Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Radiol Oncol ; 58(1): 67-77, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38378037

ABSTRACT

BACKGROUND: Gastric cancer is an epidemic malignancy that is commonly diagnosed at the late stage. Evidence has elucidated that RAD54B exerts a crucial role in the progress of various tumors, but its specific role and mechanism in gastric cancer remain gloomy. MATERIALS AND METHODS: The level of RAD54B was detected by western blot. RAD54B expression was downregulated or upregulated in both MKN45 and AGS cells by the transfection of shRAD54B or overexpression plasmid, respectively. The role of RAD54B in the growth, migration, invasion and tube formation of gastric cancer was evaluated by Edu, colony formation, transwell and tube formation assays. In addition, the molecular mechanism of RAD54B in gastric cancer was also determined by western blot. Moreover, in vivo experiment was conducted in xenografted mice. RESULTS: The expression of RAD54B was discovered to be upregulated in gastric cancer based on the ATGC and GEPIA databases, which was also confirmed in gastric cancer cell lines. Moreover, overexpression of RAD54B enhanced the growth, migration, invasion, tube formation and Wnt/ß-catenin signaling axis in AGS and MKN45 cells. As expected, knockdown of RAD54B in AGS and MKN45 cells reversed these promotions. More importantly, in vivo assay also verified that RAD54B accelerated the growth of gastric cancer and Wnt/ß-catenin signaling pathway. CONCLUSIONS: Both loss-of-function and gain-of-function assays demonstrated that RAD54B facilitated gastric cancer cell progress and angiogenesis through the Wnt/ß-catenin axis.


Subject(s)
Stomach Neoplasms , Animals , Mice , Angiogenesis , beta Catenin/metabolism , Cell Line, Tumor , Cell Movement , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Wnt Signaling Pathway , Humans
2.
Tissue Cell ; 76: 101767, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35257941

ABSTRACT

This study aims to investigate the expression levels of fibrinogen α chain (FGA) in human gastric cancer (GC) tissues and cell lines, clarify its role in gastric cancer progression, and explore its underlying mechanism. Bioinformatics analysis, Immunoblot, Immunohistochemical (IHC), and quantitative PCR assays were performed to assess the expression of FGA in human gastric cancer tissues and cell lines. CCK-8 and colony formation assays were performed to detect its role in the proliferation of gastric cancer cells. Wound healing, transwell, and Immunofluorescence were performed to detect its effects on gastric cancer cell motility and epithelial-mesenchymal transition (EMT) processes. Luciferase and CHIP assays were performed to confirm the transcriptional regulation of FGA on ITGA5. Immunoblot assays and double-label RFP-GFP-LC3 immunofluorescence analysis were conducted to detect its effects on gastric cancer cell autophagy and FAK/ERK pathway, and in vivo tumor growth assays were further performed. We found the low expression of FGA in human gastric cancer tissues and cell lines. FGA suppressed gastric cancer cell proliferation, motility, and EMT process, and stimulated cell autophagy. We further found that FGA suppressed the expression of Integrin-α5 (ITGA5) and inhibited the FAK/ERK pathway, therefore suppressing the progression of gastric cancer. The in vivo assays further confirmed that FGA suppressed tumor growth of gastric cancer cells in the BALB/c nude mice (18-22 g, female, 8-week-old) through suppressing ITGA5-mediated FAK/ERK pathway in mice. We demonstrated the mechanism underlying FGA suppressing gastric cancer progression, and therefore we thought FGA could serve as a tumor suppressor protein in gastric cancer.


Subject(s)
Autophagic Cell Death , Fibrinogen , Integrin alpha5 , MAP Kinase Signaling System , Stomach Neoplasms , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Extracellular Matrix Proteins/metabolism , Female , Fibrinogen/genetics , Fibrinogen/metabolism , Integrin alpha5/genetics , Integrin alpha5/metabolism , Mice , Mice, Nude , Neoplasm Metastasis
3.
Mol Med Rep ; 24(4)2021 Oct.
Article in English | MEDLINE | ID: mdl-34396425

ABSTRACT

Gastric cancer (GC) is one of the most common types of malignancy worldwide and is accompanied by both high mortality and morbidity rates. Homeobox B13 (HOXB13) has been reported to act as a tumor suppressor gene in multiple types of human cancer. The present study aimed to investigate the effects and potential underlying molecular mechanisms of HOXB13 in the progression of GC. The expression of HOXB13 in GC cells was first examined using the Cancer Cell Line Encyclopedia database and subsequently validated in a number of GC cell lines. Following HOXB13 overexpression (Ov­HOXB13), HGC­27 cell proliferation was evaluated by colony formation and Cell Counting Kit­8 assays. Wound healing and Matrigel assays were used to determine the migratory and invasive abilities, respectively. Additionally, cell apoptosis was assessed using TUNEL staining, and the expression of apoptosis­related proteins was detected by western blot analysis. Subsequently, TEA domain transcription factor 4 (TEAD4) was overexpressed to evaluate the effects on HGC­27 cell proliferation, migration, invasion and apoptosis following co­transfection with Ov­HOXB13. The potential binding sites of HOXB13 on the vestigial­like family member 4 (VGLL4) promoter were verified using chromatin immunoprecipitation and dual luciferase reporter assays. Moreover, the expression levels of proteins involved in the Hippo signaling pathway were analyzed using western blotting. The results revealed that the expression of HOXB13 was notably lower in GC cells compared with normal gastric cells. The overexpression of HOXB13 significantly inhibited the proliferation, migration and invasion, but promoted the apoptosis of HGC­27 cells. Moreover, Ov­HOXB13 downregulated TEAD4 expression. Notably, Ov­TEAD4 transfection partially reversed the effects of Ov­HOXB13 on the cellular behaviors of HGC­27 cells. HOXB13 was also confirmed to bind with the VGLL4 promoter. The knockdown of VGLL4 restored the inhibitory effects of Ov­HOXB13 on the expression levels of VGLL4 and Hippo pathway signaling proteins. In conclusion, the findings of the present study suggested that Ov­HOXB13 may suppress the proliferation, migration and invasion, and promote the apoptosis of GC cells through the transcriptional activation of VGLL4 to inhibit the involvement of TEAD4 in the Hippo signaling pathway. These results may provide novel and potent targets for the treatment of GC.


Subject(s)
Apoptosis/genetics , Hippo Signaling Pathway/genetics , Homeodomain Proteins/genetics , Stomach Neoplasms/genetics , TEA Domain Transcription Factors/genetics , Transcription Factors/genetics , Transcriptional Activation , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Stomach/pathology , Stomach Neoplasms/metabolism , TEA Domain Transcription Factors/metabolism , Transcription Factors/metabolism , Transcriptome
4.
Front Immunol ; 12: 769685, 2021.
Article in English | MEDLINE | ID: mdl-35003085

ABSTRACT

Both tumour-infiltrating immune cells and inflammation-related genes that can mediate immune infiltration contribute to the initiation and prognosis of patients with colon cancer. In this study, we developed a method to predict the survival outcomes among colon cancer patients and direct immunotherapy and chemotherapy. We obtained patient data from The Cancer Genome Atlas (TCGA) and captured inflammation-related genes from the GeneCards database. The package "ConsensusClusterPlus" was used to generate molecular subtypes based on inflammation-related genes obtained by differential expression analysis and univariate Cox analysis. A prognostic signature including four genes (PLCG2, TIMP1, BDNF and IL13) was also constructed and was an independent prognostic factor. Cluster 2 and higher risk scores meant worse overall survival and higher expression of human leukocyte antigen and immune checkpoints. Immune cell infiltration calculated by the estimate, CIBERSORT, TIMER, ssGSEA algorithms, tumour immune dysfunction and exclusion (TIDE), and tumour stemness indices (TSIs) were also compared on the basis of inflammation-related molecular subtypes and the risk signature. In addition, analyses of stratification, somatic mutation, nomogram construction, chemotherapeutic response prediction and small-molecule drug prediction were performed based on the risk signature. We finally used qRT-PCR to detect the expression levels of four genes in colon cancer cell lines and obtained results consistent with the prediction. Our findings demonstrated a four-gene prognostic signature that could be useful for prognostication in colon cancer patients and designing personalized treatments, which could provide new versions of personalized management for these patients.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Inflammation/genetics , Transcriptome/genetics , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Brain-Derived Neurotrophic Factor/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Gene Ontology , Gene Regulatory Networks , Humans , Immunotherapy/methods , Interleukin-13/genetics , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/metabolism , Nomograms , Phospholipase C gamma/genetics , Prognosis , Tissue Inhibitor of Metalloproteinase-1/genetics , Tumor Microenvironment/genetics
5.
Mol Neurobiol ; 54(7): 5008-5016, 2017 09.
Article in English | MEDLINE | ID: mdl-27534417

ABSTRACT

Human glioma causes substantial morbidity and mortality worldwide. However, the molecular mechanisms underlying glioma progression are still largely unknown. COP1 (constitutively photomorphogenic 1), an E3 ubiquitin ligase, is important in cell survival, development, cell growth, and cancer biology by regulating different substrates. As is well known, both tumor suppressor p53 and oncogenic protein c-JUN could be ubiquitinated and degraded by ubiquitin ligase COP1, which may be the reason that COP1 serves as an oncogene or a tumor suppressor in different cancer types. Up to now, the possible role of COP1 in human glioma is still unclear. In the present study, we found that the expression of COP1 was upregulated in human glioma tissues. The role of COP1 in glioma cell proliferation was investigated using COP1 loss- and gain-of-function. The results showed that downregulation of COP1 by short hairpin RNA (shRNA) inhibited glioma cell proliferation, while overexpression of COP1 significantly promoted it. Furthermore, we demonstrated that COP1 only interacted with and regulated p53, but not c-JUN. Taken together, these results indicate that COP1 may play a role in promoting glioma cell proliferation by interacting with and downregulating tumor suppressor p53 rather than oncogenic protein c-JUN.


Subject(s)
Cell Proliferation , Glioma/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Cell Proliferation/physiology , Down-Regulation , Glioma/pathology , Humans , RNA, Small Interfering/genetics , Transcriptional Activation
6.
J Cell Mol Med ; 20(3): 422-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26612725

ABSTRACT

The ubiquitin ligase neuregulin receptor degradation protein 1 (Nrdp1) is involved in the induction of apoptosis and suppression of tumour formation. We previously showed that it was expressed at lower levels in human glioma tissues compared with normal brain tissues. However, the mechanism underlying this is unclear. Here, we reported that a novel short variant (Nrdp1S), lacking 71 amino acids at the N-terminal, was expressed in normal human brain tissue, but absent from glioma tissues. Similar to Nrdp1, Nrdp1S could be degraded by the proteasomal pathway, but exhibited an even longer half-life than Nrdp1. Nrdp1S was also shown to form a heterodimer with Nrdp1, which increased its stability, thereby augmenting the Nrdp1-mediated ubiquitination and degradation of ErbB3. EdU incorporation, MTT assay and in vitro colony formation demonstrated that Nrdp1S significantly inhibited the cell tumourigenicity. These results together suggest that Nrdp1S is a tumour suppressor that which potentiates the Nrdp1-mediated ubiquitination and degradation of ErbB3. An Nrdp1S deficiency may also be an important factor in the loss of Nrdp1.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Receptor, ErbB-3/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitination , Base Sequence , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease Progression , Glioma/pathology , HEK293 Cells , Humans , Protein Isoforms/physiology , Protein Stability , Proteolysis
7.
J Neurooncol ; 124(3): 357-64, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26088461

ABSTRACT

We previously reported that loss of Nrdp1 contributes to human glioma progression by reducing apoptosis. However, the role of Nrdp1 in glioma migration and invasion has not been investigated. Here, we report that ErbB3, a substrate of Nrdp1, is undetectable in normal brain tissues and grade II/III glioma tissues, but is abundant in a certain percentage of grade IV glioma tissues and is associated with the loss of Nrdp1. This suggests that Nrdp1 may be involved in glioma migration and invasion by regulating ErbB3. Thus, the role of Nrdp1/ErbB3 signaling in glioma cell migration and invasion was investigated using Nrdp1 loss- and gain-of-function. The results show that down-regulation of Nrdp1 by use of short hairpin RNA promoted glioma cell migration and invasion. In contrast, overexpression of Nrdp1 significantly inhibited glioma cell migration and invasion. Further investigation on molecular targets revealed that Nrdp1 decreased the level of ErbB3, which resulted in decreasing p-AKT thereby reducing cytoplasmic p27(Kip1). Taken together, these findings suggest that Nrdp1-mediated ErbB3 degradation suppresses glioma migration and invasion and that loss of Nrdp1 may amplify ErbB3 signaling to contribute to glioma migration and invasion. These findings suggest that Nrdp1 may be a target for glioma therapy.


Subject(s)
Brain Neoplasms/pathology , Cell Movement/physiology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Glioma/pathology , Receptor, ErbB-3/metabolism , Ubiquitin-Protein Ligases/genetics , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Neoplasm Invasiveness/genetics , Receptor, ErbB-3/genetics , Retroviridae/genetics , Transfection , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...