, these results suggest a potential new therapeutic strategy for preventing and treating chronic diseases (such as idiopathic pulmonary fibrosis) as well as the fibroproliferative phase of ARDS by targeting RvD1 actions that emphasizes natural resolution signaling pathways.


Sujet(s)
Pneumocytes/métabolisme , Acide docosahexaénoïque/métabolisme , Transition épithélio-mésenchymateuse , Récepteurs couplés aux protéines G/agonistes , Transduction du signal , Facteur de croissance transformant bêta/antagonistes et inhibiteurs , Cicatrisation de plaie , Pneumocytes/cytologie , Pneumocytes/immunologie , Apoptose , Marqueurs biologiques/métabolisme , Liquide de lavage bronchoalvéolaire/composition chimique , Liquide de lavage bronchoalvéolaire/immunologie , Différenciation cellulaire , Prolifération cellulaire , Survie cellulaire , Transdifférenciation cellulaire , Cellules cultivées , Collagène/antagonistes et inhibiteurs , Collagène/génétique , Collagène/métabolisme , Régulation de l'expression des gènes , Humains , Myofibroblastes/cytologie , Myofibroblastes/immunologie , Myofibroblastes/métabolisme , Concentration osmolaire , Récepteurs couplés aux protéines G/métabolisme , Protéines recombinantes/métabolisme , /immunologie , /métabolisme , Facteur de croissance transformant bêta/génétique , Facteur de croissance transformant bêta/métabolisme
16.
ERJ Open Res ; 2(3)2016 Jul.
Article de Anglais | MEDLINE | ID: mdl-27957484

RÉSUMÉ

Therapy that promotes epithelial repair whilst protecting against fibroproliferation is critical for restoring lung function in acute and chronic respiratory diseases. Primary human alveolar type II cells were used to model the effects of lipoxin A4in vitro upon wound repair, proliferation, apoptosis and transdifferention. Effects of lipoxin A4 upon primary human lung fibroblast proliferation, collagen production, and myofibroblast differentiation were also assessed. Lipoxin A4 promoted type II cell wound repair and proliferation, blocked the negative effects of soluble Fas ligand/tumour necrosis factor α upon cell proliferation, viability and apoptosis, and augmented the epithelial cell proliferative response to bronchoaveolar lavage fluid (BALF) from acute respiratory distress syndrome (ARDS). In contrast, Lipoxin A4 reduced fibroblast proliferation, collagen production and myofibroblast differentiation induced by transforming growth factor ß and BALF from ARDS. The effects of Lipoxin A4 were phosphatidylinositol 3'-kinase dependent and mediated via the lipoxin A4 receptor. Lipoxin A4 appears to promote alveolar epithelial repair by stimulating epitheial cell wound repair, proliferation, reducing apoptosis and promoting trans-differentiation of alveolar type II cells into type I cells. Lipoxin A4 reduces fibroblast proliferation, collagen production and myofibroblast differentiation. These data suggest that targeting lipoxin actions may be a therapeutic strategy for treating the resolution phase of ARDS.

17.
Thorax ; 70(7): 617-24, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-25903964

RÉSUMÉ

RATIONALE: Vitamin D deficiency has been implicated as a pathogenic factor in sepsis and intensive therapy unit mortality but has not been assessed as a risk factor for acute respiratory distress syndrome (ARDS). Causality of these associations has never been demonstrated. OBJECTIVES: To determine if ARDS is associated with vitamin D deficiency in a clinical setting and to determine if vitamin D deficiency in experimental models of ARDS influences its severity. METHODS: Human, murine and in vitro primary alveolar epithelial cell work were included in this study. FINDINGS: Vitamin D deficiency (plasma 25(OH)D levels <50 nmol/L) was ubiquitous in patients with ARDS and present in the vast majority of patients at risk of developing ARDS following oesophagectomy. In a murine model of intratracheal lipopolysaccharide challenge, dietary-induced vitamin D deficiency resulted in exaggerated alveolar inflammation, epithelial damage and hypoxia. In vitro, vitamin D has trophic effects on primary human alveolar epithelial cells affecting >600 genes. In a clinical setting, pharmacological repletion of vitamin D prior to oesophagectomy reduced the observed changes of in vivo measurements of alveolar capillary damage seen in deficient patients. CONCLUSIONS: Vitamin D deficiency is common in people who develop ARDS. This deficiency of vitamin D appears to contribute to the development of the condition, and approaches to correct vitamin D deficiency in patients at risk of ARDS should be developed. TRIAL REGISTRATION: UKCRN ID 11994.


Sujet(s)
/étiologie , Carence en vitamine D/complications , Indice APACHE , Sujet âgé , Animaux , Calcifédiol/sang , Calcifédiol/pharmacologie , Calcitriol/sang , Cellules cultivées , Modèles animaux de maladie humaine , Cellules épithéliales/effets des médicaments et des substances chimiques , Oesophagectomie/effets indésirables , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Unités de soins intensifs , Mâle , Souris de lignée C57BL , Adulte d'âge moyen , Alvéoles pulmonaires/cytologie , Alvéoles pulmonaires/effets des médicaments et des substances chimiques , /sang , /prévention et contrôle , Facteurs de risque , Analyse de survie , Vitamine D/usage thérapeutique , Carence en vitamine D/sang , Carence en vitamine D/traitement médicamenteux
18.
Mediators Inflamm ; 2013: 964012, 2013.
Article de Anglais | MEDLINE | ID: mdl-24174713

RÉSUMÉ

Fibroblasts, far from being merely bystander cells, are known to play a specific role in inflammation resolution after an acute injury. As the endogenous "braking signal," resolvins possess potent anti-inflammatory and pro-resolution actions. We demonstrated that the expression of COX-2 protein was significantly peaked initially at 6 hours but then also at 48 hours after LPS stimulation in lung fibroblasts. PGE2 levels also peaked at 6 hours, and PGD2 levels were increased and peaked at 48 hours. However, no significant change in the protein expression of COX-1 was observed after treatment with LPS in lung fibroblasts. Exogenous resolvin D1 inhibited the first peak of COX-2 expression as well as the production of PGE2 induced by LPS. In contrast, exogenous resolvin D1 increased the second peak of COX-2 expression as well as the production of PGD2 induced by LPS. In addition, resolvin D1 inhibited COX-2 expression at 6 hours, which was partly through PI3K/AKT and ERK2 signalling pathways.


Sujet(s)
Cyclooxygenase 2/métabolisme , Acide docosahexaénoïque/pharmacologie , Fibroblastes/métabolisme , Poumon/anatomopathologie , Système de signalisation des MAP kinases , Animaux , Lignée cellulaire , Chimiokine CCL2/métabolisme , Dinoprostone/métabolisme , Régulation de l'expression des gènes , Humains , Interleukine-8/métabolisme , Lipopolysaccharides , Mitogen-Activated Protein Kinase 1/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Prostaglandine D2/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Rats , Transduction du signal
19.
Am J Respir Cell Mol Biol ; 48(5): 610-8, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23470626

RÉSUMÉ

Edema fluid resorption is critical for gas exchange, and both alveolar epithelial sodium channel (ENaC) and Na,K-ATPase are accredited with key roles in the resolution of pulmonary edema. Alveolar fluid clearance (AFC) was measured in in situ ventilated lungs by instilling isosmolar 5% BSA solution with Evans Blue-labeled albumin tracer (5 ml/kg) and measuring the change in Evans Blue-labeled albumin concentration over time. Treatment with lipoxin A4 and lipoxin receptor agonist (5(S), 6(R)-7-trihydroxymethyl 17 heptanoate) significantly stimulated AFC in oleic acid (OA)-induced lung injury, with the outcome of decreased pulmonary edema. Lipoxin A4 and 5(S), 6(R)-7-trihydroxymethyl 17 heptanoate not only up-regulated the ENaC α and ENaC γ subunits protein expression, but also increased Na,K-ATPase α1 subunit protein expression and Na,K-ATPase activity in lung tissues. There was no significant difference of intracellular cAMP level between the lipoxin A4 treatment and OA group. However, the intracellular cGMP level was significantly decreased after lipoxin A4 treatment. The beneficial effects of lipoxin A4 were abrogated by butoxycarbonyl-Phe-Leu-Phe-Leu-Ph (lipoxin A4 receptor antagonist) in OA-induced lung injury. In primary rat alveolar type II epithelial cells stimulated with LPS, lipoxin A4 increased ENaC α and ENaC γ subunits protein expression and Na,K-ATPase activity. Lipoxin A4 stimulated AFC through activation of alveolar epithelial ENaC and Na,K-ATPase.


Sujet(s)
Agonistes de canaux sodiques épithéliaux/administration et posologie , Canaux sodium épithéliaux/métabolisme , Lipoxines/administration et posologie , Sodium-Potassium-Exchanging ATPase/métabolisme , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/immunologie , Lésion pulmonaire aigüe/métabolisme , Pneumocytes/effets des médicaments et des substances chimiques , Pneumocytes/immunologie , Animaux , Cellules cultivées , AMP cyclique/métabolisme , GMP cyclique/métabolisme , Agonistes de canaux sodiques épithéliaux/pharmacologie , Bloqueurs de canaux sodiques épithéliaux/pharmacologie , Canaux sodium épithéliaux/génétique , Expression des gènes/effets des médicaments et des substances chimiques , Acides heptanoïques/pharmacologie , Lipopolysaccharides/pharmacologie , Lipoxines/pharmacologie , Mâle , Clairance mucociliaire , Oligopeptides/pharmacologie , Myeloperoxidase/métabolisme , Alvéoles pulmonaires/effets des médicaments et des substances chimiques , Alvéoles pulmonaires/métabolisme , Alvéoles pulmonaires/physiopathologie , Rats , Rat Sprague-Dawley , Facteur de nécrose tumorale alpha/métabolisme
20.
Mediators Inflamm ; 2011: 745340, 2011.
Article de Anglais | MEDLINE | ID: mdl-21765620

RÉSUMÉ

Fibroblasts are important to host defence and immunity, can also as initiators of inflammation as well. As the endogenous "braking signal", Lipoxins can regulate anti-inflammation and the resolution of inflammation. We investigated the effect of lipoxinA(4) on the expression of cyclooxygenase-2 in lipopolysaccharide-stimulated lung fibroblasts. We demonstrated that the expression of cyclooxygenase-2 protein was significantly increased and peaked initially at 6 hours, with a second increase, with maximal levels occurring 24 hours after lipopolysaccharide challenge. ProstaglandinE(2) levels also peaked at 6 hours, and prostaglandinD(2) levels were increased at both 6 and 24 hours. Exogenous lipoxinA(4) inhibited the first peak of cyclooxygenase-2 expression as well as the production of prostaglandinE(2) induced by lipopolysaccharide in a dose-dependent manner. In contrast, exogenous lipoxinA(4) increased the second peak of cyclooxygenase-2 expression as well as the production of prostaglandinD(2) induced by lipopolysaccharide in a dose-dependent manner. LipoxinA(4) receptor mRNA expression was markedly stimulated by lipopolysaccharide but inhibited by lipoxinA(4). We present evidence for a novel biphasic role of lipoxinA(4) on the expression of cyclooxygenase-2 in lipopolysaccharide-stimulated lung fibroblasts, whereby LXA(4) has an anti-inflammatory and proresolving activity in lung fibroblasts following LPS stimulation.


Sujet(s)
Cyclooxygenase 2/immunologie , Fibroblastes/immunologie , Lipopolysaccharides/pharmacologie , Lipoxines/immunologie , Poumon/cytologie , Animaux , Anti-inflammatoires/immunologie , Anti-inflammatoires/pharmacologie , Cellules cultivées , Cyclooxygenase 2/génétique , Dinoprostone/métabolisme , Relation dose-effet des médicaments , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes codant pour des enzymes/physiologie , Lipoxines/métabolisme , Lipoxines/pharmacologie , Prostaglandine D2/métabolisme , Rats , Rat Sprague-Dawley , Récepteurs de la lipoxine/immunologie , Récepteurs de la lipoxine/métabolisme , Régulation positive/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE