Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 457
Filtrer
1.
Biomed Environ Sci ; 37(6): 617-627, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38988112

RÉSUMÉ

Objective: The aim of this study was to explore the role and mechanism of ferroptosis in SiO 2-induced cardiac injury using a mouse model. Methods: Male C57BL/6 mice were intratracheally instilled with SiO 2 to create a silicosis model. Ferrostatin-1 (Fer-1) and deferoxamine (DFO) were used to suppress ferroptosis. Serum biomarkers, oxidative stress markers, histopathology, iron content, and the expression of ferroptosis-related proteins were assessed. Results: SiO 2 altered serum cardiac injury biomarkers, oxidative stress, iron accumulation, and ferroptosis markers in myocardial tissue. Fer-1 and DFO reduced lipid peroxidation and iron overload, and alleviated SiO 2-induced mitochondrial damage and myocardial injury. SiO 2 inhibited Nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant genes, while Fer-1 more potently reactivated Nrf2 compared to DFO. Conclusion: Iron overload-induced ferroptosis contributes to SiO 2-induced cardiac injury. Targeting ferroptosis by reducing iron accumulation or inhibiting lipid peroxidation protects against SiO 2 cardiotoxicity, potentially via modulation of the Nrf2 pathway.


Sujet(s)
Modèles animaux de maladie humaine , Ferroptose , Surcharge en fer , Souris de lignée C57BL , Myocytes cardiaques , Silice , Silicose , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Mâle , Souris , Surcharge en fer/métabolisme , Silice/toxicité , Silicose/métabolisme , Silicose/traitement médicamenteux , Silicose/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Déferoxamine/pharmacologie , Phénylènediamines/pharmacologie , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Stress oxydatif/effets des médicaments et des substances chimiques , Fer/métabolisme , Cyclohexylamines/pharmacologie
2.
Sci Adv ; 10(28): eadl4913, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38985878

RÉSUMÉ

The pathophysiology of silicosis is poorly understood, limiting development of therapies for those who have been exposed to the respirable particle. We explored mechanisms of silica-induced pulmonary fibrosis in human lung samples collected from patients with occupational exposure to silica and in a longitudinal mouse model of silicosis using multiple modalities including whole-lung single-cell RNA sequencing and histological, biochemical, and physiologic assessments. In addition to pulmonary inflammation and fibrosis, intratracheal silica challenge induced osteoclast-like differentiation of alveolar macrophages and recruited monocytes, driven by induction of the osteoclastogenic cytokine, receptor activator of nuclear factor κΒ ligand (RANKL) in pulmonary lymphocytes, and alveolar type II cells. Anti-RANKL monoclonal antibody treatment suppressed silica-induced osteoclast-like differentiation in the lung and attenuated pulmonary fibrosis. We conclude that silica induces differentiation of pulmonary osteoclast-like cells leading to progressive lung injury, likely due to sustained elaboration of bone-resorbing proteases and hydrochloric acid. Interrupting osteoclast-like differentiation may therefore constitute a promising avenue for moderating lung damage in silicosis.


Sujet(s)
Différenciation cellulaire , Ostéoclastes , Fibrose pulmonaire , Silice , Silicose , Silice/toxicité , Animaux , Humains , Ostéoclastes/métabolisme , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/anatomopathologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Souris , Silicose/anatomopathologie , Silicose/métabolisme , Silicose/étiologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Ligand de RANK/métabolisme , Modèles animaux de maladie humaine , Mâle , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Macrophages alvéolaires/métabolisme , Macrophages alvéolaires/anatomopathologie , Macrophages alvéolaires/effets des médicaments et des substances chimiques , Femelle
3.
Sci Total Environ ; 945: 174028, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38889818

RÉSUMÉ

Silicosis, recognized as a severe global public health issue, is an irreversible pulmonary fibrosis caused by the long-term inhalation of silica particles. Given the intricate pathogenesis of silicosis, there is no effective intervention measure, which poses a severe threat to public health. Our previous study reported that dysbiosis of lung microbiota is associated with the development of pulmonary fibrosis, potentially involving the lipopolysaccharides/toll-like receptor 4 pathway. Similarly, the process of pulmonary fibrosis is accompanied by alterations in metabolic pathways. This study employed a combined approach of 16S rDNA sequencing and metabolomic analysis to investigate further the role of lung microbiota in silicosis delving deeper into the potential pathogenesis of silicosis. Silica exposure can lead to dysbiosis of the lung microbiota and the occurrence of pulmonary fibrosis, which was alleviated by a combination antibiotic intervention. Additionally, significant metabolic disturbances were found in silicosis, involving 85 differential metabolites among the three groups, which are mainly focused on amino acid metabolic pathways. The changed lung metabolites showed a substantial correlation with lung microbiota. The relative abundance of Pseudomonas negatively correlated with L-Aspartic acid, L-Glutamic acid, and L-Threonine levels. These results indicate that dysbiosis in pulmonary microbiota exacerbates silica-induced fibrosis through impacts on amino acid metabolism, providing new insights into the potential mechanisms and interventions of silicosis.


Sujet(s)
Acides aminés , Poumon , Microbiote , Fibrose pulmonaire , Silice , Silicose , Microbiote/effets des médicaments et des substances chimiques , Poumon/microbiologie , Animaux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/microbiologie , Fibrose pulmonaire/métabolisme , Acides aminés/métabolisme , Silicose/métabolisme , Dysbiose/induit chimiquement , Mâle
4.
Respir Res ; 25(1): 242, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38877465

RÉSUMÉ

BACKGROUND: Silicosis represents a paramount occupational health hazard globally, with its incidence, morbidity, and mortality on an upward trajectory, posing substantial clinical dilemmas due to limited effective treatment options available. Trigonelline (Trig), a plant alkaloid extracted mainly from coffee and fenugreek, have diverse biological properties such as protecting dermal fibroblasts against ultraviolet radiation and has the potential to inhibit collagen synthesis. However, it's unclear whether Trig inhibits fibroblast activation to attenuate silicosis-induced pulmonary fibrosis is unclear. METHODS: To evaluate the therapeutic efficacy of Trig in the context of silicosis-related pulmonary fibrosis, a mouse model of silicosis was utilized. The investigation seeks to elucidated Trig's impact on the progression of silica-induced pulmonary fibrosis by evaluating protein expression, mRNA levels and employing Hematoxylin and Eosin (H&E), Masson's trichrome, and Sirius Red staining. Subsequently, we explored the mechanism underlying of its functions. RESULTS: In vivo experiment, Trig has been demonstrated the significant efficacy in mitigating SiO2-induced silicosis and BLM-induced pulmonary fibrosis, as evidenced by improved histochemical staining and reduced fibrotic marker expressions. Additionally, we showed that the differentiation of fibroblast to myofibroblast was imped in Trig + SiO2 group. In terms of mechanism, we obtained in vitro evidence that Trig inhibited fibroblast-to-myofibroblast differentiation by repressing TGF-ß/Smad signaling according to the in vitro evidence. Notably, our finding indicated that Trig seemed to be safe in mice and fibroblasts. CONCLUSION: In summary, Trig attenuated the severity of silicosis-related pulmonary fibrosis by alleviating the differentiation of myofibroblasts, indicating the development of novel therapeutic approaches for silicosis fibrosis.


Sujet(s)
Alcaloïdes , Différenciation cellulaire , Fibroblastes , Souris de lignée C57BL , Myofibroblastes , Fibrose pulmonaire , Silice , Silicose , Animaux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/prévention et contrôle , Alcaloïdes/pharmacologie , Silice/toxicité , Souris , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Silicose/anatomopathologie , Silicose/métabolisme , Silicose/traitement médicamenteux , Mâle
5.
Redox Biol ; 74: 103224, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38865904

RÉSUMÉ

BACKGROUND: Silicosis, characterized by interstitial lung inflammation and fibrosis, poses a significant health threat. ATII cells play a crucial role in alveolar epithelial repair and structural integrity maintenance. Inhibiting ATII cell senescence has shown promise in silicosis treatment. However, the mechanism behind silica-induced senescence remains elusive. METHODS: The study employed male C57BL/6 N mice and A549 human alveolar epithelial cells to investigate silicosis and its potential treatment. Silicosis was induced in mice via intratracheal instillation of crystalline silica particles, with honokiol administered intraperitoneally for 14 days. Silica-induced senescence in A549 cells was confirmed, and SIRT3 knockout and overexpression cell lines were generated. Various analyses were conducted, including immunoblotting, qRT-PCR, histology, and transmission electron microscopy. Statistical significance was determined using one-way ANOVA with Tukey's post-hoc test. RESULTS: This study elucidates how silica induces ATII cell senescence, emphasizing mtDNA damage. Notably, honokiol (HKL) emerges as a promising anti-senescence and anti-fibrosis agent, acting through sirt3. honokiol effectively attenuated senescence in ATII cells, dependent on sirt3 expression, while mitigating mtDNA damage. Sirt3, a class III histone deacetylase, regulates senescence and mitochondrial stress. HKL activates sirt3, protecting against pulmonary fibrosis and mitochondrial damage. Additionally, HKL downregulated cGAS expression in senescent ATII cells induced by silica, suggesting sirt3's role as an upstream regulator of the cGAS/STING signaling pathway. Moreover, honokiol treatment inhibited the activation of the NF-κB signaling pathway, associated with reduced oxidative stress and mtDNA damage. Notably, HKL enhanced the activity of SOD2, crucial for mitochondrial function, through sirt3-mediated deacetylation. Additionally, HKL promoted the deacetylation activity of sirt3, further safeguarding mtDNA integrity. CONCLUSIONS: This study uncovers a natural compound, HKL, with significant anti-fibrotic properties through activating sirt3, shedding light on silicosis pathogenesis and treatment avenues.


Sujet(s)
Pneumocytes , Dérivés du biphényle , Vieillissement de la cellule , Lignanes , Transduction du signal , Silicose , Sirtuine-3 , Animaux , Silicose/métabolisme , Silicose/traitement médicamenteux , Silicose/anatomopathologie , Silicose/étiologie , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Souris , Pneumocytes/métabolisme , Pneumocytes/effets des médicaments et des substances chimiques , Dérivés du biphényle/pharmacologie , Humains , Lignanes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Cellules A549 , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Modèles animaux de maladie humaine , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris de lignée C57BL , Altération de l'ADN/effets des médicaments et des substances chimiques , Composés allyliques , Phénols
6.
Int Immunopharmacol ; 136: 112368, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38823175

RÉSUMÉ

Silicosis is a chronic fibroproliferative lung disease caused by long-term inhalation of crystalline silica dust, characterized by the proliferation of fibroblasts and pulmonary interstitial fibrosis. Currently, there are no effective treatments available. Recent research suggests that the Integrin ß1/ILK/PI3K signaling pathway may be associated with the pathogenesis of silicosis fibrosis. In this study, we investigated the effects of Echistatin (Integrin ß1 inhibitor) and BYL-719 (PI3K inhibitor) on silicosis rats at 28 and 56 days after silica exposure. Histopathological analysis of rat lung tissue was performed using H&E staining and Masson staining. Immunohistochemistry, Western blotting, and qRT-PCR were employed to assess the expression of markers associated with epithelial-mesenchymal transition (EMT), fibrosis, and the Integrin ß1/ILK/PI3K pathway in lung tissue. The results showed that Echistatin, BYL 719 or their combination up-regulated the expression of E-cadherin and down-regulated the expression of Vimentin and extracellular matrix (ECM) components, including type I and type III collagen. The increase of Snail, AKT and ß-catenin in the downstream Integrin ß1/ILK/PI3K pathway was inhibited. These results indicate that Echistatin and BYL 719 can inhibit EMT and pulmonary fibrosis by blocking different stages of Integrinß1 /ILK/PI3K signaling pathway. This indicates that the Integrin ß1/ILK/PI3K signaling pathway is associated with silica-induced EMT and may serve as a potential therapeutic target for silicosis.


Sujet(s)
Transition épithélio-mésenchymateuse , Antigènes CD29 , Phosphatidylinositol 3-kinases , Protein-Serine-Threonine Kinases , Fibrose pulmonaire , Transduction du signal , Silice , Silicose , Animaux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Antigènes CD29/métabolisme , Antigènes CD29/génétique , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/anatomopathologie , Mâle , Silice/toxicité , Silicose/métabolisme , Silicose/anatomopathologie , Silicose/traitement médicamenteux , Phosphatidylinositol 3-kinases/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Rats , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Rat Sprague-Dawley
7.
Front Immunol ; 15: 1380628, 2024.
Article de Anglais | MEDLINE | ID: mdl-38774866

RÉSUMÉ

Introduction: TAM receptor-mediated efferocytosis plays an important function in immune regulation and may contribute to antigen tolerance in the lungs, a site with continuous cellular turnover and generation of apoptotic cells. Some studies have identified failures in efferocytosis as a common driver of inflammation and tissue destruction in lung diseases. Our study is the first to characterize the in vivo function of the TAM receptors, Axl and MerTk, in the innate immune cell compartment, cytokine and chemokine production, as well as the alveolar macrophage (AM) phenotype in different settings in the airways and lung parenchyma. Methods: We employed MerTk and Axl defective mice to induce acute silicosis by a single exposure to crystalline silica particles (20 mg/50 µL). Although both mRNA levels of Axl and MerTk receptors were constitutively expressed by lung cells and isolated AMs, we found that MerTk was critical for maintaining lung homeostasis, whereas Axl played a role in the regulation of silica-induced inflammation. Our findings imply that MerTk and Axl differently modulated inflammatory tone via AM and neutrophil recruitment, phenotype and function by flow cytometry, and TGF-ß and CXCL1 protein levels, respectively. Finally, Axl expression was upregulated in both MerTk-/- and WT AMs, confirming its importance during inflammation. Conclusion: This study provides strong evidence that MerTk and Axl are specialized to orchestrate apoptotic cell clearance across different circumstances and may have important implications for the understanding of pulmonary inflammatory disorders as well as for the development of new approaches to therapy.


Sujet(s)
Axl Receptor Tyrosine Kinase , Homéostasie , Poumon , Macrophages alvéolaires , Souris knockout , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Silicose , c-Mer Tyrosine kinase , Animaux , Souris , c-Mer Tyrosine kinase/métabolisme , c-Mer Tyrosine kinase/génétique , Cytokines/métabolisme , Modèles animaux de maladie humaine , Poumon/immunologie , Poumon/métabolisme , Poumon/anatomopathologie , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/métabolisme , Souris de lignée C57BL , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/génétique , Silicose/métabolisme , Silicose/immunologie , Silicose/anatomopathologie , Mâle
8.
BMC Pulm Med ; 24(1): 224, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38720270

RÉSUMÉ

BACKGROUND: Simvastatin (Sim), a hydroxy-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, has been widely used in prevention and treatment of cardiovascular diseases. Studies have suggested that Sim exerts anti-fibrotic effects by interfering fibroblast proliferation and collagen synthesis. This study was to determine whether Sim could alleviate silica-induced pulmonary fibrosis and explore the underlying mechanisms. METHODS: The rat model of silicosis was established by the tracheal perfusion method and treated with Sim (5 or 10 mg/kg), AICAR (an AMPK agonist), and apocynin (a NOX inhibitor) for 28 days. Lung tissues were collected for further analyses including pathological histology, inflammatory response, oxidative stress, epithelial mesenchymal transformation (EMT), and the AMPK-NOX pathway. RESULTS: Sim significantly reduced silica-induced pulmonary inflammation and fibrosis at 28 days after administration. Sim could reduce the levels of interleukin (IL)-1ß, IL-6, tumor necrosis factor-α and transforming growth factor-ß1 in lung tissues. The expressions of hydroxyproline, α-SMA and vimentin were down-regulated, while E-cad was increased in Sim-treated rats. In addition, NOX4, p22pox, p40phox, p-p47phox/p47phox expressions and ROS levels were all increased, whereas p-AMPK/AMPK was decreased in silica-induced rats. Sim or AICAR treatment could notably reverse the decrease of AMPK activity and increase of NOX activity induced by silica. Apocynin treatment exhibited similar protective effects to Sim, including down-regulating of oxidative stress and inhibition of the EMT process and inflammatory reactions. CONCLUSIONS: Sim attenuates silica-induced pulmonary inflammation and fibrosis by downregulating EMT and oxidative stress through the AMPK-NOX pathway.


Sujet(s)
AMP-Activated Protein Kinases , Fibrose pulmonaire , Silice , Simvastatine , Animaux , Mâle , Rats , Acétophénones/pharmacologie , 5-Amino-imidazole-4-carboxamide/analogues et dérivés , 5-Amino-imidazole-4-carboxamide/pharmacologie , AMP-Activated Protein Kinases/métabolisme , Modèles animaux de maladie humaine , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , NADPH Oxidase 4/métabolisme , NADPH oxidase/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Pneumopathie infectieuse/induit chimiquement , Pneumopathie infectieuse/prévention et contrôle , Pneumopathie infectieuse/traitement médicamenteux , Pneumopathie infectieuse/métabolisme , Pneumopathie infectieuse/anatomopathologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Ribonucléotides/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Silicose/traitement médicamenteux , Silicose/anatomopathologie , Silicose/métabolisme , Simvastatine/pharmacologie , Facteur de croissance transformant bêta-1/métabolisme
9.
Int Immunopharmacol ; 133: 112004, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38613881

RÉSUMÉ

Silicosis is a hazardous occupational disease caused by inhalation of silica, characterized by persistent lung inflammation that leads to fibrosis and subsequent lung dysfunction. Moreover, the complex pathophysiology of silicosis, the challenges associated with early detection, and the unfavorable prognosis contribute to the limited availability of treatment options. Daphnetin (DAP), a natural lactone, has demonstrated various pharmacological properties, including anti-inflammatory, anti-fibrotic, and pulmonary protective effects. However, the effects of DAP on silicosis and its molecular mechanisms remain uncover. This study aimed to evaluate the therapeutic effects of DAP against pulmonary inflammation and fibrosis using a silica-induced silicosis mouse model, and investigate the potential mechanisms and targets through network pharmacology, proteomics, molecular docking, and cellular thermal shift assay (CETSA). Here, we found that DAP significantly alleviated silica-induced lung injury in mice with silicosis. The results of H&E staining, Masson staining, and Sirius red staining indicated that DAP effectively reduced the inflammatory response and collagen deposition over a 28-day period following lung exposure to silica. Furthermore, DAP reduced the number of TUNEL-positive cells, increased the expression levels of Bcl-2, and decreased the expression of Bax and cleaved caspase-3 in the mice with silicosis. More importantly, DAP suppressed the expression levels of NLRP3 signaling pathway-related proteins, including NLRP3, ASC, and cleaved caspase-1, thereby inhibiting silica-induced lung inflammation. Further studies demonstrated that DAP possesses the ability to inhibit the epithelial mesenchymal transition (EMT) induced by silica through the inhibition of the TGF-ß1/Smad2/3 signaling pathway. The experimental results of proteomic analysis found that the PI3K/AKT1 signaling pathway was the key targets of DAP to alleviate lung injury induced by silica. DAP significantly inhibited the activation of the PI3K/AKT1 signaling pathway induced by silica in lung tissues. The conclusion was also verified by the results of molecular and CETSA. To further verify this conclusion, the activity of PI3K/AKT1 signaling pathway was inhibited in A549 cells using LY294002. When the A549 cells were pretreated with LY294002, the protective effect of DAP on silica-induced injury was lost. In conclusion, the results of this study suggest that DAP alleviates pulmonary inflammation and fibrosis induced by silica by modulating the PI3K/AKT1 signaling pathway, and holds promise as a potentially effective treatment for silicosis.


Sujet(s)
Souris de lignée C57BL , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Fibrose pulmonaire , Transduction du signal , Silice , Silicose , Ombelliférones , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Ombelliférones/pharmacologie , Ombelliférones/usage thérapeutique , Silicose/traitement médicamenteux , Silicose/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/induit chimiquement , Phosphatidylinositol 3-kinases/métabolisme , Souris , Humains , Pneumopathie infectieuse/traitement médicamenteux , Pneumopathie infectieuse/induit chimiquement , Pneumopathie infectieuse/anatomopathologie , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Mâle , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Simulation de docking moléculaire
10.
Mol Cell Proteomics ; 23(6): 100770, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38641226

RÉSUMÉ

Inhalation of crystalline silica dust induces incurable lung damage, silicosis, and pulmonary fibrosis. However, the mechanisms of the lung injury remain poorly understood, with limited therapeutic options aside from lung transplantation. Posttranslational modifications can regulate the function of proteins and play an important role in studying disease mechanisms. To investigate changes in posttranslational modifications of proteins in silicosis, combined quantitative proteome, acetylome, and succinylome analyses were performed with lung tissues from silica-injured and healthy mice using liquid chromatography-mass spectrometry. Combined analysis was applied to the three omics datasets to construct a protein landscape. The acetylation and succinylation of the key transcription factor STAT1 were found to play important roles in the silica-induced pathophysiological changes. Modulating the acetylation level of STAT1 with geranylgeranylacetone effectively inhibited the progression of silicosis. This report revealed a comprehensive landscape of posttranslational modifications in silica-injured mouse and presented a novel therapeutic strategy targeting the posttranslational level for silica-induced lung diseases.


Sujet(s)
Lysine , Maturation post-traductionnelle des protéines , Protéome , Facteur de transcription STAT-1 , Silicose , Animaux , Silicose/métabolisme , Silicose/traitement médicamenteux , Silicose/anatomopathologie , Facteur de transcription STAT-1/métabolisme , Protéome/métabolisme , Lysine/métabolisme , Acétylation/effets des médicaments et des substances chimiques , Souris , Silice , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Souris de lignée C57BL , Protéomique/méthodes , Mâle , Acide succinique/métabolisme
11.
Environ Toxicol ; 39(7): 3808-3819, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38523403

RÉSUMÉ

Silicon dioxide (SiO2)-induced pulmonary fibrosis is potentially associated with the impairment of mitochondrial function. Previous research found that inhibition of macrophage receptor with collagenous structure (MARCO) could alleviate particle-induced lung injury by regulating phagocytosis and mitigating mitochondrial damage. The present study aims to explore the underlying anti-fibrosis mechanism of polyguanylic acid (PolyG, MARCO inhibitor) in a silicotic rat model. Hematoxylin and eosin and Masson staining were performed to visualize lung tissue pathological changes. Confocal microscopy, transmission electron microscope, western blot analysis, quantitative real-time PCR (qPCR), and adenosine triphosphate (ATP) content assay were performed to evaluate collagen content, mitochondrial function, and morphology changes in SiO2-induced rat pulmonary fibrosis. The results suggested that SiO2 exposure contributed to reactive oxygen species aggregation and the reduction of respiratory complexes and ATP synthesis. PolyG treatment could effectively reduce MARCO expression and ameliorate lung injury and fibrosis by rectifying the imbalance of mitochondrial respiration and energy synthesis. Furthermore, PolyG could maintain mitochondrial homeostasis by promoting peroxisome proliferator-activated receptor-coactivator 1 α (PGC1α)-mediated mitochondrial biogenesis and regulating fusion and fission. Together, PolyG could ameliorate SiO2-induced pulmonary fibrosis via inhibiting MARCO to protect mitochondrial function.


Sujet(s)
Mitochondries , Fibrose pulmonaire , Silice , Silicose , Animaux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Silicose/traitement médicamenteux , Silicose/anatomopathologie , Silicose/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Silice/toxicité , Mâle , Rats , Rat Sprague-Dawley , Modèles animaux de maladie humaine , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Espèces réactives de l'oxygène/métabolisme
12.
Int J Biol Macromol ; 266(Pt 1): 131058, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38522707

RÉSUMÉ

Long-term exposure to inhalable silica particles may lead to severe systemic pulmonary disease, such as silicosis. Exosomes have been demonstrated to dominate the pathogenesis of silicosis, but the underlying mechanisms remain unclear. Therefore, this study aimed to explore the roles of exosomes by transmitting miR-107, which has been linked to the toxic pulmonary effects of silica particles. We found that miR-107, miR-122-5p, miR-125a-5p, miR-126-5p, and miR-335-5p were elevated in exosomes extracted from the serum of patients with silicosis. Notably, an increase in miR-107 in serum exosomes and lung tissue was observed in the experimental silicosis mouse model, while the inhibition of miR-107 reduced pulmonary fibrosis. Moreover, exosomes helped the migration of miR-107 from macrophages to lung fibroblasts, triggering the transdifferentiation of cell phenotypes. Further experiments demonstrated that miR-107 targets CDK6 and suppresses the expression of retinoblastoma protein phosphorylation and E2F1, resulting in cell-cycle arrest. Overall, micron-grade silica particles induced lung fibrosis through exosomal miR-107 negatively regulating the cell cycle signaling pathway. These findings may open a new avenue for understanding how silicosis is regulated by exosome-mediated cell-to-cell communication and suggest the prospect of exosomes as therapeutic targets.


