Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell Biochem Biophys ; 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38635101

ABSTRACT

Acute respiratory distress syndrome (ARDS) is regarded as a type of respiratory failure. Emerging evidence has demonstrated the significant roles of microRNAs in various disorders. Nevertheless, the role of miR-202-3p in ARDS is unclear. Forty male C57BL/6 mice treated with phosphate buffer saline/lipopolysaccharide (PBS/LPS) and administrated with NC/miR-202-3p agomir were divided into four groups. A reverse transcription-quantitative polymerase chain reaction was used to evaluate the level of miR-202-3p, its target genes, and proinflammatory factors. Hematoxylin­eosin was utilized for histological observation of the lung tissues. The Wet/Dry ratio, myeloperoxidase activity, and total protein concentration in bronchoalveolar lavage fluid were assessed to determine pulmonary edema. Western blotting was used for quantifying protein levels of proinflammatory factors, nuclear factor kappa B (NF-κB), and NLR family pyrin domain containing 3 (NLRP3) signaling-associated proteins. Calmodulin 1 (Calm1) protein expression in murine lung tissues was evaluated by immunohistochemistry. The binding relation between miR-202-3p and Calm1 was assessed by luciferase reporter assay. The results showed that miR-202-3p was lowly expressed in the lung tissues of ARDS mice. Overexpressed miR-202-3p relieved LPS-induced edema, reduced proinflammatory factors, and inactivated NF-κB/NLRP3 signaling in murine lung tissues. Calm1 was targeted by miR-202-3p and displayed a high level of LPS-induced ARDS. In conclusion, miR-202-3p targets Calm1 and suppresses inflammation in LPS-induced ARDS, thereby inhibiting the pathogenesis of ARDS in a mouse model.

2.
Exp Lung Res ; 48(7-8): 199-212, 2022.
Article in English | MEDLINE | ID: mdl-35943053

ABSTRACT

Objective: Bromodomain-containing protein 7 (BRD7) is a key component of the switch/sucrose non-fermentable complex that participates in chromatin remodeling and transcriptional regulation. Although the emerging role of BRD7 in the pathophysiology of various diseases has been observed, its role in asthma remains unknown. Here, we assessed the function of BRD7 as a mediator of airway remodeling in asthma using an in vitro model. Methods: Airway smooth muscle cells (ASMCs) were challenged with tumor necrosis factor-α (TNF-α) to establish an in vitro airway remodeling model. Protein levels were examined using western blotting. Cell proliferation was measured using the cell counting kit-8 and 5-ethynyl-2'-deoxyuridine assays. Cell migration was assessed using a transwell migration assay. Results: Exposure to TNF-α dramatically decreased BRD7 levels in ASMCs. BRD7 remarkably decreased TNF-α-induced proliferation and migration of ASMCs. In contrast, ASMCs with BRD7 deficiency were more sensitive to TNF-α-induced pro-proliferative and pro-migratory effects. Mechanistically, BRD7 could repress the expression of Notch1 and block the Notch pathway in TNF-α-challenged cells. Notably, reactivation of Notch signaling substantially reversed the BRD7 overexpression-mediated effects, whereas restraining Notch signaling abolished BRD7-depletion-mediated effects on TNF-α-challenged cells. Conclusions: BRD7 inhibits the proliferation and migration of ASMCs elicited by TNF-α by downregulating the Notch pathway. This study indicates that BRD7 may exert a suppressive effect on airway remodeling during asthma.


Subject(s)
Airway Remodeling , Asthma , Chromosomal Proteins, Non-Histone , Myocytes, Smooth Muscle , Asthma/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Chromosomal Proteins, Non-Histone/metabolism , Humans , Myocytes, Smooth Muscle/cytology , Receptors, Notch/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
J Exp Clin Cancer Res ; 40(1): 318, 2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34645486

