Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Cell J ; 26(1): 13-27, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38351726

ABSTRACT

OBJECTIVE: This paper aimed to investigate the PI3K/Akt/mTOR signal-pathway regulator factor-related lncRNA signatures (PAM-SRFLncSigs), associated with regulators of the indicated signaling pathway in patients with lung adenocarcinoma (LUAD) undergoing immunotherapy. MATERIALS AND METHODS: In this retrospective study, we employed univariate Cox, multivariate Cox, and least absolute shrinkage and selection operator (LASSO) regression analyses to identify prognostically relevant long non-coding RNAs (lncRNAs), construct prognostic models, and perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Subsequently, immunoassay and chemotherapy drug screening were conducted. Finally, the prognostic model was validated using the Imvigor210 cohort, and tumor stem cells were analyzed. RESULTS: We identified seven prognosis-related lncRNAs (AC084757.3, AC010999.2, LINC02802, AC026979.2, AC024896.1, LINC00941 and LINC01312). We also developed prognostic models to predict survival in patients with LUAD. KEGG enrichment analysis confirmed association of LUAD with the PI3K/Akt/mTOR signaling pathway. In the analysis of immune function pathways, we discovered three good prognostic pathways (Cytolytic_activity, Inflammation-promoting, T_cell_co-inhibition) in LUAD. Additionally, we screened 73 oncology chemotherapy drugs using the "pRRophetic" algorithm. CONCLUSION: Identification of seven lncRNAs linked to regulators of the PI3K/Akt/mTOR signaling pathway provided valuable insights into predicting the prognosis of LUAD, understanding the immune microenvironment and optimizing immunotherapy strategies.

2.
Mol Carcinog ; 63(5): 912-925, 2024 May.
Article in English | MEDLINE | ID: mdl-38390729

ABSTRACT

Golgi phosphoprotein 3 (GOLPH3) has been reported as an oncogene in various tumors; however, the role and function of GOLPH3 and its relevant molecular mechanism in cholangiocarcinoma (CCA) are unclear. Herein, GOLPH3 expression in CCA tissues was observed to be significantly higher than that in paired adjacent noncancerous tissues. Clinicopathological analysis showed that GOLPH3 expression correlated positively with the tumor-node-metastasis stage. In addition, GOLPH3 expression correlated inversely with the overall survival of patients with CCA. Multivariate analysis showed that GOLPH3 was an independent prognostic factor for patients with CCA. Transcriptome analysis (RNA sequencing) of GOLPH3 knockdown cells showed that the expression levels of nine ferroptosis-related genes were significantly changed, indicating the important biological function of GOLPH3 in ferroptosis in CCA cells. Furthermore, GOLPH3 knockdown could significantly promote Erastin-induced ferroptosis in vitro and suppress tumor growth in vivo. Overexpression of GOLPH3 had the opposite effect on this phenotype. Further studies revealed that GOLPH3 knockdown was significantly associated with a decrease in cysteine content, an accumulation of the lipid peroxidation product malondialdehyde, an increase in reactive oxygen species, and sensitized CCA cells to Erastin-induced ferroptosis. Moreover, changes in GOLPH3 expression were found to be consistent with the expression of light chain subunit solute carrier family 7 member 11 (SLC7A11). Thus, our study suggested that GOLPH3 functions as an oncoprotein in CCA and may suppress ferroptosis by facilitating SLC7A11 expression, suggesting that GOLPH3 could serve as a therapeutic target for CCA treatment.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Ferroptosis , Membrane Proteins , Humans , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic/metabolism , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Ferroptosis/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Multivariate Analysis
3.
Cureus ; 15(11): e48436, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38074028

ABSTRACT

BACKGROUND: Single-cell RNA sequencing technology can provide insight into lung cancer. The purpose of this study was to analyze the relationship between long noncoding RNA (lncRNA) discovered by RNA sequencing and immunotherapy in patients with non-small cell lung cancer (NSCLC). METHODS: In this study, we utilized data from The Cancer Genome Atlas (TCGA) to extract gene expression data and prognostic information from patients with NSCLC. We employed univariate, least absolute shrinkage and selection operator (LASSO), multivariate Cox regression analyses to construct risk models, and Kaplan-Meier (KM) analysis to compare survival differences between high- and low-risk groups. To evaluate the accuracy of our risk model predictions, we utilized a nomogram, calibration curve, correlation index curve (C-index), and receiver operating characteristic (ROC). Additionally, we conducted Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis to investigate the differential expression of lncRNA genes. We also used the tumor immune dysfunction and exclusivity (TIDE) algorithm and the R package "pRRophetic" to analyze the tumor microenvironment. Finally, we utilized stem cell indices based on mRNA expression-based stemness index (mRNAsi) expression to better assess patient prognosis. RESULTS: Our analysis identified a set of 28 lncRNAs with prognostic risk profiles in patients with lung adenocarcinoma. Notably, patients in the low-risk group exhibited significantly better overall survival (OS) compared to those in the high-risk group. Kaplan-Meier (KM) survival curves revealed that these prognostic risk markers accurately predicted survival outcomes in non-small cell lung cancer (NSCLC) patients. MerCK18 and myeloid-derived suppressor cells (MDSC) were strongly associated with immune escape and immunotherapy in high- and low-risk subgroups. In our investigation of potential chemotherapeutic agents for the treatment of NSCLC, we screened a total of 60 agents and found that PPM1D was more effective in the low-risk group. However, we did not observe a strong correlation between the stem cell index mRNAsi and OS. CONCLUSION: Our study highlights the close association between lncRNAs and prognostic risk profiles and the prognosis of patients with non-small cell lung cancer, offering a promising avenue for the clinical implementation of immunotherapy.

4.
Front Mol Biosci ; 9: 808152, 2022.
Article in English | MEDLINE | ID: mdl-35372504

ABSTRACT

Background: Golgi phosphoprotein 3 (GOLPH3) overexpression was recently reported to be associated with a poor clinical outcome in patients with colorectal cancer (CRC). However, the underlying molecular mechanism through which GOLPH3 promotes CRC metastasis remains poorly understood. Methods: In vitro genetic ablation of GOLPH3 was performed using siRNA transfection, and a stably overexpressed GOLPH3 colon cancer cell line was constructed using the lentivirus system. Cell invasion and migration assays were conducted with or without Matrigel. Immunoblotting, qRT-PCR, immunofluorescence and immunohistochemistry were utilized to study the expression level of GOLPH3, ZEB1, integrin α3 and phosphorylation level of STAT3, AKT/mTOR and Raf/MEK/ERK pathways. Co-immunoprecipitation was used to investigate the interaction between GOLPH3 and p-STAT3 (Tyr705) or total STAT3. Results: Overexpression of GOLPH3 was found in CRC tissues and colon cancer cell lines. Knockdown of GOLPH3 using siRNAs significantly suppressed the invasion and migration of HCT116 and HCT8 cells. In contrast, the overexpression of GOLPH3 promoted the migratory and invasive ability of colon cancer cells. The phosphorylation level of STAT3 as well as the protein and mRNA levels of ZEB1 and integrin α3, were significantly decreased after GOLPH3 knockdown. Moreover, Integrin α3 expression was correlated with GOLPH3 expression in CRC tissues. Co-immunoprecipitation assay revealed that GOLPH3 interacted with pSTAT3 (Tyr705) and total STAT3. Our further experiments suggested that GOLPH3 facilitated IL-6 induced STAT3 activation and subsequently induced transcription of integrin α3 and ZEB1, which promoted the metastasis and progression of CRC. Conclusion: Our current work demonstrates that GOLPH3 facilitates STAT3 activation and regulates the expression of EMT transcription factor ZEB1 and Integrin α3 in colon cancer cells. These findings indicate that GOLPH3 plays a critical role in CRC metastasis and might be a new therapeutic target for CRC treatment.

5.
Heliyon ; 8(12): e12518, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36590511

ABSTRACT

Patient-derived xenograft (PDX) models are more faithful in maintaining the characteristics of human tumors than cell lines and are widely used in drug development, although they have some disadvantages, including their relative low success rate, long turn-around time, and high costs. The collagen gel droplet embedded culture drug sensitivity test (CD-DST) has been used as an in-vitro drug sensitivity test for patients with cancer because of its high success rate of primary cell culture, high sensitivity, and good clinical relevance, but it is based on an in-vitro cell culture and may not simulate the tumor microenvironment accurately. This study aims to combine a PDX model with CD-DST to evaluate the efficiency of antitumor agents. KRpep-2d, a small peptide targeting KRAS (G12D), and oxaliplatin were used to verify the feasibility of this approach. Whole-exome sequencing and Sanger sequencing were first applied to test and validate the KRAS mutation status of a panel of colorectal cancer PDX tissues. One PDX model was verified to carry KRAS (G12D) mutation and was used for in-vivo and the CD-DST drug tests. We then established the PDX mouse model from the patient with the KRAS (G12D) mutation and obtained viable cancer cells derived from the same PDX model. Next, the antitumor abilities of KRpep-2d and oxaliplatin were estimated in the PDX model and the CD-DST. We found that KRpep-2d showed no significant antitumor effect on the xenograft model or on cancer cells derived from the same PDX model. In contrast, oxaliplatin showed significant inhibitory effects in both tests. In conclusion, the PDX model in combination with the CD-DST assay is a comprehensive and feasible method of evaluating the antitumor properties of compounds and could be applied for new drug discovery.

6.
BMC Cancer ; 18(1): 658, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29907144

ABSTRACT

BACKGROUND: CHD5 is a conventional tumour-suppressing gene in many tumours. The aim of this study was to determine whether CHD5 variants contribute to the risk of hepatocellular carcinoma (HCC). METHODS: Gene variants were identified using next-generation sequencing targeted on referenced mutations followed by TaqMan genotyping in two case-control studies. RESULTS: We discovered a rare variant (haplotype AG) in CHD5 (rs12564469-rs9434711) that was markedly associated with the risk of HCC in a Chinese population. A logistical regression model and permutation test confirmed the association. Indeed, the association quality increased in a gene dose-dependent manner as the number of samples increased. In the stratified analysis, this haplotype risk effect was statistically significant in a subgroup of alcohol drinkers. The false-positive report probability and multifactor dimensionality reduction further supported the finding. CONCLUSIONS: Our results suggest that the rare CHD5 gene haplotype and alcohol intake contribute to the risk of HCC. Our findings can be valuable to researchers of cancer precision medicine looking to improve diagnosis and treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Helicases/genetics , Liver Neoplasms/genetics , Nerve Tissue Proteins/genetics , Adult , Aged , Alcohol Drinking/adverse effects , Asian People/genetics , Case-Control Studies , Female , Gene-Environment Interaction , Genetic Predisposition to Disease , Haplotypes , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
7.
Biomed Pharmacother ; 103: 1632-1642, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29864952

ABSTRACT

Several microRNAs (miRNAs) have been closely correlated with the development of hepatocellular carcinoma (HCC). However, the involvement of miR-300 in the development of HCC remains unknown. This study elucidated the potential molecular mechanisms of miR-300 in the modulation of the epithelial-mesenchymal transition (EMT) and invasion of HCC. The expression levels of miR-300 in HCC cells and clinical samples were detected by quantitative real-time PCR and in situ hybridization. The in vitro function of miR-300 in HCC was evaluated using a migration/invasion assay. Quantitative real-time PCR, western blotting, immunofluorescence and immunohistochemistry were used to validate the roles of miR-300 and FAK/PI3K/AKT in EMT progression. A dual-luciferase reporter assay was performed to confirm the target gene. miR-300 was down-regulated in HCC and significantly correlated with a poor prognosis in HCC patients. The down-regulation of miR-300 increased the invasiveness of the HCC cells, and promoted the EMT in both HCC tissues and HCC cells. In contrast, up-regulation of miR-300 led to the opposite results. Ectopic overexpression of miR-300 reversed TGF-ß1-induced EMT in SMMC-7721 cells, and according to a dual-luciferase reporter assay and rescue assay, miR-300 inhibits the EMT-mediated migration and invasion of HCC cells via the targeted modulation of FAK and the downstream PI3K/AKT signaling pathway. miR-300 targeting modulates FAK, and the PI3K/AKT signaling pathway inhibits the EMT and suppresses the migration and invasion of HCC cells. Thus, miR-300 represents a promising therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Liver Neoplasms/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Base Sequence , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/pathology , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Phosphorylation/drug effects , Proportional Hazards Models , Regression Analysis , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology
8.
BMC Cancer ; 17(1): 745, 2017 Nov 10.
Article in English | MEDLINE | ID: mdl-29126392

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is the malignancy with the worst outcome among all breast cancer subtypes. We reported that ETV1 is a significant oncogene in TNBC tumourigenesis. Consequently, investigating the critical regulatory microRNAs (miRNAs) of ETV1 may be beneficial for TNBC targeted therapy. METHODS: We performed in situ hybridization (ISH) and immunohistochemistry (IHC) to detect the location of miR-17-5p and ETV1 in TNBC patient samples, respectively. miR-17-5p expression in TNBC tissues and cell lines was assessed by quantitative real-time PCR (qRT-PCR). ETV1 expression was evaluated by qRT-PCR, western blotting and IHC. Cell Counting Kit-8 (CCK-8), colony formation, Transwell and wound closure assays were utilized to determine the TNBC cell proliferation and migration capabilities. In vivo tumour metastatic assays were performed in a zebra fish model. RESULTS: The abundance of miR-17-5p was significantly decreased in TNBC cell lines and clinical TNBC tissues. The miR-17-5p expression levels were closely correlated with tumour size (P < 0.05) and TNM stage (P < 0.05). By contrast, the expression of ETV1 was significantly up-regulated in TNBC cell lines and tissues. There is an inverse correlation between the expression status of miR-17-5p and ETV1 (r = -0.28, P = 3.88 × 10-3). Luciferase reporter assay confirmed that ETV1 was a direct target of miR-17-5p. Forced expression of miR-17-5p in MDA-MB-231 or BT549 cells significantly decreased ETV1 expression and suppressed cell proliferation, migration in vitro and tumour metastasis in vivo. However, rescuing the expression of ETV1 in the presence of miR-17-5p significantly recovered the cell phenotype. High miR-17-5p expression was associated with a significantly favourable prognosis, in either the ETV1-positive or ETV1-negative groups (log-rank test, P < 0.001; P < 0.001). Both univariate and multivariate analyses showed that miR-17-5p and ETV1 were independent risk factors in the prognosis of TNBC patient. CONCLUSIONS: Our data indicate that miR-17-5p acts as a tumour suppressor in TNBC by targeting ETV1, and a low-abundance of miR-17-5p may be involved in the pathogenesis of TNBC. These findings indicate that miR-17-5p may be a therapeutic target for TNBC.


Subject(s)
DNA-Binding Proteins/genetics , MicroRNAs/genetics , Prognosis , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Aged , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Invasiveness/genetics , Triple Negative Breast Neoplasms/pathology , Zebrafish
9.
Oncotarget ; 8(42): 71418-71429, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-29069716

ABSTRACT

Down-regulation of the miRNA miR-338-3p correlates with the invasive ability of hepatocellular carcinoma (HCC) cells. However, it is currently unclear whether down-regulation of miR-338-3p induces epithelial-mesenchymal transition (EMT), which may be the underlying mechanism governing HCC invasion. Here, we demonstrate that restoration of miR-338-3p expression via transfection of a miR-338-3p mimic reversed EMT and inhibited the motility and invasiveness of HCC cells. Conversely, silencing of endogenous miR-338-3p expression with a miR-338-3p-specific inhibitor induced EMT and enhanced HCC cell motility. Additionally, Snail1 (an upstream regulatory protein of EMT) and Gli1 (a key transcription factor in the sonic hedgehog (SHH) signaling pathway) expression was up-regulated in cells treated with the miR-338-3p inhibitor and down-regulated by the miR-338-3p mimic. Further analyses demonstrated that miR-338-3p inhibitor-induced EMT in HCC cells was blocked by treatment with a small interfering RNA (siRNA) targeting Snail1, that the SHH signaling pathway was required for both miR-338-3p inhibitor-induced EMT and up-regulation of Snail1, and that miR-338-3p targeted a sequence within the 3'-untranslated region of N-cadherin mRNA. Notably, miR-338-3p expression was significantly down-regulated in HCC samples from patients with metastases and was associated with poor metastasis-free survival rates. Lastly, correlations between the expression levels of miR-338-3p and E-cadherin, Smoothened (SMO), Gli1, Snail1, N-cadherin, and vimentin were confirmed in HCC xenograft tumors and HCC patient specimens. Our findings suggest that miR-338-3p suppresses EMT and metastasis via both inhibition of the SHH/Gli1 pathway and direct binding of N-cadherin. miR-338-3p is a potential therapeutic target for metastatic HCC.

10.
Biomed Pharmacother ; 84: 1113-1118, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27780140

ABSTRACT

MicroRNA-24 (miR-24), a member of the miRNA family, functions as an oncogene in various types of human cancer. However, the underlying mechanisms of miR-24 involvement in the development and progression of hepatocellular carcinoma (HCC) remain poorly understood. The present study revealed that miRNA-24 down-regulates p53 through binding to the 3'-UTR of p53 mRNA based on a luciferase reporter assay, and that the expression level of miR-24 could affect the invasion of HCC lines via p53. Down-regulation of p53 significantly attenuated the inhibitory effects of miR-24 knockdown on the invasion of HCC cells, suggesting that miR-24 could be a potential target for HCC treatment. Moreover, our results revealed that miR-24 expression was significantly increased in HCC metastatic tumor tissues compared with matched non-metastatic tumor tissues, and that the up-regulation of miR-24 was significantly associated with down-regulation of p53 in the HCC tissues. In conclusion, this study demonstrates that miR-24 functions as an oncogene in HCC, at least partly by promoting cell invasion through down-regulation of p53. Therefore, miR-24 may be a potential therapeutic target for treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Movement , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Tumor Suppressor Protein p53/genetics , 3' Untranslated Regions , Binding Sites , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Cell Line, Tumor , Down-Regulation , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/surgery , MicroRNAs/metabolism , Neoplasm Invasiveness , Proto-Oncogenes , RNA Interference , Signal Transduction , Transfection , Tumor Suppressor Protein p53/metabolism , Up-Regulation
11.
Elife ; 52016 07 07.
Article in English | MEDLINE | ID: mdl-27383270

ABSTRACT

Increasing evidence highlights the important roles of microRNAs in mediating p53's tumor suppression functions. Here, we report miR-139-5p as another new p53 microRNA target. p53 induced the transcription of miR-139-5p, which in turn suppressed the protein levels of phosphodiesterase 4D (PDE4D), an oncogenic protein involved in multiple tumor promoting processes. Knockdown of p53 reversed these effects. Also, overexpression of miR-139-5p decreased PDE4D levels and increased cellular cAMP levels, leading to BIM-mediated cell growth arrest. Furthermore, our analysis of human colorectal tumor specimens revealed significant inverse correlation between the expression of miR-139-5p and that of PDE4D. Finally, overexpression of miR-139-5p suppressed the growth of xenograft tumors, accompanied by decrease in PDE4D and increase in BIM. These results demonstrate that p53 inactivates oncogenic PDE4D by inducing the expression of miR-139-5p.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/biosynthesis , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Models, Animal , Heterografts , Humans , Mice, Inbred BALB C
12.
Biochem Biophys Res Commun ; 461(4): 618-23, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25912876

ABSTRACT

MiR-129-5p is deregulated in various human cancers and has been associated with hepatocellular carcinoma (HCC) progression. However, the underlying mechanisms of miR-129-5p involvement in the development and progression of HCC and the effects of miR-129-5p deregulation on the clinical characteristics observed in HCC patients remain poorly understood. We therefore investigated the correlation between low miR-129-5p expression and vascular invasion, intrahepatic metastasis, and poor patient survival. Ectopic restoration of miR-129-5p expression in HCC cells suppressed cellular migration and invasion and the expression of v-ets erythroblastosis virus E26 oncogene homolog 1 (ETS1), while inhibition of endogenous miR-129-5p caused an increase in these parameters. We identified the ETS1 gene as a novel direct target of miR-129-5p. SiRNA-mediated ETS1 knockdown rescued the effects of anti-miR-129-5p inhibitor in HCC cell lines, while the effects of miR-129-5p overexpression were partially phenocopied in the knockdown model. In addition, miR-129-5p levels inversely correlated with those of ETS1 in HCC cells and tissues. Taken together, our findings indicate an important role for miR-129-5p in the molecular etiology of invasive HCC and suggest that miR-129-5p could have potential therapeutic applications in HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/secondary , Gene Targeting/methods , MicroRNAs/administration & dosage , MicroRNAs/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/genetics , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Tumor Cells, Cultured
13.
J Atheroscler Thromb ; 22(4): 424-32, 2015.
Article in English | MEDLINE | ID: mdl-25445891

ABSTRACT

AIM: RhoA is a critical factor in regulating the proliferation and migration of arterial smooth muscle cells (ASMCs) in patients with arteriosclerosis obliterans (ASO). RhoA is modulated by microRNA-133a (miR-133a) in cardiac myocytes and bronchial smooth muscle cells. However, the relationship between miR-133a and RhoA with respect to the onset of ASO in the lower extremities is uncertain. METHODS: We employed in situ hybridization (ISH) and immunohistochemistry (IHC) to detect the location of miR-133a and RhoA in ASO clinical samples, respectively. 5-ethynyl-2'-deoxyuridine (EdU), cell counting kit-8 (CCK-8), Transwell and wound closure assays were utilized to determine the features of human ASMC (HASMC) proliferation and migration. The expression of miR-133a in the HASMCs was assessed using quantitative real-time PCR (qRT-PCR), while that of RhoA was examined via qRT-PCR and Western blotting. RESULTS: We found miR-133a and RhoA to be primarily located in the ASMCs of ASO. miR-133a was significantly downregulated in the ASO tissues and proliferating HASMCs. In contrast, RhoA was upregulated in the ASO samples. The proliferation and migration of HASMCs was markedly promoted by the downregulation of miR-133a and inhibited by the upregulation of miR-133a. The Luciferase assay confirmed that RhoA was a direct target of miR-133a. The upregulation of miR-133a in the HASMCs decreased the RhoA expression at the protein level. Inversely, the downregulation of miR-133a increased the RhoA protein expression. Of note, the overexpression of RhoA in the HASMCs attenuated the anti-proliferative and anti-migratory effects of miR-133a. CONCLUSIONS: Our data indicate that miR-133a regulates the functions of HASMCs by targeting RhoA and may be involved in the pathogenesis of ASO. These findings may lead to the development of potential therapeutic targets for ASO of the lower extremities.


Subject(s)
Arteriosclerosis Obliterans/etiology , Arteriosclerosis Obliterans/metabolism , Lower Extremity/physiopathology , MicroRNAs/genetics , Muscle, Smooth, Vascular/metabolism , rhoA GTP-Binding Protein/metabolism , Arteriosclerosis Obliterans/pathology , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , DNA Primers/chemistry , DNA Primers/genetics , Humans , Immunoenzyme Techniques , In Situ Hybridization , Muscle, Smooth, Vascular/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics
14.
BMC Cancer ; 14: 571, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25104140

ABSTRACT

BACKGROUND: Golgi phosphoprotein 3 (GOLPH3) has been identified as an oncoprotein in various human cancers; however, its role in pancreatic ductal adenocarcinoma (PDAC) is unknown. We examined GOLPH3 expression levels and relationship with survival in patients with PDAC to establish the significance of GOLPH3 in the development and progression of PDAC. METHODS: Real-time qPCR and Western blotting were performed to analyze the expression levels of GOLPH3 mRNA and protein in paired PDAC tumor and adjacent non-tumor tissues. Immunohistochemistry was used to analyze the expression levels of GOLPH3 protein in paraffin-embedded tissues from 109 cases of PDAC. Univariate and multivariate analyses were performed to identify correlations between the immunohistochemical data for GOLPH3 expression and the clinicopathologic characteristics in PDAC. RESULTS: Expression levels of GOLPH3 mRNA and protein were upregulated in PDAC lesions compared to paired adjacent noncancerous tissues. Expression of GOLPH3 was significantly correlated with clinical stage (P = 0.006), T classification (P = 0.021), N classification (P = 0.049) and liver metastasis (P = 0.035). Patients with high GOLPH3 expression had shorter overall survival times compared to those with low GOLPH3 expression (P = 0.007). Multivariate analysis revealed that GOLPH3 overexpression was an independent prognostic factor in PDAC. CONCLUSIONS: Our findings suggest that GOLPH3 expression status may be a potential prognostic biomarker and therapeutic target in PCAC.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Pancreatic Neoplasms/pathology , Adult , Aged , Carcinoma, Pancreatic Ductal/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Pancreatic Neoplasms/genetics , Prognosis , Survival Analysis
15.
J Transl Med ; 12: 33, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24495516

ABSTRACT

BACKGROUND: According to cancer-related microRNA (miRNA) expression microarray research available in public databases, miR-362 expression is elevated in gastric cancer. However, the expression and biological role of miR-362 in gastric progression remain unclear. METHODS: miR-362 expression levels in gastric cancer tissues and cell lines were determined using real-time PCR. The roles of miR-362, in promoting gastric cancer cell proliferation and apoptosis resistance, were assessed by different biological assays, such as colony assay, flow cytometry and TUNEL assay. The effect of miR-362 on NF-κB activation was investigated using the luciferase reporter assay, fluorescent immunostaining. RESULTS: MiR-362 overexpression induced cell proliferation, colony formation, and resistance to cisplatin-induced apoptosis in BGC-823 and SGC-7901 gastric cancer cells. MiR-362 increased NF-κB activity and relative mRNA expression of NF-κB-regulated genes, and induced nuclear translocation of p65. Expression of the tumor suppressor CYLD was inhibited by miR-362 in gastric cancer cells; miR-362 levels were inversely correlated with CYLD expression in gastric cancer tissue. MiR-362 downregulated CYLD expression by binding its 3' untranslated region. NF-κB activation was mechanistically associated with siRNA-mediated downregulation of CYLD. MiR-362 inhibitor reversed all the effects of miR-362. CONCLUSION: The results suggest that miR-362 plays an important role in repressing the tumor suppressor CYLD and present a novel mechanism of miRNA-mediated NF-κB activation in gastric cancer.


Subject(s)
Apoptosis/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Signal Transduction/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , 3' Untranslated Regions/genetics , Base Sequence , Cell Line, Tumor , Cell Proliferation , Deubiquitinating Enzyme CYLD , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Molecular Sequence Data , Tumor Suppressor Proteins/metabolism , Up-Regulation/genetics
16.
Oncol Rep ; 31(3): 1199-204, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24366206

ABSTRACT

The aim of the present study was to investigate the expression of vascular endothelial growth factor (VEGF) and macrophage migration inhibitory factor (MIF) in HCC progression and their correlation with clinicopathological factors as well as the relationship between their expression levels. The expression of serum VEGF and MIF was evaluated in 150 patients with HCC and in 30 normal volunteers by enzyme-linked immunosorbent assay (ELISA). VEGF and MIF expression levels were evaluated by immunohistochemistry on tissue microarrays containing 150 HCCs with paired adjacent non-cancer liver tissues. VEGF and MIF mRNA levels were determined by quantitative PCR in another 48 HCCs. The correlation of VEGF and MIF with clinicopathological factors was analyzed in HCC. Serum VEGF and MIF concentrations were higher in HCC patients than the levels in the controls. The expression levels of VEGF and MIF in the HCC tissues were both higher than those in the adjacent non-tumor liver tissues. Overexpression of VEGF and MIF was significantly associated with tumor size (P=0.027 and 0.022, respectively), intrahepatic metastasis (P=0.032 and 0.027, respectively), vascular invasion (P=0.044 and 0.039, respectively) and TNM stage (P=0.028 and 0.013, respectively). Furthermore, VEGF and MIF mRNA levels were higher in HCC compared to levels in the paired non-cancer liver tissues. VEGF and MIF mRNA levels were correlated with tumor stage and metastasis. The expression of VEGF was positively related with MIF expression in HCC. The expression of MIF and VEGF in HCC was markedly positively correlated, which suggests that MIF and VEGF play an important role in the progression of HCC. Both factors may concomitantly accelerate the progression of HCC.


Subject(s)
Carcinoma, Hepatocellular/blood , Intramolecular Oxidoreductases/blood , Liver Neoplasms/blood , Macrophage Migration-Inhibitory Factors/blood , Vascular Endothelial Growth Factor A/blood , Adult , Aged , Carcinoma, Hepatocellular/secondary , Case-Control Studies , Disease Progression , Female , Gene Expression , Humans , Intramolecular Oxidoreductases/genetics , Liver Neoplasms/pathology , Macrophage Migration-Inhibitory Factors/genetics , Male , Middle Aged , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Young Adult
17.
Hepatol Res ; 44(5): 551-559, 2014 May.
Article in English | MEDLINE | ID: mdl-23621665

ABSTRACT

AIM: miR-145 is a candidate tumor suppressor miRNA. However, it is unknown whether miR-145 is involved in the invasion of hepatocellular carcinoma (HCC). Therefore, we aimed to explore the effect and mechanism of miR-145 in the control of HCC cell invasion. METHODS: HCC cell invasion was evaluated by transwell assays after transfection with pre-miR-145 or anti-miR-145. A luciferase reporter assay was used to determine whether a disintegrin and metalloprotease 17 (ADAM17) were a target of miR-145. The levels of miR-145 and ADAM17 mRNA were detected by a real-time polymerase chain reaction assay, and the level of ADAM17 protein was measured by western blot analysis. Pearson's correlation test was used to assess the correlation between ADAM17 mRNA expression and miR-145 expression in 20 HCC tissue samples. RESULTS: miR-145 was significantly downregulated in HCC tissues and cell lines. The loss of miR-145 expression was associated with the tumor-node-metastasis stage, vascular invasion and intrahepatic metastasis. The overexpression of miR-145 was able to suppress tumor MHCC-97H cell invasion, whereas the knockdown of miR-145 expression induced SMMC-7721 cell invasion. We demonstrated that miR-145 bound directly to the 3'-untranslated region of ADAM17 and inhibited the expression of ADAM17. The knockdown of ADAM17 in SMMC-7721 cells could partially reverse the effects of anti-miR-145. miR-145 expression was inversely associated with ADAM17 expression in 20 HCC tissue specimens. CONCLUSION: Our findings indicate that miR-145 could inhibit HCC cell invasion by regulating the expression of ADAM17.

18.
World J Surg Oncol ; 11: 161, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23866030

ABSTRACT

BACKGROUND: Although many studies have indicated that high-mobility group box 1 protein (HMGB1) is associated with oncogenesis and a worse prognosis, the prognostic value of HMGB1 in gastric cancer (GC) remains unclear. In the present work, we aimed to evaluate the role of HMGB1 in GC and examined whether aberrant expression of both HMGB1 and vascular endothelial growth factor C (VEGF-C) increased the malignant potential of GC. METHODS: A total of 166 GC patients and 32 normal subjects were enrolled. HMGB1 and VEGF-C expression was detected by tissue microarrays (TMAs) and immunohistochemical staining. The correlation between HMGB1 and VEGF-C expression and their relationships with clinicopathological GC variables were examined. Univariate and multivariate analyses were performed using the Cox proportional hazard model to predict the factors related to the patients' overall survival rates. RESULTS: HMGB1 and VEGF-C expression were observed in 81 (48.80%) and 88 (53.01%) tumors, respectively, significantly higher than the rates among the corresponding controls. In addition, HMGB1 and VEGF-C expression were positively correlated (R2 = 0.972). HMGB1 expression was also closely associated with tumor size, pT stage, nodal status, metastasis status, TNM stage, and poor prognosis. Multivariate survival analysis indicated that patients with HMGB1 and VEGF-C coexpression had the worst prognoses and survival rates (hazard ratio, 2.78; log rank P<0.001). CONCLUSIONS: HMGB1 is commonly expressed in GC. Combined evaluation of HMGB1 and VEGF-C may serve as a valuable independent prognostic factor for GC patients.


Subject(s)
Adenocarcinoma/mortality , HMGB1 Protein/metabolism , Neoplasm Recurrence, Local/mortality , Stomach Neoplasms/mortality , Vascular Endothelial Growth Factor C/metabolism , Adenocarcinoma/secondary , Adenocarcinoma/surgery , Biomarkers, Tumor/metabolism , Case-Control Studies , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Neoplasm Staging , Prognosis , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Survival Rate , Tissue Array Analysis
19.
Oncol Rep ; 28(4): 1257-62, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22842654

ABSTRACT

Trefoil factor 1 (TFF1) is a small cysteine-rich secreted protein which is principally expressed in the superficial cells of gastric mucosa. In gastric cancer, TFF1 is downregulated and plays an important role. Gastrokine 1 (GKN1) is a secreted protein with similar expression and biological functions to TFF1. This study aimed to determine the expression and biological functions of TFF1 and the relationships between TFF1 and GKN1 in gastric cancer. RT-PCR and immunohistochemistry were performed to detect TFF1 expression in gastric cancer cell lines and tissues. The transfected and co-transfected AGS cells which stably expressed TFF1 or both TFF1 and GKN1 were generated. Phenotypic changes such as cell viability, apoptosis and cell cycle modulation were assayed in the transfected cells. We found that TFF1 expression was significantly downregulated or lost in gastric cancer cell lines, gastric dysplasia and cancer. Restoration of TFF1 expression in AGS cells suppressed tumor cell viability and arrested AGS cells in the G1-S transition phase after olomoucine treatment. However, TFF1 was unable to induce cell apoptosis. In co-transfected cells, we found that TFF1 and GKN1 did not directly interact at the protein level. GKN1 was unable to cooperate with TFF1 on cell viability suppression, cell apoptosis and differentiation. Together, these results indicate that TFF1 expression is significantly downregulated in gastric cancer. TFF1 inhibited cell proliferation by delaying G1-S phase transition but not by inducing apoptosis. TFF1 may not interact or cooperate with GKN1 at the protein and functional level.


Subject(s)
Peptide Hormones/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Aged , Apoptosis/genetics , Case-Control Studies , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gastric Mucosa/metabolism , Humans , Immunoprecipitation , Male , Middle Aged , Peptide Hormones/genetics , Stomach Neoplasms/pathology , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics
20.
Turk J Gastroenterol ; 23(3): 212-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22798109

ABSTRACT

BACKGROUND/AIMS: Gastrokine-1 is a novel protein that plays an important role in the maintenance of the integrity of the gastric mucosa. However, whether Helicobacter pylori infection and non-steroidal anti-inflammatory drugs, which are known to cause gastric mucosal injuries, affect gastrokine-1 expression in the gastric mucosa is unknown. The aim of the present study was to determine gastric mucosal expression of gastrokine-1 in patients with Helicobacter pylori infection or long-term non-steroidal anti-inflammatory drug administration. MATERIAL AND METHODS: A total of 40 patients with functional dyspepsia (20 with Helicobacter pylori-negative chronic gastritis, and 20 with Helicobacter pylori-positive chronic gastritis), and 37 Helicobacter pylori-negative long-term non-steroidal anti-inflammatory drug users (26 with aspirin, 11 with selective cyclooxygenase-2 inhibitors) were selected. In addition, 20 Helicobacter pylori-negative healthy volunteers were recruited as controls. All subjects underwent endoscopies with biopsies taken from the antrum and the sites with lesions. Gastric mucosal changes were detected endoscopically and histologically, and gastrokine-1 protein expression in the antral mucosa was analyzed by immunohistochemistry. RESULTS: Expression of gastrokine-1 protein was decreased in Helicobacter pylori-positive chronic gastritis compared with Helicobacter pylori-negative subjects and the healthy controls. Similarly, gastrokine-1 expression in non-steroidal anti-inflammatory drug users was also decreased, compared with the healthy controls, but there was no significant difference in gastrokine-1 expression between the aspirin group and selective cyclooxygenase-2 inhibitor group. Moreover, gastrokine-1 expression levels tended to be associated with the severity of chronic gastritis. CONCLUSIONS: Both Helicobacter pylori infection and long-term non-steroidal anti-inflammatory drug administration downregulate gastrokine-1 expression in the gastric mucosa, which may contribute to the gastric mucosal injuries induced by these two factors.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Gastric Mucosa/drug effects , Gastritis/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Peptide Hormones/metabolism , Aspirin/administration & dosage , Case-Control Studies , Cyclooxygenase 2 Inhibitors/administration & dosage , Down-Regulation , Dyspepsia/metabolism , Dyspepsia/microbiology , Gastric Mucosa/metabolism , Gastritis/microbiology , Gene Expression Regulation , Helicobacter Infections/microbiology , Humans , Immunohistochemistry , Peptide Hormones/genetics
SELECTION OF CITATIONS
SEARCH DETAIL