Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Int Immunopharmacol ; 133: 112067, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38608444

RESUMEN

Silicosis is one of the most common and severe types of pneumoconiosis and is characterized by lung dysfunction, persistent lung inflammation, pulmonary nodule formation, and irreversible pulmonary fibrosis. The transdifferentiation of fibroblasts into myofibroblasts is one of the main reasons for the exacerbation of silicosis. However, the underlying mechanism of transcription factors regulating silicosis fibrosis has not been clarified. The aim of this study was to investigate the potential mechanism of transcription factor FOXF1 in fibroblast transdifferentiation in silica-induced pulmonary fibrosis. Therefore, a silicosis mouse model was established, and we found that FOXF1 expression level was significantly down-regulated in the silicosis group, and after overexpression of FOXF1 by adeno-associated virus (AAV), FOXF1 expression level was up-regulated, and silicosis fibrosis was alleviated. In order to further explore the specific regulatory mechanism of FOXF1 in silicosis, we established a fibroblasts transdifferentiation model induced by TGF-ß in vitro. In the model, the expression levels of SMAD2/3 and P-SMAD2/3 were up-regulated, but the expression levels of SMAD2/3 and P-SMAD2/3 were down-regulated, inhibiting transdifferentiation and accumulation of extracellular matrix after the overexpressed FOXF1 plasmid was constructed. However, after silencing FOXF1, the expression levels of SMAD2/3 and P-SMAD2/3 were further up-regulated, aggravating transdifferentiation and accumulation of extracellular matrix. These results indicate that the activation of FOXF1 in fibroblasts can slow down the progression of silicosis fibrosis by inhibiting TGF-ß/SMAD2/3 classical pathway, which provides a new idea for further exploration of silicosis treatment.


Asunto(s)
Transdiferenciación Celular , Fibroblastos , Fibrosis Pulmonar , Transducción de Señal , Silicosis , Factor de Crecimiento Transformador beta , Animales , Humanos , Masculino , Ratones , Transdiferenciación Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Pulmón/patología , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Dióxido de Silicio , Silicosis/complicaciones , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
3.
Ecotoxicol Environ Saf ; 272: 116029, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38290312

RESUMEN

Manganese is essential trace elements, to participate in the body a variety of biochemical reactions, has important physiological functions, such as stimulate the immune cell proliferation, strengthen the cellular immunity, etc. However, excessive manganese exposure can cause damage to multiple systems of the body.The immune system is extremely vulnerable to external toxicants, however manganese research on the immune system are inadequate and biomarkers are lacking. Therefore, here we applied a manganese-exposed rat model to make preliminary observations on the immunotoxic effects of manganese. We found that manganese exposure inhibited humoral immune function in rats by decreasing peripheral blood IgG (ImmunoglobulinG, IgG), IgM (ImmunoglobulinM, IgM) and complement C3 levels; It also regulates rat cellular immune activity by influencing peripheral blood, spleen, and thymus T cell numbers and immune organ ICs (Immune Checkpoints, ICs) and cytokine expression. Furthermore, it was revealed that the impact of manganese exposure on the immune function of rats exhibited a correlation with both the dosage and duration of exposure. Notably, prolonged exposure to high doses of manganese had the most pronounced influence on rat immune function, primarily manifesting as immunosuppression.The above findings suggest that manganese exposure leads to impaired immune function and related changes in immune indicators, or may provide clues for the discovery of its biomarkers.


Asunto(s)
Manganeso , Linfocitos T , Ratas , Animales , Manganeso/toxicidad , Inmunoglobulina M , Inmunoglobulina G , Biomarcadores
4.
Front Public Health ; 11: 1289838, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38026392

RESUMEN

Mn (Manganese, Mn) is an essential trace element involved in various biological processes such as the regulation of immune, nervous and digestive system functions. However, excessive Mn exposure can lead to immune damage. Occupational workers in cement and ferroalloy manufacturing and other related industries are exposed to low levels of Mn for a long time. Mn exposure is one of the important occupational hazards, but the research on the effect of Mn on the immune system of the occupational population is not complete, and there is no reliable biomarker. Therefore, this study aimed to evaluate the immunotoxicity of Mn from the soluble immune checkpoint TIM-3 (T-cell immunoglobulin and mucin containing protein 3, TIM-3) and complement C3. A total of 144 Mn-exposed workers were recruited from a bus manufacturing company and a railroad company in Henan Province. An inductively coupled plasma mass spectrometer was used to detect the concentration of RBC Mn (Red blood cell Mn, RBC Mn), and ELISA kits were used to detect serum complement C3 and TIM-3. Finally, the subjects were statistically analyzed by dividing them into low and high Mn groups based on the median RBC Mn concentration. We found that Mn exposure resulted in elevated serum TIM-3 expression and decreased complement C3 expression in workers; that serum TIM-3 and complement C3 expression showed a dose-response relationship with RBC Mn; and that the mediating effect of complement C3 between RBC Mn and TIM-3 was found to be significant. The above findings indicate that this study has a preliminary understanding of the effect of Mn exposure on the immune system of the occupational population exposed to Mn, and complement C3 and TIM-3 may be biomarkers of Mn exposure, which may provide clues for the prevention and control of Mn occupational hazards.