ABSTRACT

BACKGROUND: SETD1A, a member of SET1/MLL family H3K4 methyltransferases, is involved in the tumorigenesis of numerous cancers. However, the biological role and mechanism of SETD1A in non-small cell lung cancer (NSCLC) remain to be elucidated. METHODS: The expression of SETD1A, NEAT1, EZH2, and ß-catenin in NSCLC tissues and cell lines was detected by qRT-PCR, immunohistochemistry and western blotting. The regulatory mechanisms were validated by chromatin immunoprecipitation, co-immunoprepitation and luciferase reporter assay. The self-renewal, cisplatin sensitivity and tumorigenesis of NSCLC cells were analyzed using sphere formation, CCK-8, colony formation assays and xenograft tumor models. RESULTS: SETD1A expression was significantly increased in NSCLC and its overexpression predicted a poor prognosis of patients with NSCLC. Functional experiments showed that SETD1A positively regulated cancer stem cell property and negatively regulated cisplatin sensitivity in NSCLC cells via the Wnt/ß-catenin pathway. Next, we found that SETD1A positively regulated the Wnt/ß-catenin pathway via interacting with and stabilizing ß-catenin. The SET domain is dispensable for the interaction between SETD1A and ß-catenin. Furthermore, we identified that SETD1A bound to the promoters of NEAT1 and EZH2 to activate gene transcription by inducing H3K4me3 enrichment. Rescue experiments showed that SETD1A promoted the Wnt/ß-catenin pathway and exerted its oncogenic functions in NSCLC, at least, partly through NEAT1 and EZH2 upregulation. In addition, SETD1A was proven to be a direct target of the Wnt/ß-catenin pathway, thus forming a positive feedback loop in NSCLC cells. CONCLUSION: SETD1A and Wnt/ß-catenin pathway form a positive feedback loop and coordinately contribute to NSCLC progression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Histone-Lysine N-Methyltransferase/metabolism , Lung Neoplasms/genetics , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Feedback , Female , Humans , Lung Neoplasms/pathology , Mice , Transfection
4.
Br J Radiol ; 94(1118): 20201030, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33237823

ABSTRACT

OBJECTIVE: We sought to explore the relationships between multiple chemokines with spirometry, inflammatory mediators and CT findings of emphysema, small airways disease and bronchial wall thickness. METHODS: All patients with COPD (n = 65) and healthy control subjects (n = 23) underwent high-resolution CT, with image analysis determining the low attenuation area (LAA), ratio of mean lung attenuation on expiratory and inspiratory scans (E/I MLD) and bronchial wall thickness of inner perimeter of a 10-mm diameter airway (Pi10). At enrollment, subjects underwent pulmonary function studies, chemokines and inflammatory mediators measurements. RESULTS: Multiple chemokines (CCL2, CCL3, CCL5, CX3CL1, CXCL8, CXCL9, CXCL10, CXCL11 and CXCL12) and inflammatory mediators (MMP-9, MMP-12, IL-18 and neutrophil count) were markedly increased in the serum of COPD patients compared with healthy controls. There were associations between small airway disease (E/I MLD) and CCL11, CXCL8, CXCL10, CXCL11, CXCL12 and CX3CL1. Especially CXCL8 and CX3CL1 are strongly associated with E/I MLD (r = 0.74, p < 0.001; r = 0.76, p < 0.001, respectively). CXCL8, CXCL12 and CX3CL1 were moderately positively correlated with emphysema (%LAA) (r = 0.49, p < 0.05; r = 0.51, p < 0.05; r = 0.54, p < 0.01, respectively). Bronchial wall thickness (Pi10)showed no significant differences between the COPD and healthy controls,,but there was an association between Pi10 and FEV1% in COPD patients (r=-0.420, p = 0.048). Our statistical results showed that there were not any associations between airway wall thickness (Pi10) and chemokines. CONCLUSION: Pulmonary chemokines levels are closely associated with the extent of gas trapping, small airways disease and emphysema identified on high-resolution chest CT scan. ADVANCES IN KNOWLEDGE: This study combines quantitative CT analysis with multiplex chemokines and inflammatory mediators to identify a new role of pathological changes in COPD.


Subject(s)
Chemokines/blood , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/diagnostic imaging , Tomography, X-Ray Computed/methods , China , Evaluation Studies as Topic , Female , Humans , Lung/diagnostic imaging , Male , Middle Aged , Reproducibility of Results
5.
Cancer Cell Int ; 20(1): 594, 2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33298086

ABSTRACT

BACKGROUND: Activating transcription factor 2 (ATF2), a member of the activator protein 1 (AP-1) transcription factor family, has been shown to be involved in the pathobiology of numerous cancers. However, the biological role and mechanism of ATF2 in lung adenocarcinoma (LUAD) remains to be elucidated. METHODS: The expression of ATF2, NEAT1 and miR-26a-5p in LUAD tissues and cell lines was detected by qRT-PCR and western blotting. The interaction between ATF2, NEAT1, and miR-26a-5p was validated by chromatin immunoprecipitation, luciferase reporter assay and RNA immunoprecipitation. Cell proliferation, invasion and tumorigenesis of LUAD cells were analyzed by using CCK8, transwell invasion assay and xenograft tumor model. RESULTS: We confirmed that ATF2 expression was increased in LUAD tissues compared with normal adjacent lung tissues. Functional experiments showed that ATF2 positively regulated cell proliferation and invasion in LUAD cells. Moreover, we identified that NEAT1 expression was increased in LUAD tissues and positively correlated with ATF2 expression. Mechanistically, ATF2 could bind to the promoter of NEAT1 to promote its transcription. Rescue experiments showed that ATF2 exerted its oncogenic function in LUAD, at least, partly through NEAT1 upregulation. In turn, NEAT1 could positively regulate ATF2 expression and form a positive feedback loop in LUAD cells. Furthermore, we demonstrated that NEAT1 positively regulated ATF2 expression via sponging miR-26a-5p. CONCLUSION: ATF2 and NEAT1 form a positive feedback loop mediated by miR-26a-5p and coordinately contribute to LUAD progression.

6.
Clin Exp Pharmacol Physiol ; 47(3): 449-458, 2020 03.
Article in English | MEDLINE | ID: mdl-31665807

ABSTRACT

Deregulation of microRNAs (miRNAs) leads to malignant growth and aggressive invasion during cancer occurrence and progression. miR-147b has emerged as one of the cancer-related miRNAs that are dysregulated in multiple cancers. Yet, the relevance of miR-147b in non-small-cell lung cancer (NSCLC) remains unclear. In the present study, we aimed to report the biological function and signalling pathways mediated by miR-147b in NSCLC. Our results demonstrate that miR-147b expression is significantly downregulated in NSCLC tissues and cell lines. Overexpression of miR-147b decreased the proliferative ability, colony-forming capability, and invasive potential of NSCLC cells. Notably, our study identified ribosomal protein S15A (RPS15A), an oncogene in NSCLC, as a target gene of miR-147b. Our results showed that miR-147b negatively modulates RPS15A expression in NSCLC cells. An inverse correlation between miR-147b and RPS15A was evidenced in NSCLC specimens. Moreover, miR-147b overexpression downregulated the activation of Wnt/ß-catenin signalling via targeting of RPS15A. Overexpression of RPS15A partially reversed the miR-147b-mediated antitumour effect in NSCLC cells. Collectively, these findings reveal that miR-147b restricts the proliferation and invasion of NSCLC cells by inhibiting RPS15A-induced Wnt/ß-catenin signalling and suggest that the miR-147b/RPS15A/Wnt/ß-catenin axis is an important regulatory mechanism for malignant progression of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/physiology , Lung Neoplasms/metabolism , MicroRNAs/biosynthesis , Ribosomal Proteins/biosynthesis , Wnt Signaling Pathway/physiology , A549 Cells , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/prevention & control , Down-Regulation/physiology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control
7.
Int J Oncol ; 55(3): 745-754, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31322202

ABSTRACT

The long non­coding RNA nuclear enriched abundant transcript 1 (NEAT1) has important roles in the regulation of multiple cell functions, such as proliferation, apoptosis and migration. However, the mechanism by which NEAT1 regulates breast cancer progression is not well elucidated. In the present study, NEAT1 and microRNA­124 (miR­124) levels were detected by reverse transcription­quantitative PCR in breast cancer tissues and cell lines. STAT3 protein levels were detected by western blot analysis. Cell proliferation and cell cycle distribution were determined using MTT and colony formation assays, and flow cytometry, respectively. The results demonstrated that NEAT1 and STAT3 expression levels were increased in breast cancer tissues compared with normal breast tissues, whereas miR­124 expression was significantly decreased. Functional analyses revealed that NEAT1 promoted cell proliferation and cell cycle progression in breast cancer cells. Additionally, NEAT1 and STAT3 expression levels were negatively correlated with miR­124 levels in breast cancer tissues. A direct interaction between miR­124, and NEAT1 and STAT3, was predicted by bioinformatics analysis and confirmed using a luciferase activity assay. NEAT1 overexpression markedly increased STAT3 protein expression levels, and this effect was reversed by miR­124 overexpression in breast cancer cells. Furthermore, miR­124 overexpression partially attenuated the effects of NEAT1 on breast cancer cell proliferation and cell cycle progression. The inhibitory effects of miR­124 overexpression on the proliferation rate and cell cycle progression were abolished by STAT3 overexpression. In turn, STAT3 silencing inhibited NEAT1 transcription in breast cancer cells. In summary, the present findings revealed that NEAT1 and STAT3 formed a feedback loop via sponging miR­124 to promote breast cancer progression.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , STAT3 Transcription Factor/genetics , Adult , Cell Line, Tumor , Cell Proliferation , Disease Progression , Feedback, Physiological , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Middle Aged , Survival Analysis
8.
Chem Biol Interact ; 300: 82-90, 2019 Feb 25.
Article in English | MEDLINE | ID: mdl-30639269

ABSTRACT

Increasing evidence illustrate that dysregulation of microRNAs (miRNAs) is involved in the pathogenesis of chronic obstructive pulmonary disease (COPD), which is mainly resulted from cigarette smoke (CS) exposure. However, the role of miR-145-5p in CS-mediated COPD remains largely unknown. Thus, the aim of this study was to investigate the expression level of miR-145-5p in 31 human lung tissues samples, and to explore its regulatory role in the apoptosis and inflammation of human bronchial epithelial cells (HBECs) following CS extract (CSE) exposure. We found that miR-145-5p was significantly down-regulated in lung tissues from smokers without or with COPD compared to non-smokers. Functional assays showed that miR-145-5p overexpression remarkably alleviated CSE-induced apoptosis and inflammation response by regulating p53-mediated apoptotic signaling and pre-inflammatory factors such as necrosis factor-α (TNF-α), interleukins (IL)-6, IL-8 in HBECs, whereas, down-regulation of miR-145-5p showed opposite effects. Furthermore, luciferase reporter assays verified that Kruppel-like 5 (KLF5) was a direct target of miR-145-5p. Western blot assay also confirmed that KLF5 was up-regulated in COPD tissues and was negatively associated with miR-145-5p expression. Restoration of miR-145-5p expression significantly abrogated the suppressive effect of miR-145-5p on CSE-stimulated apoptosis and inflammation. In addition, the CSE-induced NF-κB signaling activation was suppressed by miR-145-5p overexpression. Therefore, our data suggested that miR-145-5p conferred protection against CSE-induced airway epithelial cell apoptosis and inflammation partially via targeting KLF5, which might be a potential therapeutic biomarker in COPD treatment.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Lung/pathology , MicroRNAs/metabolism , Pulmonary Disease, Chronic Obstructive/etiology , Smoking/adverse effects , 3' Untranslated Regions , Aged , Apoptosis , Biomarkers/metabolism , Bronchi/cytology , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Inflammation/prevention & control , Interleukin-6/analysis , Interleukin-6/metabolism , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Lung/metabolism , Male , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 2/metabolism , MicroRNAs/genetics , Middle Aged , NF-kappa B/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism
9.
J Cell Mol Med ; 22(12): 6262-6274, 2018 12.
Article in English | MEDLINE | ID: mdl-30324719

ABSTRACT

Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with a poor prognosis. The microRNA-200 (miR-200) family has been associated with breast cancer metastasis. However, the epigenetic mechanisms underlying miR-200b repression in TNBC are not fully elucidated. In this study, we found that MYC proto-oncogene, bHLH transcription factor (MYC) and DNA methyltransferase 3A (DNMT3A) were highly expressed in TNBC tissues compared with other breast cancer subtypes, while miR-200b expression was inhibited significantly. We demonstrated that MYC physically interacted with DNMT3A in MDA-MB-231 cells. Furthermore, we demonstrated that MYC recruited DNMT3A to the miR-200b promoter, resulting in proximal CpG island hypermethylation and subsequent miR-200b repression. MiR-200b directly inhibited DNMT3A expression and formed a feedback loop in TNBC cells. MiR-200b overexpression synergistically repressed target genes including zinc-finger E-box-binding homeobox factor 1, Sex determining region Y-box 2 (SOX2), and CD133, and inhibited the migration, invasion and mammosphere formation of TNBC cells. Our findings reveal that MYC can collaborate with DNMT3A on inducing promoter methylation and miR-200b silencing, and thereby promotes the epithelial to mesenchymal transition and mammosphere formation of TNBC cells.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/genetics , Triple Negative Breast Neoplasms/genetics , Aged , Cell Movement/genetics , Cell Proliferation/genetics , CpG Islands/genetics , DNA Methylation/genetics , DNA Methyltransferase 3A , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Middle Aged , Proto-Oncogene Mas , Signal Transduction/genetics , Transfection , Triple Negative Breast Neoplasms/pathology
10.
Int J Oncol ; 53(4): 1601-1612, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30066905

ABSTRACT

Tamoxifen (TAM) resistance is a substantial challenge in the treatment of estrogen receptor (ER)-positive breast cancer. Previous studies have revealed an important role of microRNA (miRNA/miR)-26a in TAM resistance in breast cancer. However, the mechanism underlying the regulatory effects of miR-26a on TAM resistance remains to be elucidated. The expression levels of miR-26a in ER-positive breast cancer were detected by reverse transcription-quantitative polymerase chain reaction. E2F transcription factor 7 (E2F7) and MYC proto-oncogene, bHLH transcription factor (MYC) levels were detected by western blotting. The present study demonstrated that miR-26a expression was reduced in ER-positive breast cancer compared with in normal breast tissues, whereas E2F7 expression was significantly elevated. Furthermore, an inverse correlation between miR-26a and E2F7 expression was detected in ER-positive breast cancer. The results indicated that miR-26a directly inhibited E2F7 expression through translational inhibition and indirectly inhibited MYC expression partly via E2F7 repression. E2F7, in turn, decreased miR-26a expression via MYC-induced transcriptional inhibition of miRNAs. Furthermore, transfection with miR-26a mimics increased the expression of its host genes (CTD small phosphatase like and CTD small phosphatase 2), whereas ectopic E2F7 expression abrogated the effects of miR-26a. These findings indicated that miR-26a and E2F7 may form a double-negative feedback loop, resulting in downregulation of miR-26a and upregulation of E2F7 in ER-positive breast cancer. Both miR-26a knockdown and E2F7 overexpression conferred resistance to TAM in MCF-7 cells. Conversely, miR-26a overexpression and E2F7 silencing resensitized MCF-7 resistant cells to TAM. These findings revealed that a feedback loop between miR-26a and E2F7 may promote TAM resistance in ER-positive breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , E2F7 Transcription Factor/genetics , MicroRNAs/metabolism , Tamoxifen/pharmacology , Adult , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Down-Regulation , E2F7 Transcription Factor/metabolism , Feedback, Physiological , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism , Tamoxifen/therapeutic use , Up-Regulation
11.
Int J Oncol ; 51(4): 1077-1088, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28902370

ABSTRACT

The poor therapy response and poor prognosis of esophageal cancer has made it one of the most malignant carcinoma, and the complicated multidisciplinary treatment failed to achieve a long-term disease-free survival. To diagnose esophageal cancer at an earlier stage, and to improve the effect of anticancer therapy would improve the therapeutic efficacy. After retrospective analysis of the cancer samples of patients who received esophagectomy, we found the relevance between ratio of either ALDH1 or CD133-positive cancer stem cells and 2-year recurrence. Higher ratios of cancer stem cells indicated later clinical stages, and Wnt signaling activation was more frequent in later esophageal carcinoma. Further in bench studies, we explored the suppressive roles and the mechanisms involved in Let­7 on self-renewal in ECA­109 and ECA­9706 esophageal cancer stem cells. Isolated cancer stem cells naturally divide symmetrically and are therapy resistant. Therapy of fluorouracil and docetaxel both enriched the stem cells, proving the resistant characteristics of cancer stem cells. Wnt activation stimulated more symmetric division of stem cells, resulting in self-renewal promotion, which could be blocked by Let­7 overexpression. Furthermore, enforced Let­7 sensitized the stem cells to chemotherapies in a Wnt pathway inhibition-dependent manner, contributing to Let­7 sensitization of chemotherapeutic response. Wnt activation weakened the suppressive Let­7b through the sponge functions of CCAT-1, forming the negative feedback loop of Let­7b/Wnt/CCAT1. These results identified the crucial participation of stem cells in esophageal cancer occurrence and progression as the potent indicator, and also indicate the potential powerful agent of Let­7 nano-particles in treatment of cancer.


Subject(s)
Esophageal Neoplasms/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/cytology , Wnt Signaling Pathway , AC133 Antigen/metabolism , Aged , Aldehyde Dehydrogenase 1 Family , Cell Line, Tumor , Docetaxel , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/surgery , Female , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Isoenzymes/metabolism , Male , Middle Aged , Nanomedicine , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/genetics , Retinal Dehydrogenase/metabolism , Retrospective Studies , Taxoids/pharmacology , Wnt Signaling Pathway/drug effects
12.
Mol Med Rep ; 14(5): 4643-4649, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27748907

ABSTRACT

Pulmonary silicosis is characterized by lung fibrosis, which leads to impairment of pulmonary function; the specific mechanism remains to be fully elucidated Emodin shows antifibrotic effects in several organs with fibrosis, however, it has not been investigated in pulmonary silicosis. In the present study, the possible mechanism of lung fibrosis and the antifibrotic effect of emodin in silica inhalation­induced lung fibrosis were investigated. Pulmonary silica particle inhalation was used to induce lung fibrosis in mice. Emodin and or the sirtuin 1 (Sirt1) inhibitor, nicotinamide, were used to treat the modeled animals. Pulmonary function was assessed using an occlusion method. The deposition of collagen I and α­smooth muscle actin (SMA) in the lung tissue were detected using fluorescence staining; transforming growth factor­ß1 (TGF­ß1) in the bronchoalveolar lavage fluid (BALF) was examined using an enzyme­linked immunosorbent assay; TGF-ß1/Sirt1/small mothers against decapentaplegic (Smad) signaling activation in lung tissue was also examined. The molecular contacts between emodin were evaluated using liquid chromatography­mass spectrometry analysis. The deposition of collagen I and α­SMA in lung tissues were found to be elevated following silica exposure, however, this was relieved by emodin treatment. The pulmonary function of the animals was impaired by silica inhalation, and this was improved by emodin administration. However, the therapeutic effects of emodin on lung fibrosis were impaired by nicotinamide administration. The levels of TGF­ß1 in the BALF and lung tissue were elevated by silica inhalation, however, they were not affected by either emodin or nicotinamide treatment. Additionally, emodin was found to increase the expression level of Sirt1, which decreased the level of deacetylated Smad3 to attenuate collagen deposition. Furthermore, the data suggested that there was direct binding between emodin and Sirt1. Sirt1­regulated TGF­ß1/Smad signaling was involved in silica inhalation­induced lung fibrosis. Emodin attenuated this lung fibrosis to improve pulmonary function by targeting Sirt1, which regulated TGF-ß1/Smad fibrotic signaling.


Subject(s)
Fibrosis/drug therapy , Lung Diseases/drug therapy , Silicosis/drug therapy , Sirtuin 1/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta1/genetics , Actins/metabolism , Animals , Bronchoalveolar Lavage Fluid , Collagen Type I/metabolism , Disease Models, Animal , Emodin , Enzyme-Linked Immunosorbent Assay , Fibrosis/chemically induced , Fibrosis/genetics , Fibrosis/pathology , Gene Expression Regulation/drug effects , Humans , Lung/drug effects , Lung/pathology , Lung Diseases/chemically induced , Lung Diseases/genetics , Lung Diseases/pathology , Mice , Silicon Dioxide/toxicity , Silicosis/genetics , Silicosis/pathology , Sirtuin 1/biosynthesis , Smad3 Protein/biosynthesis , Transforming Growth Factor beta1/biosynthesis
13.
Oncol Lett ; 11(4): 2635-2643, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27073531

ABSTRACT

The E-cadherin gene (CDH1) is associated with poor prognosis and metastasis in patients with breast cancer, and methylation of its promoter is correlated with decreased gene expression. However, there is currently no direct evidence that CDH1 promoter methylation indicates poor prognosis in patients with breast cancer. In the present study, methylation-specific polymerase chain reaction (PCR) was applied to detect the methylation status of the CDH1 promoter in 137 primary breast cancer, 85 matched normal breast tissue and 13 lung metastasis specimens. Reverse transcription-quantitative PCR was used to assess the relative expression levels of CDH1 mRNA, and correlation analysis between CDH1 methylation status, and gene expression, clinicopathological characteristics and patient survival was performed. Methylation of CDH1 was identified in 40.9% (56/137) of primary breast cancer specimens, 61.5% (8/13) of lung metastasis specimens and none of the matched normal breast specimens. The downregulation of CDH1 mRNA and E-cadherin protein expression were identified to be significantly correlated with CDH1 methylation (P<0.05). In addition, CDH1 methylation was significantly associated with lymph node metastasis and estrogen receptor status of patients (P<0.05). In univariate analyses, patients with CDH1 methylation exhibited poor overall survival (OS) and disease-free survival (DFS; P<0.05). Furthermore, multivariate analyses revealed that CDH1 methylation was an independent prognostic factor predicting poor OS (HR, 1.737; 95% CI, 0.957-3.766; P=0.041) and DFS (HR, 2.018; 95% CI, 2.057-3.845; P=0.033) in patients with breast cancer. Therefore, the present study suggests that CDH1 promoter methylation may be correlated with breast carcinogenesis and indicates poor prognosis in patients with breast cancer.

14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(3): 299-303, 2016 Mar.
Article in Chinese | MEDLINE | ID: mdl-26927545

ABSTRACT

OBJECTIVE: To investigate the effect of microRNA-101 (miR-101) on the proliferation and migration of breast cancer cells and its possible mechanism. METHODS: The expressions of miR-101 and DNA methyltransferase 3a (DNMT3a) in breast cancer tissues, corresponding normal breast tissues, breast cancer cells and normal breast cells were detected by real-time quantitative PCR. The lentiviral vectors containing miR-101 and shRNA-DNMT3a sequences were transfected into MDA-MB-231 cells to regulate the expressions of miR-101 and DNMT3a. The expressions of DNMT3a and E-cadherin were determined by Western blotting. The proliferation and migration abilities of MDA-MB-231 cells were evaluated by MTT assay and wound healing assay, respectively. RESULTS: Breast cancer tissues and breast cancer cell lines (MCF-7, MDA-MB-231, MDA-MB-361, HCC70) showed lower miR-101 expression and higher DNMT3a expression than the adjacent normal breast tissues and normal breast cell line. The overexpression of miR-101 resulted in downregulation of DNMT3a and restoration of E-cadherin. Besides, knockdown of DNMT3a by shRNA increased E-cadherin expression. MTT assay and would healing assay showed that miR-101 overexpression significantly inhibited the proliferation and migration abilities of MDA-MB-231 cells. CONCLUSION: miR-101 may inhibit MDA-MB-231 cell proliferation and migration by repressing DNMT3a expression and up-regulating E-cadherin expression.


Subject(s)
Breast Neoplasms/genetics , Cell Movement/genetics , Cell Proliferation/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Survival/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Down-Regulation , Female , Humans , MCF-7 Cells , Microscopy, Fluorescence , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...