Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 9.677
Filtrer
1.
PLoS One ; 19(7): e0305058, 2024.
Article de Anglais | MEDLINE | ID: mdl-38954702

RÉSUMÉ

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.


Sujet(s)
Lésion pulmonaire aigüe , Microbiome gastro-intestinal , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Saponines , Transduction du signal , Sérine-thréonine kinases TOR , Triterpènes , Animaux , Saponines/pharmacologie , Saponines/usage thérapeutique , Triterpènes/pharmacologie , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/microbiologie , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire aigüe/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Rats , Mâle , Souris , Rat Sprague-Dawley , Inflammation/traitement médicamenteux , Liquide de lavage bronchoalvéolaire/composition chimique , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/microbiologie , Poumon/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique
2.
Respir Res ; 25(1): 263, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956592

RÉSUMÉ

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.


Sujet(s)
Lésion pulmonaire aigüe , Histone deacetylases , Lipopolysaccharides , Lysine , Souris de lignée C57BL , Animaux , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/prévention et contrôle , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/génétique , Lésion pulmonaire aigüe/anatomopathologie , Lipopolysaccharides/toxicité , Souris , Acétylation , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Histone deacetylases/déficit , Lysine/métabolisme , Souris knockout , Mâle , Séquestosome-1/métabolisme , Séquestosome-1/génétique , Cellules myéloïdes/métabolisme
3.
Gen Physiol Biophys ; 43(4): 353-366, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953577

RÉSUMÉ

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.


Sujet(s)
Lésion pulmonaire aigüe , Berbérine , Récepteur PPAR gamma , Sepsie , Transduction du signal , Berbérine/pharmacologie , Berbérine/usage thérapeutique , Animaux , Récepteur PPAR gamma/métabolisme , Sepsie/métabolisme , Sepsie/traitement médicamenteux , Rats , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/prévention et contrôle , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques , Rat Wistar , Rat Sprague-Dawley
4.
Molecules ; 29(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38998987

RÉSUMÉ

The inhibition of soluble epoxide hydrolase (sEH) can reduce the level of dihydroxyeicosatrienoic acids (DHETs) effectively maintaining endogenous epoxyeicosatrienoic acids (EETs) levels, resulting in the amelioration of inflammation and pain. Consequently, the development of sEH inhibitors has been a prominent research area for over two decades. In the present study, we synthesized and evaluated sulfonyl urea derivatives for their potential to inhibit sEH. These compounds underwent extensive in vitro investigation, revealing their potency against human and mouse sEH, with 4f showing the most promising sEH inhibitory potential. When subjected to lipopolysaccharide (LPS)-induced acute lung injury (ALI) in studies in mice, compound 4f manifested promising anti-inflammatory efficacy. We investigated the analgesic efficacy of sEH inhibitor 4f in a murine pain model of tail-flick reflex. These results validate the role of sEH inhibition in inflammatory diseases and pave the way for the rational design and optimization of sEH inhibitors based on a sulfonyl urea template.


Sujet(s)
Antienzymes , Epoxide hydrolase , Urée , Epoxide hydrolase/antagonistes et inhibiteurs , Epoxide hydrolase/métabolisme , Animaux , Souris , Humains , Urée/pharmacologie , Urée/analogues et dérivés , Urée/composition chimique , Antienzymes/pharmacologie , Antienzymes/composition chimique , Antienzymes/synthèse chimique , Lésion pulmonaire aigüe/traitement médicamenteux , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Anti-inflammatoires/synthèse chimique , Anti-inflammatoires/usage thérapeutique , Lipopolysaccharides , Relation structure-activité , Solubilité , Modèles animaux de maladie humaine , Douleur/traitement médicamenteux
5.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000242

RÉSUMÉ

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.


Sujet(s)
Lésion pulmonaire aigüe , Modèles animaux de maladie humaine , Lipopolysaccharides , Animaux , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/étiologie , Lésion pulmonaire aigüe/traitement médicamenteux , Souris , Humains , Inflammation/anatomopathologie , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Mâle , Extrait cellulaire/pharmacologie , Extrait cellulaire/usage thérapeutique , Facteur de transcription NF-kappa B/métabolisme , Liquide de lavage bronchoalvéolaire , Macrophages alvéolaires/métabolisme , Macrophages alvéolaires/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Médiateurs de l'inflammation/métabolisme , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques ,
6.
Sci Rep ; 14(1): 16071, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992150

RÉSUMÉ

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.


