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.
World J Gastroenterol ; 27(3): 240-254, 2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33519139

ABSTRACT

BACKGROUND: Recent studies have demonstrated that circular RNA AKT3 (circAKT3) plays a crucial role in regulating the malignant phenotypes of tumor cells. However, the potential effects of circAKT3 on esophageal cancer have not been investigated. AIM: To illuminate the role of circAKT3 in malignant behaviors of esophageal cancer cells and its underlying mechanism. METHODS: Clinical samples were collected to detect the expression of circAKT3. The role of circAKT3 in proliferation, migration, invasion, and apoptosis of esophageal cancer cells was evaluated using Cell Counting Kit-8, wound healing assays, Transwell assays, and fluorescence analysis, respectively. The target of circAKT3 was screened and identified using an online database and luciferase reporter assay. A xenograft nude mouse model was established to investigate the role of circAKT3 in vivo. RESULTS: In vitro assays showed that proliferative, migratory, and invasive capacities of esophageal cancer cells were significantly enhanced by circAKT3 overexpression. Furthermore, miR-17-5p was screened as the target of circAKT3, and miR-17-5p antagonized the effects of circAKT3 on esophageal cancer cells. Moreover, we identified RHOC and STAT3 as the direct target molecules of miR-17-5p, and circAKT3 facilitated expression of RHOC and STAT3 by inhibiting miR-17-5p. In vivo assays showed circAKT3 knockdown inhibited growth of esophageal cancer. CONCLUSION: CircAKT3 contributed to the malignant behaviors of esophageal cancer in vitro and in vivo by sponging miR-17-5p thus providing a potential target for treatment of esophageal cancer.


Subject(s)
Esophageal Neoplasms , MicroRNAs , Animals , Cell Line, Tumor , Cell Proliferation , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mice , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt , RNA, Circular
2.
Front Oncol ; 10: 567, 2020.
Article in English | MEDLINE | ID: mdl-32435614

ABSTRACT

Global incidence and mortality associated with hepatocellular carcinoma (HCC) is steadily increasing. Metastasis-associated 1 (MTA1) can induce tumorigenesis and metastatic progression in HCC. However, the mechanistic details of MTA1-mediated regulation of HCC has not been completely defined. Epigenetic histone modification is closely related to tumor development. Histone cluster 1 H1 family member c (H1.2) is important for epigenetic histone modification and chromatin remodeling; however, whether it has a role in HCC tumorigenesis is not known. In the current study, we confirmed that MTA1 promoted HCC cell growth and migration. Our results further show that MTA1 inhibited the phosphorylation of histone cluster 1 H1 family member c (H1.2) at threonine-146 residue (T146) (H1.2T146ph). MTA1 inhibited H1.2T146ph by mediating proteasomal degradation of the DNA protein kinase (DNA-PK). Pharmacological inhibition of proteasomal degradation of DNA-PK or genetic ablation of E3 ligase mouse double minute 2 (MDM2) rescued expression of DNA-PK, and subsequent phosphorylation of H1.2. MTA1's role in HCC was inhibited by ectopic expression of H1.2T146ph in HCC cell lines. Our results showed that H1.2T146ph can bind to MTA1 target genes. Collectively, our study confirms that MTA1 functions as an oncogene and promotes HCC progression. The epigenetic histone modifier H1.2T146ph exerts critical role in the regulation of MTA1-induced tumorigenesis. MTA1 regulates posttranslational activation of H1.2 by regulating the cognate kinase, DNA-PK, via the ubiquitin proteasome system. MTA1 expression was inversely correlated to both DNA-PK and phosphorylated H1.2 in HCC tissue specimens compared to tumor adjacent normal hepatic tissue, revealing that the MTA1/MDM2/DNA-PK/H1.2 is an important therapeutic axis in HCC.

3.
World J Gastroenterol ; 25(32): 4727-4738, 2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31528097

ABSTRACT

BACKGROUND: As the malignant tumor, pancreatic cancer with a meager 5-years survival rate has been widely concerning. However, the molecular mechanisms that result in malignant transformation of pancreatic cells remain elusive. AIM: To investigate the gene expression profiles in normal or malignant transformed pancreas development. METHODS: MaSigPro and ANOVA were performed on two pancreas development datasets downloaded from the Gene Expression Omnibus database. Six pancreatic cancer datasets collected from TCGA database were used to establish differentially expressed genes related to pancreas development and pancreatic cancer. Moreover, gene clusters with highly similar interpretation patterns between pancreas development and pancreatic cancer progression were established by self-organizing map and singular value decomposition. Additionally, the hypergeometric test was performed to compare the corresponding interpretation patterns. Abnormal regions of metabolic pathway were analyzed using the Sub-pathway-GM method. RESULTS: This study established the continuously upregulated and downregulated genes at different stages in pancreas development and progression of pancreatic cancer. Through analysis of the differentially expressed genes, we established the inverse and consistent direction development-cancer pattern associations. Based on the application of the Subpathway-GM analysis, we established 17 significant metabolic sub-pathways that were closely associated with pancreatic cancer. Of note, the most significant metabolites sub-pathway was related to glycerophospholipid metabolism. CONCLUSION: The inverse and consistent direction development-cancer pattern associations were established. There was a significant correlation in the inverse patterns, but not consistent direction patterns.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Pancreas/growth & development , Pancreatic Neoplasms/genetics , Datasets as Topic , Disease Progression , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , Male , Metabolic Networks and Pathways/genetics , Oligonucleotide Array Sequence Analysis , Pancreas/pathology , Pancreatic Neoplasms/pathology , Trans-Activators/genetics
4.
Exp Cell Res ; 371(1): 250-254, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30118695

ABSTRACT

Ninety percent of all cancer related deaths happen due to metastatic progression. One important protein facilitating metastatic progression in hepatocellular carcinoma (HCC) is the metastasis associated 1 protein (MTA-1). We have earlier shown that in the context of HCC and normal liver cell lines, HuH6 and THLE-2, respectively. MTA-1 protein is actively stabilized in HCC cell lines and actively degraded in normal liver cells. We had also shown that TRIM25 is the E3 ligase that interacts with and degrades MTA-1 protein in normal liver cells. However, the exact mechanism by which TRIM25 degrades MTA-1 protein has still not been elucidated. In the study, we used both in situ prediction algorithms and mass spectrometry based post-translational modification analysis to map the lysine residues in MTA-1 that are polyubiquitinated. Whereas UbPred algorithm revealed a combination of medium and low confidence sites, it revealed only one high confidence lysine (K98) residue. The hCKSAAP_UbSite algorithm also predicted K98 site. Mass spectrometry analysis also showed that K98 has ubiquitin modification. Immunofluorescence analysis showed that in normal liver cell line, THLE-2, which has high expression of TRIM25, ectopically expressed FLAG-tagged wild-type MTA-1 was actively degraded, but the K98R mutant MTA-1 was not. In vitro ubiquitination assay using recombinant wild-type and K98R mutant MTA-1 confirmed that MTA-1 is poly-ubiquitinated at K98 residue by TRIM25. The K98R mutant had a longer half-life than wild-type MTA-1 protein in an in vitro protein stability assay. We establish that TRIM25 ubiquitinates MTA-1 at lysine 98 and degrades it normal liver cells.


Subject(s)
Histone Deacetylases/metabolism , Lysine/metabolism , Protein Processing, Post-Translational , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , 3' Untranslated Regions , Amino Acid Sequence , Cell Line , Hepatocytes , Histone Deacetylases/genetics , Humans , Mass Spectrometry , Protein Engineering/methods , Protein Stability , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , Support Vector Machine , Trans-Activators , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination
5.
Exp Cell Res ; 368(1): 37-41, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29654742

ABSTRACT

Tumor metastasis accounts for 90% of all cancer-related deaths. Epithelial to mesenchymal transition (EMT) considered to be centrally important in acquired resistance to chemotherapy and in progression of tumors to secondary organs. One of the important mediators of metastatic progression in hepatocellular carcinoma (HCC) is the metastasis associated protein 1 (MTA-1). We have earlier shown that in the context of HCC and normal liver cell lines, MTA-1 protein is actively stabilized in HCC cell lines and actively degraded in normal liver cells. We have also shown that TRIM25 is the E3 ligase that interacts with and degrades MTA-1 protein. The identity of the factor regulating expression of TRIM25 in normal liver cells and HCC is unknown. In the current work we elucidate that microRNA (miR)- 873 targets TRIM25 in HCC cells. Both metagenomic analysis and quantification of miR-873 and TRIM25 in 25 HCC patients revealed an inverse correlation between the two in HCC patients with high miR-873 and low TRIM25 expression, respectively. The expression pattern was mimicked in the normal liver cells THLE-2 and the HCC cell line, HuH6. In vitro luciferase reporter assays confirmed TRIM25 as the target of miR-873. Transient transfection of HuH6 cells with an anti-miR-873 antagomir significantly decreased both transwell motility in these cells. Furthermore, in in vivo xenograft assays treatment with anti-miR-873 antagomir significantly decreased hepatic nodules formation. Cumulatively, our data indicate that suppression of TRIM25 expression by high levels of miR-873 dictates MTA1 protein upregulation in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Humans , Trans-Activators
6.
IUBMB Life ; 69(10): 795-801, 2017 10.
Article in English | MEDLINE | ID: mdl-28861931

ABSTRACT

Metastasis associated 1 protein (MTA1) is one of the prime facilitators of metastatic progression in all solid tumors including hepatocellular carcinoma (HCC). However, the underlying regulatory mechanism of MTA1 expression in HCC is not clear. In this study, we evaluated MTA1 transcript and protein expression in HCC and normal hepatic cell lines. The results revealed that MTA1 protein expression had a significantly increase in HCC cell line, HuH6, compared with that in normal hepatic cell line, THLE-2. Determination of protein half-life using cycloheximide (CHX) treatment did not reveal any statistically significant difference in protein turn-over rates between THLE-2 (3.3 ± 0.25 h) and HuH6 (3.6 ± 0.15 h) cell lines. MTA1 protein level was stabilized in THLE-2 cells after treatment with MG-132 to levels similar to those observed in HuH6 cells. Mass spectrometric analysis of FLAG immunoprecipitates of FLAG-MTA1 transfected THLE-2 cells after MG-132 treated revealed candidate ubiquitin ligases that were interacting with MTA1. RNAi-mediated silencing of each prospective ubiquitin ligase in THLE-2 cells indicated that knockdown of TRIM25 resulted in stabilization of MTA1 protein, indicating TRIM25 as a putative E3 ligase for MTA1. Coimmunoprecipitation of FLAG-tagged MTA1, but not IgG, in MG-132 treated and untreated THLE-2 cells cotransfected with either FLAG-MTA1 or Myc-TRIM25 revealed robust polyubiquitinated MTA1, confirming that the TRIM25 is the ubiquitin ligase for MTA1 degradation. Overexpression of TRIM25 in HuH6 and RNAi mediated silencing of TRIM25 in THLE-2 cells inhibited and increased the cell migration and invasion, respectively. Analysis of The Cancer Genome Atlas data for assessment of TRIM25 transcript level and MTA1 protein expression in 25 HCC patients confirmed an inverse correlation between the expression of TRIM25 and MTA1. Cumulatively, our data reveal a novel mechanism of post-translational to regulate MTA1 expression in normal hepatic cells, which is repressed in HCC. © 2017 IUBMB Life, 69(10):795-801, 2017.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Hepatocytes/metabolism , Histone Deacetylases/genetics , Liver Neoplasms/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Atlases as Topic , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Cell Movement , Cycloheximide/pharmacology , Disease Progression , Half-Life , Hepatocytes/drug effects , Hepatocytes/pathology , Histone Deacetylases/metabolism , Humans , Leupeptins/pharmacology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Protein Binding , Protein Stability/drug effects , Proteolysis/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/metabolism , Signal Transduction , Trans-Activators , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
7.
Mol Med Rep ; 14(6): 5205-5210, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27779720

ABSTRACT

Previous studies have demonstrated the overexpression of paired basic amino acid cleaving enzyme 4 (PACE4) mRNA in prostate cancer tissues. This overexpression is correlated with higher circulating protein levels in certain patients, however, the role of PACE4 in apoptosis and the potential molecular mechanisms of pancreatic cancer remain to be elucidated. The aim of the present study was to investigate the effect and potential molecular mechanisms of PACE4 on apoptosis in the Panc­1 pancreatic cancer cell line. Cell proliferation was assessed using a Cell Counting Kit­8 assay. Apoptotic nuclear shrinkage was monitored using Hoechst 33258 staining. Caspase­3/7 activities were measured using a colorimetric caspase­glo 3/7 assay. Alterations in protein expression were monitored using Western blot analysis. The results indicated that PACE4 small interfering (si)RNA inhibited cell proliferation and activated caspase­3/7 activities. In addition, PACE4 siRNA significantly increased apoptosis via the activation of caspase­3 and the downregulation of anti­apoptotic proteins, X­linked inhibitor of apoptosis protein and phosphorylated­Akt. In addition, the results showed deregulation of the B cell lymphoma­2 (Bcl­2)-associated X protein/Bcl­2 ratio which led to the release of cytochrome c following PACE4 siRNA transfection. In conclusion, PACE4 siRNA may exert antitumor activity through the mitochondrial pathway and is expected to be a promising therapeutic strategy for the treatment of pancreatic cancer.


Subject(s)
Apoptosis/genetics , Mitochondria/genetics , Mitochondria/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proprotein Convertases/genetics , Serine Endopeptidases/genetics , Signal Transduction , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Expression , Humans , Proprotein Convertases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Serine Endopeptidases/metabolism
8.
PLoS One ; 11(7): e0158657, 2016.
Article in English | MEDLINE | ID: mdl-27387128

ABSTRACT

Liver fibrosis results from a sustained wound healing response to chronic liver injury, and the activation of nonparenchymal hepatic stellate cells (HSCs) is the pivotal process. MicroRNA-34a (miR-34a) is the direct target gene of p53 and activates p53 through sirtuin 1 (SIRT1) simultaneously. The miR-34a/SIRT1/p53 signaling pathway thus forms a positive feedback loop wherein p53 induces miR-34a and miR-34a activates p53 by inhibiting SIRT1, playing an important role in cell proliferation and apoptosis. miR-34a expression has been found to be increased in animal models or in human patients with different liver diseases, including liver fibrosis. However, the exact role of this classical miR-34a/SIRT1/p53 signaling pathway in liver fibrosis remains unclear. In the present study, using a CCl4-induced rat liver fibrosis model, we found that the miR-34a/SIRT1/p53 signaling pathway was activated and could be inhibited by SIRT1 activator SRT1720. Further studies showed that the miR-34a/SIRT1/p53 signaling pathway was activated in hepatocytes but not in HSCs. The activation of this pathway in hepatocytes resulted in the apoptosis of hepatocytes and thus activated HSCs. Our data indicate that the miR-34a/SIRT1/p53 signaling pathway might be a promising therapeutic target for liver fibrosis.


Subject(s)
Liver Cirrhosis/metabolism , MicroRNAs/metabolism , Signal Transduction , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Line , Coculture Techniques , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...