Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Respir Res ; 25(1): 186, 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38678295

RESUMEN

BACKGROUND: Influenza A viruses (IAV) are extremely common respiratory viruses for the acute exacerbation of chronic obstructive pulmonary disease (AECOPD), in which IAV infection may further evoke abnormal macrophage polarization, amplify cytokine storms. Melatonin exerts potential effects of anti-inflammation and anti-IAV infection, while its effects on IAV infection-induced AECOPD are poorly understood. METHODS: COPD mice models were established through cigarette smoke exposure for consecutive 24 weeks, evaluated by the detection of lung function. AECOPD mice models were established through the intratracheal atomization of influenza A/H3N2 stocks in COPD mice, and were injected intraperitoneally with melatonin (Mel). Then, The polarization of alveolar macrophages (AMs) was assayed by flow cytometry of bronchoalveolar lavage (BAL) cells. In vitro, the effects of melatonin on macrophage polarization were analyzed in IAV-infected Cigarette smoking extract (CSE)-stimulated Raw264.7 macrophages. Moreover, the roles of the melatonin receptors (MTs) in regulating macrophage polarization and apoptosis were determined using MTs antagonist luzindole. RESULTS: The present results demonstrated that IAV/H3N2 infection deteriorated lung function (reduced FEV20,50/FVC), exacerbated lung damages in COPD mice with higher dual polarization of AMs. Melatonin therapy improved airflow limitation and lung damages of AECOPD mice by decreasing IAV nucleoprotein (IAV-NP) protein levels and the M1 polarization of pulmonary macrophages. Furthermore, in CSE-stimulated Raw264.7 cells, IAV infection further promoted the dual polarization of macrophages accompanied with decreased MT1 expression. Melatonin decreased STAT1 phosphorylation, the levels of M1 markers and IAV-NP via MTs reflected by the addition of luzindole. Recombinant IL-1ß attenuated the inhibitory effects of melatonin on IAV infection and STAT1-driven M1 polarization, while its converting enzyme inhibitor VX765 potentiated the inhibitory effects of melatonin on them. Moreover, melatonin inhibited IAV infection-induced apoptosis by suppressing IL-1ß/STAT1 signaling via MTs. CONCLUSIONS: These findings suggested that melatonin inhibited IAV infection, improved lung function and lung damages of AECOPD via suppressing IL-1ß/STAT1-driven macrophage M1 polarization and apoptosis in a MTs-dependent manner. Melatonin may be considered as a potential therapeutic agent for influenza virus infection-induced AECOPD.


Asunto(s)
Apoptosis , Subtipo H3N2 del Virus de la Influenza A , Melatonina , Enfermedad Pulmonar Obstructiva Crónica , Animales , Melatonina/farmacología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/virología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Ratones , Apoptosis/efectos de los fármacos , Células RAW 264.7 , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Ratones Endogámicos C57BL , Masculino , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Progresión de la Enfermedad , Polaridad Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología
2.
Oxid Med Cell Longev ; 2022: 2520348, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36425057

RESUMEN

Influenza virus infection is one of the strongest pathogenic factors for the development of acute lung injury (ALI)/ acute respiratory distress syndrome (ARDS). However, the underlying cellular and molecular mechanisms have not been clarified. In this study, we aim to investigate whether melatonin modulates macrophage polarization, oxidative stress, and pyroptosis via activating Apolipoprotein E/low-density lipoprotein receptor (ApoE/LDLR) pathway in influenza A-induced ALI. Here, wild-type (WT) and ApoE-/- mice were instilled intratracheally with influenza A (H3N2) and injected intraperitoneally with melatonin for 7 consecutive days. In vitro, WT and ApoE-/- murine bone marrow-derived macrophages (BMDMs) were pretreated with melatonin before H3N2 stimulation. The results showed that melatonin administration significantly attenuated H3N2-induced pulmonary damage, leukocyte infiltration, and edema; decreased the expression of proinflammatory M1 markers; enhanced anti-inflammatory M2 markers; and switched the polarization of alveolar macrophages (AMs) from M1 to M2 phenotype. Additionally, melatonin inhibited reactive oxygen species- (ROS-) mediated pyroptosis shown by downregulation of malonaldehyde (MDA) and ROS levels as well as inhibition of the NLRP3/GSDMD pathway and lactate dehydrogenase (LDH) release. Strikingly, the ApoE/LDLR pathway was activated when melatonin was applied in H3N2-infected macrophages and mice. ApoE knockout mostly abrogated the protective impacts of melatonin on H3N2-induced ALI and its regulatory ability on macrophage polarization, oxidative stress, and pyroptosis. Furthermore, recombinant ApoE3 (re-ApoE3) inhibited H3N2-induced M1 polarization of BMDMs with upregulation of MT1 and MT2 expression, but re-ApoE2 and re-ApoE4 failed to do this. Melatonin combined with re-ApoE3 played more beneficial protective effects on modulating macrophage polarization, oxidative stress, and pyroptosis in H3N2-infected ApoE-/- BMDMs. Our study indicated that melatonin attenuated influenza A- (H3N2-) induced ALI by inhibiting the M1 polarization of pulmonary macrophages and ROS-mediated pyroptosis via activating the ApoE/LDLR pathway. This study suggested that melatonin-ApoE/LDLR axis may serve as a novel therapeutic strategy for influenza virus-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda , Melatonina , Infecciones por Orthomyxoviridae , Animales , Ratones , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/virología , Apolipoproteína E3/farmacología , Apolipoproteínas E/metabolismo , Subtipo H3N2 del Virus de la Influenza A , Macrófagos/metabolismo , Melatonina/uso terapéutico , Ratones Noqueados para ApoE , Piroptosis , Especies Reactivas de Oxígeno/metabolismo , Infecciones por Orthomyxoviridae/tratamiento farmacológico
3.
Int Immunopharmacol ; 109: 108782, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35468366