Sujet(s)
Lésion pulmonaire aigüe , Ginsénosides , Simulation de dynamique moléculaire , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Sepsie , Transduction du signal , Ginsénosides/pharmacologie , Ginsénosides/composition chimique , Ginsénosides/usage thérapeutique , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Souris , Sepsie/traitement médicamenteux , Sepsie/métabolisme , Sepsie/complications , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire aigüe/étiologie , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Simulation de docking moléculaire , Modèles animaux de maladie humaine , Souris de lignée C57BL , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Lipopolysaccharides
7.
J Cell Mol Med ; 28(13): e18386, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38990057

RÉSUMÉ

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.


Sujet(s)
Lésion pulmonaire aigüe , Protéine-1 de type kelch associée à ECH , Lipopolysaccharides , Facteur-2 apparenté à NF-E2 , Naphtoquinones , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Animaux , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/anatomopathologie , Facteur-2 apparenté à NF-E2/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Protéine-1 de type kelch associée à ECH/métabolisme , Lipopolysaccharides/effets indésirables , Lipopolysaccharides/toxicité , Naphtoquinones/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Souris , Mâle , Cytokines/métabolisme , Heme oxygenase-1/métabolisme , Cellules RAW 264.7 , Anti-inflammatoires/pharmacologie , Heme oxygenase (decyclizing)/métabolisme , Protéines membranaires/métabolisme
8.
Chem Biol Drug Des ; 104(1): e14579, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39013775

RÉSUMÉ

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.


Sujet(s)
Lésion pulmonaire aigüe , Lipopolysaccharides , Sepsie , Transduction du signal , Récepteur de type Toll-4 , Animaux , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/métabolisme , Récepteur de type Toll-4/métabolisme , Sepsie/traitement médicamenteux , Sepsie/métabolisme , Sepsie/complications , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Lignée cellulaire , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Myeloperoxidase/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Malonaldéhyde/métabolisme , Cytokines/métabolisme , Modèles animaux de maladie humaine , Survie cellulaire/effets des médicaments et des substances chimiques , Agents protecteurs/pharmacologie , Agents protecteurs/usage thérapeutique , Sulfonamides
9.
Respir Res ; 25(1): 276, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39010105

RÉSUMÉ

BACKGROUND: The pathogenesis of acute lung injury (ALI) involves a severe inflammatory response, leading to significant morbidity and mortality. N6-methylation of adenosine (m6A), an abundant mRNA nucleotide modification, plays a crucial role in regulating mRNA metabolism and function. However, the precise impact of m6A modifications on the progression of ALI remains elusive. METHODS: ALI models were induced by either intraperitoneal injection of lipopolysaccharide (LPS) into C57BL/6 mice or the LPS-treated alveolar type II epithelial cells (AECII) in vitro. The viability and proliferation of AECII were assessed using CCK-8 and EdU assays. The whole-body plethysmography was used to record the general respiratory functions. M6A RNA methylation level of AECII after LPS insults was detected, and then the "writer" of m6A modifications was screened. Afterwards, we successfully identified the targets that underwent m6A methylation mediated by METTL3, a methyltransferase-like enzyme. Last, we evaluated the regulatory role of METTL3-medited m6A methylation at phosphatase and tensin homolog (Pten) in ALI, by assessing the proliferation, viability and inflammation of AECII. RESULTS: LPS induced marked damages in respiratory functions and cellular injuries of AECII. The m6A modification level in mRNA and the expression of METTL3, an m6A methyltransferase, exhibited a notable rise in both lung tissues of ALI mice and cultured AECII cells subjected to LPS treatment. METTL3 knockdown or inhibition improved the viability and proliferation of LPS-treated AECII, and also reduced the m6A modification level. In addition, the stability and translation of Pten mRNA were enhanced by METTL3-mediated m6A modification, and over-expression of PTEN reversed the protective effect of METTL3 knockdown in the LPS-treated AECII. CONCLUSIONS: The progression of ALI can be attributed to the elevated levels of METTL3 in AECII, as it promotes the stability and translation of Pten mRNA through m6A modification. This suggests that targeting METTL3 could offer a novel approach for treating ALI.


Sujet(s)
Lésion pulmonaire aigüe , Pneumocytes , Prolifération cellulaire , Methyltransferases , Souris de lignée C57BL , Phosphohydrolase PTEN , ARN messager , Animaux , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/génétique , Lésion pulmonaire aigüe/anatomopathologie , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Methyltransferases/métabolisme , Methyltransferases/génétique , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Pneumocytes/métabolisme , Pneumocytes/effets des médicaments et des substances chimiques , Pneumocytes/anatomopathologie , Mâle , ARN messager/métabolisme , Survie cellulaire/physiologie , Survie cellulaire/effets des médicaments et des substances chimiques , Méthylation , Adénosine/analogues et dérivés , Adénosine/métabolisme , Lipopolysaccharides/toxicité , Stabilité de l'ARN , Cellules cultivées
10.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(5): 514-519, 2024 May.
Article de Chinois | MEDLINE | ID: mdl-38845499

RÉSUMÉ

OBJECTIVE: To investigate the effect of mild hypothermia on macrophage polarization in lipopolysaccharide (LPS)-induced acute lung injury (ALI) mice and to clarify its role in lung injury. METHODS: According to a random number table method, 18 male C57BL/6 mice were divided into sham operation group (Sham group), ALI normothermic model group (NT group) and ALI mild hypothermia treatment group (HT group), with 6 mice in each group. The ALI model in mice was established by the method of tracheal instillation of LPS, and temperature control was administered at 1 hour after surgery. The anus temperature in NT group was kept at 36-38?centigrade, while the anus temperature in HT group was kept at 32-34?centigrade. The target anus temperature in both groups were maintained for 6 hours and then slowly rewarmed to 36-38 centigrade. The Sham group was infused with an equal amount of physiological saline through the trachea without temperature control. After 24 hours of modeling, serum was collected and mice were sacrificed to obtain lung tissue. Pathological changes in lung tissue were observed under light microscopy and semi-quantitative lung injury score was performed. Enzyme linked immunosorbent assay (ELISA) was used to detect the serum levels of interleukins (IL-1ß, IL-10). Real-time quantitative polymerase chain reaction (RT-qPCR) was used to test the indicators of macrophage polarization, such as the mRNA expressions of CD86, IL-6, CD206 and arginase 1 (Arg1) in the lung tissue. The protein expression of M1 macrophage marker inducible nitric oxide synthase (iNOS) and M2 macrophage marker Arg1 were detected by Western blotting. RESULTS: Compared with the Sham group, the NT group appeared significant pulmonary hemorrhage and edema, thickened lung septum, inflammatory cell infiltration, and lung injury score was significantly increased; serum IL-1ß level was significantly elevated; IL-10 level was increased without statistical significance; the expressions of CD86 mRNA, IL-6 mRNA and iNOS protein were significantly elevated, and CD206 mRNA was significantly decreased; the mRNA and protein expressions of Arg1 decreased, but there were no significant differences. Compared with the NT group, the pathological injury of lung tissue in HT group was significantly reduced, and the lung injury score was significantly decreased (4.78±0.96 vs. 8.56±1.98, P < 0.01); serum IL-1ß level was decreased (ng/L: 13.52±1.95 vs. 27.18±3.87, P < 0.01), and IL-10 level was significantly increased (ng/L: 42.59±15.79 vs. 14.62±4.47, P < 0.01); IL-6 mRNA expression was decreased in lung tissue (2-ΔΔCt: 3.37±0.92 vs. 10.04±0.91, P < 0.05), the expression of M1 macrophage markers CD86 mRNA and iNOS protein were significantly decreased [CD86 mRNA (2-ΔΔCt): 0.52±0.16 vs. 1.95±0.33, iNOS protein (iNOS/ß-actin): 0.57±0.19 vs. 1.11±0.27, both P < 0.05], the expression of M2 macrophage markers CD206 mRNA, Arg1 mRNA and Arg1 protein were significantly increased [CD206 mRNA (2-ΔΔCt): 3.99±0.17 vs. 0.34±0.17, Arg1 mRNA (2-ΔΔCt): 2.33±0.73 vs. 0.94±0.23, Arg1 protein (Arg1/ß-actin): 0.96±0.09 vs. 0.31±0.11, all P < 0.05]. CONCLUSIONS: Mild hypothermia can alleviate the inflammatory response and protect lung tissue in ALI mice, which may be related to the inhibition of M1 macrophage polarization and promotion of M2 macrophage polarization.


Sujet(s)
Lésion pulmonaire aigüe , Lipopolysaccharides , Macrophages , Souris de lignée C57BL , Animaux , Lésion pulmonaire aigüe/thérapie , Mâle , Souris , Macrophages/métabolisme , Lipopolysaccharides/effets indésirables , Nitric oxide synthase type II/métabolisme , Interleukine-10/métabolisme , Interleukine-6/métabolisme , Hypothermie provoquée , Interleukine-1 bêta/métabolisme , Modèles animaux de maladie humaine
11.
Molecules ; 29(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38930884

RÉSUMÉ

Acute lung injury (ALI) remains a significant global health issue, necessitating novel therapeutic interventions. In our latest study, we pioneered the use of D-mannitol-cerium-quercetin/rutin coordination polymer nanoparticles (MCQ/R NPs) as a potential treatment for ALI. The MCQ/R NPs, which integrate rutin and quercetin for their therapeutic potential and D-mannitol for its pulmonary targeting, displayed exceptional efficacy. By utilizing cerium ions for optimal nanoparticle assembly, the MCQ/R NPs demonstrated an average size of less than 160 nm. Impressively, these nanoparticles outperformed conventional treatments in both antioxidative capabilities and biocompatibility. Moreover, our in vivo studies on LPS-induced ALI mice showed a significant reduction in lung tissue inflammation. This groundbreaking research presents MCQ/R NPs as a promising new approach in ALI therapeutics.


Sujet(s)
Lésion pulmonaire aigüe , Cérium , Mannitol , Nanoparticules , Polymères , Quercétine , Lésion pulmonaire aigüe/traitement médicamenteux , Quercétine/pharmacologie , Quercétine/composition chimique , Animaux , Mannitol/composition chimique , Mannitol/usage thérapeutique , Nanoparticules/composition chimique , Souris , Polymères/composition chimique , Cérium/composition chimique , Cérium/pharmacologie , Cérium/usage thérapeutique , Rutoside/composition chimique , Rutoside/pharmacologie , Rutoside/usage thérapeutique , Antioxydants/pharmacologie , Antioxydants/composition chimique , Humains , Synergie des médicaments , Modèles animaux de maladie humaine , Lipopolysaccharides
12.
PLoS One ; 19(6): e0302721, 2024.
Article de Anglais | MEDLINE | ID: mdl-38935660

RÉSUMÉ

OBJECTIVE: To investigate the therapeutic effect and mechanism of sivelestat sodium on acute lung injury (AIL). METHODS: A rat model for ALI/acute respiratory distress syndrome (ALI/ARDS) was established. Pathological examination of lung tissue was conducted to assess lung injury. Blood gas in the arteries was measured using a blood analyzer. Changes in PaO2, PaO2/FiO2, and lung wet/dry (W/D) weight ratio were carefully compared. ELISA assay was conducted to estimate cell adhesion and inflammation response. Finally, real-time reverse transcription polymerase chain reaction and western blotting assay was used to determine the activation of PI3K/AKT/mTOR pathway. RESULTS: ARDS in vivo model was successfully constructed by LPS injection. Compared with the sham group, PaO2 and PaO2/FiO2 were significantly lower in the vehicle group, while the lung W/D ratio, the lung injury score, NE, VCAM-1, IL-8 andTNF-αwere significantly increased. After treatment with different doses of sivelestat sodium, we found PaO2, PaO2/FiO2 were prominently increased, while the lung W/D ratio, the lung injury score, NE, VCAM-1, IL-8, TNF-α levels were decreased in the dose-dependent manner. Meanwhile, compared with the vehicle group, the expression levels of Bax, PI3K, Akt and mTOR were significantly lower, and the expression of Bcl-2 was significantly higher after injection with sivelestat sodium. CONCLUSION: Sivelestat sodium has an interventional effect on ALI in sepsis by inhibiting the PI3K/AKT/mTOR signalling pathway.


Sujet(s)
Lésion pulmonaire aigüe , Glycine , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Rat Sprague-Dawley , Transduction du signal , Sulfonamides , Sérine-thréonine kinases TOR , Animaux , Sérine-thréonine kinases TOR/métabolisme , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Rats , Mâle , Glycine/analogues et dérivés , Glycine/pharmacologie , Glycine/usage thérapeutique , Sulfonamides/pharmacologie , Sulfonamides/usage thérapeutique , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/anatomopathologie , Modèles animaux de maladie humaine
13.
Ann Med ; 56(1): 2362871, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38902986

RÉSUMÉ

The lung is an important site of extramedullary platelet formation, and megakaryocytes in the lung participate in immune responses in addition to platelet production. In acute lung injury and chronic lung injury, megakaryocytes and platelets play a promoting or protective role through different mechanisms. The authors reviewed the role of megakaryocytes and platelets in common clinical lung injuries with different course of disease and different pathogenic factors in order to provide new thinking for the diagnosis and treatment of lung injuries.


What is the context?Platelets are specialized non-nucleated blood cells produced by cytoplasmic lysis of megakaryocytes.HSCs differentiate into granular mature megakaryocytes and produce platelets.Lung is a reservoir of megakaryocytes and a site where platelets are produced in addition to bone marrow and spleen.Lung injury can be divided into acute lung injury and chronic lung injury, and characterized by different pathogenesis.Platelets and megakaryocytes are involved in hemostasis and regulation of the body 's inflammatory response.The disease state of the lung affects the functions of megakaryocytes and platelets.The role of megakaryocytes and platelets in acute and chronic lung injury is poorly studied.What is new?Platelets in the lung are derived not only from the spleen and bone marrow, but also from megakaryocytes in the pulmonary circulation. In this study, we demonstrated that pulmonary megakaryocytes not only produce platelets to play a hemostatic role in lung injury, but also participate in inflammation and immune response with platelets to promote the process of lung injury or play a protective role.Therefore, it was suggested in our analysis that targeting lung megakaryocytes and platelets is currently a new direction for the treatment of a variety of lung injuries.What is the impact?This review intends to explain the relationship between megakaryocytes, platelets and many types of lung injury from the mechanism of platelet production in the lung, and make a prospect in the new progress in the diagnosis and treatment of lung injury.


Sujet(s)
Lésion pulmonaire aigüe , Plaquettes , Mégacaryocytes , Humains , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire , Poumon/anatomopathologie , Animaux , /anatomopathologie , /immunologie
14.
Phytomedicine ; 130: 155738, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38824825

RÉSUMÉ

BACKGROUND: Respiratory diseases pose a grave threat to human life. Therefore, understanding their pathogenesis and therapeutic strategy is important. Ferroptosis is a novel type of iron-dependent programmed cell death, distinct from apoptosis, necroptosis, and autophagy, characterised by iron, reactive oxygen species, and lipid peroxide accumulation, as well as glutathione (GSH) depletion and GSH peroxidase 4 (GPX4) inactivation. A close association between ferroptosis and the onset and progression of respiratory diseases, including chronic obstructive pulmonary disease, acute lung injury, bronchial asthma, pulmonary fibrosis, and lung cancer, has been reported. Recent studies have shown that traditional Chinese medicine (TCM) compounds exhibit unique advantages in the treatment of respiratory diseases owing to their natural properties and potential efficacy. These compounds can effectively regulate ferroptosis by modulating several key signalling pathways such as system Xc- -GSH-GPX4, NCOA4-mediated ferritinophagy, Nrf2-GPX4, and Nrf2/HO-1, thus playing a positive role in improving respiratory diseases. PURPOSE: This comprehensive review systematically outlines the regulatory role of ferroptosis in the onset and progression of respiratory diseases and provides evidence for treating respiratory diseases by targeting ferroptosis with TCM compounds. These insights aim to offer potential remedies for the clinical prevention and treatment of respiratory diseases. STUDY DESIGN AND METHODS: We searched scientific databases PubMed, Web of Science, Scopus, and CNKI using keywords such as "ferroptosis","respiratory diseases","chronic obstructive pulmonary disease","bronchial asthma","acute lung injury","pulmonary fibrosis","lung cancer","traditional Chinese medicine","traditional Chinese medicine compound","monomer", and "natural product" to retrieve studies on the therapeutic potential of TCM compounds in ameliorating respiratory diseases by targeting ferroptosis. The retrieved data followed PRISMA criteria (preferred reporting items for systematic review). RESULTS: TCM compounds possess unique advantages in treating respiratory diseases, stemming from their natural origins and proven clinical effectiveness. TCM compounds can exert therapeutic effects on respiratory diseases by regulating ferroptosis, which mainly involves modulation of pathways such as system Xc- -GSH-GPX4,NCOA4-mediated ferritinophagy, Nrf2-GPX4, and Nrf2/HO-1. CONCLUSION: TCM compounds have demonstrated promising potential in improving respiratory diseases through the regulation of ferroptosis. The identification of specific TCM-related inducers and inhibitors of ferroptosis holds great significance in developing more effective strategies. However, current research remains confined to animal and cellular studies, emphasizing the imperative for further verifications through high-quality clinical data.


Sujet(s)
Médicaments issus de plantes chinoises , Ferroptose , Ferroptose/effets des médicaments et des substances chimiques , Humains , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Lésion pulmonaire aigüe/traitement médicamenteux , Médecine traditionnelle chinoise/méthodes , Maladies de l'appareil respiratoire/traitement médicamenteux , Espèces réactives de l'oxygène/métabolisme , Fibrose pulmonaire/traitement médicamenteux
15.
Nutrients ; 16(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38892531

RÉSUMÉ

Propolis has potential anti-inflammatory properties, but little is known about its efficacy against inflammatory reactions caused by drug-resistant bacteria, and the difference in efficacy between propolis and tree gum is also unclear. Here, an in vivo study was performed to study the effects of ethanol extract from poplar propolis (EEP) and poplar tree gum (EEG) against heat-inactivated methicillin-resistant Staphylococcus aureus (MRSA)-induced acute lung injury (ALI) in mice. Pre-treatment with EEP and EEG (100 mg/kg, p.o.) resulted in significant protective effects on ALI in mice, and EEP exerted stronger activity to alleviate lung tissue lesions and ALI scores compared with that of EEG. Furthermore, EEP significantly suppressed the levels of pro-inflammatory mediators in the lung, including TNF-α, IL-1ß, IL-6, and IFN-γ. Gut microbiota analysis revealed that both EEP and EEG could modulate the composition of the gut microbiota, enhance the abundance of beneficial microbiota and reduce the harmful ones, and partly restore the levels of short-chain fatty acids. EEP could modulate more serum metabolites and showed a more robust correlation between serum metabolites and gut microbiota. Overall, these results support the anti-inflammatory effects of propolis in the treatment of ALI, and the necessity of the quality control of propolis.


Sujet(s)
Lésion pulmonaire aigüe , Microbiome gastro-intestinal , Médiateurs de l'inflammation , Staphylococcus aureus résistant à la méticilline , Propolis , Propolis/pharmacologie , Animaux , Staphylococcus aureus résistant à la méticilline/effets des médicaments et des substances chimiques , Lésion pulmonaire aigüe/microbiologie , Lésion pulmonaire aigüe/traitement médicamenteux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Souris , Mâle , Médiateurs de l'inflammation/sang , Médiateurs de l'inflammation/métabolisme , Anti-inflammatoires/pharmacologie , Infections à staphylocoques/traitement médicamenteux , Cytokines/sang , Cytokines/métabolisme , Température élevée , Modèles animaux de maladie humaine
16.
J Nanobiotechnology ; 22(1): 321, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38849841

RÉSUMÉ

Acute lung injury (ALI) is a life threatening disease in critically ill patients, and characterized by excessive reactive oxygen species (ROS) and inflammatory factors levels in the lung. Multiple evidences suggest that nanozyme with diversified catalytic capabilities plays a vital role in this fatal lung injury. At present, we developed a novel class of polydopamine (PDA) coated cerium dioxide (CeO2) nanozyme (Ce@P) that acts as the potent ROS scavenger for scavenging intracellular ROS and suppressing inflammatory responses against ALI. Herein, we aimed to identify that Ce@P combining with NIR irradiation could further strengthen its ROS scavenging capacity. Specifically, NIR triggered Ce@P exhibited the most potent antioxidant and anti-inflammatory behaviors in lipopolysaccharide (LPS) induced macrophages through decreasing the intracellular ROS levels, down-regulating the levels of TNF-α, IL-1ß and IL-6, up-regulating the level of antioxidant cytokine (SOD-2), inducing M2 directional polarization (CD206 up-regulation), and increasing the expression level of HSP70. Besides, we performed intravenous (IV) injection of Ce@P in LPS induced ALI rat model, and found that it significantly accumulated in the lung tissue for 6 h after injection. It was also observed that Ce@P + NIR presented the superior behaviors of decreasing lung inflammation, alleviating diffuse alveolar damage, as well as promoting lung tissue repair. All in all, it has developed the strategy of using Ce@P combining with NIR irradiation for the synergistic enhanced treatment of ALI, which can serve as a promising therapeutic strategy for the clinical treatment of ROS derived diseases as well.


Sujet(s)
Lésion pulmonaire aigüe , Cérium , Indoles , Polymères , Espèces réactives de l'oxygène , Cérium/composition chimique , Cérium/pharmacologie , Animaux , Lésion pulmonaire aigüe/traitement médicamenteux , Polymères/composition chimique , Polymères/pharmacologie , Indoles/composition chimique , Indoles/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Rats , Souris , Mâle , Cellules RAW 264.7 , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Antioxydants/pharmacologie , Antioxydants/composition chimique , Rat Sprague-Dawley , Lipopolysaccharides/pharmacologie , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Rayons infrarouges , Piégeurs de radicaux libres/pharmacologie , Piégeurs de radicaux libres/composition chimique , Piégeurs de radicaux libres/usage thérapeutique , Nanoparticules/composition chimique , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Cytokines/métabolisme
17.
J Clin Invest ; 134(11)2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38828725

RÉSUMÉ

Although antibody-mediated lung damage is a major factor in transfusion-related acute lung injury (ALI), autoimmune lung disease (for example, coatomer subunit α [COPA] syndrome), and primary graft dysfunction following lung transplantation, the mechanism by which antigen-antibody complexes activate complement to induce lung damage remains unclear. In this issue of the JCI, Cleary and colleagues utilized several approaches to demonstrate that IgG forms hexamers with MHC class I alloantibodies. This hexamerization served as a key pathophysiological mechanism in alloimmune lung injury models and was mediated through the classical pathway of complement activation. Additionally, the authors provided avenues for exploring therapeutics for this currently hard-to-treat clinical entity that has several etiologies but a potentially focused mechanism.


Sujet(s)
Lésion pulmonaire aigüe , Activation du complément , Immunoglobuline G , Humains , Immunoglobuline G/immunologie , Lésion pulmonaire aigüe/immunologie , Lésion pulmonaire aigüe/anatomopathologie , Activation du complément/immunologie , Animaux , Alloanticorps/immunologie , Multimérisation de protéines/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Complexe antigène-anticorps/immunologie
18.
Int J Mol Med ; 54(1)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38874017

RÉSUMÉ

In paraquat (PQ)­induced acute lung injury (ALI)/ acute respiratory distress syndrome, PQ disrupts endothelial cell function and vascular integrity, which leads to increased pulmonary leakage. Anthrahydroquinone­2,6­disulfonate (AH2QDS) is a reducing agent that attenuates the extent of renal injury and improves survival in PQ­intoxicated Sprague­Dawley (SD) rats. The present study aimed to explore the beneficial role of AH2QDS in PQ­induced ALI and its related mechanisms. A PQ­intoxicated ALI model was established using PQ gavage in SD rats. Human pulmonary microvascular endothelial cells (HPMECs) were challenged with PQ. Superoxide dismutase, malondialdehyde, reactive oxygen species and nitric oxide (NO) fluorescence were examined to detect the level of oxidative stress in HPMECs. The levels of TNF­α, IL­1ß and IL­6 were assessed using an ELISA. Transwell and Cell Counting Kit­8 assays were performed to detect the migration and proliferation of the cells. The pathological changes in lung tissues and blood vessels were examined by haematoxylin and eosin staining. Evans blue staining was used to detect pulmonary microvascular permeability. Western blotting was performed to detect target protein levels. Immunofluorescence and immunohistochemical staining were used to detect the expression levels of target proteins in HPMECs and lung tissues. AH2QDS inhibited inflammatory responses in lung tissues and HPMECs, and promoted the proliferation and migration of HPMECs. In addition, AH2QDS reduced pulmonary microvascular permeability by upregulating the levels of vascular endothelial­cadherin, zonula occludens­1 and CD31, thereby attenuating pathological changes in the lungs in rats. Finally, these effects may be related to the suppression of the phosphatidylinositol­3­kinase (PI3K)/protein kinase B (AKT)/endothelial­type NO synthase (eNOS) signalling pathway in endothelial cells. In conclusion, AH2QDS ameliorated PQ­induced ALI by improving alveolar endothelial barrier disruption via modulation of the PI3K/AKT/eNOS signalling pathway, which may be an effective candidate for the treatment of PQ­induced ALI.


Sujet(s)
Lésion pulmonaire aigüe , Perméabilité capillaire , Poumon , Nitric oxide synthase type III , Paraquat , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Rat Sprague-Dawley , Transduction du signal , Animaux , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Nitric oxide synthase type III/métabolisme , Perméabilité capillaire/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Humains , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Paraquat/effets indésirables , Paraquat/toxicité , Rats , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques
19.
Cell Death Dis ; 15(6): 425, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890311

RÉSUMÉ

Neutrophil reverse migration (rM) is a recently identified phenomenon in which neutrophils migrate away from the inflammatory site back into the vasculature following initial infiltration, which involved in the resolution of loci inflammatory response or dissemination of inflammation. Present study was aimed to explore the mechanisms in neutrophil rM. By scRNA-seq on the white blood cells in acute lung injury model, we found rM-ed neutrophils exhibited increased gene expression of C-C motif chemokine receptor-like 2 (Ccrl2), an atypical chemokine receptor. Furthermore, an air pouch model was established to directly track rM-ed neutrophils in vivo. Air pouches were generated by 3 ml filtered sterile air injected subcutaneously for 3 days, and then LPS (2 mg/kg) was injected into the pouches to mimic the inflammatory state. For the rM-ed neutrophil tracking system, cell tracker CMFDA were injected into the air pouch to stain the inflammatory loci cells, and after 6 h, stained cells in blood were regarded as the rM-ed neutrophil. Based on this tracking system, we confirmed that rM-ed neutrophils showed increased CCRL2. We also found that the concentrations of the CCRL2 ligand chemerin in plasma was increased in the late stage. Neutralizing chemerin decreased the rM-ed neutrophil ratio in the blood. These results suggest that circulating chemerin attracts neutrophils to leave inflammatory sites by interacting with CCRL2, which might involve in the dissemination of inflammation.


Sujet(s)
Mouvement cellulaire , Chimiokines , Protéines et peptides de signalisation intercellulaire , Granulocytes neutrophiles , Granulocytes neutrophiles/métabolisme , Chimiokines/métabolisme , Animaux , Souris , Protéines et peptides de signalisation intercellulaire/métabolisme , Souris de lignée C57BL , Mâle , Humains , Récepteurs CCR/métabolisme , Inflammation/anatomopathologie , Inflammation/métabolisme , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/anatomopathologie
20.
J Nanobiotechnology ; 22(1): 362, 2024 Jun 23.
Article de Anglais | MEDLINE | ID: mdl-38910259

RÉSUMÉ

Patients who suffer from sepsis typically experience acute lung injury (ALI). Extracellular vesicles (EVs) contain miRNAs, which are potentially involved in ALI. However, strategies to screen more effective EV-miRNAs as therapeutic targets are yet to be elucidated. In this study, functional EV-miRNAs were identified based on multiomics analysis of single-cell RNA sequencing of targeted organs and serum EV (sEV) miRNA profiles in patients with sepsis. The proportions of neutrophils and macrophages were increased significantly in the lungs of mice receiving sEVs from patients with sepsis compared with healthy controls. Macrophages released more EVs than neutrophils. MiR-125a-5p delivery by sEVs to lung macrophages inhibited Tnfaip3, while miR-221-3p delivery to lung neutrophils inhibited Fos. Macrophage membrane nanoparticles (MM NPs) loaded with an miR-125a-5p inhibitor or miR-221-3p mimic attenuated the response to lipopolysaccharide (LPS)-induced ALI. Transcriptome profiling revealed that EVs derived from LPS-stimulated bone marrow-derived macrophages (BMDMs) induced oxidative stress in neutrophils. Blocking toll-like receptor, CXCR2, or TNFα signaling in neutrophils attenuated the oxidative stress induced by LPS-stimulated BMDM-EVs. This study presents a novel method to screen functional EV-miRNAs and highlights the pivotal role of macrophage-derived EVs in ALI. MM NPs, as delivery systems of key sEV-miRNA mimics or inhibitors, alleviated cellular responses observed in sepsis-induced ALI. This strategy can be used to reduce septic organ damage, particularly lung damage, by targeting EVs.


Sujet(s)
Lésion pulmonaire aigüe , Vésicules extracellulaires , Macrophages , Souris de lignée C57BL , microARN , Nanoparticules , Sepsie , Animaux , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/traitement médicamenteux , Sepsie/métabolisme , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/composition chimique , microARN/métabolisme , Souris , Nanoparticules/composition chimique , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Humains , Mâle , Lipopolysaccharides , Granulocytes neutrophiles/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/anatomopathologie , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Multi-omique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...