Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Heliyon ; 10(9): e30651, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38765063

ABSTRACT

Silicosis is a progressive pulmonary fibrosis disease caused by long-term inhalation of silica. The early diagnosis and timely implementation of intervention measures are crucial in preventing silicosis deterioration further. However, the lack of screening and diagnostic measures for early-stage silicosis remains a significant challenge. In this study, silicosis models of varying severity were established through a single exposure to silica with different doses (2.5mg/mice or 5mg/mice) and durations (4 weeks or 12 weeks). The diagnostic performance of computed tomography (CT) quantitative analysis was assessed using lung density biomarkers and the lung density distribution histogram, with a particular focus on non-aerated lung volume. Subsequently, we developed and evaluated a stacking learning model for early diagnosis of silicosis after extracting and selecting features from CT images. The CT quantitative analysis reveals that while the lung densitometric biomarkers and lung density distribution histogram, as traditional indicators, effectively differentiate severe fibrosis models, they are unable to distinguish early-stage silicosis. Furthermore, these findings remained consistent even when employing non-aerated areas, which is a more sensitive indicator. By establishing a radiomics stacking learning model based on non-aerated areas, we can achieve remarkable diagnostic performance to distinguish early-stage silicosis, which can provide a valuable tool for clinical assistant diagnosis. This study reveals the potential of using non-aerated lung areas as a region of interest in stacking learning for early diagnosis of silicosis, providing new insights into early detection of this disease.

2.
Sci Total Environ ; 912: 168948, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38048996

ABSTRACT

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.


Subject(s)
Microbiota , Pulmonary Fibrosis , Silicosis , Humans , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Silicon Dioxide/toxicity , Toll-Like Receptor 4 , Lipopolysaccharides , Dysbiosis/chemically induced , Lung/pathology , Silicosis/genetics , Silicosis/metabolism , Silicosis/pathology , Inflammation/chemically induced , Fibrosis , Signal Transduction
3.
Ecotoxicol Environ Saf ; 268: 115693, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37976936

ABSTRACT

Macrophage pyroptosis has recently been involved in some inflammatory and fibrosis diseases, however, the role of macrophage pyroptosis in silica-induced pulmonary fibrosis has not been fully elucidated. In this study, we explored the role of macrophage pyroptosis in silicosis in vivo and in vitro. A mouse model of silicosis was established and mice were sacrificed at 7, 14, and 28 days after exposure of silica. The results revealed that the expression of GSDMD and other pyroptosis-related indicators was up-regulated obviously at 14 days after silica exposure, indicating that silica induced pyroptosis in vivo. In vitro, human monocytic leukemia cells (THP-1) and human lung fibroblasts (MRC-5) were used to detect the relationship between macrophage pyroptosis and lung fibroblasts. It showed that silica increased the levels of GSDMD and other pyroptosis-related indicators remarkably in macrophages and the supernatant of macrophage stimulated by silica could promote the upregulation of fibrosis markers in fibroblasts. However, GSDMD knockdown suppressed silica-induced macrophage pyroptosis and alleviated the upregulation of fibrosis markers in fibroblasts, suggesting the important role of macrophage pyroptosis in the activation of myofibroblasts during the progression of silicosis. Taken together, it showed that silica could induce macrophage pyroptosis and inhibiting macrophage pyroptosis could be a feasible clinical strategy to alleviate silicosis.


Subject(s)
Pulmonary Fibrosis , Silicosis , Mice , Humans , Animals , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Silicon Dioxide/toxicity , Pyroptosis , Macrophages/metabolism , Silicosis/metabolism , Fibrosis
4.
Int J Mol Sci ; 24(19)2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37833927

ABSTRACT

Silicosis is a fatal occupational respiratory disease caused by the prolonged inhalation of respirable silica. The core event of silicosis is the heightened activity of fibroblasts, which excessively synthesize extracellular matrix (ECM) proteins. Our previous studies have highlighted that human umbilical cord mesenchymal stem cell-derived extracellular vesicles (hucMSC-EVs) hold promise in mitigating silicosis and the significant role played by microRNAs (miRNAs) in this process. Delving deeper into this mechanism, we found that miR-148a-3p was the most abundant miRNA of the differential miRNAs in hucMSC-EVs, with the gene heat shock protein 90 beta family member 1 (Hsp90b1) as a potential target. Notably, miR-148a-3p's expression was downregulated during the progression of silica-induced pulmonary fibrosis both in vitro and in vivo, but was restored after hucMSC-EVs treatment (p < 0.05). Introducing miR-148a-3p mimics effectively hindered the collagen synthesis and secretion of fibroblasts induced by transforming growth factor-ß1 (TGF-ß1) (p < 0.05). Confirming our hypothesis, Hsp90b1 was indeed targeted by miR-148a-3p, with significantly reduced collagen activity in TGF-ß1-treated fibroblasts upon Hsp90b1 inhibition (p < 0.05). Collectively, our findings provide compelling evidence that links miR-148a-3p present in hucMSC-EVs with the amelioration of silicosis, suggesting its therapeutic potential by specifically targeting Hsp90b1, thereby inhibiting fibroblast collagen activities. This study sheds light on the role of miR-148a-3p in hucMSC-EVs, opening avenues for innovative therapeutic interventions targeting molecular pathways in pulmonary fibrosis.


Subject(s)
Extracellular Vesicles , MicroRNAs , Pulmonary Fibrosis , Silicosis , Humans , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/therapy , Transforming Growth Factor beta1/metabolism , Silicon Dioxide/pharmacology , MicroRNAs/metabolism , Silicosis/genetics , Silicosis/therapy , Silicosis/pathology , Fibroblasts/metabolism , Collagen/pharmacology , Extracellular Vesicles/metabolism
5.
Ecotoxicol Environ Saf ; 257: 114950, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37099959

ABSTRACT

Silicosis is one of several potentially fatal occupational pathologies caused by the prolonged inhalation of respirable crystalline silica. Previous studies have shown that lung epithelial-mesenchymal transition (EMT) plays a significant role in the fibrosis effect of silicosis. Human umbilical cord mesenchymal stem cells-derived Extracellular vesicles (hucMSC-EVs) have attracted great interest as a potential therapy of EMT and fibrosis-related diseases. However, the potential effects of hucMSC-EVs in inhibiting EMT in silica-induced fibrosis, as well as its underlying mechanisms, remain largely unknown. In this study, we used the EMT model in MLE-12 cells and observed the effects and mechanism of hucMSC-EVs inhibition of EMT. The results revealed that hucMSC-EVs can indeed inhibit EMT. MiR-26a-5p was highly enriched in hucMSC-EVs but was down-regulated in silicosis mice. We found that miR-26a-5p in hucMSC-EVs was over-expressed after transfecting miR-26a-5p expressing lentivirus vectors into hucMSCs. Subsequently, we explored if miR-26a-5p, attained from hucMSC-EVs, was involved in inhibiting EMT in silica-induced lung fibrosis. Our findings suggested that hucMSC-EVs could deliver miR-26a-5p into MLE-12 cells and cause the inhibition of the Adam17/Notch signalling pathway to ameliorate EMT in silica-induced pulmonary fibrosis. These findings might represent a novel insight into treating silicosis fibrosis.


Subject(s)
Extracellular Vesicles , MicroRNAs , Pulmonary Fibrosis , Silicosis , Humans , Mice , Animals , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Epithelial-Mesenchymal Transition , Silicon Dioxide/toxicity , Fibrosis , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Silicosis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , ADAM17 Protein/genetics
6.
Ecotoxicol Environ Saf ; 251: 114537, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36646008

ABSTRACT

Silicosis is a progressive inflammatory disease with poorly defined mechanisms and limited therapeutic options. Recent studies found that microRNAs (miRNAs) and circular RNAs (circRNAs) were involved in the development of respiratory diseases; however, the function of non-coding RNAs in silicosis was still needed to be further explored. We found that miR-223-3p was significantly decreased in macrophages and lung tissues of mice after silica treatment, which were consistent with the results of GEO database microarray analysis. Notably, NLRP3 is a target gene downstream of miR-223-3p. And circular RNA PWWP2A (circPWWP2A) was significantly elevated after silica stimulation. To elucidate the role of these RNAs in silica-induced inflammation in macrophages and lung tissues, we investigated the upstream molecular mechanisms of circPWWP2A on the inflammatory response. The inhibitory effect of miR-223-3p on its target NLRP3 was suppressed by circPWWP2A, which led to lung fibrosis. Our study found that circPWWP2A could adsorb miR-223-3p to regulate NLRP3 after silica stimulation in pulmonary fibrosis. And our results revealed that the circPWWP2A-miR-223-3p-NLRP3 axis was potentially instrumental in managing silica-induced inflammation and fibrosis. Previous studies have demonstrated that human umbilical cord mesenchymal stem cell-derived extracellular vesicles (hucMSC-EVs) exhibit anti-inflammatory and anti-fibrotic effects in multiple organs. However, the potential effectiveness of hucMSC-EVs against silicosis or the underlying mechanisms of their biological outcomes remains unclear. Therefore, we used 3D culture technology to extract hucMSC-EVs and observed their effects in macrophages and lung tissues, respectively. According to the EVmiRNA database, miR-223-3p was abundant in MSC-EVs. In addition, hucMSC-EVs may modulate lung function, reduce the secretion of inflammatory factors (NLRP3, IL-1ß, IL-18 and cleaved Caspase-1) and attenuate the deposition of fibrosis-related factors (Collagen Ⅰ, Collagen Ⅲ, fibronectin and α-SMA). In vitro results evinced that hucMSC-EVs reduced the inflammatory response of macrophages and restricted the activation and proliferation of fibroblasts. Moreover, our study showed that hucMSCs-EVs acted as a mediator to transfer miR-223-3p to suppress circPWWP2A, thereby alleviating pulmonary fibrosis through the NLRP3 signaling pathway. These data may provide potentially novel strategies for investigating the pathogenesis of silicosis and developing novel treatments for this disease.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Pneumonia , Pulmonary Fibrosis , Silicosis , Humans , Mice , Animals , RNA, Circular/genetics , RNA, Circular/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/therapy , Silicon Dioxide/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Fibrosis , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Silicosis/genetics , Silicosis/therapy , Silicosis/metabolism , Pneumonia/metabolism , Immunologic Factors/metabolism , Extracellular Vesicles/metabolism , Umbilical Cord , Mesenchymal Stem Cells/metabolism
7.
Sci Total Environ ; 831: 154974, 2022 Jul 20.
Article in English | MEDLINE | ID: mdl-35378184

ABSTRACT

Airborne fine particulate matter (PM2.5) is considered to be a risk factor for lung fibrosis, and therefore, it has attracted public attention due to its various physicochemical features and its adverse effects on health. However, little remains to be known regarding the mechanism of PM2.5-induced pulmonary fibrosis. The lung microbiota may be a potential factor involved in the adverse outcomes of pulmonary fibrosis. Meanwhile, miRNAs are thought to be key regulators that participate in the complex interplay between the host and the microbiota. Hence, to investigate the potential mechanisms of pulmonary fibrosis, and to explore the impact of PM2.5-induced alterations in miRNAs and the lung microbiota and possible interaction patterns in mice models, we took advantage of 16S rDNA gene sequencing, miRNAs sequencing (miRNAs-Seq), and mining of public databases profiling. The results of 16S rDNA analysis showed that PM2.5 interfered with the microbial community composition, resulting in Proteobacteria becoming an additional dominant phylum. In addition, differentially expressed miRNAs were enriched in HIF-1 signaling, the IL-17 signaling, as well as Th17 cell differentiation pathways, which are closely related to microbial functional pathways. Significantly, a target miRNA, miR-149-5p, may be a key factor triggering the MAPK signal pathway related to pulmonary fibrosis and disturbing the homeostasis of lung bacterial flora. These results indicate that PM2.5 may lead to interaction between lung microbiota dysbiosis and an imbalance of miRNA levels to form a vicious cycle that promotes lung fibrogenesis. The current study provides new insights into the progression of pulmonary fibrosis.


Subject(s)
MicroRNAs , Microbiota , Pulmonary Fibrosis , Animals , DNA, Ribosomal , Lung/pathology , Mice , MicroRNAs/genetics , Particulate Matter/metabolism , Particulate Matter/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Transcriptome
8.
Ecotoxicol Environ Saf ; 233: 113302, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35189518

ABSTRACT

Silicosis of pulmonary fibrosis (PF) is related to long-term excessive inhalation of silica. The activation of fibroblasts into myofibroblasts is the main terminal effect leading to lung fibrosis, which is of great significance to the study of the occurrence and development of silicosis fibrosis and its prevention and treatment. Exosomes derived from human umbilical cord mesenchymal stem cells (hucMSC-Exos) are considered to be a potential therapy of silica-induced PF, however, their exact mechanism remains unknown. Therefore, this study aims to explore whether hucMSC-Exos affect the activation of fibroblasts to alleviate PF. In this study, a three-dimensional (3D) method was applied to culture hucMSCs and MRC-5 cells (human embryonic lung fibroblasts), and exosomes were isolated from serum-free media, identified by nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM) and Western blotting analysis. Then, the study used an animal model of silica-induced PF to observe the effects of hucMSC-Exos and MRC-5-Exos on activation of fibroblasts. In addition, the activation of fibroblasts was analyzed by Western blotting analysis, wound healing, and migration assay with the treatment of hucMSC-Exos and MRC-5-Exos in NIH-3T3 cells (mouse embryonic fibroblasts). Furthermore, differential expression of microRNAs (DE miRNAs) was measured between hucMSCs-Exos and MRC-5-Exos by high throughput sequence. HucMSC-Exos inhibited the activation of fibroblasts in mice and NIH-3T3 cells. Let-7i-5p was significantly up-regulated in hucMSCs-Exos compared to MRC-5-Exos, which was related to silica-induced PF. Let-7i-5p of hucMSCs-Exos was responsible for the activation of fibroblasts by targeting TGFBR1. Meanwhile, Smad3 was also an important role in the activation of fibroblasts. The study demonstrates that hucMSCs-Exos act as a mediator that transfers let-7i-5p to inhibit the activation of fibroblasts, which alleviates PF through the TGFBR1/Smad3 signaling pathway. The mechanism has potential value for the treatment of silica-induced PF.


Subject(s)
Mesenchymal Stem Cells , Silicosis , Animals , Fibroblasts , Humans , Mice , MicroRNAs , Receptor, Transforming Growth Factor-beta Type I/metabolism , Silicosis/metabolism , Umbilical Cord
9.
Environ Sci Pollut Res Int ; 29(27): 41567-41576, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35094267

ABSTRACT

The impact of PM2.5 on epithelial cells is a pivotal process leading to many lung pathological changes and pulmonary diseases. In addition to PM2.5 direct interaction with epithelia, macrophages that engulf PM2.5 may also influence the function of epithelial cells. However, among the toxic researches of PM2.5, there is a lack of evaluation of direct or indirect exposure model on human bronchial epithelial cell against PM2.5. In this present research, PM2.5-exposed human bronchial epithelial cell line (BEAS-2B) serves as the direct interaction model. By contrast, a PM2.5-stimulated co-culture model of macrophages and epithelial cells based on the transwell system was adopted as indirect stimulation model. By comparing these two models of interaction, we examined the viability of BEAS-2B and mRNA/protein expression profile of oxidative stress and inflammatory response-related transcription factors Nrf2, NF-kB, and according inflammatory indicators such as IL-1, IL-6, and IL-8, with a view to evaluating the effects of different interaction models of PM2.5 on epithelial cell damage in vitro. Our results indicated that under the same doses, the direct stimulation model of PM2.5 could inhibit the viability of BEAS-2B. Furthermore, the indirect stimulation model strengthen inflammation response of epithelia under the higher concentration of PM2.5 and induce epithelia to undergo EMT under the lower concentration of PM2.5. Overall, we have found that macrophage involvement may protect epithelia from PM2.5 cytotoxic effect, while it strengthens the inflammation response and induce epithelia to undergo EMT.


Subject(s)
Air Pollutants , Particulate Matter , Air Pollutants/analysis , Bronchi , Epithelial Cells , Humans , Inflammation/metabolism , Particulate Matter/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...