Sujet(s)
Exosomes , microARN , Fibrose pulmonaire , Silice , Exosomes/métabolisme , Exosomes/génétique , microARN/génétique , microARN/métabolisme , Animaux , Silice/toxicité , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/génétique , Fibrose pulmonaire/anatomopathologie , Souris , Humains , Silicose/métabolisme , Silicose/anatomopathologie , Silicose/génétique , Silicose/étiologie , Communication cellulaire , Mâle , Modèles animaux de maladie humaine , Fibroblastes/métabolisme , Macrophages/métabolisme , Poumon/anatomopathologie , Poumon/métabolisme
13.
Biomolecules ; 14(2)2024 Jan 24.
Article de Anglais | MEDLINE | ID: mdl-38397383

RÉSUMÉ

Long-term silica particle exposure leads to interstitial pulmonary inflammation and fibrosis, called silicosis. Silica-activated macrophages secrete a wide range of cytokines resulting in persistent inflammation. In addition, silica-stimulated activation of fibroblast is another checkpoint in the progression of silicosis. The pathogenesis after silica exposure is complex, involving intercellular communication and intracellular signaling pathway transduction, which was ignored previously. Exosomes are noteworthy because of their crucial role in intercellular communication by delivering bioactive substances, such as lncRNA. However, the expression profile of exosomal lncRNA in silicosis has not been reported yet. In this study, exosomes were isolated from the peripheral serum of silicosis patients or healthy donors. The exosomal lncRNAs were profiled using high-throughput sequencing technology. Target genes were predicted, and functional annotation was performed using differentially expressed lncRNAs. Eight aberrant expressed exosomal lncRNAs were considered to play a key role in the process of silicosis according to the OPLS-DA. Furthermore, the increased expression of lncRNA MSTRG.43085.16 was testified in vitro. Its target gene PARP1 was critical in regulating apoptosis based on bioinformatics analysis. In addition, the effects of exosomes on macrophage apoptosis and fibroblast activation were checked based on a co-cultured system. Our findings suggested that upregulation of lncRNA MSTRG.43085.16 could regulate silica-induced macrophage apoptosis through elevating PARP1 expression, and promote fibroblast activation, implying that the exosomal lncRNA MSTRG.43085.16 might have potential as a biomarker for the early diagnosis of silicosis.


Sujet(s)
Exosomes , ARN long non codant , Silicose , Humains , Silice , ARN long non codant/génétique , ARN long non codant/métabolisme , Exosomes/génétique , Exosomes/métabolisme , Silicose/génétique , Silicose/métabolisme , Silicose/anatomopathologie , Macrophages/métabolisme , Fibroblastes/métabolisme , Apoptose/génétique
14.
Article de Chinois | MEDLINE | ID: mdl-38403426

RÉSUMÉ

Inhalation of crystalline silicon dioxide particles can induce silicosis, and the development of silicosis is closely related to the occurrence and development of pulmonary inflammation and pulmonary fibrosis. NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome has been established as a major proinflammatory receptor for sensing environmental danger signals. Activation of NLRP3 inflammasomes after phagocytosis of silicon dioxide particles by pulmonary macrophages may be an important mechanism to induce oxidative stress and sustained inflammatory response in the lung. This article summarizes the role of NLRP3 inflammasome in the inflammatory response and pulmonary fibrosis in silicosis, and analyzes it as a potential target for silicosis treatment.


Sujet(s)
Fibrose pulmonaire , Silicose , Humains , Inflammasomes/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Fibrose pulmonaire/métabolisme , Silicose/métabolisme , Silice , Fibrose
15.
Molecules ; 29(2)2024 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-38276616

RÉSUMÉ

Silicosis is a complex occupational disease without recognized effective treatment. Celastrol, a natural product, has shown antioxidant, anti-inflammatory, and anti-fibrotic activities, but the narrow therapeutic window and high toxicity severely limit its clinical application. Through structural optimization, we have identified a highly efficient and low-toxicity celastrol derivative, CEL-07. In this study, we systematically investigated the therapeutic potential and underlying mechanisms of CEL-07 in silicosis fibrosis. By constructing a silicosis mouse model and analyzing with HE, Masson, Sirius Red, and immunohistochemical staining, CEL-07 significantly prevented the progress of inflammation and fibrosis, and it effectively improved the lung respiratory function of silicosis mice. Additionally, CEL-07 markedly suppressed the expression of inflammatory factors (IL-6, IL-1α, TNF-α, and TNF-ß) and fibrotic factors (α-SMA, collagen I, and collagen III), and promoted apoptosis of fibroblasts by increasing ROS accumulation. Moreover, bioinformatics analysis combined with experimental validation revealed that CEL-07 inhibited the pathways associated with inflammation (PI3K-AKT and JAK2-STAT3) and the expression of apoptosis-related proteins. Overall, these results suggest that CEL-07 may serve as a potential candidate for the treatment of silicosis.


Sujet(s)
Triterpènes pentacycliques , Silice , Silicose , Souris , Animaux , Espèces réactives de l'oxygène/pharmacologie , Silice/pharmacologie , Phosphatidylinositol 3-kinases , Silicose/traitement médicamenteux , Silicose/métabolisme , Silicose/prévention et contrôle , Fibrose , Collagène/pharmacologie , Inflammation , Apoptose , Fibroblastes
16.
Sci Total Environ ; 912: 168948, 2024 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-38048996

RÉSUMÉ

The widespread manufacture of silica and its extensive use, and potential release of silica into the environment pose a serious human health hazard. Silicosis, a severe global public health issue, is caused by exposure to silica, leading to persistent inflammation and fibrosis of the lungs. The underlying pathogenic mechanisms of silicosis remain elusive. Lung microbiota dysbiosis is associated with the development of inflammation and fibrosis. However, limited information is currently available regarding the role of lung microbiota in silicosis. The study therefore is designed to conduct a comprehensive analysis of the role of lung microbiota dysbiosis and establish a basis for future investigations into the potential mechanisms underlying silicosis. Here, the pathological and biochemical parameters were used to systematically assessed the degree of inflammation and fibrosis following silica exposure and treatment with combined antibiotics. The underlying mechanisms were studied via integrative multi-omics analyses of the transcriptome and microbiome. Analysis of 16S ribosomal DNA revealed dysbiosis of the microbial community in silicosis, characterized by a predominance of gram-negative bacteria. Exposure to silica has been shown to trigger lung inflammation and fibrosis, leading to an increased concentration of lipopolysaccharides in the bronchoalveolar lavage fluid. Furthermore, Toll-like receptor 4 was identified as a key molecule in the lung microbiota dysbiosis associated with silica-induced lung fibrosis. All of these outcomes can be partially controlled through combined antibiotic administration. The study findings demonstrate that the dysbiosis of lung microbiota enhances silica-induced fibrosis associated with the lipopolysaccharides/Toll-like receptor 4 pathway and provided a promising target for therapeutic intervention of silicosis.


Sujet(s)
Microbiote , Fibrose pulmonaire , Silicose , Humains , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Silice/toxicité , Récepteur de type Toll-4 , Lipopolysaccharides , Dysbiose/induit chimiquement , Poumon/anatomopathologie , Silicose/génétique , Silicose/métabolisme , Silicose/anatomopathologie , Inflammation/induit chimiquement , Fibrose , Transduction du signal
17.
Ecotoxicol Environ Saf ; 269: 115767, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38039851

RÉSUMÉ

Inhaling silica causes the occupational illness silicosis, which mostly results in the gradual fibrosis of lung tissue. Previous research has demonstrated that hypoxia-inducible factor-1α (HIF-1α) and glycolysis-related genes are up-regulated in silicosis. The role of 2-deoxy-D-glucose (2-DG) as an inhibitor of glycolysis in silicosis mouse models and its molecular mechanisms remain unclear. Therefore, we used 2-DG to observe its effect on pulmonary inflammation and fibrosis in a silicosis mouse model. Furthermore, in vitro cell experiments were conducted to explore the specific mechanisms of HIF-1α. Our study found that 2-DG down-regulated HIF-1α levels in alveolar macrophages induced by silica exposure and reduced the interleukin-1ß (IL-1ß) level in pulmonary inflammation. Additionally, 2-DG reduced silica-induced pulmonary fibrosis. From these findings, we hypothesize that 2-DG reduced glucose transporter 1 (GLUT1) expression by inhibiting glycolysis, which inhibits the expression of HIF-1α and ultimately reduces transcription of the inflammatory cytokine, IL-1ß, thus alleviating lung damage. Therefore, we elucidated the important regulatory role of HIF-1α in an experimental silicosis model and the potential defense mechanisms of 2-DG. These results provide a possible effective strategy for 2-DG in the treatment of silicosis.


Sujet(s)
Pneumopathie infectieuse , Fibrose pulmonaire , Silicose , Animaux , Souris , Désoxyglucose/pharmacologie , Désoxyglucose/métabolisme , Glucose/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/effets des médicaments et des substances chimiques , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Inflammation/métabolisme , Macrophages alvéolaires , Pneumopathie infectieuse/métabolisme , Fibrose pulmonaire/métabolisme , Silice/toxicité , Silicose/traitement médicamenteux , Silicose/métabolisme
18.
Inflammation ; 47(1): 45-59, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37938462

RÉSUMÉ

Long-term exposure to silica dust can cause silicosis, which is characterized by chronic progressive inflammatory injury, fibroblast activation, and the deposition of extracellular matrix. IRF4 is involved in immune response. However, the potential regulation of IRF4 in silicosis and pulmonary fibrosis remains largely unexplored. In this study, RNA-seq analysis identified the upregulated expression of IRF4 in fibrotic lung tissues of mice exposed to silica particles. And we verified the increased expression of IRF4 in SiO2-treated macrophages and TGF-ß1-treated fibroblasts. We further found that the down-regulation of IRF4 impeded the macrophage polarization and the release of pro-fibrotic factors. Moreover, the down-regulation of IRF4 alleviated the migration, invasion, and the expression of fibrotic molecules in fibroblasts. Using ChIP-qPCR assay, we confirmed that IRF4 regulated the transcriptional activity of the IL-17A promoter, thus stimulated fibroblast activation, migration and invasion. In vivo experiment, the AAV-siIRF4 was designed to interfere with the expression of IRF4 in lung tissues of mice exposed to silica particles. Whole blood, bronchoalveolar lavage fluid and lung tissues were obtained from mice at 7, 14, 28 and 56 days after silica exposure. The results showed that the leukocyte content and inflammatory factors reached a peak at day 14 and remained peak for a long time after IRF4 knockdown. Furthermore, the fibrotic responses of mouse lung tissues were alleviated after IRF4 knockdown. Our study explored the important roles of IRF4 in inflammatory and fibrotic responses, which provided a new target for the treatment of silicosis and pulmonary fibrosis.


Sujet(s)
Fibrose pulmonaire , Silicose , Souris , Animaux , Fibrose pulmonaire/métabolisme , Silice/toxicité , Silice/métabolisme , Poumon/métabolisme , Silicose/métabolisme , Silicose/anatomopathologie , Inflammation/métabolisme , Fibrose , Macrophages/métabolisme , Fibroblastes/métabolisme , Souris de lignée C57BL
19.
J Hazard Mater ; 465: 133199, 2024 03 05.
Article de Anglais | MEDLINE | ID: mdl-38103296

RÉSUMÉ

Long term exposure to silica particles leads to various diseases, among which silicosis is of great concern. Silicosis is an interstitial lung disease caused by inhalation of silica particles in production environments. However, the mechanisms underlying silicosis remains unclear. Our previous studies revealed that progranulin (Pgrn) promoted the expression of pro-inflammatory factors in alveolar macrophages treated with silica particles and the secretion of extracellular matrix of pulmonary fibroblasts. Nevertheless, the role of Pgrn in silica particles-induced silicosis in vivo was unknown. This study found that silica particles increased Pgrn expression in silicosis patients. Pgrn deficiency reduced lung inflammation and fibrosis in silica particles-induced silicosis mouse models. Subsequently, based on transcriptional sequencing and interleukin (Il) -6 knockout mouse models, results demonstrated that Pgrn deficiency might decrease silicosis inflammation by reducing the production of Il-6, thereby modulating pulmonary fibrosis in the early stage of silicosis mouse models. Furthermore, another mechanism through which Pgrn deficiency reduced fibrosis in silicosis mouse models was the regulation of the transforming growth factor (Tgf) -ß1/Smad signaling pathway. Conclusively, Pgrn contributed to silicosis inflammation and fibrosis induced by silica particles, indicating that Pgrn could be a promising therapeutic target.


Sujet(s)
Pneumopathie infectieuse , Silicose , Animaux , Humains , Souris , Fibrose , Inflammation , Interleukine-6 , Progranulines/usage thérapeutique , Silice , Silicose/traitement médicamenteux , Silicose/étiologie , Silicose/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/usage thérapeutique
20.
J Appl Toxicol ; 44(1): 86-95, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37468209

RÉSUMÉ

Several epidemiologic and toxicological studies have widely regarded that mitochondrial dysfunction is a popular molecular event in the process of silicosis from different perspectives, but the details have not been systematically summarized yet. Thus, it is necessary to investigate how silica dust leads to pulmonary fibrosis by damaging the mitochondria of macrophages. In this review, we first introduce the molecular mechanisms that silica dust induce mitochondrial morphological and functional abnormalities and then introduce the main molecular mechanisms that silica-damaged mitochondria induce pulmonary fibrosis. Finally, we conclude that the mitochondrial abnormalities of alveolar macrophages caused by silica dust are involved deeply in the pathogenesis of silicosis through these two sequential mechanisms. Therefore, reducing the silica-damaged mitochondria will prevent the potential occurrence and fatality of the disease in the future.


Sujet(s)
Fibrose pulmonaire , Silicose , Humains , Fibrose pulmonaire/métabolisme , Silice/métabolisme , Macrophages , Silicose/métabolisme , Macrophages alvéolaires , Mitochondries , Poussière
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE