Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.769
Filter
1.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3837-3847, 2024 Jul.
Article in Chinese | MEDLINE | ID: mdl-39099357

ABSTRACT

The study investigates the therapeutic effects and mechanisms of ginsenoside Rg_1(GRg_1) on sepsis-induced acute lung injury(SALI). A murine model of SALI was created using cecal ligation and puncture(CLP) surgery, and mice were randomly assigned to groups for GRg_1 intervention. Survival and body weight changes were recorded, lung function was assessed with a non-invasive lung function test system, and lung tissue damage was evaluated through HE staining. The content and expression of inflammatory factors were measured by ELISA and qRT-PCR. Apoptosis was examined using flow cytometry and TUNEL staining. The activation and expression of apoptosis-related molecules cysteinyl aspartate specific proteinase 3(caspase-3), B-cell lymphoma-2(Bcl-2), Bcl-2 associated X protein(Bax), and endoplasmic reticulum stress-related molecules protein kinase R-like endoplasmic reticulum kinase(PERK), eukaryotic initiation factor 2α(eIF2α), activating transcription factor 4(ATF4), and C/EBP homologous protein(CHOP) were studied using Western blot and qRT-PCR. In addition, an in vitro model of lipopolysaccharide(LPS)-induced lung alveolar epithelial cell injury was used, with the application of the endoplasmic reticulum stress inducer tunicamycin to validate the action mechanism of GRg_1. RESULTS:: indicated that, when compared to the model group, GRg_1 intervention significantly enhanced the survival time of CLP mice, mitigated body weight loss, and improved impaired lung function indices. The GRg_1-treated mice also displayed reduced lung tissue pathological scores, a reduced lung tissue wet-to-dry weight ratio, and lower protein content in the bronchoalveolar lavage fluid. Serum levels of interleukin-6(IL-6), interleukin-1ß(IL-1ß), and tumor necrosis factor-α(TNF-α), as well as the mRNA expressions of these cytokines in lung tissues, were decreased. There was a notable decrease in the proportion of apopto-tic alveolar epithelial cells, and down-regulated expressions of caspase-3, Bax, PERK, eIF2α, ATF4, and CHOP and up-regulated expression of Bcl-2 were observed. In vitro findings showed that the apoptosis-lowering and apoptosis-related protein down-regulating effects of GRg_1 were significantly inhibited with the co-application of tunicamycin. Altogether, GRg_1 reduces apoptosis of alveolar epithelial cells, inhibits inflammation in the lungs, alleviates lung injury, and enhances lung function, possibly through the PERK/eIF2α/ATF4/CHOP pathway.


Subject(s)
Activating Transcription Factor 4 , Acute Lung Injury , Alveolar Epithelial Cells , Apoptosis , Eukaryotic Initiation Factor-2 , Ginsenosides , Sepsis , Transcription Factor CHOP , eIF-2 Kinase , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Ginsenosides/pharmacology , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Mice , Apoptosis/drug effects , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Sepsis/drug therapy , Sepsis/complications , Sepsis/metabolism , Sepsis/genetics , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/genetics , Male , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Humans , Endoplasmic Reticulum Stress/drug effects , Mice, Inbred C57BL
2.
Clin Transl Med ; 14(8): e1785, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39090662

ABSTRACT

BACKGROUND: Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a common acute respiratory failure due to diffuse pulmonary inflammation and oedema. Elaborate regulation of macrophage activation is essential for managing this inflammatory process and maintaining tissue homeostasis. In the past decades, metabolic reprogramming of macrophages has emerged as a predominant role in modulating their biology and function. Here, we observed reduced expression of carnitine palmitoyltransferase 1A (CPT1A), a key rate-limiting enzyme of fatty acid oxidation (FAO), in macrophages of lipopolysaccharide (LPS)-induced ALI mouse model. We assume that CPT1A and its regulated FAO is involved in the regulation of macrophage polarization, which could be positive regulated by interleukin-10 (IL-10). METHODS: After nasal inhalation rIL-10 and/or LPS, wild type (WT), IL-10-/-, Cre-CPT1Afl/fl and Cre+CPT1Afl/fl mice were sacrificed to harvest bronchoalveolar lavage fluid, blood serum and lungs to examine cell infiltration, cytokine production, lung injury severity and IHC. Bone marrow-derived macrophages (BMDMs) were extracted from mice and stimulated by exogenous rIL-10 and/or LPS. The qRT-PCR, Seahorse XFe96 and FAO metabolite related kits were used to test the glycolysis and FAO level in BMDMs. Immunoblotting assay, confocal microscopy and fluorescence microplate were used to test macrophage polarization as well as mitochondrial structure and function damage. RESULTS: In in vivo experiments, we found that mice lacking CPT1A or IL-10 produced an aggravate inflammatory response to LPS stimulation. However, the addition of rIL-10 could alleviate the pulmonary inflammation in mice effectively. IHC results showed that IL-10 expression in lung macrophage decreased dramatically in Cre+CPT1Afl/fl mice. The in vitro experiments showed Cre+CPT1Afl/fl and IL-10-/- BMDMs became more "glycolytic", but less "FAO" when subjected to external attacks. However, the supplementation of rIL-10 into macrophages showed reverse effect. CPT1A and IL-10 can drive the polarization of BMDM from M1 phenotype to M2 phenotype, and CPT1A-IL-10 axis is also involved in the process of maintaining mitochondrial homeostasis. CONCLUSIONS: CPT1A modulated metabolic reprogramming and polarisation of macrophage under LPS stimulation. The protective effects of CPT1A may be partly attributed to the induction of IL-10/IL-10 receptor expression.


Subject(s)
Acute Lung Injury , Carnitine O-Palmitoyltransferase , Interleukin-10 , Macrophages , Animals , Male , Mice , Acute Lung Injury/metabolism , Acute Lung Injury/drug therapy , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Disease Models, Animal , Interleukin-10/metabolism , Lipopolysaccharides , Macrophages/metabolism , Macrophages/drug effects , Mice, Inbred C57BL , Phenotype , Mice, Knockout
3.
Respir Res ; 25(1): 303, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112999

ABSTRACT

BACKGROUND: Acute lung injury (ALI) following pneumonia involves uncontrolled inflammation and tissue injury, leading to high mortality. We previously confirmed the significantly increased cargo content and extracellular vesicle (EV) production in thrombin-preconditioned human mesenchymal stromal cells (thMSCs) compared to those in naïve and other preconditioning methods. This study aimed to investigate the therapeutic efficacy of EVs derived from thMSCs in protecting against inflammation and tissue injury in an Escherichia coli (E. coli)-induced ALI mouse model. METHODS: In vitro, RAW 264.7 cells were stimulated with 0.1 µg/mL liposaccharides (LPS) for 1 h, then were treated with either PBS (LPS Ctrl) or 5 × 107 particles of thMSC-EVs (LPS + thMSC-EVs) for 24 h. Cells and media were harvested for flow cytometry and ELISA. In vivo, ICR mice were anesthetized, intubated, administered 2 × 107 CFU/100 µl of E. coli. 50 min after, mice were then either administered 50 µL saline (ECS) or 1 × 109 particles/50 µL of thMSC-EVs (EME). Three days later, the therapeutic efficacy of thMSC-EVs was assessed using extracted lung tissue, bronchoalveolar lavage fluid (BALF), and in vivo computed tomography scans. One-way analysis of variance with post-hoc TUKEY test was used to compare the experimental groups statistically. RESULTS: In vitro, IL-1ß, CCL-2, and MMP-9 levels were significantly lower in the LPS + thMSC-EVs group than in the LPS Ctrl group. The percentages of M1 macrophages in the normal control, LPS Ctrl, and LPS + thMSC-EV groups were 12.5, 98.4, and 65.9%, respectively. In vivo, the EME group exhibited significantly lower histological scores for alveolar congestion, hemorrhage, wall thickening, and leukocyte infiltration than the ECS group. The wet-dry ratio for the lungs was significantly lower in the EME group than in the ECS group. The BALF levels of CCL2, TNF-a, and IL-6 were significantly lower in the EME group than in the ECS group. In vivo CT analysis revealed a significantly lower percentage of damaged lungs in the EME group than in the ECS group. CONCLUSION: Intratracheal thMSC-EVs administration significantly reduced E. coli-induced inflammation and lung tissue damage. Overall, these results suggest therapeutically enhanced thMSC-EVs as a novel promising therapeutic option for ARDS/ALI.


Subject(s)
Acute Lung Injury , Extracellular Vesicles , Mesenchymal Stem Cells , Mice, Inbred ICR , Thrombin , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Mice , Mesenchymal Stem Cells/metabolism , RAW 264.7 Cells , Thrombin/metabolism , Escherichia coli , Male , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli Infections/therapy , Treatment Outcome , Disease Models, Animal , Humans
4.
Gen Physiol Biophys ; 43(5): 399-409, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39140680

ABSTRACT

Acute lung injury (ALI) is a significant health condition with notable rates of morbidity and mortality globally. Long non-coding ribose nucleic acids (lncRNAs) play vital roles in mitigating various inflammation-related diseases, including ALI. The study aimed to investigate the functional role and molecular mechanisms of lncRNA SNHG1 on ALI in lipopolysaccharide (LPS)-treated A549 cells and in LPS-induced ALI mice. The expression of SNHG1 was initially examined in LPS-treated A549 cells. We further demonstrated the critical function of SNHG1 through various cellular assessments following SNHG1 knockdown, including cell counting kit (CCK)-8 assay, flow cytometry analysis, as well as enzyme-linked immunosorbent assay (ELISA). Reducing SNHG1 levels hindered the negative effects of LPS on cell viability, apoptosis, and inflammation. Moreover, SNHG1 acted as a negative regulator for miR-199a-3p, which targeted downstream ROCK2. Depletion of miR-199a-3p or enhanced expression of ROCK2 abolished the protective effects of SNHG1 knockdown on LPS-induced apoptosis and inflammation. Consistently, silencing SNHG1 alleviated LPS-induced lung injury in mice, demonstrating its potential therapeutic benefits in managing ALI. Overall, this study sheds light on the role of SNHG1 in modulating inflammation and apoptosis in ALI through the miR-199a-3p/ROCK2 pathway, offering new insights for the treatment of this condition.


Subject(s)
Acute Lung Injury , Gene Knockdown Techniques , Lipopolysaccharides , MicroRNAs , RNA, Long Noncoding , rho-Associated Kinases , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Mice , Humans , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Male , A549 Cells , Signal Transduction , Mice, Inbred C57BL , Apoptosis/genetics
5.
ACS Nano ; 18(32): 21009-21023, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39087239

ABSTRACT

Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), induce high morbidity and mortality rates, which challenge the present approaches for the treatment of ALI/ARDS. The clinically used photosensitizer verteporfin (VER) exhibits great potential in the treatment of acute lung injury and acute respiratory distress syndrome (ALI/ARDS) by regulating macrophage polarization and reducing inflammation. Nevertheless, its hydrophobic characteristics, nonspecificity, and constrained bioavailability hinder its therapeutic efficacy. In this work, we developed a type of VER-cored artificial exosome (EVM), which was produced by using mesoporous silica nanoparticles (MSNs) to load VER, followed by the exocytosis of internalized VER-MSNs from mouse bone marrow-derived mesenchymal stem cells (mBMSCs) without further modification. Both in vitro and in vivo assessments confirmed the powerful anti-inflammation induced by EVM. EVM also showed significant higher accumulation to inflammatory lungs compared with healthy ones, which was beneficial to the treatment of ALI/ARDS. EVM improved pulmonary function, attenuated lung injury, and reduced mortality in ALI mice with high levels of biocompatibility, exhibiting a 5-fold higher survival rate than the control. This type of artificial exosome emitted near-infrared light in the presence of laser activation, which endowed EVM with trackable ability both in vitro and in vivo. Our work developed a type of clinically used photosensitizer-loaded artificial exosome with membrane integrity and traceability. To the best of our knowledge, this kind of intracellularly synthesized artificial exosome was developed and showed great potential in ALI/ARDS therapy.


Subject(s)
Acute Lung Injury , Exosomes , Silicon Dioxide , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/therapy , Mice , Exosomes/metabolism , Exosomes/chemistry , Silicon Dioxide/chemistry , Verteporfin/pharmacology , Verteporfin/chemistry , Verteporfin/therapeutic use , Nanoparticles/chemistry , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Male , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Porosity
6.
J Cell Mol Med ; 28(15): e18589, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39135202

ABSTRACT

Sepsis causes systemic inflammatory responses and acute lung injury (ALI). Despite modern treatments, sepsis-related ALI mortality remains high. Aqueous extract of Descuraniae Semen (AEDS) exerts anti-endoplasmic reticulum (ER) stress, antioxidant and anti-inflammatory effects. AEDS alleviates inflammation and oedema in ALI. Sodium-potassium-chloride co-transporter isoform 1 (NKCC1) is essential for regulating alveolar fluid and is important in ALI. The NKCC1 activity is regulated by upstream with-no-lysine kinase-4 (WNK4) and STE20/SPS1-related proline/alanine-rich kinase (SPAK). This study aimed to investigate the effects of AEDS on lipopolysaccharide (LPS)-induced ALI model in A549 cells, considering the regulation of ER stress, WNK4-SPAK-NKCC1 cascades, inflammation and apoptosis. Cell viability was investigated by the CCK-8 assay. The expressions of the proteins were assessed by immunoblotting analysis assays. The levels of pro-inflammatory cytokines were determined by ELISA. The expression of cytoplasmic Ca2+ in A549 cells was determined using Fluo-4 AM. AEDS attenuates LPS-induced inflammation, which is associated with increased pro-inflammatory cytokine expression and activation of the WNK4-SPAK-NKCC1 pathway. AEDS inhibits the WNK4-SPAK-NKCC1 pathway by regulating of Bcl-2, IP3R and intracellular Ca2+. WNK4 expression levels are significantly higher in the WNK4-overexpressed transfected A549 cells and significantly decrease after AEDS treatment. AEDS attenuates LPS-induced inflammation by inhibiting the WNK4-SPAK-NKCC1 cascade. Therefore, AEDS is regarded as a potential therapeutic agent for ALI.


Subject(s)
Endoplasmic Reticulum Stress , Inflammation , Lipopolysaccharides , Protein Serine-Threonine Kinases , Signal Transduction , Solute Carrier Family 12, Member 2 , Humans , Protein Serine-Threonine Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , A549 Cells , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Solute Carrier Family 12, Member 2/metabolism , Solute Carrier Family 12, Member 2/genetics , Signal Transduction/drug effects , Apoptosis/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Plant Extracts/pharmacology , Cell Survival/drug effects , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology
7.
Zhongguo Zhong Yao Za Zhi ; 49(12): 3295-3301, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-39041092

ABSTRACT

This study aims to reveal the effects of the herb pair Astragali Radix-Salviae Miltiorrhizae Radix et Rhizoma(AR-SMRR) on phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin(PI3K/Akt/mTOR) pathway and autophagy in the lung tissue of the rat model of acute lung injury(ALI). Fifty adult male SD rats were randomized into sham, model, autophagy inhibition(intraperitoneal injection of chloroquine at 10 mg·kg~(-1)), autophagy induction(intraperitoneal injection of rapamycin at 15 mg·kg~(-1)), and AR-SMRR(5 g·kg~(-1), gavage) groups. The rats in the sham group received intratracheal instillation of normal saline, and those in other groups received intratracheal instillation of lipopolysaccharide(LPS, 5 mg·kg~(-1)) for the modeling of ALI. Seven days before the operation, the rats in the sham and model groups were administrated with normal saline, and those in other groups with corresponding drugs. Specimens were collected 24 h after modeling. The pathological changes of the lung tissue were observed under a light microscope. The lung wet/dry weight ratio and the lactate dehydrogenase(LDH) activity and total protein concentration in the bronchoalveolar lavage fluid(BALF) were measured. Western blot was employed to measure the protein levels of microtubule-associated protein 1-light chain 3(LC3), beclin-1, p62, PI3K, Akt, and mTOR. Compared with the sham group, the model group showed increased histopathological score of the lung tissue, lung wet/dry weight ratio, and LDH activity and protein concentration in BALF. Autophagy inhibition further increased these indicators compared with the model group, while autophagy induction and AR-SMRR lowered the levels. In addition, AR-SMRR up-regulated the protein levels of LC3-Ⅱ and beclin-1, down-regulated the expression of p62, and inhibited the expression of p-PI3K, p-Akt, and p-mTOR in the lung tissue of ALI rats. The findings suggest that AR-SMRR can alleviate the lung injury and edema in the rat model of ALI induced by LPS by enhancing autophagy via down-regulating PI3K/Akt/mTOR signaling pathway.


Subject(s)
Acute Lung Injury , Autophagy , Drugs, Chinese Herbal , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Male , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Rats , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Autophagy/drug effects , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Salvia miltiorrhiza/chemistry , Astragalus propinquus/chemistry , Rhizome/chemistry , Lung/drug effects , Lung/metabolism , Lung/pathology , Humans
8.
Respir Res ; 25(1): 263, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956592

ABSTRACT

BACKGROUND: Aberrant activation of macrophages is associated with pathogenesis of acute lung injury (ALI). However, the potential pathogenesis has not been explored. OBJECTIVES: We aimed to identify whether histone deacetylase (HDAC) 10 is involved in lipopolysaccharide (LPS)-exposed ALI and reveal the underlying pathogenesis by which it promotes lung inflammation in LPS-exposed ALI via modifying P62 with deacetylation. METHODS: We constructed an ALI mice model stimulated with LPS to determine the positive effect of Hdac10 deficiency. Moreover, we cultured murine alveolar macrophage cell line (MH-S cells) and primary bone marrow-derived macrophages (BMDMs) to explore the pro-inflammatory activity and mechanism of HDAC10 after LPS challenge. RESULTS: HDAC10 expression was increased both in mice lung tissues and macrophage cell lines and promoted inflammatory cytokines production exposed to LPS. Hdac10 deficiency inhibited autophagy and inflammatory response after LPS stimulation. In vivo, Hdac10fl/fl-LysMCre mice considerably attenuated lung inflammation and inflammatory cytokines release exposed to LPS. Mechanistically, HDAC10 interacts with P62 and mediates P62 deacetylation at lysine 165 (K165), by which it promotes P62 expression and increases inflammatory cytokines production. Importantly, we identified that Salvianolic acid B (SAB), an HDAC10 inhibitor, reduces lung inflammatory response in LPS-stimulated ALI. CONCLUSION: These results uncover a previously unknown role for HDAC10 in regulating P62 deacetylation and aggravating lung inflammation in LPS-induced ALI, implicating that targeting HDAC10 is an effective therapy for LPS-exposed ALI.


Subject(s)
Acute Lung Injury , Histone Deacetylases , Lipopolysaccharides , Lysine , Mice, Inbred C57BL , Animals , Acute Lung Injury/chemically induced , Acute Lung Injury/prevention & control , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Lipopolysaccharides/toxicity , Mice , Acetylation , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/deficiency , Lysine/metabolism , Mice, Knockout , Male , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Myeloid Cells/metabolism
9.
Chem Biol Drug Des ; 104(1): e14579, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39013775

ABSTRACT

Sepsis-induced acute lung injury (ALI) is a severe complication of sepsis. Karanjin, a natural flavonoid compound, has been proved to have anti-inflammatory function, but its role in sepsis-stimulated ALI is uncertain. Herein, the effect of karanjin on sepsis-stimulated ALI was investigated. We built a mouse model of lipopolysaccharide (LPS)-stimulated ALI. The histopathological morphology of lung tissues was scrutinized by hematoxylin-eosin (H&E) staining. The lung injury score and lung wet/dry weight ratio were detected. The myeloperoxidase (MPO) activity and malondialdehyde (MDA) content were scrutinized by commercial kits. Murine alveolar lung epithelial (MLE-12) cells were treated with LPS to mimic a cellular model of ALI. The cell viability was scrutinized by the CCK-8 assay. The contents of proinflammatory cytokines were scrutinized by qRT-PCR and ELISA. The TLR4 and MyD88 contents were scrutinized by qRT-PCR and western blotting. Results showed that karanjin alleviated LPS-stimulated ALI in mice by inhibiting lung tissue lesions, edema, and oxidative stress. Moreover, karanjin inhibited LPS-stimulated inflammation and TLR4 pathway activation in mice. However, treatment with GSK1795091, an agonist of TLR4, attenuated the effects of karanjin on LPS-induced ALI. Furthermore, karanjin repressed LPS-stimulated inflammatory response and TLR4 pathway activation in MLE-12 cells. Overexpression of TLR4 attenuated karanjin effects on LPS-stimulated inflammatory responses in MLE-12 cells. In conclusion, karanjin repressed sepsis-stimulated ALI in mice by suppressing the TLR4 pathway.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Sepsis , Signal Transduction , Toll-Like Receptor 4 , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Toll-Like Receptor 4/metabolism , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/complications , Mice , Signal Transduction/drug effects , Male , Cell Line , Lung/pathology , Lung/metabolism , Lung/drug effects , Peroxidase/metabolism , Myeloid Differentiation Factor 88/metabolism , Malondialdehyde/metabolism , Cytokines/metabolism , Disease Models, Animal , Cell Survival/drug effects , Protective Agents/pharmacology , Protective Agents/therapeutic use , Sulfonamides
10.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000242

ABSTRACT

Acute lung injury (ALI) is a condition associated with acute respiratory failure, resulting in significant morbidity and mortality. It involves cellular changes such as disruption of the alveolar-capillary membrane, excessive neutrophil migration, and release of inflammatory mediators. Broncho-Vaxom® (BV), a lyophilized product containing cell membrane components derived from eight bacteria commonly found in the respiratory tract, is known for its potential to reduce viral and bacterial lung infections. However, the specific effect of BV on ALI has not been clearly defined. This study explored the preventive effects of BV and its underlying mechanisms in a lipopolysaccharide (LPS)-induced ALI mouse model. Oral BV (1 mg/kg) gavage was administered one hour before the intratracheal injection of LPS to evaluate its preventive effect on the ALI model. The pre-administration of BV significantly mitigates inflammatory parameters, including the production of inflammatory mediators, macrophage infiltration, and NF-κB activation in lung tissue, and the increase in inflammatory cells in bronchoalveolar lavage fluid (BALF). Moreover, BV (3 µg/mL) pretreatment reduced the expression of M1 macrophage markers, interleukins (IL-1ß, IL-6), tumor necrosis factor α, and cyclooxygenase-2, which are activated by LPS, in both mouse alveolar macrophage MH-S cells and human macrophage THP-1 cells. These findings showed that BV exhibits anti-inflammatory effects by suppressing inflammatory mediators through the NF-κB pathway, suggesting its potential to attenuate bronchial and pulmonary inflammation.


Subject(s)
Acute Lung Injury , Disease Models, Animal , Lipopolysaccharides , Animals , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/drug therapy , Mice , Humans , Inflammation/pathology , Inflammation/metabolism , Inflammation/drug therapy , Male , Cell Extracts/pharmacology , Cell Extracts/therapeutic use , NF-kappa B/metabolism , Bronchoalveolar Lavage Fluid , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/drug effects , Cytokines/metabolism , Inflammation Mediators/metabolism , Lung/pathology , Lung/metabolism , Lung/drug effects , Bacterial Lysates
11.
PLoS One ; 19(7): e0305058, 2024.
Article in English | MEDLINE | ID: mdl-38954702

ABSTRACT

OBJECTIVES: Astragaloside IV (AS-IV) is a natural triterpenoid saponin compound with a variety of pharmacological effects, and several studies have clarified its anti-inflammatory effects, which may make it an effective alternative treatment against inflammation. In the study, we aimed to investigate whether AS-IV could attenuate the inflammatory response to acute lung injury and its mechanisms. METHODS: Different doses of AS-IV (20mg·kg-1, 40mg·kg-1, and 80mg·kg-1) were administered to the ALI rat model, followed by collection of serum and broncho alveolar lavage fluid (BALF) for examination of the inflammatory response, and HE staining of the lung and colon tissues, and interpretation of the potential molecular mechanisms by quantitative real-time PCR (qRT-PCR), Western blotting (WB). In addition, fecal samples from ALI rats were collected and analyzed by 16S rRNA sequencing. RESULTS: AS-IV decreased the levels of TNF-α, IL-6, and IL-1ß in serum and BALF of mice with Acute lung injury (ALI). Lung and colon histopathology confirmed that AS-IV alleviated inflammatory infiltration, tissue edema, and structural changes. qRT-PCR and WB showed that AS-IV mainly improved inflammation by inhibiting the expression of PI3K, AKT and mTOR mRNA, and improved the disorder of intestinal microflora by increasing the number of beneficial bacteria and reducing the number of harmful bacteria. CONCLUSION: AS-IV reduces the expression of inflammatory factors by inhibiting the PI3K/AKT/mTOR pathway and optimizes the composition of the gut microflora in AIL rats.


Subject(s)
Acute Lung Injury , Gastrointestinal Microbiome , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Saponins , Signal Transduction , TOR Serine-Threonine Kinases , Triterpenes , Animals , Saponins/pharmacology , Saponins/therapeutic use , Triterpenes/pharmacology , Acute Lung Injury/drug therapy , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , TOR Serine-Threonine Kinases/metabolism , Gastrointestinal Microbiome/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Rats , Male , Mice , Rats, Sprague-Dawley , Inflammation/drug therapy , Bronchoalveolar Lavage Fluid/chemistry , Lung/pathology , Lung/drug effects , Lung/microbiology , Lung/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
12.
Gen Physiol Biophys ; 43(4): 353-366, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38953577

ABSTRACT

This study aimed to assess the prophylactic effects of Berberine on experimentally induced lung sepsis and examine its effects on selected cytokines, genes, and protein expression besides the histopathological evaluation. Berberine significantly reduced the wet/dry lung ratio, the broncho-alveolar lavage fluid (BALF) protein, cells, neutrophils percentage, and cytokines levels. In addition, pretreatment with Berberine decreased the myeloperoxidase (MPO) and malondialdehyde (MDA) levels and decreased gene expression of nuclear factor kappa B (NF-κB), monocyte chemoattractant protein-1 (MCP-1), and the intracellular adhesion molecule 1 (ICAM-1) by RT-qPCR analysis, revealing Berberine's antioxidant and anti-inflammatory mode of action. Western blot analysis revealed increased peroxisome proliferator-activated receptor gamma (PPAR-γ) expression in the Berberine pretreated group compared to the cecal ligation and puncture (CLP) group, in which the histopathological examination evidenced this improvement. In conclusion, Berberine improved lung sepsis via its PPAR-γ mediated antioxidant and anti-inflammatory effects.


Subject(s)
Acute Lung Injury , Berberine , PPAR gamma , Sepsis , Signal Transduction , Berberine/pharmacology , Berberine/therapeutic use , Animals , PPAR gamma/metabolism , Sepsis/metabolism , Sepsis/drug therapy , Rats , Acute Lung Injury/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/prevention & control , Male , Signal Transduction/drug effects , Up-Regulation/drug effects , Rats, Wistar , Rats, Sprague-Dawley
13.
Respir Res ; 25(1): 291, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080660

ABSTRACT

Acute lung injury (ALI) is characterized by an unregulated inflammatory reaction, often leading to severe morbidity and ultimately death. Excessive inflammation caused by M1 macrophage polarization and pyroptosis has been revealed to have a critical role in ALI. Recent study suggests that glycolytic reprogramming is important in the regulation of macrophage polarization and pyroptosis. However, the particular processes underlying ALI have yet to be identified. In this study, we established a Lipopolysaccharide(LPS)-induced ALI model and demonstrated that blocking glycolysis by using 2-Deoxy-D-glucose(2-DG) significantly downregulated the expression of M1 macrophage markers and pyroptosis-related genes, which was consistent with the in vitro results. Furthermore, our research has revealed that Phosphoglycerate Kinase 1(PGK1), an essential enzyme in the glycolysis pathway, interacts with NOD-, LRR- and pyrin domain-containing protein 3(NLRP3). We discovered that LPS stimulation improves the combination of PGK1 and NLRP3 both in vivo and in vitro. Interestingly, the absence of PGK1 reduces the phosphorylation level of NLRP3. Based on in vitro studies with mice bone marrow-derived macrophages (BMDMs), we further confirmed that siPGK1 plays a protective role by inhibiting macrophage pyroptosis and M1 macrophage polarization. The PGK1 inhibitor NG52 suppresses the occurrence of excessive inflammation in ALI. In general, it is plausible to consider a therapeutic strategy that focuses on modulating the relationship between PGK1 and NLRP3 as a means to mitigate the activation of inflammatory macrophages in ALI.


Subject(s)
Acute Lung Injury , Macrophages , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphoglycerate Kinase , Pyroptosis , Pyroptosis/physiology , Pyroptosis/drug effects , Animals , Phosphoglycerate Kinase/metabolism , Phosphoglycerate Kinase/genetics , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Mice , Macrophages/metabolism , Macrophages/drug effects , Macrophages/enzymology , Glycolysis/physiology , Glycolysis/drug effects , Male , Lipopolysaccharides/toxicity , Mice, Knockout , Cells, Cultured
15.
Sci Rep ; 14(1): 16071, 2024 07 11.
Article in English | MEDLINE | ID: mdl-38992150

ABSTRACT

Sepsis-induced acute lung injury (SALI) poses a significant threat with high incidence and mortality rates. Ginsenoside Rg1 (GRg1), derived from Ginseng in traditional Chinese medicine, has been found to reduce inflammation and protect lung epithelial cells against tissue damage. However, the specific roles and mechanisms by which GRg1 mitigates SALI have yet to be fully elucidated. In this context, we employed a relevant SALI mouse model, alongside network pharmacology, molecular docking, and molecular dynamics simulation to pinpoint GRg1's action targets, complemented by in vitro assays to explore the underlying mechanisms. Our research shows that GRg1 alleviates CLP-induced SALI, decreasing lung tissue damage and levels of serum proinflammatory factor IL-6, TNF-α, and IL-1ß, also enhancing the survival rate of CLP mice. A total of 116 common targets between GRg1 and ALI, with specific core targets including AKT1, VEGFA, SRC, IGF1, ESR1, STAT3, and ALB. Further in vitro experiments assessed GRg1's intervention effects on MLE-12 cells exposed to LPS, with qRT-PCR analysis and molecular dynamics simulations confirming AKT1 as the key target with the favorable binding activity for GRg1. Western blot results indicated that GRg1 increased the Bcl-2/Bax protein expression ratio to reduce apoptosis and decreased the high expression of cleaved caspase-3 in LPS-induced MLE-12 cells. More results showed significant increases in the phosphorylation of PI3K and AKT1. Flow cytometric analysis using PI and Annexin-V assays further verified that GRg1 decreased the apoptosis rate in LPS-stimulated MLE-12 cells (from 14.85 to 6.54%, p < 0.05). The employment of the AKT1 inhibitor LY294002 confirmed these trends, indicating that AKT1's inhibition negates GRg1's protective effects on LPS-stimulated MLE-12 cells. In conclusion, our research highlights GRg1's potential as an effective adjunct therapy for SALI, primarily by inhibiting apoptosis in alveolar epithelial cells and reducing pro-inflammatory cytokine secretion, thus significantly enhancing the survival rates of CLP mice. These beneficial effects are mediated through targeting AKT1 and activating the PI3K-AKT pathway.


Subject(s)
Acute Lung Injury , Ginsenosides , Molecular Dynamics Simulation , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Sepsis , Signal Transduction , Ginsenosides/pharmacology , Ginsenosides/chemistry , Ginsenosides/therapeutic use , Animals , Proto-Oncogene Proteins c-akt/metabolism , Mice , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/complications , Acute Lung Injury/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/etiology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Male , Molecular Docking Simulation , Disease Models, Animal , Mice, Inbred C57BL , Apoptosis/drug effects , Cell Line , Lipopolysaccharides
16.
J Ethnopharmacol ; 334: 118584, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39019418

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Fuzheng-Qushi decoction (FZQS) is a practical Chinese herbal formula for relieving cough and fever. Therefore, the action and specific molecular mechanism of FZQS in the treatment of lung injury with cough and fever as the main symptoms need to be further investigated. AIMS OF THE STUDY: To elucidate the protective effects of FZQS against lung injury in mice and reveal its potential targets and key biological pathways for the treatment of lung injury based on transcriptomics, microbiomics, and untargeted metabolomics analyses. MATERIALS AND METHODS: Lipopolysaccharide (LPS) was used to induce a mouse model of lung injury, followed by the administration of FZQS. ELISA was used to detect IL-1ß, IL-6, IL-17A, IL-4, IL-10, and TNF-α, in mouse lung tissues. Macrophage polarization and neutrophil activation were measured by flow cytometry. RNA sequencing (RNA-seq) was applied to screen for differentially expressed genes (DEGs) in lung tissues. RT-qPCR and Western blot assays were utilized to validate key DEGs and target proteins in lung tissues. 16S rRNA sequencing was employed to characterize the gut microbiota of mice. Metabolites in the gut were analyzed using untargeted metabolomics. RESULTS: FZQS treatment significantly ameliorated lung histopathological damage, decreased pro-inflammatory cytokine levels, and increased anti-inflammatory cytokine levels. M1 macrophage levels in the peripheral blood decreased, M2 macrophage levels increased, and activated neutrophils were inhibited in mice with LPS-induced lung injury. Importantly, transcriptomic analysis showed that FZQS downregulated macrophage and neutrophil activation and migration and adhesion pathways by reversing 51 DEGs, which was further confirmed by RT-qPCR and Western blot analysis. In addition, FZQS modulated the dysbiosis of the gut microbiota by reversing the abundance of Corynebacterium, Facklamia, Staphylococcus, Paenalcaligenes, Lachnoclostridium, norank_f_Muribaculaceae, and unclassified_f_Lachnospiraceae. Meanwhile, metabolomics analysis revealed that FZQS significantly regulated tryptophan metabolism by reducing the levels of 3-Indoleacetonitrile and 5-Hydroxykynurenine. CONCLUSION: FZQS effectively ameliorated LPS-induced lung injury by inhibiting the activation, migration, and adhesion of macrophages and neutrophils and modulating gut microbiota and its metabolites.


Subject(s)
Drugs, Chinese Herbal , Lipopolysaccharides , Lung Injury , Metabolomics , Animals , Drugs, Chinese Herbal/pharmacology , Mice , Lung Injury/drug therapy , Lung Injury/metabolism , Lung Injury/chemically induced , Male , Gastrointestinal Microbiome/drug effects , Transcriptome/drug effects , Lung/drug effects , Lung/pathology , Lung/metabolism , Mice, Inbred C57BL , Cytokines/metabolism , Disease Models, Animal , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism
17.
Int Immunopharmacol ; 139: 112719, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39032470

ABSTRACT

Autophagy in alveolar macrophages (AMs) is an important mechanism for maintaining immune homeostasis and normal lung tissue function, and insufficient autophagy in AMs may mediate the development of sepsis-induced acute lung injury (SALI). Insufficient autophagy in AMs and the activation of the NLRP3 inflammasome were observed in a mouse model with SALI induced by cecal ligation and puncture (CLP), resulting in the release of a substantial quantity of proinflammatory factors and the formation of SALI. However, after andrographolide (AG) intervention, autophagy in AMs was significantly promoted, the activation of the NLRP3 inflammasome was inhibited, the release of proinflammatory factors and pyroptosis were suppressed, and SALI was then ameliorated. In the MH-S cell model stimulated with LPS, insufficient autophagy was discovered to promote the overactivation of the NLRP3 inflammasome. AG was found to significantly promote autophagy, inhibit the activation of the NLRP3 inflammasome, and attenuate the release of proinflammatory factors. The primary mechanism of AG promoting autophagy was to inhibit the activation of the PI3K/AKT/mTOR pathway by binding RAGE to the membrane. In addition, it inhibited the activation of the NLRP3 inflammasome to ameliorate SALI. Our findings suggest that AG promotes autophagy in AMs through the RAGE/PI3K/AKT/mTOR pathway to inhibit the activation of the NLRP3 inflammasome, remodel the functional homeostasis of AMs in SALI, and exert anti-inflammatory and lung-protective effects. It has also been the first to suggest that RAGE is likely a direct target through which AG regulates autophagy, providing theoretical support for a novel therapeutic strategy in sepsis.


Subject(s)
Acute Lung Injury , Autophagy , Diterpenes , Macrophages, Alveolar , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Receptor for Advanced Glycation End Products , Sepsis , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , TOR Serine-Threonine Kinases/metabolism , Autophagy/drug effects , Sepsis/drug therapy , Sepsis/complications , Sepsis/metabolism , Sepsis/immunology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Diterpenes/pharmacology , Diterpenes/therapeutic use , Mice , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Male , Receptor for Advanced Glycation End Products/metabolism , Inflammasomes/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal
18.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167354, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39004378

ABSTRACT

Acute lung injury (ALI) is a serious disorder characterized by the release of pro-inflammatory cytokines and cascade activation of macrophages. Ferroptosis, a form of iron-dependent cell death triggered by intracellular phospholipid peroxidation, has been implicated as an internal mechanism underlying ALI. In this study, we investigated the effects of m6A demethylase fat mass and obesity-associated protein (FTO) on the inhibition of macrophage ferroptosis in ALI. Using a mouse model of lipopolysaccharide (LPS)-induced ALI, we observed the induction of ferroptosis and its co-localization with the macrophage marker F4/80, suggesting that ferroptosis might be induced in macrophages. Ferroptosis was promoted during LPS-induced inflammation in macrophages in vitro, and the inflammation was counteracted by the ferroptosis inhibitor ferrostatin-1 (fer-1). Given that FTO showed lower expression levels in the lung tissue of mice with ALI and inflammatory macrophages, we further dissected the regulatory capacity of FTO in ferroptosis. The results demonstrated that FTO alleviated macrophage inflammation by inhibiting ferroptosis. Mechanistically, FTO decreased the stability of ACSL4 mRNA via YTHDF1, subsequently inhibiting ferroptosis and inflammation by interrupting polyunsaturated fatty acid consumption. Moreover, FTO downregulated the synthesis and secretion of prostaglandin E2, thereby reducing ferroptosis and inflammation. In vivo, the FTO inhibitor FB23-2 aggravated lung injury, the inflammatory response, and ferroptosis in mice with ALI; however, fer-1 therapy mitigated these effects. Overall, our findings revealed that FTO may function as an inhibitor of the inflammatory response driven by ferroptosis, emphasizing its potential as a target for ALI treatment.


Subject(s)
Acute Lung Injury , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Coenzyme A Ligases , Ferroptosis , Inflammation , Lipopolysaccharides , Macrophages , Animals , Ferroptosis/drug effects , Mice , Macrophages/metabolism , Macrophages/drug effects , Macrophages/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Male , Mice, Inbred C57BL , Phenylenediamines/pharmacology , Disease Models, Animal , RAW 264.7 Cells , Cyclohexylamines
19.
Cell Mol Life Sci ; 81(1): 325, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39079969

ABSTRACT

Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. The inflammatory cytokine storm causes systemic organ damage, especially acute lung injury in sepsis. In this study, we found that the expression of S-phase kinase-associated protein 2 (Skp2) was significantly decreased in sepsis-induced acute lung injury (ALI). Sepsis activated the MEK/ERK pathway and inhibited Skp2 expression in the pulmonary epithelium, resulting in a reduction of K48 ubiquitination of solute carrier family 3 member 2 (SLC3A2), thereby impairing its membrane localization and cystine/glutamate exchange function. Consequently, the dysregulated intracellular redox reactions induced ferroptosis in pulmonary epithelial cells, leading to lung injury. Finally, we demonstrated that intravenous administration of Skp2 mRNA-encapsulating lipid nanoparticles (LNPs) inhibited ferroptosis in the pulmonary epithelium and alleviated lung injury in septic mice. Taken together, these data provide an innovative understanding of the underlying mechanisms of sepsis-induced ALI and a promising therapeutic strategy for sepsis.


Subject(s)
Acute Lung Injury , Ferroptosis , Mice, Inbred C57BL , S-Phase Kinase-Associated Proteins , Sepsis , Ubiquitination , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/etiology , Sepsis/metabolism , Sepsis/complications , Sepsis/pathology , Animals , Mice , Humans , Male , S-Phase Kinase-Associated Proteins/metabolism , S-Phase Kinase-Associated Proteins/genetics , Lung/pathology , Lung/metabolism , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics
20.
J Cell Mol Med ; 28(13): e18386, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38990057

ABSTRACT

Acute lung injury (ALI) is a major pathophysiological problem characterized by severe inflammation, resulting in high morbidity and mortality. Plumbagin (PL), a major bioactive constituent extracted from the traditional Chinese herb Plumbago zeylanica, has been shown to possess anti-inflammatory and antioxidant pharmacological activities. However, its protective effect on ALI has not been extensively studied. The objective of this study was to investigate the protective effect of PL against ALI induced by LPS and to elucidate its possible mechanisms both in vivo and in vitro. PL treatment significantly inhibited pathological injury, MPO activity, and the wet/dry ratio in lung tissues, and decreased the levels of inflammatory cells and inflammatory cytokines TNF-α, IL-1ß, IL-6 in BALF induced by LPS. In addition, PL inhibited the activation of the PI3K/AKT/mTOR signalling pathway, increased the activity of antioxidant enzymes CAT, SOD, GSH and activated the Keap1/Nrf2/HO-1 signalling pathway during ALI induced by LPS. To further assess the association between the inhibitory effects of PL on ALI and the PI3K/AKT/mTOR and Keap1/Nrf2/HO-1 signalling, we pretreated RAW264.7 cells with 740Y-P and ML385. The results showed that the activation of PI3K/AKT/mTOR signalling reversed the protective effect of PL on inflammatory response induced by LPS. Moreover, the inhibitory effects of PL on the production of inflammatory cytokines induced by LPS also inhibited by downregulating Keap1/Nrf2/HO-1 signalling. In conclusion, the results indicate that the PL ameliorate LPS-induced ALI by regulating the PI3K/AKT/mTOR and Keap1-Nrf2/HO-1 signalling, which may provide a novel therapeutic perspective for PL in inhibiting ALI.


Subject(s)
Acute Lung Injury , Kelch-Like ECH-Associated Protein 1 , Lipopolysaccharides , NF-E2-Related Factor 2 , Naphthoquinones , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Acute Lung Injury/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , NF-E2-Related Factor 2/metabolism , TOR Serine-Threonine Kinases/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Lipopolysaccharides/adverse effects , Lipopolysaccharides/toxicity , Naphthoquinones/pharmacology , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Mice , Male , Cytokines/metabolism , Heme Oxygenase-1/metabolism , RAW 264.7 Cells , Anti-Inflammatory Agents/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Membrane Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL