Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Infect Dis (Lond) ; 55(12): 839-846, 2023 12.
Article in English | MEDLINE | ID: mdl-37624684

ABSTRACT

BACKGROUND: Data on the coincidence of Tuberculosis (TB) and Coronavirus disease 2019 (COVID-19) are limited. We sought to investigate the clinical characteristics and outcomes of coinfected patients in Henan and identify whether TB disease is associated with an increased risk of intensive care unit (ICU) admission and mortality. METHOD: We conducted a retrospective matched cohort study of COVID-19 inpatients involving 41 TB-positive patients with 82 patients without TB. Leveraging data was collected from electronic medical records. RESULTS: There were no significant differences in clinical manifestations, the need for mechanical ventilation and vasopressors, ICU admission, or in-hospital mortality between 2 groups. TB-positive patients had a lower lymphocyte counts (1.24 ± 0.54 vs. 1.59 ± 0.58, p = 0.01), B cells (99/µl vs. 201/µl, p < 0.01), CD4+ T cells (382/µl vs. 667/µl, p < 0.01), CD8+ T cells (243/µl vs. 423/µl, p < 0.01), NK cells (145/µl vs. 216/µl, p = 0.01), IL-2 (14.18 ± 11.23 vs. 31.86 ± 34.55, p < 0.01) and TNF-α (3.42 ± 2.93 vs. 5.62 ± 3.69, p < 0.01). Notably, the TB-positive group had a longer duration of SARS-CoV-2 shedding (67 days vs. 22 days, p < 0.01). CONCLUSIONS: Concomitant TB does not significantly impact clinical outcomes of hospitalised patients with acute COVID-19. However, TB-positive patients had longer duration of SARS-COV-2-RNA positivity.


Subject(s)
COVID-19 , Coinfection , Tuberculosis , Humans , COVID-19/complications , COVID-19/therapy , SARS-CoV-2 , Retrospective Studies , Cohort Studies , Coinfection/epidemiology , Tuberculosis/complications
3.
Pathol Res Pract ; 235: 153913, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35552087

ABSTRACT

BACKGROUND: Circular RNA 0000514 (circ_0000514) has a modulatory function in the progression of several cancers, but its role and mechanism in non-small cell lung cancer (NSCLC) are obscure. METHODS: The relative expressions of circ_0000514, microRNA-330-5p (miR-330-5p) and high mobility group AT-hook 2 (HMGA2) mRNA in 67 cases of NSCLC and adjacent lung tissues were examined by quantitative real-time polymerase chain reaction (qRT-PCR). The changes in cell phenotypes were examined by cell counting kit-8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU), Transwell and flow cytometry assays, respectively. Bioinformatics analysis, dual-luciferase reporter gene experiment and RNA immunoprecipitation (RIP) experiment were adopted to predict and validate the relationship between circ_0000514 and miR-330-5p, and miR-330-5p and HMGA2 3'UTR, respectively. In addition, Western blot was performed to examine the effects of circ_0000514 overexpression or knockdown on HMGA2 protein expression. RESULTS: Circ_0000514 expression was up-regulated in NSCLC tissues. Circ_0000514 significantly facilitated the growth, cell cycle progression, metastatic potential, and repressed the apoptosis of NSCLC cells. Circ_0000514 was identified as a molecular sponge to decoy miR-330-5p, and HMGA2 was proven to be a target gene of miR-330-5p. Moreover, circ_0000514 positively regulated HMGA2 expression, and its biological effect was dependent on miR-330-5p/HMGA2 axis. CONCLUSION: Circ_0000514 is an oncogenic circRNA in NSCLC progression, and it modulates miR-330-5p/HMGA2 axis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , HMGA2 Protein , Lung Neoplasms , MicroRNAs , RNA, Circular , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/genetics , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism
4.
Front Cardiovasc Med ; 9: 814557, 2022.
Article in English | MEDLINE | ID: mdl-35509273

ABSTRACT

Background: Previous studies have suggested that pregnant women with pulmonary hypertension (PH) have high maternal mortality. However, indexes or factors that can predict maternal death are lacking. Methods: We retrospectively reviewed pregnant women with PH admitted for delivery from 2012 to 2020 and followed them for over 6 months. The patients were divided into two groups according to 10-day survival status after delivery. Predictive models and predictors for maternal death were identified using four machine learning algorithms: naïve Bayes, random forest, gradient boosting decision tree (GBDT), and support vector machine. Results: A total of 299 patients were included. The most frequent PH classifications were Group 1 PH (73.9%) and Group 2 PH (23.7%). The mortality within 10 days after delivery was 9.4% and higher in Group 1 PH than in the other PH groups (11.7 vs. 2.6%, P = 0.016). We identified 17 predictors, each with a P-value < 0.05 by univariable analysis, that were associated with an increased risk of death, and the most notable were pulmonary artery systolic pressure (PASP), platelet count, red cell distribution width, N-terminal brain natriuretic peptide (NT-proBNP), and albumin (all P < 0.01). Four prediction models were established using the candidate variables, and the GBDT model showed the best performance (F1-score = 66.7%, area under the curve = 0.93). Feature importance showed that the three most important predictors were NT-proBNP, PASP, and albumin. Conclusion: Mortality remained high, particularly in Group 1 PH. Our study shows that NT-proBNP, PASP, and albumin are the most important predictors of maternal death in the GBDT model. These findings may help clinicians provide better advice regarding fertility for women with PH.

5.
Respir Res ; 23(1): 4, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34996461

ABSTRACT

BACKGROUND: The exact risk assessment is crucial for the management of connective tissue disease-associated interstitial lung disease (CTD-ILD) patients. In the present study, we develop a nomogram to predict 3­ and 5-year mortality by using machine learning approach and test the ILD-GAP model in Chinese CTD-ILD patients. METHODS: CTD-ILD patients who were diagnosed and treated at the First Affiliated Hospital of Zhengzhou University were enrolled based on a prior well-designed criterion between February 2011 and July 2018. Cox regression with the least absolute shrinkage and selection operator (LASSO) was used to screen out the predictors and generate a nomogram. Internal validation was performed using bootstrap resampling. Then, the nomogram and ILD-GAP model were assessed via likelihood ratio testing, Harrell's C index, integrated discrimination improvement (IDI), the net reclassification improvement (NRI) and decision curve analysis. RESULTS: A total of 675 consecutive CTD-ILD patients were enrolled in this study, during the median follow-up period of 50 (interquartile range, 38-65) months, 158 patients died (mortality rate 23.4%). After feature selection, 9 variables were identified: age, rheumatoid arthritis, lung diffusing capacity for carbon monoxide, right ventricular diameter, right atrial area, honeycombing, immunosuppressive agents, aspartate transaminase and albumin. A predictive nomogram was generated by integrating these variables, which provided better mortality estimates than ILD-GAP model based on the likelihood ratio testing, Harrell's C index (0.767 and 0.652 respectively) and calibration plots. Application of the nomogram resulted in an improved IDI (3- and 5-year, 0.137 and 0.136 respectively) and NRI (3- and 5-year, 0.294 and 0.325 respectively) compared with ILD-GAP model. In addition, the nomogram was more clinically useful revealed by decision curve analysis. CONCLUSIONS: The results from our study prove that the ILD-GAP model may exhibit an inapplicable role in predicting mortality risk in Chinese CTD-ILD patients. The nomogram we developed performed well in predicting 3­ and 5-year mortality risk of Chinese CTD-ILD patients, but further studies and external validation will be required to determine the clinical usefulness of the nomogram.


Subject(s)
Connective Tissue Diseases/complications , Lung Diseases, Interstitial/mortality , Machine Learning , Risk Assessment/methods , China/epidemiology , Connective Tissue Diseases/diagnosis , Connective Tissue Diseases/mortality , Female , Follow-Up Studies , Humans , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/etiology , Male , Middle Aged , Retrospective Studies , Risk Factors , Survival Rate/trends , Time Factors
6.
Onco Targets Ther ; 14: 3555-3565, 2021.
Article in English | MEDLINE | ID: mdl-34113122

ABSTRACT

BACKGROUND: Long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) plays a crucial role in non-small cell lung cancer (NSCLC). Nonetheless, regulatory effects of PVT1 on functions of NSCLC cells remain blurry. METHODS: Relative expression levels of PVT1, miR-551b and FGFR1 mRNA in tumor tissues and cells were examined employing quantitative real-time polymerase chain reaction (qRT-PCR); CCK-8 and BrdU assays were utilized for measuring cell viability and proliferation of H1299 and A549 cells; cell migration and invasion were detected deploying Transwell assay; dual-luciferase assay was used for the validation of binding sequence between PVT1 and miR-551b. FGFR1 expression in protein level was quantified employing Western blot. RESULTS: PVT1 was highly expressed in NSCLC tissues and cell lines, whereas miR-551b expression was down-regulated. Overexpression of PVT1 potentiated viability, proliferation, migration and invasion of NSCLC cells while miR-551b inhibited the biological behaviors mentioned above. MiR-551b was predicted and then confirmed as a direct downstream target of PVT1. Meanwhile, a negative correlation was observed between PVT1 expression and miR-551b expression in NSCLC tissues. Besides, PVT1 could increase FGFR1 expression by repressing miR-551b expression. CONCLUSION: PVT1 promotes the proliferation, migration and invasion of NSCLC cells by indirectly mediating FGFR1 via targeting miR-551b.

7.
Int Immunopharmacol ; 97: 107690, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33940323

ABSTRACT

It has recently been shown that expression levels of tissue factor (TF) are high in the serum and peripheral blood mononuclear cells of patients with asthma. However, whether TF impacts airway inflammation and remodelling in asthma remains unknown. The aim of this study was to investigate the effect of TF in asthma airway inflammation and remodelling using a house dust mite (HDM)-induced chronic asthma model and human bronchial epithelial (16HBE) cells. A chronic asthma model was constructed in BALB/c mice by the intranasal instillation of HDM. Mice were treated with short hairpin TF (shTF), and airway inflammation and remodelling features of asthma and epithelial-mesenchymal transition (EMT) were assessed. 16HBE cells were induced by transforming growth factor-ß1 (TGF-ß1) and HDM in the presence or absence of shTF; then, EMT markers and invasion and migration ability were determined. TF expression increased in the lung tissue and 16HBE cells when exposed to HDM. TF downregulation in the lung significantly reduced airway hyperresponsiveness, eosinophil inflammation, the EMT process, and levels of interleukin (IL)-4, IL-6, IL-13, and TGF-ß1 in bronchoalveolar lavage fluid of asthmatic mice. Moreover, TF downregulation inhibited migration and incursion and decreased the expression levels of fibronectin 1 and TGF-ß1, but increased the expression of E-cadherin in HDM- and TGF-ß1-stimulated 16HBE cells. These results demonstrated that TF promoted airway pathological features by enhancing the EMT of bronchial epithelial cells both in vitro and in mice with house dust mite-induced asthma.


Subject(s)
Allergens/immunology , Asthma/immunology , Dermatophagoides pteronyssinus/immunology , Thromboplastin/metabolism , Airway Remodeling/immunology , Animals , Asthma/pathology , Bronchi/cytology , Bronchi/immunology , Bronchi/pathology , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/immunology , HEK293 Cells , Humans , Mice , Specific Pathogen-Free Organisms , Thromboplastin/genetics , Up-Regulation/immunology
8.
PLoS One ; 15(8): e0238416, 2020.
Article in English | MEDLINE | ID: mdl-32857826

ABSTRACT

Fangcang shelter hospitals were established in China during the coronavirus disease 2019 (COVID-19) pandemic as a countermeasure to stop the spread of the disease. To our knowledge, no research has been conducted on mental health problems among patients in Fangcang shelter hospitals. This study aimed to determine the prevalence and major influencing factors of anxiety and depressive symptoms among COVID-19 patients admitted to Fangcang shelter hospitals. From February 23, 2020, to February 26, 2020, we obtained sociodemographic and clinical characteristics information of COVID-19 patients in Jianghan Fangcang Shelter Hospital (Wuhan, China) and assessed their mental health status and sleep quality. Data were obtained with an online questionnaire. The questionnaire consisted of a set of items on demographic characteristics, a set of items on clinical characteristics, the Self-Rating Anxiety Scale, Self-Rating Depression Scale, and Pittsburgh Sleep Quality Index. Three hundred seven COVID-19 patients who were admitted to Jianghan Fangcang Shelter Hospital participated in this study. The prevalence of anxiety and depressive symptoms were 18.6% and 13.4%, respectively. Poor sleep quality and having ≥ two current physical symptoms were independent risk factors for anxiety symptoms. Female sex, having a family member with confirmed COVID-19, and having ≥ two current physical symptoms were independent risk factors for depressive symptoms. Anxiety and depressive symptoms were found to be common among COVID-19 patients in Fangcang Shelter Hospital, with some patients being at high risk.


Subject(s)
Anxiety/epidemiology , Coronavirus Infections/psychology , Depression/epidemiology , Pneumonia, Viral/psychology , Adult , Betacoronavirus , COVID-19 , China/epidemiology , Coronavirus Infections/epidemiology , Cross-Sectional Studies , Female , Hospitals, Special , Humans , Male , Middle Aged , Mobile Health Units , Pandemics , Pneumonia, Viral/epidemiology , Prevalence , Risk Factors , SARS-CoV-2
9.
Oncol Rep ; 44(3): 1025-1036, 2020 09.
Article in English | MEDLINE | ID: mdl-32705261

ABSTRACT

Emerging evidence has demonstrated that abnormally expressed long non­coding (lnc) RNAs contribute to drug resistance in various types of malignancy. LncRNA antisense non­coding RNA in the inhibitor of cyclin­dependent kinase 4 locus (ANRIL) exerts oncogenic activity and acts as a key player in a variety of carcinomas, including non­small cell lung cancer. The present study aimed to investigate the functional roles of ANRIL in cisplatin (DDP) resistance of lung cancer and the underlying mechanism involved in the competing endogenous RNA regulatory network. The expression levels of ANRIL and microRNA (miR)­98 in lung cancer tissues and DDP­resistant lung cancer cells were assessed by reverse transcription­quantitative (RT­q) PCR. The Pearson's correlation coefficient was used to determine the correlation between the expression levels of ANRIL and miR­98 in lung cancer tissues. Cell proliferation and apoptosis were detected by MTT assay and flow cytometry analysis, respectively. The expression levels of proliferation­ and apoptosis­associated proteins were detected by western blot analysis. The potential interaction between ANRIL and miR­98 was confirmed by RNA immunoprecipitation (RIP), dual luciferase reporter assay and RT­qPCR. The results of the present study demonstrated that ANRIL was upregulated and miR­98 was downregulated in lung cancer tissues and A549/DDP cells. In addition, the Pearson's correlation analysis revealed that the expression of ANRIL was significantly negatively correlated with that of miR­98 in lung cancer tissues. ANRIL overexpression reversed the DDP­induced cell proliferation suppression and apoptosis in lung cancer cells, whereas ANRIL knockdown exhibited the opposite effects. RIP, dual luciferase reporter assay and RT­qPCR analysis results demonstrated that ANRIL directly interacted with miR­98 and suppressed miR­98 expression. Furthermore, transfection with the miR­98 mimics promoted DDP­induced cell proliferation inhibition and apoptosis in lung cancer cells, and these effects were partially reversed by ANRIL overexpression. In conclusion, ANRIL knockdown inhibited the development of DDP resistance by upregulating miR­98, providing novel insights into the molecular mechanism of ANRIL involvement in DDP resistance of lung cancer cells.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Adult , Aged , Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cisplatin/therapeutic use , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung/pathology , Lung/surgery , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Middle Aged , Pneumonectomy , RNA, Long Noncoding/genetics , Up-Regulation
10.
Mol Cell Biol ; 39(12)2019 06 15.
Article in English | MEDLINE | ID: mdl-30988156

ABSTRACT

Long noncoding RNAs (lncRNAs) are involved in various human diseases. Recently, H19 was reported to be upregulated in fibrotic rat lung and play a stimulative role in bleomycin (BLM)-induced pulmonary fibrosis in mice. However, its expression in human fibrotic lung tissues and mechanism of action remain unclear. Here, our observations showed that H19 expression was significantly upregulated and that of microRNA 140 (miR-140) was markedly reduced in pulmonary fibrotic tissues from idiopathic pulmonary fibrosis (IPF) patients and transforming growth factor ß1 (TGF-ß1)-induced HBE and A549 cells. Moreover, the expression of H19 was negatively correlated with the expression of miR-140 in IPF tissues. H19 knockdown attenuated TGF-ß1-induced pulmonary fibrosis in vitro Furthermore, animal experiments showed that H19 knockdown attenuated BLM-induced pulmonary fibrosis in mice. The study of molecular mechanisms showed that H19 functioned via reduction of miR-140 expression by binding to miR-140. The increase of miR-140 inhibited TGF-ß1-induced pulmonary fibrosis, and H19 upregulation diminished the inhibitory effects of miR-140 on TGF-ß1-induced pulmonary fibrosis, which was involved in the TGF-ß/Smad3 pathway. Taken together, our findings showed that H19 knockdown attenuated pulmonary fibrosis via the regulatory network of lncRNA H19-miR-140-TGF-ß/Smad3 signaling, and H19 and miR-140 might represent therapeutic targets and early diagnostic and prognostic biomarkers for patients with pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Signal Transduction , A549 Cells , Animals , Bleomycin/adverse effects , Cell Line , Disease Models, Animal , Early Diagnosis , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Markers/genetics , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Mice , Smad3 Protein/metabolism , Transforming Growth Factor beta1/pharmacology
11.
Cell Physiol Biochem ; 47(4): 1482-1496, 2018.
Article in English | MEDLINE | ID: mdl-29940571

ABSTRACT

BACKGROUND/AIMS: Thymic stromal lymphopoietin (TSLP) is a cytokine that plays diverse roles in the regulation of immune responses. However, a detailed understanding of the TSLP signaling pathway in asthma remains elusive. In this study, we aimed to investigate the role of the TSLP signaling pathway in asthma and its effect on airway inflammation and remodeling. METHODS: Forty Sprague Dawley (SD) rats were evenly classified into control, asthma, IgG2a mAb and anti-TSLP mAb groups. Ovalbumin (OVA)-induced asthma models were successfully established. Blood, bronchoalveolar lavage fluid (BALF) and lung tissue samples were prepared. Total BALF leukocytes were counted, and the proportions of different leukocyte types were determined. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry were performed to determine the mRNA and protein levels of TSLP, OX40L, α-smooth muscle actin (α-SMA, a marker of airway remodeling in asthma) and collagen I in the plasma. Enzyme-linked immunosorbent assay (ELISA) was carried out to measure the concentrations of TSLP, OX40L, and other inflammatory factors, such as interferon (IFN)-γ, interleukin (IL)-4, IL-5 and IL-13, in the plasma. RESULTS: Compared with the control group, there were more leukocytes, increased EOS and LYM proportions, higher Underwood and PAS scores, increased WTt, WTm, WAt/A0, WAm/WAt, WTt/R0, WTm/WTt, TSLP, OX40L, a-SMA and collagen I mRNA and protein levels, and higher SLP, OX40L, IL-4, IL-5 and IL-13 levels, but lower MON proportions and IFN-γ levels in the asthma and IgG2a mAb groups. Compared with the asthma and IgG2a mAb groups, there were less leukocytes, decreased EOS and LYM proportions, lower Underwood and PAS scores, decreased WTt, WTm, WAt/A0, WAm/WAt, WTt/R0, WTm/WTt, TSLP, OX40L, a-SMA and Collagen I mRNA and protein levels, and lower levels of SLP, OX40L, IL-4, IL-5 and IL-13, but higher MON proportions and IFN-γ levels in the anti-TSLP mAb group. WTm and WTt were positively associated with the TSLP, OX40L, α-SMA and collagen-I levels in the rat lung tissues. CONCLUSION: The results indicate that TSLP may be an important contributor for asthma development as TSLP signaling blockade attenuates airway inflammation and remodeling in asthmatic rats.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Cytokines/immunology , Signal Transduction/immunology , Animals , Asthma/pathology , Rats , Rats, Sprague-Dawley , Thymic Stromal Lymphopoietin
12.
Genet Test Mol Biomarkers ; 22(6): 333-342, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29812958

ABSTRACT

BACKGROUND: Noncoding RNAs (ncRNAs), primarily microRNAs and long ncRNAs, play important roles in lung cancer. However, the role of long ncRNA (lncRNA)-X-inactive specific transcript (XIST) in non-small-cell lung cancer (NSCLC) is unclear. The purpose of this study was to explore the biologic function and potential mechanism of XIST in NSCLC progression. MATERIALS AND METHODS: XIST, miR-137, and Notch-1 expression were detected by quantitative real-time PCR (qRT-PCR). Levels of proliferation- and epithelial-mesenchymal transition (EMT)-related proteins were assessed by Western blot. The correlations between XIST and miR-137, as well as miR-137 and Notch-1, were evaluated by bioinformatic analysis and luciferase reporter assays. RESULTS: We confirmed that XIST is aberrantly upregulated in NSCLC tissues and cell lines. XIST depletion inhibited cell proliferation and TGF-ß1-induced EMT in A549 and H1299 cells. Spearman's correlation analysis showed an inverse correlation between miR-137 and XIST in NSCLC tissues, and miR-137 levels were found to be aberrantly reduced in A549 and H1299 cells. Furthermore, XIST could act as an endogenous sponge by directly binding to miR-137, negatively regulating its expression. miR-137 overexpression inhibited proliferation and TGF-ß1-induced EMT in A549 and H1299 cells, whereas XIST could reverse the inhibitory effect of miR-137 on proliferation and TGF-ß1-induced EMT. In addition, Notch-1 was identified as a direct target gene of miR-137, with the XIST-miR-137 axis regulating activation of the Notch-1 pathway. CONCLUSION: We identified a branch of the XIST/miR-137/Notch-1 pathway that regulates proliferation and TGF-ß1-induced EMT in NSCLC, which could be involved in NSCLC progression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Knockdown Techniques , Lung Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Receptor, Notch1/metabolism , Transforming Growth Factor beta1/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line , Female , Gene Regulatory Networks , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Signal Transduction , Up-Regulation
13.
Exp Cell Res ; 368(2): 174-183, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29733812

ABSTRACT

OBJECTIVES: This study aimed to investigate whether autophagy mediated cell proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) under hypoxia and to validate the underlying mechanism. METHODS: The rat model of chronic hypoxia-induced pulmonary vascular remodeling was established. Rat primary PASMCs were isolated and cultured in vitro in hypoxia or normoxia to explore the underlying mechanism. RESULTS: Hypoxia exposure for 21 days induced pulmonary vascular remodeling in rats, accompanied by enhanced proliferation and decreased apoptosis, as well as induced expression of autophagic marker LC3II in their lungs. Furthermore, in vitro assays revealed that hypoxia exposure significantly promoted cell proliferation and migration, but inhibited apoptosis of PASMCs compared with normoxia treatment. Besides, hypoxia exposure led to an increase of cell fractions in S phase and a decrease in G0/G1 of PASMCs, whereas had no significant effect on cell population in G2/M phase. Moreover, the autophagy inhibitor 3-MA significantly decreased the hypoxia-induced upregulated expression of LC3II in PASMCs and abrogated the effect of hypoxia on cell proliferation, cell apoptosis, cell cycle, and cell migration of PASMCs. Additionally, inhibition of NF-κB pathway by PDTC suppressed the hypoxia-induced upregulation of NF-κB activity, LC3II, and cell cycle regulators (cyclin D1, CDK4, and CDK6) in PASMCs, and abolished the hypoxia-mediated regulation of cell proliferation, apoptosis, cycle, and migration of PASMCs. CONCLUSION: In summary, hypoxia could induce autophagy activation through NF-κB pathway, and thereby regulate cell proliferation and migration to induce pulmonary vascular remodeling.


Subject(s)
Autophagy/physiology , Cell Movement/physiology , Cell Proliferation/physiology , Hypoxia/metabolism , Pulmonary Artery/metabolism , Signal Transduction/physiology , Vascular Remodeling/physiology , Animals , Apoptosis/physiology , Cell Cycle/physiology , Cells, Cultured , Disease Models, Animal , Hypoxia/physiopathology , Male , Microtubule-Associated Proteins/metabolism , Pulmonary Artery/physiopathology , Rats , Up-Regulation/physiology
14.
Biochem J ; 475(12): 1995-2008, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29626160

ABSTRACT

Activation of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome received substantial attention recently in inflammatory diseases. Macrophages contribute to allergic inflammation in asthma. The present study was aimed to investigate the effect of NLRP3 inflammasome on the polarization of macrophages. We utilized human primary monocytes and monocyte-derived macrophages to study the expression of NLRP3 inflammasome components (NLRP3, apoptosis-associated specklike protein, and caspase-1) and its downstream cytokine interleukin-1ß (IL-1ß). By gain- or loss-of-function assays, we next explored the effects of NLRP3 inflammasome on M1/M2 polarization and secretion of IL-4, interferon-γ, tumor necrosis factor-α, and IL-1ß. The results showed increased numbers of M2 cells in asthma. And NLRP3 inflammasome was activated and involved in the inflammation of asthma. Furthermore, silence of NLRP3 down-regulated IL-4 secretion and up-regulated M1/M2. In contrast, overexpression of NLRP3 increased IL-4 and decreased M1/M2. As expected, IL-4 was involved in NLRP3-mediated down-regulation of Ml/M2 ratio. Moreover, NLRP3 interacted with IRF4 and was required for optimal IRF4-dependent IL-4 transcription. Subsequently, deficiency of NLRP3 in ovalbumin-induced allergic asthmatic mice impaired lung inflammation and up-regulated M1/M2, and diminished IL-4 in bronchoalveolar lavage fluid. Collectively, we demonstrated here that activation of NLRP3 was engaged in the promotion of asthma. NLRP3, but not the inflammasome adaptor ASC or caspase-1, promoted the polarization of M2 macrophages through up-regulating the expression of IL-4, thereby contributing to its regulation of asthma.


Subject(s)
Asthma/metabolism , Inflammasomes/metabolism , Interleukin-4/biosynthesis , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Up-Regulation , Asthma/pathology , Caspase 1/metabolism , Humans , Interferon Regulatory Factors/metabolism , Macrophages/pathology
15.
Connect Tissue Res ; 59(6): 581-592, 2018 11.
Article in English | MEDLINE | ID: mdl-29649906

ABSTRACT

BACKGROUND: Acute lung injury (ALI) is a type of severe pulmonary inflammatory disease with high rates of morbidity and mortality. Now, an increasing number of studies suggest that lncRNAs may act as key regulators of the inflammatory response and play a crucial role in the pathogenesis of many inflammatory diseases. Our study firstly explored the function and underlying mechanism of lncRNA metastasis-associated lung adenocarcinoma transcription 1 (MALAT1) in regulating the inflammatory response of lipopolysaccharide (LPS)-induced ALI in rats. METHODS: The ALI rats were constructed by intratracheal instillation with LPS. Hematoxylin and eosin (HE) for histological examination were performed to detect histopathological changes in the lung tissues. Enzyme-linked immunosorbent assay (ELISA) was used to determine the concentrations of cytokines TNF-α, IL-6, and IL-1ß in the supernatants of the bronchoalveolar lavage fluid (BALF). Quantitative real-time PCR (qRT-PCR) analysis was employed to assess the expression of MALAT1, miR-146a, TNF-α, IL-6, and IL-1ß in lung tissues. Luciferase reporter assay and RNA immunoprecipitation (RIP) assay were used to detect the relationship between MALAT1 and miR-146a. RESULTS: The results revealed that MALAT1 knockdown played a protective role in the LPS-induced ALI rat model. In addition, knockdown of MALAT1 in vitro inhibited LPS-induced inflammatory response in murine alveolar macrophages cell line MH-S and murine alveolar epithelial cell line MLE-12. This study found that MALAT1 acts as a molecular sponge for miR-146a and MALAT1 negatively regulated miR-146a expression. Mechanistically, MALAT1 overexpression alleviated the inhibitory effect of miR-146a on LPS-induced inflammatory response in MH-S. CONCLUSIONS: Together, our study provided the first evidence that MALAT1 knockdown could suppress inflammatory response by up-regulating miR-146a in LPS-induced ALI, which provided a potential therapeutic target for the treatment of ALI.


Subject(s)
Acute Lung Injury/metabolism , Lipopolysaccharides/toxicity , MicroRNAs/biosynthesis , RNA, Long Noncoding/metabolism , Up-Regulation/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Gene Knockdown Techniques , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Rats , Rats, Sprague-Dawley
16.
Clin Respir J ; 12(3): 986-990, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28127951

ABSTRACT

OBJECTIVE: To explore the diagnostic value of joint detection of soluble B7-H4 (sB7-H4) and carcinoembryonic antigen (CEA) in identifying malignant pleural effusion (MPE) from benign pleural effusion (BPE). METHODS: A total of 97 patients with pleural effusion specimens were enrolled from The First Affiliated Hospital of Zhengzhou University between June 2014 and December 2015. All cases were categorized into malignant pleural effusion group (n = 55) and benign pleural effusion group (n = 42) according to etiologies. Enzyme-linked immunosorbent assay was applied to examine the levels of sB7-H4 in pleural effusion and meanwhile CEA concentrations were detected by electro-chemiluminescence immunoassays. Receiver operating characteristic (ROC) curve was established to assess the diagnostic value of sB7-H4 and CEA in pleural effusion. The correlation between sB7-H4 and CEA levels was analyzed by Pearson's product-moment. RESULTS: The concentrations of sB7-H4 and CEA in MPE exhibited obviously higher than those of BPE ([60.08 ± 35.04] vs. [27.26 ± 9.55] ng/ml, P = .000; [41.49 ± 37.16] vs. [2.41 ± 0.94] ng/ml, P = .000). The AUC area under ROC curve of sB7-H4 and CEA was 0.884 and 0.954, respectively. Two cutoff values by ROC curve analysis of sB7-H4 36.5 ng/ml and CEA 4.18 ng/ml were obtained, with a corresponding sensitivity (81.82%, 87.28%), specificity (90.48%, 95.24%), accuracy (85.57%, 90.72%), positive predictive value (PPV) (91.84%, 96.0%), negative predictive value (NPV) (79.17%, 85.11%), positive likelihood ratio (PLR) (8.614, 18.327), and negative likelihood ratio (NLR) (0.201, 0.134). When sB7-H4 and CEA were combined to detect pleural effusion, it obtained a higher sensitivity 90.91% and specificity 97.62%. Furthermore, correlation analysis result showed that the level of sB7-H4 was correlated with CEA level (r = .770, P = .000). CONCLUSIONS: sB7-H4 was a potentially valuable tumor marker in the differentiation between BPE and MPE. The combined detection of sB7-H4 and CEA could improve the diagnostic sensitivity and specificity for MPE.


Subject(s)
Carcinoembryonic Antigen/metabolism , Pleura/metabolism , Pleural Effusion, Malignant/metabolism , Pleural Effusion/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Aged , Biomarkers, Tumor/metabolism , Carcinoembryonic Antigen/immunology , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Pleura/pathology , Pleural Effusion/pathology , Pleural Effusion, Malignant/pathology , Predictive Value of Tests
17.
Clin Respir J ; 12(5): 1882-1890, 2018 May.
Article in English | MEDLINE | ID: mdl-29227032

ABSTRACT

PURPOSE: The aim of this study was to assess the incidence of pulmonary thromboembolism (PTE) in patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD),and to evaluate the efficacy and safety of low-dose urokinase (UK) thrombolysis therapy when treating hemodynamically stable AECOPD patients with acute PTE (AECOPD-PTE). METHODS: A total of 419 AECOPD patients, including 96 AECOPD-PTE, were enrolled. A total of 30 AECOPD-PTE patients were collected retrospectively, and 66 AECOPD-PTE patients were prospectively divided into anticoagulation-only, low-dose UK and standard-dose UK groups. Follow-up 1 year, we evaluated the efficacy and safety of low-dose UK therapy for hemodynamically stable AECOPD-PTE patients. RESULTS: The incidence of PTE in AECOPD patients was 22.9% (96/419), which increased with COPD severity degree ranging from 3.5% (2/57) in mild, 13.6% (19/140) in moderate and 33.8% (75/222) in severe subgroups (P < .05). In the prospective study, the total effective rate of low-dose UK group 97.2% (35/36) was higher than that in anticoagulation 75.0% (12/16) and standard-dose UK group 78.6% (11/14) respectively (P < .05). In the follow-up, the adverse events rate in low-dose UK group 8.3% (3/36) was significantly lower than that in anticoagulation group 25.0% (4/16) and standard-dose UK group 71.4% (10/14) respectively (P < .05). In addition, the mean PTE recurrence time of low-dose UK group (9.0 ± 0) months was longer than anticoagulation group (2.0 ± 1.41) months (P < .05). AECOPD relapse time in anticoagulation, low-dose UK and standard-dose UK groups corresponding to (8.5 ± 2.12), (9.0 ± 0) and (8.8 ± 3.40) months were compared with no significant difference (P > .05). CONCLUSIONS: The incidence of PTE in AECOPD patients was 22.9%, especially with higher occurrence rate in severe COPD. Compared with anticoagulation-only therapy, low-dose UK treatment (500 000 IU/day for 5-7 days) could obtain a better efficacy and safety in hemodynamically stable AECOPD patients with acute PTE, corresponding with a higher effective rate (97.2%) and lower adverse events rate (8.3%) respectively.


Subject(s)
Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Embolism/epidemiology , Urokinase-Type Plasminogen Activator/pharmacology , Aged , Anticoagulants/pharmacology , Disease Progression , Female , Humans , Incidence , Male , Middle Aged , Prospective Studies , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/metabolism , Retrospective Studies , Risk Factors , Severity of Illness Index , Urokinase-Type Plasminogen Activator/administration & dosage
18.
Oncotarget ; 8(40): 67933-67941, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978085

ABSTRACT

Injury to airway smooth muscle (ASM) cells hallmarks the pathological progression of asthma, a chronic inflammatory airway disease. MicroRNAs (miRNAs) play essential roles in the development of asthma as well as airway remodeling. Here we studied the involvement of miRNAs in the regulation of autophagic survival of ASM cells and airway disorder. We analyzed autophagy-associated factors LC3 and Beclin-1 by RT-qPCR and protein blotting in purified airway smooth muscle cells from ovalbumin (OVA)-induced asthmatic mice. The biological activity of miRNA targeting Beclin-1 was explored by bioinformatics method and confirmed in a dual luciferase reporter assay. Loss of function experiment was performed via transplantation of miRNA in OVA-induced asthmatic mice. We detected high autophagy levels in ASM cells, which appeared to result from augmentation of Beclin-1 protein, rather than Beclin-1 mRNA, suggesting presence of post-transcriptional control of Beclin-1. Next, miR-384 was figured out to be a Belcin-1-targeting miRNA, which significantly decreased after OVA treatment. Mechanistically, binding of miR-384 to 3'-UTR of Beclin-1 mRNA potently suppressed Beclin-1 protein translation in ASM cells, similar to previous finding in another cell type. In vivo, transplantation of miR-384 significantly attenuated Belcin-1 protein levels in ASM cells, resulting in reduced autophagy of ASM cells and attenuation of asthmatic features by OVA. Together, these data suggest that re-expression of miR-384 may reduce augmentation of Beclin-1-dependent autophagy of ASM cells, as a novel promising treatment for asthma.

19.
Oncotarget ; 8(28): 46312-46325, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28564633

ABSTRACT

This study aims to explore how microRNA-145 (miR-145) affects airway remodeling and cytokine expression by targeting epidermal growth factor receptor (EGFR) to regulate mucin 5AC (MUC5AC).Mice alveolar epithelial cells (AECs) were divided into a control, blank, miR-145 mimics, mimic control, miR-145 inhibitors, inhibitor control, si-EGFR and miR-145 inhibitors + si-EGFR group. Asthma mice models with airway remodeling were induced with an Ovalbumin (OVA) solution and randomly divided into a normal, asthma, asthma + miR-145 mimics, asthma + miR-145 mimic control, asthma + si-EGFR or asthma + si-EGFR NC group. Airway remodeling degree and histomorphology was measured using hematoxylin-eosin (HE), Masson and periodic acid-Schiff (PAS) staining. Flow cytometry was used to detect Th2 and Th17 cells in peripheral blood, ELISA was used to measure inflammatory factors. qRT-PCR and western blotting was adapted to detect the expressions of EGFR and the relevant cytokines that are regulated by miR-145.The control, miR-145 mimics and si-EGFR groups showed a higher expression of miR-145 and a lower expression of EGFR and cytokines than the blank, mimic control, inhibitor control and miR-145 inhibitor + si-EGFR groups. Mice in the asthma + miR-145 mimics and asthma + si-EGFR groups showed lower WAt/Pbm, WAi/Pbm and WAm/Pbm, less inflammatory cells, less airway modeling and alleviated goblet cell hyperplasia and mucus obstruction than the asthma group. Furthermore, the expressions of EGFR and cytokines of transfected cells and lung tissues were negatively related to those of miR-145. MiR-145 can down-regulate MUC5AC by negatively targeting EGFR and thereby relieving airway remodeling.


Subject(s)
Airway Remodeling/genetics , Alveolar Epithelial Cells/metabolism , Cytokines/genetics , ErbB Receptors/genetics , Gene Expression Regulation , MicroRNAs/genetics , Mucin 5AC/genetics , RNA Interference , Animals , Cytokines/metabolism , ErbB Receptors/metabolism , Gene Expression , Genes, Reporter , Immunohistochemistry , Male , Mice , Models, Biological , Mucin 5AC/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
20.
Oncol Rep ; 36(4): 2282-8, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27574108

ABSTRACT

Forkhead box P3 (Foxp3) is a member of forkhead box transcription factor family and it was identified as a tumor suppressor in various solid tumors. This study evaluated the expression of Foxp3 in non-small cell lung cancer (NSCLC) and investigated its role in epithelial­mesenchymal transition (EMT) of cancer cells. qRT-PCR and western blot analysis were used for examining the expression of Foxp3 in NSCLC tissues and the non-tumor tissues. A tissue microarray was constructed and scored for evaluating the clinical significance of Foxp3 expression in NSCLC tissues. RNAi was employed for downregulating Foxp3 expression and cell proliferation was done with MTT assay. Transwell with or without basement membrane matrix was used for cell migration and invasion assay respectively. Foxp3 was found downregulated in NSCLC tissues compared with non-tumoral tissues; downregulation of Foxp3 predicted adverse tumor stage and overall survival; silencing of FOXP3 promoted the proliferation, migration and invasion ability of NSCLC cells and influenced the expression level of EMT-associated proteins. However, forced expression of Foxp3 could reverse this effect. Moreover, Foxp3 could interact with LMO2 and affect the expression level of TAL1, which was in accordance with the findings in T-cell acute lymphoblastic leukemia. By screening the signalling pathways, we observed an obvious upregulation of phosphorylated NF-κB in A549 and H520 cells after silencing of FOXP3. Our results suggest that Foxp3 suppressed NSCLC cell metastasis, at least partially, via NF-κB signaling.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Epithelial-Mesenchymal Transition/genetics , Forkhead Transcription Factors/biosynthesis , Transcription Factor RelA/biosynthesis , A549 Cells , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Prognosis , Signal Transduction/genetics , Transcription Factor RelA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...