Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Allergol Immunopathol (Madr) ; 50(6): 84-92, 2022.
Article in English | MEDLINE | ID: mdl-36335450

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is a common chronic intestinal disease. Protopine isolated from different plants has been investigated to understand its special functions on varied diseases. However, the regulatory effects of protopine on the progression of IBD remain unclear. Our study is aimed to explore the effects of protopine on the progression of IBD and its underlying regulatory mechanism of action. METHODS: The cell viability was assessed through MTT colorimetric assay. The protein expressions of genes were examined by Western blot analysis. The cell apoptosis and reactive oxygen species level were measured using flow cytometry. The levels of inflammation and oxidative stress-related proteins were tested through enzyme-linked-immunosorbent serologic assay. The intracellular Ca2+ concentration and mitochondrial membrane potential were measured through immunofluorescence assay. RESULTS: First, different concentrations of lipopolysaccharide (LPS) were treated with NCM460 cells to establish IBD cell model, and 5-µg/mL LPS was chosen for followed experiments. In this study, we discovered that protopine relieved the LPS-induced inhibited intestinal epithelial cell viability and enhanced cell apoptosis. Moreover, protopine attenuated LPS-stimulated inflammation activation and oxidative stress. Further experiments illustrated that the increased intracellular Ca2+ concentration and decreased mitochondrial membrane potential stimulated by LPS were reversed by protopine treatment. Finally, through Western blot analysis, it was demonstrated that protopine retarded the activated NLR family pyrin domain containing 3 (NLRP3) and nuclear factor kappa B (NF-κB) signaling pathways mediated by LPS. CONCLUSION: Protopine alleviated LPS-triggered intestinal epithelial cell injury by inhibiting NLRP3 and NF-κB signaling pathways to reduce inflammation and oxidative stress. This discovery may provide a useful drug for treating IBD.


Subject(s)
Inflammatory Bowel Diseases , NF-kappa B , Humans , NF-kappa B/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , Inflammation/drug therapy , Oxidative Stress , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/drug therapy
2.
Histol Histopathol ; 37(11): 1143-1150, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35852131

ABSTRACT

It has been reported that HITT can inhibit colon cancer. However, the role of HITT in gastric cancer (GC) is unknown. Our preliminary sequencing data revealed the altered expression of HITT in GC and its close correlation with miR-602, suggesting the involvement of HITT and its potential interaction with miR-602 in GC. This study explored the role of HITT and its crosstalk with miR-602 in GC. In this study, the expression of HITT, premature and mature miR-602 in paired GC and normal tissues (62 patients) was detected by RT-qPCR. RNA pull-down assay was performed to evaluate the direct interaction between HITT and mature miR-602. The subcellular location of HITT was assessed by nuclear fractionation assay. The role of HITT in regulating miR-602 maturation was explored by overexpression assay. Cell apoptosis was analyzed by flow cytometry. Our data illustrated that HITT was highly upregulated and mature miR-602 was downregulated in GC. No alteration in premature miR-602 in GC was observed. HITT was located in both nucleus and cytoplasm, and it can directly interact with miR-602. In addition, overexpression of HITT in GC cells increased the expression levels of mature miR-602 but not premature miR-602. Overexpression of HITT further increased GC cell apoptosis and suppressed the role of miR-602 in inhibiting GC cell apoptosis. In conclusion, HITT may promote GC cell apoptosis by suppressing the maturation of miR-602.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Stomach Neoplasms , Humans , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Apoptosis , MicroRNAs/genetics
3.
Arab J Gastroenterol ; 23(3): 188-194, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35514011

ABSTRACT

BACKGROUND AND STUDY AIMS: This study aimed to investigate the effect of Forkhead Box A2 (FOXA2) on migration, invasion, and epithelial-mesenchymal transition (EMT) of colorectal cancer (CRC) cells in hypoxia and explore its related molecular mechanisms. PATIENTS AND METHODS: A cellular hypoxia model was established, and the FOXA2 overexpression vector was transfected into SW480 and HCT116 cells. Cell apoptosis, migration, and invasion were examined by flow cytometry, scratch test, and transwell-invasion assay. Next, the hsa-let-7 g gene expression was detected by quantitative reverse transcription-polymerase chain reaction. Relative protein levels of HIF-1, FOXA2, c14orf28, E-cadherin, N-cadherin, and Vimentin were detected by western blot. RESULTS: Hypoxia-exposed CRC cells showed a significantly increased cell apoptosis rate, as well as enhanced cell invasion and migration abilities compared with the cells in normoxia. FOXA2 overexpression induced apoptosis and inhibited hypoxia-exposed CRC cell migration and invasion. Additionally, FOXA2 overexpression led to the significantly increased hsa-let-7 g and E-cadherin expression, as well as the decreased c14orf28, N-cadherin, and Vimentin expression in hypoxic CRC cells. CONCLUSIONS: This study demonstrated that FOXA2 could affect the apoptosis, migration, invasion, and EMT of CRC cells under hypoxia conditions. FOXA2 transcriptionally activates hsa-let-7 g to inhibit hypoxia-induced EMT by targeting c14orf28.


Subject(s)
Colorectal Neoplasms , Epithelial-Mesenchymal Transition , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/genetics , Vimentin/genetics , Vimentin/metabolism , Vimentin/pharmacology
4.
Am J Transl Res ; 14(2): 1376-1383, 2022.
Article in English | MEDLINE | ID: mdl-35273740

ABSTRACT

OBJECTIVE: This study was designed to investigate the expression of serum autophagy-related protein P62 in patients with severe acute pancreatitis (AP) and its correlation with prognosis. METHODS: Eighty patients with AP treated in the First Affiliated Hospital of Gannan Medical University from January 2020 to January 2021 were enrolled as study subjects in this retrospective analysis, and they were placed into the mild AP group (n=52) or the severe AP group (n=28). According to clinical outcomes, these 80 patients were divided into a good prognosis group (GP group, n=51, surviving without serious complications such as organ failure) and a poor prognosis group (PP group, n=29, death or developing organ failure). The differences in C-reactive protein (CRP), P62 and the Acute Physiology and Chronic Health Evaluation II (APACHE-II) were compared upon admission. The changes of CRP, P62 and APACHE-II within 1-7 h after admission were dynamically analyzed in the two groups. Spearman correlation analysis was performed to explore the correlation between P62 and APACHE-II scores, and the receiver operating characteristic (ROC) curve of P62 related to poor AP outcome was plotted. RESULTS: CRP, P62 and APACHE-II in the mild AP group were significantly higher than those in the severe AP group, and these in the PP group were also significantly higher than those in the GP group (P<0.05). Dynamic monitoring showed that within 1-7 h after admission, CRP, P62, and APACHE-II in the severe AP group were significantly higher than those in the mild AP group (P<0.05), and these in the PP group were significantly higher than those in the GP group (P<0.05). Spearman correlation analysis showed that P62 level was significantly positively correlated with both CRP and APACHE-II (r=0.9331, r=0.9500, P<0.0001). ROC curve showed that AUC of P62 was 0.9570 in AP patients with poor prognosis (95% CI=0.8939-1.000, P<0.0001). CONCLUSION: Serum autophagy-related protein P62 was closely related to the condition and prognosis of AP patients, and P62 could be used as a potential indicator to assess the condition and prognosis of AP patients.

5.
Genet Mol Biol ; 45(1): e20210067, 2022.
Article in English | MEDLINE | ID: mdl-35167648

ABSTRACT

Gastric carcinoma (GC) is a malignant tumor that has high mortality and morbidity worldwide. Although many efforts have been focused on the development and progression of GC, the underlying functional regulatory mechanism of GC needs more clarification. Metallothionein 1G (MT1G) is a member of the metallothionein family (MTs), and hypermethylation of MT1G occurred in a variety of cancers, including gastric cancer. However, the functional mechanism of MT1G in GC remains unclear. Here, we demonstrated that MT1G was down-regulated in GC tissues and cells. Overexpression of MT1G inhibited cell proliferation, foci formation and cell invasion, while knockdown of MT1G increased cell proliferation, foci formation and cell invasion. In addition, MT1G overexpression inhibited cell cycle progression and MT1G deficiency exerted opposite phenotype. p-AKT was negatively regulated by MT1G. In summary, our study reveals that MT1G exerts crucial role in regulating of cell proliferation and migration of gastric cancer, providing new insights for MT1G-related pathogenesis and a basis for developing new strategies for treatment of GC.

6.
Tissue Cell ; 73: 101598, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34333382

ABSTRACT

Colorectal cancer (CRC) is one of the most common tumors that has a high incidence worldwide. Targeted therapy for CRC has received much attention recently. It is still necessary to develop novel and promising therapeutic targets to improve the prognosis. SYNPO2, also known as synapsopoprotein 2 or myopod, encodes actin binding proteins and has been characterized as a tumor suppressor for aggressive cancers. SYNPO2 has been reported to inhibit the activity of YAP/TAZ. However, whether SYNPO2 could regulate the progression of CRC through the YAP/YAZ signaling pathway remains unclear. Herein, it was found that the expression of SYNPO2 was low in hypoxia-exposed CRC cells, consistent with the data from TCGA database. SYNPO2 inhibited the growth of CRC cells upon hypoxia treatment and promoted the cell apoptosis. Additionally, SYNPO2 inhibited the migration and epithelial-mesenchymal transformation (EMT) CRC cell upon hypoxia treatment. Mechanically, the results demonstrated that SYNPO2 suppressed hypoxia-induced progression of CRC by regulating YAP-Kruppel like factor 5 (KLF5) axis. Therefore, SYNPO2 can serve as a promising therapeutic target for CRC treatment.


Subject(s)
Cell Movement , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Kruppel-Like Transcription Factors/metabolism , Microfilament Proteins/metabolism , Signal Transduction , Tumor Hypoxia , YAP-Signaling Proteins/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Tumor Hypoxia/genetics
7.
Bioengineered ; 12(1): 1311-1323, 2021 12.
Article in English | MEDLINE | ID: mdl-33896384

ABSTRACT

Gastric cancer is a considerable health burden worldwide. DNA methylation, a major epigenetic phenomenon, is closely related to the pathogenesis of cancer. Neuronal pentraxin II (NPTX2) has been found to be hypermethylated in several cancers such as glioblastoma and pancreatic cancer. However, the roles of NPTX2 in gastric cancer have not been reported. To explore this issue, NPTX2 expression in gastric cancer cells was assessed by western blot and quantitative real-time polymerase chain reaction (qRT-PCR). The methylation analysis of NPTX2 was performed by qRT-PCR as well as methylation-specific PCR (MS-PCR). The effects of NPTX2 on gastric cancer cell proliferation, apoptosis and cell cycle were detected by colony formation, CCK-8 and flow cytometry assays, respectively. The interaction of NPTX2 with the p53 signaling pathway was evaluated by western blot. Our study found the down-regulated expression of NPTX2 in gastric cancer cells compared with human gastric mucosal cells. In addition, the hypermethylation of NPTX2 was observed in gastric cancer cells, which was correlated with the low expression of NPTX2. Moreover, NPTX2 inhibited gastric cancer cell proliferation, inhibited apoptosis and induced cell cycle arrest. Furthermore, NPTX2 enhanced the protein expression of p53, p21 and PTEN to activate the p53 signaling pathway. Therefore, NPTX2 hypermethylation caused the downregulation of NPTX2 expression, which could promote cell proliferation, inhibit apoptosis and cause cell cycle arrest in gastric cancer cells by suppressing the p53 signaling pathway. Therefore, NPTX2 may be crucial for the progression of gastric cancer.


Subject(s)
Apoptosis/genetics , C-Reactive Protein/metabolism , Cell Cycle Checkpoints/genetics , DNA Methylation/genetics , Nerve Tissue Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , C-Reactive Protein/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
8.
Drug Des Devel Ther ; 9: 3989-4104, 2015.
Article in English | MEDLINE | ID: mdl-26347199

ABSTRACT

NADPH oxidases (NOXs) are a predominant mediator of redox homeostasis in hepatic stellate cells (HSCs), and oxidative stress plays an important role in the pathogenesis of liver fibrosis. Ursolic acid (UA) is a pentacyclic triterpenoid with various pharmacological activities, but the molecular targets and underlying mechanisms for its antifibrotic effect in the liver remain elusive. This study aimed to computationally predict the molecular interactome and mechanistically investigate the antifibrotic effect of UA on oxidative stress, with a focus on NOX4 activity and cross-linked signaling pathways in human HSCs and rat liver. Drug-drug interaction via chemical-protein interactome tool, a server that can predict drug-drug interaction via chemical-protein interactome, was used to predict the molecular targets of UA, and Database for Annotation, Visualization, and Integrated Discovery was employed to analyze the signaling pathways of the predicted targets of UA. The bioinformatic data showed that there were 611 molecular proteins possibly interacting with UA and that there were over 49 functional clusters responding to UA. The subsequential benchmarking data showed that UA significantly reduced the accumulation of type I collagen in HSCs in rat liver, increased the expression level of MMP-1, but decreased the expression level of TIMP-1 in HSC-T6 cells. UA also remarkably reduced the gene expression level of type I collagen in HSC-T6 cells. Furthermore, UA remarkably attenuated oxidative stress via negative regulation of NOX4 activity and expression in HSC-T6 cells. The employment of specific chemical inhibitors, SB203580, LY294002, PD98059, and AG490, demonstrated the involvement of ERK, PI3K/Akt, and p38 MAPK signaling pathways in the regulatory effect of UA on NOX4 activity and expression. Collectively, the antifibrotic effect of UA is partially due to the oxidative stress attenuating effect through manipulating NOX4 activity and expression. The results suggest that UA may act as a promising antifibrotic agent. More studies are warranted to evaluate the safety and efficacy of UA in the treatment of liver fibrosis.


Subject(s)
Antioxidants/pharmacology , Computational Biology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Experimental/drug therapy , Liver/drug effects , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Triterpenes/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Dimethylnitrosamine , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Gene Expression Regulation , Hepatic Stellate Cells/enzymology , Hepatic Stellate Cells/pathology , Humans , Liver/enzymology , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/enzymology , Liver Cirrhosis, Experimental/pathology , Male , Matrix Metalloproteinase 1/metabolism , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Interaction Maps , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors , Tissue Inhibitor of Metalloproteinase-1/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Ursolic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...