Asunto(s)
Complemento C3 , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Manganeso/toxicidad , Biomarcadores
5.
Ecotoxicol Environ Saf ; 249: 114410, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36516619

RESUMEN

Silicosis, a disease characterized by diffuse fibrosis of the lung tissue, is caused by long-term inhalation of free silica (SiO2) dust in the occupational environment and is currently the most serious occupational diseases of pneumoconiosis. Several studies have suggested that alveolar type Ⅱ epithelial cells (AEC Ⅱ) undergo epithelial-mesenchymal transition (EMT) as one of the crucial components of silicosis in lung fibroblasts. A2aR can play a critical regulatory role in fibrosis-related diseases by modulating the Wnt/ß-catenin pathway, but its function in the EMT process of silicosis has not been explained. In this study, an EMT model of A549 cells was established. The results revealed that A2aR expression is reduced in the EMT model. Furthermore, activation of A2aR or suppression of the Wnt/ß-catenin pathway reversed the EMT process, while the opposite result was obtained by inhibiting A2aR. In addition, activation of A2aR in a mouse silicosis model inhibited the Wnt/ß-catenin pathway and ameliorated the extent of silica-induced lung fibrosis in mice. To sum up, we uncovered that A2aR inhibits fibrosis and the EMT process in silicosis by regulating the Wnt/ß-catenin pathway. Our study can provide an experimental basis for elucidating the role of A2aR in the development of silicosis and offer new ideas for further exploration of interventions for silicosis.


Asunto(s)
Transición Epitelial-Mesenquimal , Fibrosis Pulmonar , Receptor de Adenosina A2A , Silicosis , beta Catenina , Animales , Ratones , beta Catenina/genética , beta Catenina/metabolismo , Dióxido de Silicio/toxicidad , Silicosis/metabolismo , Silicosis/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Vía de Señalización Wnt , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo , Agonistas del Receptor de Adenosina A2/farmacología
6.
Ecotoxicol Environ Saf ; 249: 114401, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36508789

RESUMEN

Silicosis caused by long-term inhalation of crystalline silica during occupational activities seriously threatens the health of occupational populations. Imbalances in T helper 1(Th1), Th2, Th17, and regulatory T cells (Tregs) promote the development of pulmonary silicosis. Exosomes and their contents, especially microRNAs (miRNAs), represent a new type of intercellular signal transmission mediator related to various diseases including pulmonary fibrosis. However, whether exosomal miRNAs can affect the progression of silicosis by regulating T cell differentiation remains to be determined. To test this hypothesis, we established a miR-125a-5p antagomir mouse model and examined changes in miR-125a-5p levels and T cell subtypes. We found that miR-125a-5p levels were increased in lung tissues and serum exosomes in the silica group at 7 days and 28 days. Downregulation of miR-125a-5p attenuated α-smooth muscle actin (α-SMA), collagen I, fibronectin, p-p65, and p-inhibitor of nuclear factor kappa B (NF-κB) kinase (IKK) protein expression, while tumor necrosis factor receptor-associated factor 6 (TRAF6) and p-inhibitor of κBα (IKBα) expression were increased. MiR-125a-5p anta-miR treatment contributes to the maintenance of Th1/Th2 balance during the progression of pulmonary fibrosis. Our findings indicated that knockdown miR-125a-5p could regulate T lymphocyte subsets and significantly reduce pulmonary fibrosis by targeting TRAF6.


Asunto(s)
Exosomas , MicroARNs , Fibrosis Pulmonar , Dióxido de Silicio , Silicosis , Animales , Ratones , Proliferación Celular , MicroARNs/genética , MicroARNs/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Dióxido de Silicio/toxicidad , Silicosis/genética , Silicosis/patología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Exosomas/genética , Exosomas/metabolismo
7.
Ecotoxicol Environ Saf ; 249: 114392, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36508811

RESUMEN

Epithelial-mesenchymal transdifferentiation of alveolar type Ⅱ epithelial cells is a vital source of pulmonary myofibroblasts, and myofibroblasts formation is recognized as an important phase in the pathological process of silicosis. miR-30c-5p has been determined to be relevant in the activation of the epithelial-mesenchymal transition (EMT) in numerous disease processes. However, elucidating the role played by miR-30c-5p in the silicosis-associated EMT process remains a great challenge. In this work, based on the establishment of mouse silicosis and A549 cells EMT models, miR-30c-5p was interfered with in vivo and in vitro models to reveal its effects on EMT and autophagy. Moreover, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), connective tissue growth factor (CTGF), autophagy-related gene 5 (ATG5), and autophagy were further interfered with in the A549 cells models to uncover the possible molecular mechanism through which miR-30c-5p inhibits silicosis associated EMT. The results demonstrated the targeted binding of miR-30c-5p to CTGF, ATG5, and MALAT1, and showed that miR-30c-5p could prevent EMT in lung epithelial cells by acting on CTGF and ATG5-associated autophagy, thereby inhibiting the silicosis fibrosis process. Furthermore, we also found that lncRNA MALAT1 might competitively absorb miR-30c-5p and affect the EMT of lung epithelial cells. In a word, interfering with miR-30c-5p and its related molecules (MALAT1, CTGF, and ATG5-associated autophagy) may provide a reference point for the application of silicosis intervention-related targets.


Asunto(s)
Células Epiteliales Alveolares , Proteína 5 Relacionada con la Autofagia , Factor de Crecimiento del Tejido Conjuntivo , Transición Epitelial-Mesenquimal , MicroARNs , ARN Largo no Codificante , Silicosis , Animales , Ratones , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Proteína 5 Relacionada con la Autofagia/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Dióxido de Silicio/toxicidad , Silicosis/genética , Silicosis/metabolismo
8.
Toxicol Lett ; 360: 11-19, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35271943

RESUMEN

Silicosis is a systemic disease characterized by diffuse fibrosis of lung tissue. However, its pathogenesis has not been fully elucidated. Previous studies have demonstrated that there is a close relationship between EMT and pulmonary fibrosis. However, LncRNA XIST and miR-101-3p regulate the expression of ZEB1 which is a key transcription factor in the process of EMT through competitive endogenous RNA, thus affecting the process of EMT has not been reported. In this work, an experimental silicosis mouse model and cell model of TGF-ß1 stimulated lung epithelial cells (A549) for 48 h are established to investigate the biological effects of LncRNA XIST/ miR-101-3p/ZEB1 axis in the EMT process. The results reveal that LncRNA XIST and ZEB1 are up-regulated while the miR-101-3p expression is down-regulated in vivo and vitro models. Furthermore, the knockdown of LncRNA XIST prevents the EMT process and the inhibition of miR-101-3p markedly promotes EMT stimulated by TGF-ß1. Moreover, the results also illustrate that LncRNA XIST is mainly localized in the cytoplasm used FISH and possesses binding site with miR-101-3p which was identified as the target of ZEB1 used bioinformatics prediction website and Dual-luciferase reporter assay. The above demonstrated that LncRNA-XIST regulates ZEB1 by directly sponging miR-101-3p. To sum up, we uncovered that the up-regulated LncRNA XIST can modulate miR-101-3p and then up-regulate the expression of ZEB1, thus promoting the EMT process of alveolar epithelial cells in the process of silicosis-related pulmonary fibrosis EMT. Our study provides a new research idea for related targets of silicosis treatment.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Silicosis , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Células A549 , Animales , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Silicosis/genética
9.
Toxicol Appl Pharmacol ; 441: 115977, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35288145

RESUMEN

The main clinical manifestations are pulmonary fibrosis, silicosis, is one of the most common types of pneumoconiosis, and its pathogenesis is still unclear. The proliferation and transdifferentiation of fibroblasts are considered to be the key link leading to pulmonary fibrosis. Type II alveolar epithelial cells can be transformed into lung fibroblasts through epithelial-mesenchymal transition (EMT) to promote lung fibrosis. Involved in the EMT process of a variety of cancers, lncRNA UCA1 (UCA1) has been shown to competitively adsorb miR-204-5p, and play an effect on the downstream target gene E-box binding zinc finger protein 1 (ZEB1), thereby promoting EMT to facilitate the invasion and migration of cancer cells. This is an important potential intervention target that affects the process of EMT, but it has not been reported in the study of EMT related to silicosis. Therefore, this study established a SiO2 dust-treated mouse silicosis model and an in vitro EMT model of A549 cells to observe the changes and effects of UCA1 and miR-204-5p, and intervene on the two respectively. The results showed that the EMT process existed in the aforementioned models, while UCA1 was upregulated in the in vitro model. Double luciferase reporter assay demonstrated the targeted binding of UCA1 and miR-204-5p. Silencing UCA1 can up-regulate the expression of miR-204-5p and reduce the level of ZEB1, thus inhibiting EMT process, while intervention of miR-204-5p can change the level of ZEB1 and regulate EMT. Therefore, UCA1 may release its target gene ZEB1 through competitive adsorption of miR-204-5p to regulate EMT process.


Asunto(s)
MicroARNs , Fibrosis Pulmonar , ARN Largo no Codificante , Silicosis , Células A549 , Adsorción , Animales , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Pulmón/patología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Dióxido de Silicio/metabolismo , Silicosis/genética , Silicosis/patología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
10.
Environ Pollut ; 292(Pt A): 118272, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34718086

RESUMEN

Silicosis is a disease mainly caused by pulmonary interstitial fibrosis caused by long-term inhalation of dust with excessively high content of free SiO2. Transdifferentiation of lung fibroblasts into myofibroblasts is an important cellular basis for silicosis, but the key transcription factors (TFs) involved in this process are still unclear. In order to explore the biological regulation of transcription factor PPARγ/LXRα in silica-induced pulmonary fibrosis, this study explored the molecular mechanism of PPARγ/LXRα involved in regulating transcription factors related to SiO2-induced lung injury at the cellular level and in animal models. ChIP-qPCR detected that PPARγ directly regulated the transcriptional activity of the LXRα gene promoter, while the PPARγ agonist RSG increased the expression of LXRα. In addition, we demonstrated in the cell model that upregulation of LXRα can inhibit silica-mediated fibroblast transdifferentiation, accompanied by an increase in the expression of SREBF1, PLTP and ABCA1. The results of LXRα silencing experiment matched those of overexpression experiment. These studies explored the role of LXRα in plasticity and phenotypic transformation between lung fibroblasts and myofibroblasts. Therefore, inhibiting or reversing the transdifferentiation of lung fibroblasts to myofibroblasts by intervening PPARγ/LXRα may provide a new therapeutic target for the treatment of silicosis.


Asunto(s)
Dióxido de Silicio , Silicosis , Adaptación Fisiológica , Animales , Fibroblastos , Receptores X del Hígado/metabolismo , Pulmón , PPAR gamma/genética , PPAR gamma/metabolismo , Dióxido de Silicio/toxicidad
11.
J Cell Mol Med ; 2021 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-34076355

RESUMEN

Silicosis is a devastating occupational disease caused by long-term inhalation of silica particles, inducing irreversible lung damage and affecting lung function, without effective treatment. Mesenchymal stem cells (MSCs) are a heterogeneous subset of adult stem cells that exhibit excellent self-renewal capacity, multi-lineage differentiation potential and immunomodulatory properties. The aim of this study was to explore the effect of bone marrow-derived mesenchymal stem cells (BMSCs) in a silica-induced rat model of pulmonary fibrosis. The rats were treated with BMSCs on days 14, 28 and 42 after perfusion with silica. Histological examination and hydroxyproline assays showed that BMSCs alleviated silica-induced pulmonary fibrosis in rats. Results from ELISA and qRT-PCR indicated that BMSCs inhibited the expression of inflammatory cytokines TNF-α, IL-1ß and IL-6 in lung tissues and bronchoalveolar lavage fluid of rats exposed to silica particles. We also performed qRT-PCR, Western blot and immunohistochemistry to examine epithelial-mesenchymal transition (EMT)-related indicators and demonstrated that BMSCs up-regulate E-cadherin and down-regulate vimentin and extracellular matrix (ECM) components such as fibronectin and collagen Ⅰ. Additionally, BMSCs inhibited the silica-induced increase in TGF-ß1, p-Smad2 and p-Smad3 and decrease in Smad7. These results suggested that BMSCs can inhibit inflammation and reverse EMT through the inhibition of the TGF-ß/Smad signalling pathway to exhibit an anti-fibrotic effect in the rat silicosis model. Our study provides a new and meaningful perspective for silicosis treatment strategies.

12.
Biomed Res Int ; 2020: 3075729, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33381546

RESUMEN

BACKGROUND: Esophageal cancer is one of the most deadly malignant tumors. Among the common malignant tumors in the world, esophageal cancer is ranked seventh, which has a high mortality rate. Long noncoding RNAs (lncRNAs) play an important role in the occurrence and development of various tumors. lncRNAs can competitively bind microRNAs (miRNAs) with mRNA, which can regulate the expression level of the encoded gene at the posttranscriptional level. This regulatory mechanism is called the competitive endogenous RNA (ceRNA) hypothesis, and ceRNA has important research value in tumor-related research. However, the regulation of lncRNAs is less studied in the study of esophageal cancer. METHODS: The Cancer Genome Atlas (TCGA) database was used to download transcriptome profiling data of esophageal cancer. Gene expression quantification data contains 160 cancer samples and 11 normal samples. These data were used to identify differentially expressed lncRNAs and mRNAs. miRNA expression data includes 185 cancer samples and 13 normal samples. The differentially expressed RNAs were identified using the edgeR package in R software. Then, the miRcode database was used to predict miRNAs that bind to lncRNAs. MiRTarBase, miRDB, and TargetScan databases were used to predict the target genes of miRNAs. Cytoscape software was used to draw ceRNA network. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using DAVID 6.8. Finally, multifactor cox regression was used to screen lncRNAs related to prognosis. RESULTS: We have screened 1331 DElncRNAs, 3193 DEmRNAs, and 162 DEmiRNAs. Among them, the ceRNA network contains 111 lncRNAs, 11 miRNAs, and 63 DEmRNAs. Finally, we established a prediction model containing three lncRNAs through multifactor Cox regression analysis. CONCLUSIONS: Our research screened out three independent prognostic lncRNAs from the ceRNA network and constructed a risk assessment model. This is helpful to understand the regulatory role of lncRNAs in esophageal cancer.


Asunto(s)
Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Biomarcadores de Tumor/genética , Perfilación de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Genoma Humano , Humanos , Estimación de Kaplan-Meier , MicroARNs/metabolismo , Pronóstico , Modelos de Riesgos Proporcionales , Transcriptoma
13.
J Cell Mol Med ; 24(20): 12219-12224, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32929850

RESUMEN

Silicosis is an incurable occupational disease, and its pathological feature is diffuse pulmonary fibrosis. Pulmonary epithelial-mesenchymal transition (EMT) is one of the important events in the pathogenesis of silicosis. Previous studies found that abnormal expression of various microRNAs (miRNAs) involved in the development of lung fibrosis. However, their roles in silicosis have not been elucidated. To research the biological effects of miR-34a in EMT process in silica-induced lung fibrosis, we established the silicosis model in mouse and miR-34a intervention in a cell model of TGF-ß1 stimulated lung epithelial cells (A549). The results showed that miR-34a expression was down-regulated in the fibrotic lung tissue after silica treatment, and it was similarly expressed in A549 cells stimulated by TGF-ß1. Meanwhile, silica-induced EMT process can increase expression of two mesenchymal markers, α-SMA and vimentin. Furthermore, overexpression miR-34a markedly inhibited EMT stimulated by TGF-ß1. Mechanistically, SMAD4 was identified as the target of miR-34a. SMAD4 levels decreased in mRNA and protein levels in A549 cells upon miR-34a overexpression. In addition, the knockdown of SMAD4 blocked the EMT process. Taken together, miR-34a regulated EMT, which might be partially realized by targeting SMAD4. Our data might provide new insight into treatment targets for silica-induced pulmonary fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , MicroARNs/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Proteína Smad4/metabolismo , Células A549 , Animales , Regulación hacia Abajo/genética , Silenciador del Gen , Humanos , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , Modelos Biológicos , Fibrosis Pulmonar/patología , Dióxido de Silicio
14.
Sci Total Environ ; 747: 141531, 2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-32791419

RESUMEN

Silicosis, a severe and irreversible form of pulmonary fibrosis (PF) caused by long-term exposure to dust particles in production environments, is the biggest occupational health concern in China and most low-income countries. The transdifferentiation of pulmonary fibroblasts is the terminal event in silicosis, and specific transcription factors (TFs) play a crucial role in this condition. However, the relationship between TF-mediated regulation and silicosis remains unknown. We performed a transcriptomic analysis to elucidate this relationship, and our results revealed that two TFs, EGR2 and BHLHE40, were upregulated and five, i.e., TBX2, NR1H3 (LXRα), NR2F1, PPARG (PPARγ), and EPAS1, were downregulated in activated fibroblasts. Notably, PPARγ and LXRα expression was also decreased in an experimental mouse model of silicosis. The mechanism underlying these changes may involve TGF-ß1 secretion from silica-exposed alveolar macrophages, causing PPARγ and LXRα downregulation, which in turn would result in aberrant α-SMA transcription. Our results suggest that LXRα is a potential target for the prevention of silicosis and PF.


Asunto(s)
Dióxido de Silicio , Silicosis , Animales , China , Perfilación de la Expresión Génica , Receptores X del Hígado/genética , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA