Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Eur J Med Res ; 28(1): 198, 2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37370148

ABSTRACT

BACKGROUND: Urinary pan-cancer system is a general term for tumors of the urinary system including renal cell carcinoma (RCC), prostate cancer (PRAD), and bladder cancer (BLCA). Their location, physiological functions, and metabolism are closely related, making the occurrence and outcome of these tumors highly similar. Cuproptosis is a new type of cell death that is different from apoptosis and plays an essential role in tumors. Therefore, it is necessary to study the molecular mechanism of cuproptosis-related lncRNAs to urinary system pan-cancer for the prognosis, clinical diagnosis, and treatment of urinary tumors. METHOD: In our study, we identified 35 co-expression cuproptosis-related lncRNAs (CRLs) from the urinary pan-cancer system. 28 CRLs were identified as prognostic-related CRLs by univariate Cox regression analysis. Then 12 CRLs were obtained using lasso regression and multivariate cox analysis to construct a prognostic model. We divided patients into high- and low-risk groups based on the median risk scores. Next, Kaplan-Meier analysis, principal component analysis (PCA), functional rich annotations, and nomogram were used to compare the differences between the high- and low-risk groups. Finally, the prediction of tumor immune dysfunction and rejection, gene mutation, and drug sensitivity were discussed. CONCLUSION: Finally, the candidate molecules of the urinary system pan-cancer were identified. This CRLs risk model may be promising for clinical prediction of prognosis and immunotherapy response in urinary system pan-cancer patients.


Subject(s)
Kidney Neoplasms , RNA, Long Noncoding , Male , Humans , RNA, Long Noncoding/genetics , Prognosis , Immunotherapy , Nomograms , Apoptosis
2.
Front Endocrinol (Lausanne) ; 14: 1188547, 2023.
Article in English | MEDLINE | ID: mdl-37351106

ABSTRACT

Background: Coffee is one of the most consumed beverages worldwide, but the effects on the thyroid are unknown. This study aims to examine the association between coffee and thyroid function. Methods: Participant data (≥ 20 years, n = 6578) for the observational study were obtained from NHANES 2007-2012. Analysis was performed using weighted linear regression models and multiple logistic regression models. Genetic datasets for Hyperthyroidism and Hypothyroidism were obtained from the IEU database and contained 462,933 European samples. Mendelian randomization (MR) was used for the analysis, inverse variance weighting (IVW) was the main method of analysis. Results: In the model adjusted for other covariates, participants who drank 2-4 cups of coffee per day had significantly lower TSH concentrations compared to non-coffee drinkers (b=-0.23, 95% CI: -0.30, -0.16), but no statistically significant changes in TT4, FT4, TT3 and FT3. In addition, participants who drank <2 cups of coffee per day showed a low risk of developing subclinical hypothyroidism. (OR=0.60, 95% CI: 0.41, 0.88) Observational studies and MR studies have demonstrated both that coffee consumption has no effect on the risk of hyperthyroidism and hypothyroidism. Conclusions: Our study showed that drinking <2 cups of coffee per day reduced the risk of subclinical hypothyroidism and drinking 2-4 cups of coffee reduced serum TSH concentrations. In addition, coffee consumption was not associated with the risk of hyperthyroidism and hypothyroidism.


Subject(s)
Hyperthyroidism , Hypothyroidism , Humans , Mendelian Randomization Analysis/methods , Nutrition Surveys , Hypothyroidism/epidemiology , Hypothyroidism/genetics , Hyperthyroidism/epidemiology , Hyperthyroidism/genetics , Thyrotropin
3.
Front Oncol ; 12: 812786, 2022.
Article in English | MEDLINE | ID: mdl-35574385

ABSTRACT

Digestive system pan-cancer is a general term for digestive system tumors including colorectal carcinoma (CRC), esophageal carcinoma (ESCA), stomach adenocarcinoma (STAD), and liver hepatocellular carcinoma (LIHC). Since the anatomical location, function and metabolism are closely related, there may be similarities in development and progression of these tumors. Hypoxia is the consequence of an imbalance between oxygen demand and supply, and intracellular hypoxia is associated with malignant progression, treatment resistance, and poor prognosis in tumors. Therefore, an urgent and challenging task is to investigate the molecular mechanisms associated with hypoxia in digestive system pan-cancer for the prognosis and treatment of digestive tract tumors. In this study, we identified 18 hypoxia-related lncRNAs (HRlncRNAs) by co-expression analysis between hypoxia genes and lncRNAs from digestive system pan-cancer. Six HRlncRNAs were then obtained using lasso regression and multivariate cox analysis to construct a prognostic model. Next, the Akaike information criterion (AIC) values for 3-year receiver operating curve (ROC) were counted to determine the cut-off point and establish an optimal model to distinguish between high- or low-risk groups among patients with digestive system pan-cancer. To evaluate the stability of the prognosis model, we validated it in terms of survival outcomes, clinicopathological stage, tumor-infiltrating immune cells, immune checkpoint inhibitors (ICIs) and anticancer drugs sensitivity. The results suggested that high- risk group had a worse prognosis and a more positive association with tumor-infiltrating immune cells such as B cells, cancer-associated fibroblasts, endothelial cells, monocytes, macrophages and bone marrow dendritic cells in digestive system pan-cancer. Immune checkpoint inhibitors (ICIs) related biomarkers discovered that high-risk group was positively correlated with high expression of HAVCR2 in digestive system pan-cancer. The anticancer drugs sensitivity analysis showed that the high-risk group was associated with the lower half-inhibitory centration (IC50) of Imatinib in digestive system pan-cancer. In conclusion, the prognostic model of HRlncRNAs showed a promising clinical prediction value and may provide a useful reference for the diagnosis and treatment of the digestive system tumors.

4.
Front Oncol ; 11: 718761, 2021.
Article in English | MEDLINE | ID: mdl-34490117

ABSTRACT

PURPOSE: Breast cancer type 1 susceptibility (BRCA) mutations not only increase breast cancer (BC) risk but also result in poor survival and prognosis for BC patients. This study will analyze the effect and safety of therapeutic regimens for the treatment of BC patients with germline BRCA (gBRCA) mutations by network meta-analysis. METHODS: Public databases were searched from inception to 29 April 2021. Frequentist network meta-analysis was conducted to analyze the benefit of chemotherapy and targeted drug-related strategies. RESULTS: Seventeen articles were included in the analysis. For progression-free survival (PFS), olaparib (hazard ratio (HR): 0.58; 95% confidence interval (CI): 0.43 - 0.79), platinum (HR: 0.45; 95% CI: 0.22 - 0.89), and talazoparib (HR: 0.54; 95% CI: 0.41 - 0.71) were significantly better than platinum-free chemotherapy (Chemo). The results based on indirect comparisons showed that veliparib (Vel) + platinum + Chemo was also significantly better than Chemo (HR: 0.37; 95% CI: 0.20 - 0.69). For overall survival (OS), olaparib was significantly better than Chemo only in the population who did not receive prior chemotherapy. For pathologic complete response (pCR), bevacizumab+Chemo had a significant advantage over platinum agents (OR: 3.64; 95% CI: 1.07 - 12.39). Olaparib and talazoparib both showed significantly higher objective response rates (ORRs) than Chemo. CONCLUSION: The PFS results suggested that olaparib, talazoparib, and Vel+platinum agent+Chemo were ideal regimens for overall, TNBC, and advanced BC patients with gBRCA mutations. Whether PARPis are suitable for patients with gBRCA mutations who have received prior platinum therapy still needs to be clarified.

5.
PeerJ ; 9: e12114, 2021.
Article in English | MEDLINE | ID: mdl-34557356

ABSTRACT

Cervical cancer is one of the most common malignant tumors in women, and its morbidity and mortality are increasing year by year worldwide. Therefore, an urgent and challenging task is to identify potential biomarkers for cervical cancer. This study aims to identify the hub genes based on the GEO database and then validate their prognostic values in cervical cancer by multiple databases. By analysis, we obtained 83 co-expressed differential genes from the GEO database (GSE63514, GSE67522 and GSE39001). GO and KEGG enrichment analysis showed that these 83 co-expressed it mainly involved differential genes in DNA replication, cell division, cell cycle, etc.. The PPI network was constructed and top 10 genes with protein-protein interaction were selected. Then, we validated ten genes using some databases such as TCGA, GTEx and oncomine. Survival analysis demonstrated significant differences in CDC45, RFC4, TOP2A. Differential expression analysis showed that these genes were highly expressed in cervical cancer tissues. Furthermore, univariate and multivariate cox regression analysis indicated that CDC45 and clinical stage IV were independent prognostic factors for cervical cancer. In addition, the HPA database validated the protein expression level of CDC45 in cervical cancer. Further studies investigated the relationship between CDC45 and tumor-infiltrating immune cells via CIBERSORT. Finally, gene set enrichment analysis (GSEA) showed CDC45 related genes were mainly enriched in cell cycle, chromosome, catalytic activity acting on DNA, etc. These results suggested CDC45 may be a potential biomarker associated with the prognosis of cervical cancer.

7.
Vet Microbiol ; 251: 108913, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33166843

ABSTRACT

As a severe disease characterized by reproductive failure and respiratory distress, porcine reproductive and respiratory syndrome (PRRS) is one of the most leading threats to the swine industry worldwide. Highly evolving porcine reproductive and respiratory syndrome virus (PRRSV) strains with distinct genetic diversity make the current vaccination strategy much less cost-effective and thus urge alternative protective host directed therapeutic approaches. RACK1-PKC-NF-κB signalling axis was suggested as a potential therapeutic target for PRRS control, therefore we tested the inhibitory effect of PKC inhibitor dequalinium chloride (DECA) on the PRRSV infection in vitro. RT-qPCR, western blot, Co-IP and cytopathic effect (CPE) observations revealed that DECA suppressed PRRSV infection and protected Marc-145 cells and porcine alveolar macrophages (PAMs) from severe cytopathic effects, by repressing the PKCα expression, the interaction between RACK1 and PKCα, and subsequently the NF-κB activation. In conclusion, the data presented in this study shed more light on deeper understanding of the molecular pathogenesis upon PRRSV infection and more importantly suggested DECA as a potential promising drug candidate for PRRS control.


Subject(s)
Dequalinium/pharmacology , Porcine respiratory and reproductive syndrome virus/drug effects , Protein Kinase C-alpha/antagonists & inhibitors , Virus Replication/drug effects , Animals , Cell Line , Cells, Cultured , Cytopathogenic Effect, Viral , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/virology , Signal Transduction , Swine
8.
Epigenomics ; 12(15): 1257-1271, 2020 08.
Article in English | MEDLINE | ID: mdl-32811161

ABSTRACT

Aim: To investigate the role and mechanisms of AC245100.4 in prostate cancer. Materials & methods: The expression and location of AC245100.4 were examined using real-time PCR and in situ hybridization. Cell Counting Kit-8, clone formation, flow cytometry and in vivo assays were conducted to determine the role of AC245100.4. RNA antisense purification with mass spectrometry and RNA immunoprecipitation were performed to identify proteins that bind to AC245100.4. Western blotting was performed to quantify the expression of protein. Results:AC245100.4 expression was upregulated in prostate cancer and mainly located in the cytoplasm. Knockdown of AC245100.4 inhibited proliferation of prostate cancer. Mechanistically, AC245100.4 bound to HSP90 and altered its chaperone function, increased the stability of IκB kinase and activated the NFκB signaling pathway. Conclusion:AC245100.4 promotes the proliferation of prostate cancer via binding of HSP90.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Prostatic Neoplasms , RNA, Long Noncoding , Animals , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Signal Transduction
9.
Front Genet ; 11: 361, 2020.
Article in English | MEDLINE | ID: mdl-32362911

ABSTRACT

[This corrects the article DOI: 10.3389/fgene.2019.01377.].

10.
J Mol Endocrinol ; 65(2): 1-10, 2020 08.
Article in English | MEDLINE | ID: mdl-32302969

ABSTRACT

miR-146b-5p is overexpressed in papillary thyroid carcinoma (PTC) and is thought to be a related diagnostic marker. Previous studies have indicated the effects of iodine on oncogenic activation. However, the effect of iodine on the proliferation of PTC cells and the associated underlying mechanisms remain unclear. We found that miR-146b-5p was downregulated and smad4 was upregulated in patients exposed to high iodine concentration by in situ hybridisation (ISH) and immunohistochemical (IHC). NaI (10-3 M) treatment downregulated miR-146b-5p and upregulated Smad4 in PTC cell lines. Luciferase assay was used to confirm that Smad4 is a target of miR-146b-5p. Furthermore, MTT assay and cell cycle analysis indicated that 10-3 M NaI suppressed cell proliferation and caused G0/G1 phase arrest. Real-time PCR and Western blotting demonstrated that 10-3 M NaI increased p21, p27, and p57 levels and reduced cyclin D1 levels in PTC cells. Our findings suggest that 10-3 M NaI increases Smad4 levels through repression of miR-146b-5p expression, curbing the proliferation in PTC.


Subject(s)
Down-Regulation , Iodine/metabolism , MicroRNAs/metabolism , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Adult , Base Sequence , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/genetics , Middle Aged , Smad4 Protein/genetics , Smad4 Protein/metabolism
11.
Onco Targets Ther ; 12: 6471-6480, 2019.
Article in English | MEDLINE | ID: mdl-31496744

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) play an important role in the pathogenesis of multiple tumors. However, the roles of lncRNAs during colon adenocarcinoma and cancer progression remain unclear. This study aimed identify new lncRNAs that act as molecular markers for the prevention and diagnosis of colon adenocarcinoma. METHODS: RNA sequencing (RNA-Seq) data associated with colon adenocarcinoma were retrieved from the Cancer Genome Atlas (TCGA). Biological processes in Gene Ontology (Go) and the Kyoto Encyclopedia of Genomes (KEGG) were searched for pathways at the significance level. The expression of LINC00491 and its downstream targets were assessed by real-time PCR, Western blotting and dual-luciferase assays. Biological functions of LINC00491 during cell proliferation, migration and invasion were assessed using CCK-8, colony formation assays, wound healing, and transwell invasion assays in colon adenocarcinoma HT-29 and HCT116 cells. RESULTS: Bioinformatics analysis with the TCGA colon adenocarcinoma dataset showed that LINC00491 was significantly up-regulated in colon adenocarcinoma. Furthermore, we found that LINC00491 positively regulates SERPINE1 expression through sponging miR-145 and promoting the proliferation, migration, and invasion of colon adenocarcinoma cells, thus playing an oncogenic role during colon adenocarcinoma pathogenesis. CONCLUSION: LINC00491 functions as a ceRNA to promote SERPINE1 expression by sponging miR-145. LINC00491 serves as a therapeutic target and prognostic biomarker in colon adenocarcinoma.

12.
Cancer Med ; 8(14): 6358-6369, 2019 10.
Article in English | MEDLINE | ID: mdl-31466138

ABSTRACT

BACKGROUND: As an oncogene, long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) can promote tumor metastasis. Hyperexpression of MALAT1 has been observed in many malignant tumors, including hepatocellular carcinoma (HCC). However, the role and mechanism of MALAT1 in HCC remain unclear. METHODS: Thirty human HCC and paracancerous tissue specimens were collected, and the human hepatoma cell lines Huh7 and HepG2 were cultured according to standard methods. MALAT1 and Snail family zinc finger (Slug) expression were measured by real-time PCR, immunohistochemistry, and western blotting. Luciferase reporter assay and RNA immunoprecipitation (RIP) assay verified the direct interaction between miR-124-3p and Slug(SNAI2) or MALAT1. Wound healing and transwell assays were performed to examine invasion and migration, and a subcutaneous tumor model was established to measure tumor progression in vivo. RESULTS: MALAT1 expression was upregulated in HCC tissues and positively correlated with Slug expression. MALAT1 and miR-124-3p bind directly and reversibly to each other. MALAT1 silencing inhibited cell migration and invasion. miR-124-3p inhibited HCC metastasis by targeting Slug. CONCLUSIONS: MALAT1 regulates Slug through miR-124-3p, affecting HCC cell metastasis. Thus, the MALAT1/miR-124-3p/Slug axis plays an important role in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Snail Family Transcription Factors/genetics , Adult , Aged , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , RNA Interference , ROC Curve , Snail Family Transcription Factors/metabolism , Xenograft Model Antitumor Assays
13.
Cancer Med ; 8(5): 2392-2403, 2019 05.
Article in English | MEDLINE | ID: mdl-30932362

ABSTRACT

BACKGROUND: MiRNAs can regulate gene expression directly or indirectly, and long noncoding RNAs as competing endogenous RNA (ceRNAs) can bind to miRNAs competitively and affect mRNA expression. The ceRNA network is still unclear in breast cancer. In this study, a ceRNA network was constructed, and new treatment and prognosis targets and biomarkers for breast cancer were explored. METHODS: A total of 1 096 cancer tissues and 112 adjacent normal tissues to cancer from the TCGA database were used to screen out significant differentially expressed mRNAs (DEMs), lncRNAs (DELs), and miRNAs (DEMis) to construct a ceRNA network. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were used to predict potential functions. Survival analysis was performed to predict which functions were significant for prognosis. RESULTS: From the analysis, 2 139 DEMs, 1 059 DELs, and 84 DEMis were obtained. Targeting predictions for DEMis-DELs and DEMis-DEMs can yield 26 DEMs, 90 DELs, and 18 DEMis. We performed GO enrichment analysis, and the results showed that the upregulated DEMs were involved in nucleosomes, extracellular regions, and nucleosome assembly, while the downregulated DEMs were mainly involved in Z disk, muscle contraction, and structural constituents of muscle. KEGG pathway analysis was performed on all DEMs, and the pathways were enriched in retinol metabolism, steroid hormone biosynthesis, and tyrosine metabolism. Through survival analysis of the ceRNA network, we identified four DEMs, two DELs, and two DEMis that were significant for poor prognosis. CONCLUSIONS: This study suggested that constructing a ceRNA network and performing survival analysis on the network could screen out new significant treatment and prognosis targets and biomarkers.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Biomarkers, Tumor , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , Computational Biology/methods , Databases, Genetic , Female , Gene Expression Profiling , Gene Ontology , Humans , Prognosis , ROC Curve
14.
Front Genet ; 10: 1377, 2019.
Article in English | MEDLINE | ID: mdl-32153626

ABSTRACT

Long noncoding RNAs (lncRNAs) play important roles in the regulation of gene expression by acting as competing endogenous RNAs (ceRNAs). However, the roles of lncRNA-associated ceRNAs in oncogenesis are not fully understood. The present study aims to determine whether a ceRNA network can serve as a prognostic marker in human prostate cancer (PCa). In order to identify a ceRNA network and the key lncRNAs in PCa, we constructed a differentially expressed lncRNAs (DELs)-differentially expressed miRNAs (DEMis)-differentially expressed mRNAs (DEMs) regulatory network based on the ceRNA theory using data from the Cancer Genome Atlas (TCGA). We found that the DELs-DEMis-DEMs network was composed of 27 DELs nodes, seven DEMis nodes, and three DEMs nodes. The 27 DELs were further analyzed with several public databases to provide meaningful information for understanding the functional roles of lncRNAs in regulatory networks in PCa. We selected ADAMTS9-AS1 to determine its role in PCa and found that ADAMTS9-AS1 significantly influences tumor cell growth and proliferation, suggesting that it plays a tumor suppressive role. In addition, ADAMTS9-AS1 functioned as ceRNA, effectively becoming a sponge for hsa-mir-96 and modulating the expression of PRDM16. These results suggest that ceRNAs could accelerate biomarker discovery and therapeutic strategies for PCa.

15.
Cell Physiol Biochem ; 50(1): 108-120, 2018.
Article in English | MEDLINE | ID: mdl-30278452

ABSTRACT

BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) has a high morbidity as well as mortality and is believed to be one of the most prevalent cancers worldwide. The long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is involved in numerous cancers, including HCC. This study aimed to explore the role of MALAT1 in HCC progression. METHODS: The expression levels of MALAT1 and Vimentin in HCC tissues and relative pair-matched adjacent normal liver tissues were analyzed by RT-PCR, and immunohistochemistry. Using bioinformatics analysis and dual-luciferase assay, we examined the correlation between MALAT1 and miR-30a-5p. Dual-luciferase assay and western blotting suggested that Vimentin was a target of miR-30a-5p. A wound healing assay and transwell assays were employed to determine the effect of MALAT1 and miR-30a-5p on cell migration and invasion in HCC. RESULTS: Our data demonstrated that the levels of MALAT1 and Vimentin were upregulated in HCC tissues and that miR-30a-5p was a direct target of MALAT1. Silenced MALAT1 and overexpressed miR-30a-5p each inhibited cell migration and invasion. Additionally, dual-luciferase assay and western blotting demonstrated that MALAT1 could competitively sponge miR-30a-5p and thereby regulate Vimentin. CONCLUSION: Our data suggest that MALAT1 acts as an oncogenic lncRNA that promotes HCC migration and invasion. Therefore, the MALAT1-miR-30a-5p-Vimentin axis is a potential therapeutic target and molecular biomarker in HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Vimentin/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Base Sequence , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Sequence Alignment , Vimentin/chemistry , Vimentin/genetics
16.
Oncol Lett ; 15(6): 9436-9442, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29805667

ABSTRACT

MicroRNAs (miRNAs/miRs), which are endogenous non-coding single-stranded RNAs 19-25 nucleotides in length, regulate gene expression by blocking translation or transcription repression. The present study revealed that miR-3160-5p was widely expressed in prostate cancer cells by reverse transcription-quantitative polymerase chain reaction. There was a negative association between the expression of miR-3160-5p and F-box and WD repeat domain containing 8 (Fbxw8) in prostate cancer DU145 cells. A luciferase activity assay was used to verify that Fbxw8 is the target of miR-3160-5p. In the present study, using MTT assay and cell cycle analysis, it was demonstrated that DU145 cell proliferation was repressed and the cell cycle was arrested in the G2/M cell cycle phase with upregulation of miR-3160-5p. Subsequent studies demonstrated that miR-3160-5p regulated the progression of the cell cycle in DU145 prostate cancer cells when the expression levels of phosphorylated cell division cycle (CDC)2, CDC25C and cyclin B1 were directly inhibited. Taken together, these findings revealed the mechanism underlying the role of miR-3160-5p in regulating the proliferation of DU145 cells and indicated that miR-3160-5p may serve as a promising novel therapeutic tool for prostate cancer.

17.
Oncotarget ; 7(40): 65553-65567, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27566588

ABSTRACT

EMT (epithelial-mesenchymal transition) occurs in a wide range of tumor types, and has been shown to be crucial for metastasis. Epigenetic modifications of histones contribute to chromatin structure and result in the alterations in gene expression. Tri-methylation of histone H3 lysine 4 (H3K4me3) is associated with the promoters of actively transcribed genes and can serve as a transcriptional on/off switch. RbBP5 is a component of the COMPASS/ -like complex, which catalyzes H3K4me3 formation. In this study, we found that in the process of TGF-Beta1 induced EMT in the prostate cancer cell line DU145, H3K4me3 enrichment and RbBP5 binding increased in the vicinity of Snail (SNAI1) transcription start site. Knocking-down of RbBP5 notably decreased Snail expression and EMT. Recruitment of RbBP5 and formation of H3K4me3 at Snail TSS during EMT depend on binding of SMAD2/3 and CBP at Snail TSS. This study links the SMAD2/3 signal with Snail transcription via a histone modification - H3K4me3. Furthermore, our research also demonstrates that RbBP5 and even WRAD may be a promising therapeutic candidates in treating prostate cancer metastasis, and that DU145 cells maintain their incomplete mesenchymal state in an auto/ paracrine manner.


Subject(s)
Histones/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Snail Family Transcription Factors/genetics , Cell Line, Tumor , DNA-Binding Proteins , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Humans , Male , Methylation , Nuclear Proteins/genetics , Peptide Fragments/metabolism , Prostatic Neoplasms/genetics , Protein Binding , RNA, Small Interfering/genetics , Sialoglycoproteins/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Initiation Site , Transforming Growth Factor beta/metabolism
18.
Oncol Rep ; 35(2): 1153-62, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26718770

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) endows cancer cells with enhanced invasive and metastatic potential during cancer progression. Fractalkine, also known as chemokine (C-X3-C motif) ligand 1 (CX3CL1), the only member recognized so far that belongs to the CX3C chemokine subfamily, was reported to participate in the molecular events that regulate cell adhesion, migration and survival of human prostate cancer cells. However, the relationship between CX3CL1 and EMT remains unknown. We treated DU145 and PC-3 cells with CX3CL1 under hypoxic conditions. The migration and invasion abilities of DU145 and PC-3 cells were detected by Transwell assays. Induction of EMT was verified by morphological changes in the DU145 and PC-3 cells and analysis of protein expression of EMT markers such as E-cadherin and vimentin. To identify the involved signaling pathway in CX3CL1-induced EMT, activation of epidermal growth factor receptor (EGFR) was measured using western blot analysis, and Slug expression was detected with or without an EGFR inhibitor prior to CX3CL1 treatment. Concentrations of soluble and total TGF-α in the CX3CL­treated DU145 cells were detected by ELISA. Additionally, we determined the involvement of the TACE/TGF-α/EGFR pathway in CX3CL1­induced EMT using RNA interference and specific inhibitors. CX3CL1 increased the migration and invasiveness of the DU145 and PC-3 cells, and resulted in characteristic alterations of EMT. Our results demonstrated that TACE/TGF-α/EGFR pathway activation and subsequent upregulation of Slug expression were responsible for CX3CL1­induced EMT, and contributed to the migration and inva-sion of prostate cancer cells. Inhibition of TACE/TGF-α/EGFR signaling reversed EMT and led to reduced migration and invasion abilities of the prostate cancer cells. We provide initial evidence that CX3CL1 exposure resulted in EMT occurrence and enhancement of cell migration and invasion through a mechanism involving activation of TACE/TGF-α/EGFR signaling. These findings revealed that CX3CL1 may serve as a new target for the treatment of prostate cancer.


Subject(s)
ADAM Proteins/physiology , Adenocarcinoma/pathology , Chemokine CX3CL1/physiology , Epithelial-Mesenchymal Transition/physiology , ErbB Receptors/physiology , Neoplasm Proteins/physiology , Prostatic Neoplasms/pathology , Signal Transduction/physiology , Transforming Growth Factor alpha/physiology , ADAM Proteins/genetics , ADAM17 Protein , Adenocarcinoma/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Humans , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Prostatic Neoplasms/metabolism , RNA Interference , RNA, Small Interfering/genetics , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation
19.
Mol Med Rep ; 12(6): 8201-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26458406

ABSTRACT

Gastric cancer is the fourth most common cancer type and the second leading cause of cancer­associated mortality worldwide. Metastasis is a crucial feature of its progression. DNA methylation provides a key epigenetic signature in the epigenetic regulation pathway, and is implicated in transcriptional regulation. CpG sites, which are associated with gene transcriptional activity, are underrepresented in the mammalian genome and tend to be clustered within CpG islands (CGIs) located in the vicinity of the transcription start sites of the majority of the protein­coding genes in humans. The DNA methylation inhibitor, decitabine (DAC), has been demonstrated to be active in hematological disorders. The majority of previous studies in cancer cells demonstrated that DAC inhibits cell proliferation and the motility of the cells. However, since demethylation across the entire genome alters the expression of a large number of genes, the effects of DAC in different tumor cell types are difficult to accurately predict. Neural precursor cell­expressed, developmentally downregulated (NEDD)4­1, a member of the NEDD4 family, which belongs to the E3­ubiquitin ligase family, was reported to be highly expressed in a wide range of tumor types, and it activates the phosphoinositide 3­kinase/Akt pathway by degrading phosphatase and tensin homolog. NEDD4­1 promotes the migration and invasion of glioma cells via the ubiquitination and subsequent degradation of cyclic nucleotide­Ras guanine nucleotide exchange factors (CNrasGEFs). In gastric cardia adenocarcinoma, NEDD4­1 acts as an exceptional prognostic biomarker. In the present study, DAC was revealed to promote the invasive properties of MGC803 gastric cancer cells. NEDD4­1 targeted the CNrasGEF­mediated DAC invasion­promoting activity in MGC803 cells, and CGI methylation in neither the NEDD4 promoter nor the first intron was demonstrated to be associated with this effect. The results of the present study revealed that DAC exerts variable effects in different gastric cancer cell lines and may provide a reference for DAC administration in the clinic.


Subject(s)
Azacitidine/analogs & derivatives , Cell Movement/drug effects , Endosomal Sorting Complexes Required for Transport/genetics , Stomach Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Up-Regulation , Azacitidine/pharmacology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Methylation/drug effects , Decitabine , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomal Sorting Complexes Required for Transport/physiology , Humans , Nedd4 Ubiquitin Protein Ligases , Neoplasm Invasiveness/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology
20.
Mol Med Rep ; 12(6): 7907-14, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26496926

ABSTRACT

Hypoxia is a common phenomenon in prostate cancer, which leads to cell proliferation and tumor growth. Fractalkine (FKN) is a membrane­bound chemokine, which is implicated in the progression of human prostate cancer and skeletal metastasis. However, the association between FKN and hypoxia­induced prostate cancer cell proliferation remains to be elucidated. The present study demonstrated that hypoxia induced the expression and secretion of FKN in the DU145 prostate cancer cell line. Furthermore, inhibiting the activity of FKN with the anti­FKN FKN­specific antibody markedly inhibited hypoxia­induced DU145 cell proliferation. Under normoxic conditions, DU145 cell proliferation markedly increased following exogenous administration of human recombinant FKN protein, and the increase was significantly alleviated by anti­FKN, indicating the importance of FKN in DU145 cell proliferation. In addition, subsequent determination of cell cycle distribution and expression levels of two core cell cycle regulators, cyclin E and cyclin­dependent kinase (CDK)2, suggested that FKN promoted the G1/S phase transition by upregulating the expression levels of cyclin E and CDK2. The results of the present study demonstrated that hypoxia led to the upregulation of the secretion and expression of FKN, which enhanced cell proliferation by promoting cell cycle progression in the prostate cancer cells. These findings provide evidence of a novel function for FKN, and suggest that FKN may serve as a potential target for treating androgen­independent prostate cancer.


Subject(s)
Cell Proliferation/genetics , Chemokine CX3CL1/physiology , Prostatic Neoplasms/pathology , Up-Regulation , Cell Hypoxia , Cell Line, Tumor , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Male , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...