Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Ann Transl Med ; 9(7): 563, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33987261

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a fatal chronic pulmonary fibrosis disease and pathological mechanisms of fibrogenesis in IPF are still to be elucidated. Here, we investigated the potential role of Nogo-B in pulmonary fibrogenesis. METHODS: A mouse model of pulmonary fibrosis was established by intratracheal injection of bleomycin (BLM). Lung epithelial cells MLE-12 and TC-1 JHU-1 were cultured for TGF-ß treatment. The extent of lung fibrosis was evaluated using hematoxylin and eosin (HE) staining and Masson staining in model mice and Nogo-B knockout mice. The protein levels of Nogo-B, endoplasmic reticulum stress (ERS) sensors including PERK, IRE1α, ATF6 and epithelial-mesenchymal transition (EMT) markers including E-cadherin and N-cadherin, vimentin were assayed by Western blotting respectively after Nogo-B knockdown or overexpression with lentivirus. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate cytokine levels of TGF-ß, TNF-α, IL-1ß, IL-6 and IL-10 in bronchoalveolar lavage fluid (BALF). RESULTS: Nogo-B expression was up-regulated in lung tissues of fibrosis model mice and alveolar epithelial cells. Nogo-B knockdown significantly attenuated lung fibrogenesis, downregulated the levels of inflammatory cytokines, inhibited EMT as well as decreased the level of phosphor-PERK/PERK but not the levels of phosphor-IRE1α/IRE1α and c-ATF6. Additionally, a potential efficacy of PERK blockade was demonstrated in improving the extent of lung fibrosis in model mice. CONCLUSIONS: This study discovered that involvement of Nogo-B in pulmonary fibrogenesis was associated with the PERK branch of ERS pathway and EMT. Nogo-B could be considered as a potential therapeutic target for the treatment of IPF.

2.
Free Radic Biol Med ; 147: 159-166, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31874250

ABSTRACT

OBJECTIVE: To investigate the role of geranylgeranyl diphosphate synthase 1 (GGPPS1) in ventilator-induced lung injury along with the underlying mechanism. METHODS: A murine VILI model was induced by high-tidal volume ventilation in both wild-type and GGPPS1 knockout mice. GGPPS1 expression was detected in the bronchoalveolar lavage fluid (BALF) supernatants of acute respiratory distress syndrome (ARDS) patients and healthy volunteers, as well as in lung tissues and BALF supernatants of the VILI mice using enzyme-linked immunosorbent assay (ELISA), quantitative reverse transcription polymerase chain reaction (qRT-PCR), western bolt and immunohistochemical (IHC). The wet/dry ratio, total BALF proteins, and lung injury score were analyzed. The percentage of neutrophils was detected by flow cytometry and IHC. Inflammatory cytokine levels were measured by ELISA and qRT-PCR. The related expression of Toll-like receptor (TLR)2/4 and its downstream proteins was evaluated by western blot. RESULTS: GGPPS1 in BALF supernatants was upregulated in ARDS patients and the VILI mice. Depletion of GGPPS1 significantly alleviated the severity of ventilator induced lung injury in mice. Total cell count, neutrophils and inflammatory cytokines (interleukin [IL]-6, IL-1ß, IL-18 and tumor necrosis factor-α) levels in BALF were reduced after GGPPS1 depletion. Moreover, addition of exogenous GGPP in GGPPS-deficient mice significantly exacerbated the severity of ventilator induced lung injury as compared to the PBS treated controls. Mechanistically, the expression of TLR2/4, as well as downstream proteins including activator protein-1 (AP-1) was suppressed in lung tissues of GGPPS1-deficient mice. CONCLUSION: GGPPS1 promoted the pathogenesis of VILI by modulating the TLR2/4-AP-1 signaling pathway, and GGPPS1 knockout significantly alleviated the lung injury and inflammation in the VILI mice.


Subject(s)
Transcription Factor AP-1 , Ventilator-Induced Lung Injury , Animals , Bronchoalveolar Lavage Fluid , Farnesyltranstransferase , Humans , Lung , Mice , Multienzyme Complexes , Signal Transduction , Toll-Like Receptor 2/genetics , Ventilator-Induced Lung Injury/genetics
3.
Am J Physiol Lung Cell Mol Physiol ; 316(3): L567-L577, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30652497

ABSTRACT

Inhibition of the mevalonate pathway using statins has been shown to be beneficial in the treatment of acute lung injury (ALI). Here, we investigated whether partial inhibition of this pathway by targeting geranylgeranyl pyrophosphate synthase large subunit 1 (GGPPS1), a catalase downstream of the mevalonate pathway, was effective at treating lung inflammation in ALI. Lipopolysaccharide (LPS) was intratracheally instilled to induce ALI in lung-specific GGPPS1-knockout and wild-type mice. Expression of GGPPS1 in lung tissues and alveolar epithelial cells was examined. The severity of lung injury and inflammation was determined in lung-specific GGPPS1 knockout and wild-type mice by measuring alveolar exudate, neutrophil infiltration, lung injury, and cell death. Change in global gene expression in response to GGPPS1 depletion was measured using mRNA microarray and verified in vivo and in vitro. We found that GGPPS1 levels increased significantly in lung tissues and alveolar epithelial cells in LPS-induced ALI mice. Compared with wild-type and simvastatin treated mice, the specific deletion of pulmonary GGPPS1 attenuated the severity of lung injury by inhibiting apoptosis of AECs. Furthermore, deletion of GGPPS1 inhibited LPS-induced inflammasome activation, in terms of IL-1ß release and pyroptosis, by downregulating NLRP3 expression. Finally, downregulation of GGPPS1 reduced the membrane expression of Ras-related protein Rab10 and Toll-like receptor 4 (TLR4) and inhibited the phosphonation of IκB. This effect might be attributed to the downregulation of GGPP levels. Our results suggested that inhibition of pulmonary GGPPS1 attenuated LPS-induced ALI predominantly by suppressing the NLRP3 inflammasome through Rab10-mediated TLR4 replenishment.


Subject(s)
Acute Lung Injury/drug therapy , Lipopolysaccharides/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Pneumonia/drug therapy , Acute Lung Injury/metabolism , Animals , Inflammasomes/drug effects , Inflammasomes/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Mice, Transgenic , Pneumonia/metabolism
4.
Cytokine ; 110: 381-388, 2018 10.
Article in English | MEDLINE | ID: mdl-29803659

ABSTRACT

OBJECTIVE: We investigated the effect of topotecan on injury and inflammation in a model of ventilator-inducedlunginjury (VILI). METHODS: Acute lung injury (ALI) was induced in mice by high-tidal volume ventilation, and the mice were then treated with topotecan or PBS. Lung tissue and bronchoalveolar lavage fluid were collected to assess pulmonary vascular leaks, inflammation, and cell apoptosis. RESULTS: Compared to PBS treatment, topotecan significantly decreased the ALI score, myeloperoxidase (MPO) content, total protein concentration, and presence of inflammatory cells and inflammatory cytokines in bronchoalveolar lavage fluid. Topotecan also reduced caspase-3 activation and type Ⅱ alveolar epithelial cell apoptosis. Moreover, topotecan inhibited NF-κB expression and activation in the VILI model. CONCLUSION: Topotecan alleviates acute lung injury in the model of VILI through the inhibition of the NF-κB pathway.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , NF-kappa B/metabolism , Topotecan/pharmacology , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/metabolism , Animals , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid/chemistry , Caspase 3/metabolism , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Peroxidase/metabolism
5.
Clin Respir J ; 12(4): 1607-1614, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28960939

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) regulate a variety of genes and biological processes. Lnc-IL7R plays a considerable role in the regulation of inflammation, but its prognostic potential in acute respiratory distress syndrome (ARDS) has not been fully explained. In this study, the role of lnc-IL7R as a potential biomarker in ARDS was examined. OBJECTIVE: Role of lnc-IL7R as potential biomarker in ARDS. METHODS: LncRNA-IL7R was isolated from the plasma of patients with ARDS and healthy controls and clinical indexes were obtained within 24 h after admission. The relative expression of lnc-IL7R was obtained by quantitative real-time PCR. The correlations between lnc-IL7R and continuous variables in ARDS were tested using Spearman's coefficients. RESULTS: A total of 85 ARDS patients and 49 healthy controls were included. Plasma lnc-IL7R was significantly down-regulated in ARDS compared with the levels in healthy control individuals, especially in severe ARDS (P < .01). The area under the curve (AUC) of lnc-IL7R for ARDS diagnosis was 0.87 (sensitivity 75.3%, specificity 93.9%). The lnc-IL7R levels were correlated with the severity of ARDS (ρ = -0.31, P = .0215), oxygenation index (ρ = 0.61, P < .001), APACHE II score (ρ = -0.04, P = .0230), CRP (ρ = -0.26, P = .0148) and WBC (ρ = -0.29, P = .0064). Lnc-IL7R relative value ≥ 0.33 showed the lower 28-day mortality in the patients with ARDS(P < .05).The survivors showed higher lnc-IL7R level and lower APACHE II score, SOFA score and length of mechanical ventilation than in the non-survivors (P = .0109, P < .001, P < .001 and P = .017, respectively). CONCLUSIONS: Lnc-IL7R is a novel biomarker for the diagnosis of ARDS and predicts the severity of ARDS and 28-day mortality in this patients cohort. TRIAL REGISTRATION: The study was registered with the Chinese Clinical Trial Registry (ChiCTR-DOD-16008657).


Subject(s)
Gene Expression Regulation , RNA, Long Noncoding/blood , Receptors, Interleukin-7/blood , Respiration, Artificial/methods , Respiratory Distress Syndrome/blood , Biomarkers/blood , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Prospective Studies , RNA, Long Noncoding/genetics , ROC Curve , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-7/genetics , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/therapy
6.
J Thorac Dis ; 9(7): 2054-2060, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28840006

ABSTRACT

BACKGROUND: Protein regulator of cytokinesis-1 (PRC1) has been shown to participate in the completion of cytokinesis, and it is dysregulated in cancer processes. However, its relevance in lung squamous cell carcinoma (SCC) remained largely unknown. We aimed to study the expression pattern of PRC1 and assess its clinical significance in lung SCC. METHODS: PRC1 protein expression in human lung SCC and adjacent normal lung tissues was detected by immunohistochemistry. PRC1 expression was assessed in association with clinicopathological features and clinical outcomes of lung SCC patients. RESULTS: In lung SCC tissues, PRC1 protein expression was significantly higher than those in paired normal lung tissues. The lung SCC patients with PRC1 overexpression had an advanced pathological stage (TNM stage), positive lymph node metastasis, and a shorter overall survival (OS) time more frequently than patients with low PRC1 expression. Additional, PRC1 expression was also shown to be poor as a prognostic factor for OS in patients with lung SCC. CONCLUSIONS: Our study indicated that aberrant expression of PRC1 may point to biochemical recurrence in lung SCC. This highlights its potential as a valuable prognostic marker for lung SCC.

7.
Mol Cancer ; 16(1): 108, 2017 06 24.
Article in English | MEDLINE | ID: mdl-28646916

ABSTRACT

BACKGROUND: Protein regulator of cytokinesis-1 (PRC1) belongs to the microtubule-associated proteins (MAPs) family, and is involved in cytokinesis. Recent investigations suggest PRC1 involvement in human carcinogenesis, including breast carcinoma, hepatocellular carcinoma and etc. However, whether PRC1 contributes to lung adenocarcinoma tumorigenesis remains unknown. METHODS: Quantitative reverse-transcription polymerase chain reaction (qRT-PCR), Western blotting and Immunohistochemical staining (IHC) were used to evaluate and contrast the PRC1 expression profile in lung adenocarcinoma and adjacent normal lung tissues. We examined the clinical use of PRC1 in lung adenocarcinoma prognosis. Additionally, the tumorigenesis impact of PRC1 in lung adenocarcinoma cells was verified via in vitro and in vivo metastasis and tumorigenesis assays. Notably, Next Generation Sequencing (NGS) was performed to investigate the molecular mechanism underlying the oncogenic role of PRC1 in lung adenocarcinoma. RESULTS: PRC1 mRNA and protein expressions were upregulated in lung adenocarcinoma tissues compared to adjacent normal lung tissues. PRC1 protein overexpression correlated with lymph node metastasis and was an independent poor prognostic factor for lung adenocarcinoma patients. Our data implied that PRC1 depletion limited the proliferation and invasion of lung adenocarcinoma cells in vitro and lowered tumor development and lung metastasis in vivo. Remarkably, limiting PRC1 substantially prompted G2/M phase cell cycle arrest and apoptosis. Mechanistically, by conducting NGS on PRC1-depleted A549 cells and control cells, we discovered that PRC1 expression was significantly correlated with the Wnt signaling pathway. CONCLUSIONS: This investigation offers confirmation that PRC1 is a prognostic and promising therapeutic biomarker for people with lung adenocarcinoma and takes on a key part in the activation of the Wnt/ß-catenin pathway in lung adenocarcinoma development.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Wnt Signaling Pathway/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Aged , Animals , Apoptosis/genetics , Cell Line, Tumor , Female , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice, Inbred BALB C , Middle Aged , Prognosis , Xenograft Model Antitumor Assays , beta Catenin/genetics , beta Catenin/metabolism
8.
J Cell Mol Med ; 21(4): 665-676, 2017 04.
Article in English | MEDLINE | ID: mdl-27862966

ABSTRACT

NCAPG2 is a component of the condensin II complex and contributes to chromosome segregation via microtubule-kinetochore attachment during mitosis. It is well known that NCAPG2 plays a critical role in cell mitosis; however, the role of altered NCAPG2 expression and its transcriptional regulatory function in cancer development remains mostly unknown. Here, for the first time we reported that NCAPG2 was evidently increased in non-small cell lung cancer tissues compared to adjacent normal lung tissues. Clinicopathological data analysis showed that NCAPG2 overexpression was significantly correlated with lymph node metastasis and pathologic-Tumour Nodes Metastasen stages, and was an independent prognostic factor in lung adenocarcinoma patients. Moreover, siRNA-mediated knockdown of NCAPG2 could inhibit tumour cell growth of lung adenocarcinoma cells (A549 and H1299) in vitro and could significantly lead to cell cycle arrest in the G2 phase. Furthermore, we found that NCAPG2 silencing significantly decreased the expression levels of G2/M phase cell cycle-related protein expressions (Cyclin B1, Cdc2) and increased the expression levels of p27 and p21 through Western blot analysis. Taken together, we demonstrated that increased NCAPG2 expression could regulate cell proliferation and identified as a poor prognostic biomarker in lung adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Chromosomal Proteins, Non-Histone/genetics , G2 Phase , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mitosis , Adenocarcinoma of Lung , Aged , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Chromosomal Proteins, Non-Histone/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing , Humans , Male , Multivariate Analysis , Prognosis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/metabolism , Survival Analysis , Up-Regulation/genetics , Polo-Like Kinase 1
9.
Sheng Li Xue Bao ; 65(4): 445-50, 2013 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-23963076

ABSTRACT

Nogo-B is a major family member of the reticulon protein family 4. It is widely expressed in the central nervous system and peripheral tissues, and is mainly located in endoplasmic reticulum and cell membrane. Previous studies have revealed that Nogo-B plays a key role in vascular injury, tissue repair and inflammation process. It also may be critical for apoptosis of tumor cells and central diseases. Further investigation of the molecular characteristics and biological function of Nogo-B might be of great help to understand its role in diverse diseases.


Subject(s)
Myelin Proteins/physiology , Animals , Apoptosis , Cell Membrane/physiology , Endoplasmic Reticulum/physiology , Humans , Inflammation , Nogo Proteins
10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 28(10): 1046-50, 2012 Oct.
Article in Chinese | MEDLINE | ID: mdl-23046936

ABSTRACT

AIM: To investigate the effect of co-stimulation of mimic viral infection [polyinosinic: polycytidylic acid, poly(I:C)] and endotoxin (lipopolysaccharide, LPS) on chemotactic factors production of human bronchial epithelial cells and explore its related mechanism. METHODS: Human bronchial epithelial cells (16HBE) were challenged by co-stimulation of different concentrations of poly(I:C) and LPS. Some other 16HBE cells, before the co-stimulation of poly(I:C) and LPS, were pretreated with dexamethasone and p38MAPK specific inhibitor (SB203580), respectively. The levels of IL-8/CXCL8 and IP-10/CXCL1 mRNA transcription were detected by RT-PCR after 6 h challenge. The contents of IL-8 and IP-10 proteins were detected by ELISA after 24 h challenge. RESULTS: (1) The mRNA and protein expression of IL-8, not IP-10, increased under 10 µg/mL LPS stimulation compared with control group. The mRNA and protein expressions of both IL-8 and IP-10 were elevated significantly by the co-stimulation of LPS and 0.1 µg/mL poly (I:C) compared with control group and simple LPS groups. (2) The mRNA and protein expressions of IL-8 and IP-10 increased under the challenge of different concentrations of poly(I:C) (0.001, 0.01, 0.1 µg/mL) in a concentration-dependent manner, which showed no change after adding 10 µg/mL LPS. (3) Dexamethasone (1 µmol/L) and SB203580 (20 µmol/L) significantly decreased both the mRNA and protein production of IL-8 and IP-10 induced by co-stimulation of 0.1 µg/mL poly (I:C) and 10 µg/mL LPS compared with the control group and control+DMSO group respectively (P<0.01 and P<0.05). Inhibitive effect of dexamethasone was stronger than that of p38MAPK inhibitor. CONCLUSIONS: The co-stimulation of poly(I:C) and LPS can induce chemokine expression of airway epithelial cells. Poly(I:C) enhances the IL-8 expression induced by single LPS challenge, which suggests that viral infection could enhance the inflammation resulted from bacterial colonization in airway. Glucocorticoid has the greater effect on the inhibition of chemotactic factors production than p38MAPK inhibitor. These two drugs have a potential therapeutic effect on AECOPD caused by viral infection.


Subject(s)
Chemotactic Factors/genetics , Epithelial Cells/immunology , Gene Expression Regulation , Lipopolysaccharides/immunology , Poly I-C/immunology , Respiratory Mucosa/immunology , Cell Line , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemotactic Factors/immunology , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Interleukin-8/biosynthesis , Interleukin-8/genetics , Interleukin-8/immunology , Lipopolysaccharides/pharmacology , Poly I-C/pharmacology , Respiratory Mucosa/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...