RESUMEN

Acute lung injury (ALI)/ acute respiratory distress syndrome (ARDS) is featured by intensive inflammatory responses and oxidative stress, which lead to cytokine storms and pyroptosis. Here, we aimed to investigate whether melatonin was capable of alleviating LPS-induced ALI via activating the nuclear factor erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) signaling axis and inhibiting pyroptosis. Mice were injected with melatonin (30 mg/kg) intraperitoneally for consecutive five days before LPS instillation intratracheally, and human alveolar epithelial cell (AECⅡ) A549 cell lines and murine macrophages Raw264.7 cell lines were pretreated with melatonin (400 µM) before LPS (10 µg/ml) stimulation. The result demonstrated that LPS induced obvious lung injury characterized by alveolar damage, neutrophil infiltration and lung edema as well as the reduction of the survival rate of mice, which were totally reversed by melatonin pretreatment. Mechanistically, melatonin pretreatment activated nuclear factor erythroid2-related factor (Nrf) 2 signaling, subsequently, drove antioxidant pathways including significant increases in the expression of Nrf2, HO-1, NQO1, Mn-SOD and Catalase in vivo and in vitro. Simultaneously, melatonin inhibited ROS and MDA overproduction, iNOS expression as well as TNF-α and IL-1ß expression and release. Furthermore, melatonin inhibited LPS-induced pyroptosis by reversing the overexpression of NLRP3, Caspase-1, IL-1ß, IL-18 and GSDMD-N, as well as LDH release and TUNEL-positive cells in A549 cells and Raw264.7 cells. Overall, the current study suggests that melatonin exerts protective roles on LPS-induced ALI and pyroptosis by inhibiting NLRP3-GSDMD pathway via activating Nrf2/HO-1 signaling axis.


Asunto(s)
Lesión Pulmonar Aguda , Melatonina , Piroptosis , Síndrome de Dificultad Respiratoria , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Animales , Hemo-Oxigenasa 1/metabolismo , Lipopolisacáridos , Melatonina/farmacología , Melatonina/uso terapéutico , Proteínas de la Membrana/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/metabolismo
4.
J Inflamm Res ; 15: 2491-2507, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35479834

RESUMEN

Purpose: Emphysema is the main cause of the progression of chronic obstructive pulmonary disease (COPD). This study aimed to identify the key genes involved in COPD-related emphysema. Patients and Methods: GSE76925 was downloaded from Gene Expression Omnibus database. Protein-protein interaction networks of differentially expressed genes (DEGs) between control and COPD groups were constructed to identify hub genes using Cytoscape. Diagnostic performance of hub genes was evaluated using receiver operating characteristic analysis. Correlation analysis was performed to identify the key genes by analyzing the relationship between the hub genes and lung function and computed tomography (CT) indexes of emphysema. COPD patients were then divided into two groups based on the median expression of key genes and DEGs between these two groups were identified. Enrichment analysis of DEGs and correlation analysis between key genes and the infiltration of the immune cells were also analyzed. Finally, the role of key genes was evaluated in a lung tissues dataset (GSE47460) and a blood dataset (GSE76705). Additionally, the expression of key genes was validated by quantitative real-time polymerase chain reaction and immunohistochemistry. Results: CD19 and POU2AF1 had diagnostic efficacy for COPD and were significantly correlated with lung function and CT indexes of emphysema. Enrichment and immune analyses revealed that CD19 and POU2AF1 were correlated with the B cells in COPD. These results were consistent in GSE47460. The expression of CD19 and POU2AF1 in blood was the opposite of that in lung tissues, and CD19 and POU2AF1 were both significantly upregulated in COPD lung tissues at both the mRNA and protein levels. Conclusion: CD19 and POU2AF1 may serve as key regulators of emphysema and contribute to the progression of COPD by regulating the B-cell immunology. Targeting B cells may be a promising strategy for treating COPD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...