Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Int Immunopharmacol ; 102: 108348, 2022 Jan.
Article En | MEDLINE | ID: mdl-34920958

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.


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.
Respir Res ; 22(1): 193, 2021 Jul 03.
Article En | MEDLINE | ID: mdl-34217286

BACKGROUND: Endothelial glycocalyx loss is integral to increased pulmonary vascular permeability in sepsis-related acute lung injury. Protectin conjugates in tissue regeneration 1 (PCTR1) is a novel macrophage-derived lipid mediator exhibiting potential anti-inflammatory and pro-resolving benefits. METHODS: PCTR1 was administrated intraperitoneally with 100 ng/mouse after lipopolysaccharide (LPS) challenged. Survival rate and lung function were used to evaluate the protective effects of PCTR1. Lung inflammation response was observed by morphology and inflammatory cytokines level. Endothelial glycocalyx and its related key enzymes were measured by immunofluorescence, ELISA, and Western blot. Afterward, related-pathways inhibitors were used to identify the mechanism of endothelial glycocalyx response to PCTR1 in mice and human umbilical vein endothelial cells (HUVECs) after LPS administration. RESULTS: In vivo, we show that PCTR1 protects mice against lipopolysaccharide (LPS)-induced sepsis, as shown by enhanced the survival and pulmonary function, decreased the inflammatory response in lungs and peripheral levels of inflammatory cytokines such as tumor necrosis factor-α, interleukin-6, and interleukin-1ß. Moreover, PCTR1 restored lung vascular glycocalyx and reduced serum heparin sulphate (HS), syndecan-1 (SDC-1), and hyaluronic acid (HA) levels. Furthermore, we found that PCTR1 downregulated heparanase (HPA) expression to inhibit glycocalyx degradation and upregulated exostosin-1 (EXT-1) protein expression to promote glycocalyx reconstitution. Besides, we observed that BAY11-7082 blocked glycocalyx loss induced by LPS in vivo and in vitro, and BOC-2 (ALX antagonist) or EX527 (SIRT1 inhibitor) abolished the restoration of HS in response to PCTR1. CONCLUSION: PCTR1 protects endothelial glycocalyx via ALX receptor by regulating SIRT1/NF-κB pathway, suggesting PCTR1 may be a significant therapeutic target for sepsis-related acute lung injury.


Adaptor Proteins, Signal Transducing/metabolism , Anti-Inflammatory Agents/pharmacology , Glycocalyx/metabolism , NF-kappa B/metabolism , Respiratory Mucosa/metabolism , Sirtuin 1/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Docosahexaenoic Acids/pharmacology , Glycocalyx/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipopolysaccharides/toxicity , Male , Mice , NF-kappa B/antagonists & inhibitors , Respiratory Mucosa/drug effects , Sirtuin 1/antagonists & inhibitors
3.
J Cell Physiol ; 235(10): 7283-7294, 2020 10.
Article En | MEDLINE | ID: mdl-32037554

Endothelial glycocalyx degradation, critical for increased pulmonary vascular permeability, is thought to facilitate the development of sepsis into the multiple organ failure. Maresin conjugates in tissue regeneration 1 (MCTR1), a macrophage-derived lipid mediator, which exhibits potentially beneficial effects via the regulation of bacterial phagocytosis, promotion of inflammation resolution, and regeneration of tissue. In this study, we show that MCTR1 (100 ng/mouse) enhances the survival of mice with lipopolysaccharide (LPS)-induced (15 mg/kg) sepsis. MCTR1 alleviates LPS (10 mg/kg)-induced lung dysfunction and lung tissue inflammatory response by decreasing inflammatory cytokines (tumor necrosis factor-α, interleukin-1ß [IL-1ß], and IL-6) expression in serum and reducing the serum levels of heparan sulfate (HS) and syndecan-1. In human umbilical vein endothelial cells (HUVECs) experiments, MCTR1 (100 nM) was added to the culture medium with LPS for 6 hr. MCTR1 treatment markedly inhibited HS degradation by downregulating heparanase (HPA) protein expression in vivo and in vitro. Further analyses indicated that MCTR1 upregulates sirtuin 1 (SIRT1) expression and decreases NF-κB p65 phosphorylation. In the presence of BOC-2 or EX527, the above effects of MCTR1 were abolished. These results suggest that MCTR1 protects against LPS-induced sepsis in mice by attenuating pulmonary endothelial glycocalyx injury via the ALX/SIRT1/NF-κB/HPA pathway.


Acute Lung Injury/drug therapy , Docosahexaenoic Acids/pharmacology , Acute Lung Injury/blood , Acute Lung Injury/chemically induced , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cytokines/blood , Endothelium/drug effects , Endothelium/pathology , Glucuronidase/metabolism , Glycocalyx/drug effects , Glycocalyx/pathology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation Mediators/blood , Lipopolysaccharides/toxicity , Lung/drug effects , Lung/physiology , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Sepsis/drug therapy , Sepsis/pathology , Sepsis/physiopathology , Signal Transduction , Sirtuin 1/metabolism , Transcription Factor RelA/metabolism
4.
Inflammation ; 41(4): 1297-1303, 2018 Aug.
Article En | MEDLINE | ID: mdl-29654431

Saikosaponin a (SSa), a triterpenoid saponin, has numerous pharmacological properties, including anti-inflammatory and antioxidant effects. The purpose of this study was to investigate whether and how SSa protected against cigarette smoke (CS)-induced lung inflammation in mice. The mice were exposed to CS and SSa was administered by an intraperitoneal (i.p.) injection 1 h before CS treatment for 5 consecutive days. The results showed that SSa significantly inhibited CS-induced inflammatory cell infiltration, NO, TNF-α, and IL-1ß production in BALF. SSa also inhibited CS-induced MPO and MDA contents in lung tissues. Furthermore, SSa significantly inhibited CS-induced NF-κB and upregulated the expression of Nrf2 and HO-1. In conclusion, these results support a therapeutic potential for SSa in CS-induced lung inflammation.


NF-E2-Related Factor 2/metabolism , Oleanolic Acid/analogs & derivatives , Oxidative Stress/drug effects , Pneumonia/drug therapy , Saponins/pharmacology , Smoke/adverse effects , Animals , Cigarette Smoking/adverse effects , Heme Oxygenase-1 , Membrane Proteins/agonists , Mice , NF-E2-Related Factor 2/drug effects , Oleanolic Acid/pharmacology , Pneumonia/etiology , Smoke/prevention & control
5.
Microb Pathog ; 110: 208-213, 2017 Sep.
Article En | MEDLINE | ID: mdl-28666844

Acute kidney injury (AKI) is a major clinical problem associated with high morbidity and mortality. Esculentoside A (EsA), a kind of saponin isolated from the root of the Chinese herb Phytolaca esculenta, has been reported to have anti-inflammatory effect. In this study, we aimed to investigate the protective effects of EsA on LPS-induced AKI in mice. The protective effects of EsA was evaluated by detecting kidney histological change, blood urea nitrogen (BUN) and creatinine levels, and inflammatory cytokines production. The results showed that EsA significantly attenuated LPS-induced kidney histological change, as well as BUN and creatinine levels. EsA also inhibited LPS-induced TNF-α, IL-1ß, and IL-6 production. LPS-induced NF-κB activation was significantly suppressed by treatment of EsA. In addition, EsA up-regulated the expression of PPAR-γ in a dose-dependent manner. In conclusion, EsA protected mice effectively from LPS-induced AKI by PPAR-γ, which subsequently inhibited LPS-induced inflammatory response.


Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Lipopolysaccharides/adverse effects , Oleanolic Acid/analogs & derivatives , PPAR gamma/metabolism , Saponins/antagonists & inhibitors , Acute Kidney Injury/pathology , Animals , Blood Urea Nitrogen , Creatinine/blood , Cytokines/metabolism , Disease Models, Animal , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Oleanolic Acid/administration & dosage , Oleanolic Acid/antagonists & inhibitors , Saponins/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
6.
Inflammation ; 39(6): 1876-1882, 2016 Dec.
Article En | MEDLINE | ID: mdl-27581277

Angelicin, a furocoumarin found in Psoralea corylifolia L. fruit, has been reported to have anti-inflammatory activity. The purpose of this study was to determine the protective effects of angelicin on allergic asthma induced by ovalbumin (OVA) in mice. Mice were sensitized to OVA (on days 0 and 14) and challenged with OVA three times (on days 21 to 23). Angelicin (2.5, 5, 10 mg/kg) was given intraperitoneally 1 h before OVA treatment after the initial OVA sensitization. The production of IL-4, IL-5, and IL-13 in BALF and IgE in the serum were measured by ELISA. Lung histological changes were detected by using hematoxylin and eosin (H&E) stain. The results showed that angelicin significantly inhibited inflammatory cells infiltration into the lungs. Histological studies showed that angelicin significantly attenuated OVA-induced lung injury. Meanwhile, treatment of angelicin dose-dependently inhibited OVA-induced the production of IL-4, IL-5, and IL-13 in BALF and IgE in the serum. Furthermore, angelicin was found to inhibit airway hyperresponsiveness and NF-kB activation. In conclusion, our results suggested that angelicin inhibited allergic airway inflammation and hyperresponsiveness by inhibiting NF-kB activation.


Asthma/drug therapy , Furocoumarins/pharmacology , Inflammation/prevention & control , Animals , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/prevention & control , Bronchoalveolar Lavage Fluid/chemistry , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Furocoumarins/therapeutic use , Immunoglobulin E/blood , Inflammation/diet therapy , Lung/pathology , Mice , NF-kappa B/antagonists & inhibitors , Ovalbumin
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(10): 2249-51, 2010 Oct.
Article Zh | MEDLINE | ID: mdl-20965816

OBJECTIVE: To observe the dynamic changes of CD3+, CD4+, and CD8+ T lymphocytes in the peripheral blood of patients with sepsis and discuss the clinical significance. METHODS: Sixty-four patients admitted in the Emergency Center and Emergency Intensive Care Unit of the Second Hospital of Wenzhou Medical University between August, 2007 and July, 2009 were enrolled in this study. CD3+, CD4+, and CD8+ T lymphocytes in the peripheral blood were detected by flow cytometry on days 1, 7 and 14 after admission, and the results were compared between the patients with improvement of the condition and those without improvement, with 20 healthy subjects as the control group. RESULTS: On day 1 after admission, CD3+ and CD4+ T lymphocytes and CD4+/CD8+ T cell ratio were obviously lower in the 2 groups of patients with sepsis than in the control group (P<0.05), but no significant difference was found in CD8+ T lymphocytes. The sepsis patients with clinical improvement showed significant higher CD3+ and CD4+ T lymphocyte percentages and CD4+/CD8+ T cell ratio than those without improvement on day 1. In the patients with clinical improvement, CD3+ and CD4+T lymphocytes and CD4+/CD8+ T cell ratio increased gradually with time and till day 14, they were comparable with the control levels; in the patients without improvement, CD3+ and CD4+ T lymphocytes and CD4+/CD8+ T cell ratio showed no obvious alterations in the course of observation. CONCLUSION: Immune imbalance occurs in patients with sepsis represented by lowered CD3+ and CD4+T lymphocyte percentages and CD4+/CD8+ T cell ratio in relation to the severity of the condition. CD3+ and CD4+ T lymphocytes and CD4+/CD8+ T cell ratio can be used as the indicators for assessing the severity of sepsis.


Sepsis/blood , T-Lymphocyte Subsets/cytology , Adult , Aged , Aged, 80 and over , CD4-CD8 Ratio , Case-Control Studies , Female , Flow Cytometry , Humans , Male , Middle Aged , Sepsis/immunology
...