Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Int Immunopharmacol ; 102: 108348, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34920958

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Docosahexaenoic Acids/therapeutic use , Respiratory Distress Syndrome/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Cyclooxygenase 2/metabolism , Docosahexaenoic Acids/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/metabolism , Male , Prostaglandin D2/metabolism , Rats, Sprague-Dawley , Receptors, Prostaglandin/metabolism , Respiratory Distress Syndrome/metabolism
2.
Cell Death Discov ; 7(1): 339, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34750369

ABSTRACT

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.

3.
J Pharmacol Exp Ther ; 379(2): 156-165, 2021 11.
Article in English | MEDLINE | ID: mdl-34465632

ABSTRACT

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.


Subject(s)
Acute Lung Injury/metabolism , Docosahexaenoic Acids/pharmacology , Epithelial Sodium Channel Agonists/pharmacology , Epithelial Sodium Channels/metabolism , Pulmonary Alveoli/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Animals , Docosahexaenoic Acids/analogs & derivatives , Docosahexaenoic Acids/therapeutic use , Lipopolysaccharides/toxicity , Male , Pulmonary Alveoli/drug effects , Rats , Rats, Sprague-Dawley
4.
J Cell Mol Med ; 24(23): 14001-14012, 2020 12.
Article in English | MEDLINE | ID: mdl-33098250

ABSTRACT

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.


Subject(s)
Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Anaplastic Lymphoma Kinase/metabolism , Docosahexaenoic Acids/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Angiotensin II/metabolism , Animals , Apoptosis/drug effects , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators , Lipopolysaccharides/adverse effects , Mice , Rats
5.
J Cell Mol Med ; 24(17): 9646-9657, 2020 09.
Article in English | MEDLINE | ID: mdl-32757380

ABSTRACT

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.


Subject(s)
Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Cell Polarity/physiology , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/metabolism , Oncogene Proteins/metabolism , STAT6 Transcription Factor/metabolism , Animals , Bronchoalveolar Lavage Fluid , Inflammation/metabolism , Lung/metabolism , Macrophage Activation/physiology , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Respiratory Distress Syndrome/metabolism , Signal Transduction/physiology
6.
J Cell Mol Med ; 24(18): 10604-10614, 2020 09.
Article in English | MEDLINE | ID: mdl-32735065

ABSTRACT

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.


Subject(s)
Acute Lung Injury/pathology , Docosahexaenoic Acids/therapeutic use , Macrophages/drug effects , Acute Lung Injury/chemically induced , Administration, Intranasal , Animals , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CXCL2/biosynthesis , Chemokine CXCL2/genetics , Chemokine CXCL2/physiology , Chemotaxis, Leukocyte/drug effects , Clodronic Acid/administration & dosage , Clodronic Acid/pharmacology , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/physiology , Inflammation , Injections, Intraperitoneal , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/toxicity , Liposomes , Macrophages/physiology , Matrix Metalloproteinase 9/physiology , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Receptors, CCR2/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Signal Transduction/drug effects , Transendothelial and Transepithelial Migration/drug effects , Tumor Necrosis Factor-alpha/physiology
7.
Int Immunopharmacol ; 76: 105877, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31522017

ABSTRACT

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.


Subject(s)
Acute Lung Injury/drug therapy , Chemokine CXCL2/antagonists & inhibitors , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Macrophages, Alveolar/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Lipopolysaccharides , Lung/drug effects , Lung/immunology , Lung/pathology , Macrophages, Alveolar/immunology , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects
8.
Respir Res ; 20(1): 192, 2019 Aug 22.
Article in English | MEDLINE | ID: mdl-31438948

ABSTRACT

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.


Subject(s)
Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Apoptosis/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Lipoxins/therapeutic use , Respiratory Distress Syndrome/drug therapy , Acute Lung Injury/metabolism , Animals , Cells, Cultured , Humans , Injections, Intraperitoneal , Lipopolysaccharides , Lipoxins/adverse effects , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/therapeutic use , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Respiratory Distress Syndrome/chemically induced
9.
Arch Gynecol Obstet ; 298(1): 89-96, 2018 07.
Article in English | MEDLINE | ID: mdl-29777348

ABSTRACT

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.


Subject(s)
Analgesia, Epidural/adverse effects , Analgesia, Obstetrical/adverse effects , Labor, Obstetric/drug effects , Adult , Analgesia, Epidural/methods , Analgesia, Obstetrical/methods , Cohort Studies , Female , Humans , Infant, Newborn , Pregnancy , Retrospective Studies
10.
Am J Respir Cell Mol Biol ; 48(5): 610-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23470626

ABSTRACT

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.


Subject(s)
Epithelial Sodium Channel Agonists/administration & dosage , Epithelial Sodium Channels/metabolism , Lipoxins/administration & dosage , Sodium-Potassium-Exchanging ATPase/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/immunology , Animals , Cells, Cultured , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Epithelial Sodium Channel Agonists/pharmacology , Epithelial Sodium Channel Blockers/pharmacology , Epithelial Sodium Channels/genetics , Gene Expression/drug effects , Heptanoic Acids/pharmacology , Lipopolysaccharides/pharmacology , Lipoxins/pharmacology , Male , Mucociliary Clearance , Oligopeptides/pharmacology , Peroxidase/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/physiopathology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL