Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Exp Cell Res ; 391(2): 111817, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32179097

ABSTRACT

Overexpression of the c-Myc oncogene has been implicated in cancer stem cell - like (CSC) phenotypes and epithelial-to-mesenchymal transition (EMT) in cancer. However, the underlying molecular mechanism by which c-Myc regulates EMT and CSC potential in remains unclear. In the present study, we showed that the expression of c-Myc protein is inversely correlated with microRNA (miR)-200c expression in primary tumor samples from nasopharyngeal cancer (NPC) patients. We further demonstrated that Myc and miR-200c negatively regulate the expression each other in NPC cell lines. c-Myc transcriptionally repressed expression of miR-200c by directly binding to two E-box sites located within a 1 kb segment upstream of TSS of the miR-200c. In addition, miR-200c post-transcriptionally repressed expression of c-Myc by binding to its 3'-untranslated region, suggesting the existence of a negative feedback loop between Myc and miR-200c. Overexpression of c-Myc interfered with this feedback loop and activated the EMT program, induced CSC phenotypes, and enhanced drug sensitivity, whereas miR-200c could counteract these biological effects of c-Myc. Our results provide a novel mechanism governing c-Myc and miR-200c expression and indicate that either targeting c-Myc or restoring miR-200c expression would be a promising approach to overcome oncogenic role of c-Myc in NPC.


Subject(s)
Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Nasopharyngeal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-myc/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Cisplatin/pharmacology , Humans , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Prognosis , Proto-Oncogene Proteins c-myc/genetics , Survival Rate , Tumor Cells, Cultured
2.
Bioinformatics ; 2019 Nov 04.
Article in English | MEDLINE | ID: mdl-31681951

ABSTRACT

SUMMARY: We present a web server, GenCLiP 3, which is an updated version of GenCLiP 2.0 to enhance analysis of human gene functions and regulatory networks, with the following improvements: i) accurate recognition of molecular interactions with polarity and directionality from the entire PubMed database; ii) support for Boolean search to customize multiple-term search and to quickly retrieve function related genes; iii) strengthened association between gene and keyword by a new scoring method; and iv) daily updates following literature release at PubMed FTP. AVAILABILITY: The server is freely available for academic use at: http://ci.smu.edu.cn/genclip3/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

3.
Cancer Cell Int ; 19: 348, 2019.
Article in English | MEDLINE | ID: mdl-31889900

ABSTRACT

BACKGROUND: Casticin, an isoflavone compound extracted from the herb Fructus Viticis, has demonstrated anti-inflammatory and anticancer activities and properties. The aim of this study was to investigate the effects and mechanisms of casticin in nasopharyngeal carcinoma (NPC) cells and to determine its potential for targeted use as a medicine. METHODS: NPC cells were used to perform the experiments. The CCK­8 assay and colony formation assays were used to assess cell viability. Flow cytometry was used to measure the cell cycle and apoptosis analysis (annexin V/PI assay). A three-dimensional (3D) tumour sphere culture system was used to characterize the effect of casticin on NPC stem cells. In silico molecular docking prediction and high-throughput KINOME scan assays were used to evaluate the binding of casticin to phosphoinositide 3-kinase (PI3K), including wild-type and most of mutants variants. We also used the SelectScreen assay to detect the IC50 of ATP activity in the active site of the target kinase. Western blotting was used to evaluate the changes in key proteins involved cell cycle, apoptosis, stemness, and PI3K/protein kinase B (AKT) signalling. The effect of casticin treatment in vivo was determined by using a xenograft mouse model. RESULTS: Our results indicate that casticin is a new and novel selective PI3K inhibitor that can significantly inhibit NPC proliferation and that it induces G2/GM arrest and apoptosis by upregulating Bax/BCL2 expression. Moreover, casticin was observed to affect the self-renewal ability of the nasopharyngeal carcinoma cell lines, and a combination of casticin with BYL719 was observed to induce a decrease in the level of the phosphorylation of mTORC1 downstream targets in BYL719-insensitive NPC cell lines. CONCLUSION: Casticin is a newly emerging selective PI3K inhibitor with potential for use as a targeted therapeutic treatment for nasopharyngeal carcinoma. Accordingly, casticin might represent a novel and effective agent against NPC and likely has high potential for combined use with pharmacological agents targeting PI3K/AKT.

4.
PLoS Genet ; 12(10): e1006308, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27711113

ABSTRACT

miR-155 plays critical roles in numerous physiological and pathological processes, however, its function in the regulation of blood glucose homeostasis and insulin sensitivity and underlying mechanisms remain unknown. Here, we reveal that miR-155 levels are downregulated in serum from type 2 diabetes (T2D) patients, suggesting that miR-155 might be involved in blood glucose control and diabetes. Gain-of-function and loss-of-function studies in mice demonstrate that miR-155 has no effects on the pancreatic ß-cell proliferation and function. Global transgenic overexpression of miR-155 in mice leads to hypoglycaemia, improved glucose tolerance and insulin sensitivity. Conversely, miR-155 deficiency in mice causes hyperglycemia, impaired glucose tolerance and insulin resistance. In addition, consistent with a positive regulatory role of miR-155 in glucose metabolism, miR-155 positively modulates glucose uptake in all cell types examined, while mice overexpressing miR-155 transgene show enhanced glycolysis, and insulin-stimulated AKT and IRS-1 phosphorylation in liver, adipose tissue or skeletal muscle. Furthermore, we reveal these aforementioned phenomena occur, at least partially, through miR-155-mediated repression of important negative regulators (i.e. C/EBPß, HDAC4 and SOCS1) of insulin signaling. Taken together, these findings demonstrate, for the first time, that miR-155 is a positive regulator of insulin sensitivity with potential applications for diabetes treatment.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Hyperglycemia/genetics , Insulin Resistance/genetics , Insulin/genetics , MicroRNAs/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Cell Proliferation/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Glucose/metabolism , Humans , Hyperglycemia/blood , Hyperglycemia/pathology , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Proto-Oncogene Proteins c-akt/genetics
5.
J Transl Med ; 14(1): 131, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27170279

ABSTRACT

BACKGROUND: The receptor for activated C kinase 1 (RACK1) is involved in various cancers, but its roles in nasopharyngeal carcinoma (NPC) have not yet been fully elucidated. METHODS: Initially, RACK1 expression was analyzed by immunohistochemistry in NPC and normal nasopharyngeal (NP) tissues. It was also detected by qPCR and Western blot in NPC cells. Confocal microscope and immunofluorescence were performed to detect the subcellular compartmentalization of RACK1. Subsequently, after up- or down-regulating RACK1 in NPC cells, cell proliferation and migration/invasion were tested using in vitro assays including MTT, EdU, colony formation, Transwell and Boyden assays. Furthermore, several key molecules were detected by Western blot to explore underlying mechanism. Finally, clinical samples were analyzed to confirm the relationship between RACK1 expression and clinical features. RESULTS: Receptor for activated C kinase 1 expression was much higher in NPC than NP tissues. And RACK1 was mainly located in the cytoplasm. Overexpression of RACK1 promoted NPC cell proliferation and metastasis/invasion, whereas depletion of this protein suppressed NPC cell proliferation and metastasis/invasion. Mechanistically, RACK1 deprivation obviously suppressed the activation of Akt and FAK, suggesting the PI3K/Akt/FAK pathway as one of functional mechanisms of RACK1 in NPC. Furthermore, clinical sample analysis indicated a positive correlation between in vivo expression of RACK1 with lymph node invasion and clinical stage of NPC. CONCLUSION: Our results demonstrate that RACK1 protein plays an important role in NPC development and progression. The upregulation of RACK1 can promote the proliferation and invasion of NPC by regulating the PI3K/Akt/FAK signal pathway. Thus, this study contributes to the discovery of a potential therapeutic target for NPC.


Subject(s)
Carcinoma/metabolism , Carcinoma/pathology , Disease Progression , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Receptors, Cell Surface/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Female , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors for Activated C Kinase , Signal Transduction
6.
Am J Pathol ; 185(9): 2412-20, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26362716

ABSTRACT

Gastric cancer is one of the most common malignancies in developing countries. We examined the possible role of miR-506 in gastric cancer, investigated its associations with the clinical outcomes of gastric cancer patients, and explored its potential role in angiogenesis and the metastasis of gastric cancer cells. We found that miR-506 expression was a useful marker for stratifying patients from early to advanced clinical stages and for overall survival prediction. miR-506 overexpression inhibited the epithelial-to-mesenchymal transition of gastric cancer cells; however, depletion of miR-506 promoted it. In addition, miR-506 suppressed gastric cancer angiogenesis and was associated with decreased matrix metalloproteinase-9 expression. We also found that ETS1 was a miR-506 target, and it was expressed in 71.10% of gastric cancer tissue samples. Moreover, ETS1 expression was associated with matrix metalloproteinase-9 expression (P < 0.001). In conclusion, miR-506 was identified as an ETS1 targeting suppressor of metastatic invasion and angiogenesis in gastric cancer.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Stomach Neoplasms/genetics , Adult , Aged , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Stomach Neoplasms/blood supply , Stomach Neoplasms/pathology
8.
Genome ; 59(10): 816-826, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27643679

ABSTRACT

The Cre/loxP system has become an important tool for the conditional gene knockout and conditional gene expression in genetically engineered mice. The applications of this system depend on transgenic reporter mouse lines that provide Cre recombinase activity with a defined cell type-, tissue-, or developmental stage-specificity. To develop a sensitive assay for monitoring Cre-mediated DNA excisions in mice, we generated Cre-mediated excision reporter mice, designated R/L mice (R/L: mRFP(monomeric red fluorescent protein)/luciferase), express mRFP throughout embryonic development and adult stages, while Cre-mediated excision deletes a loxP-flanked mRFP reporter gene and STOP sequence, thereby activating the expression of the second reporter gene luciferase, as assayed by in vivo and ex vivo bioluminescence imaging. After germ line deletion of the floxed mRFP and STOP sequence in R/L mice by EIIa-Cre mice, the resulting luciferase transgenic mice in which the loxP-mRFP-STOP-loxP cassette is excised from all cells express luciferase in all tissues and organs examined. The expression of luciferase transgene was activated in liver of RL/Alb-Cre double transgenic mice and in brain of RL/Nestin-Cre double transgenic mice when R/L reporter mice were mated with Alb-Cre mice and Nestin-Cre mice, respectively. Our findings reveal that the double reporter R/L mouse line is able to indicate the occurrence of Cre-mediated excision from early embryonic to adult lineages. Taken together, these findings demonstrate that the R/L mice serve as a sensitive reporter for Cre-mediated DNA excision both in living animals and in organs, tissues, and cells following necropsy.


Subject(s)
Gene Expression , Gene Knockout Techniques , Genes, Reporter , Integrases/metabolism , Luciferases/genetics , Luminescent Proteins/genetics , Animals , Female , Homologous Recombination , Liver/metabolism , Male , Mice , Mice, Transgenic , Phenotype , Transcriptional Activation , Red Fluorescent Protein
9.
Carcinogenesis ; 36(1): 41-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25416560

ABSTRACT

Junctional adhesion molecule-A (JAM-A) is preferentially concentrated at tight junctions and influences epithelial cell morphology and migration. Epithelial-to-mesenchymal transition (EMT) is the conversion process of epithelial cells into mesenchymal cells, and it plays an important role in the invasiveness and metastasis of various cancers. However, the role of JAM-A in regulating the invasive behaviours of human nasopharyngeal carcinoma (NPC) is unknown. In this study, we found that JAM-A upregulation induced EMT, whereas silencing of endogenous JAM-A expression reversed EMT. Furthermore, upregulation of JAM-A led to EMT via activation phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) pathway. PI3K inhibitors blocked JAM-A-induced EMT, suggesting that the kinase acts downstream of JAM-A. Finally, results from 172 human patients with NPC showed that high expression levels of JAM-A correlated with metastasis and poor prognosis in NPC. Taken together, these results suggest that high JAM-A expression positively correlates with poor prognosis in patients with NPC, and induces EMT of NPC cells in vitro and in vivo via the PI3K/Akt pathway. These data indicate novel functions in the JAM-A repertoire, and have clinical implications for the treatment of patients with NPC.


Subject(s)
Cell Adhesion Molecules/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/secondary , Nasopharyngeal Neoplasms/pathology , Receptors, Cell Surface/metabolism , Adult , Aged , Carcinoma , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Proliferation/radiation effects , Female , Fluorescent Antibody Technique , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/mortality , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Radiation Tolerance , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Signal Transduction , Survival Rate , Tumor Cells, Cultured
10.
Lab Invest ; 95(9): 1056-70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26098000

ABSTRACT

The miR-19 family (miR-19a and miR-19b-1) are key oncogenic components of the miR-17-92 cluster. Overexpression of miR-19 is strongly associated with cancer invasion and metastasis, and poor prognosis of cancer patients. However, the underlying mechanisms remain largely unknown. In the present study, we found that enforced expression of miR-19 including miR-19a and miR-19b-1 triggered epithelial-mesenchymal transition (EMT) of lung cancer cells A549 and HCC827 as shown by mesenchymal-like morphological conversion, downregulation of epithelial proteins (e.g., E-cadherin, ZO-1 (zona occludens 1), and α-catenin), upregulation of mesenchymal proteins (e.g., vimentin, fibronectin 1, N-cadherin, and snail1), formation of stress fibers, and reduced cell adhesion. In addition, enhanced migration and invasion were observed in the cancer cells A549 and HCC827 undergoing EMT. In contrast, silencing of endogenous miR-19 reversed EMT and reduced the migration and invasion abilities of A549 and HCC827 cells. DNA microarray results revealed significant changes of the expression of genes related to EMT, migration, and metastasis of miR-19-expressing A549 cells. Moreover, siRNA-mediated knockdown of PTEN, a target of miR-19, also resulted in EMT, migration, and invasion of A549 and HCC827 cells, suggesting that PTEN is involved in miR-19-induced EMT, migration and invasion of lung cancer cells. Furthermore, lung cancer cells undergoing EMT induced by miR-19 demonstrated reduced proliferation in vitro and in vivo, and enhanced resistance to apoptosis caused by TNF-α. Taken together, these findings suggest that miR-19 triggers EMT, which has an important role in the invasion and migration of lung cancer cells, accompanied by the reduced proliferation of cells.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/physiology , Lung Neoplasms/physiopathology , MicroRNAs/metabolism , Animals , Antigens, CD/metabolism , Blotting, Western , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Luciferases , Mice , Mice, Inbred BALB C , MicroRNAs/pharmacology , Oligonucleotide Array Sequence Analysis , RNA Interference , Snail Family Transcription Factors , Tetrazolium Salts , Thiazoles , Transcription Factors/metabolism , Tumor Stem Cell Assay , Vimentin/metabolism , Zonula Occludens-1 Protein/metabolism , alpha Catenin/metabolism
11.
Bioinformatics ; 30(17): 2534-6, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24764463

ABSTRACT

UNLABELLED: Identifying biological functions and molecular networks in a gene list and how the genes may relate to various topics is of considerable value to biomedical researchers. Here, we present a web-based text-mining server, GenCLiP 2.0, which can analyze human genes with enriched keywords and molecular interactions. Compared with other similar tools, GenCLiP 2.0 offers two unique features: (i) analysis of gene functions with free terms (i.e. any terms in the literature) generated by literature mining or provided by the user and (ii) accurate identification and integration of comprehensive molecular interactions from Medline abstracts, to construct molecular networks and subnetworks related to the free terms. AVAILABILITY AND IMPLEMENTATION: http://ci.smu.edu.cn. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Data Mining/methods , Gene Regulatory Networks , Genes , Software , Cluster Analysis , Humans , Internet , MEDLINE
12.
BMC Cancer ; 15: 1, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25971837

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is the most common primary central nervous system neoplasm in adults. Radioactive (125)I seed implantation has been widely applied in the treatment of cancers. Moreover, previous clinical trials have confirmed that (125)I seeds treatment was an effective therapy in GBM. We sought to investigate the effect of (125)I seed on GBM cell growth and Epithelial-mesenchymal transition (EMT). METHODS: Cells were exposed to irradiation at different doses. Colony-formation assay, EdU assay, cell cycle analysis, and TUNEL assay were preformed to investigate the radiation sensitivity. The effects of (125)I seeds irradiation on EMT were measured by transwell, Boyden and wound-healing assays. The levels of reactive oxygen species (ROS) were measured by DCF-DA assay. Moreover, the radiation sensitivity and EMT were investigated with or without pretreatment with glutathione. Additionally, nude mice with tumors were measured after treated with radiation. RESULTS: Radioactive (125)I seeds are more effective than X-ray irradiation in inhibiting GBM cell growth. Moreover, EMT was effectively inhibited by (125)I seed irradiation. A mechanism study indicated that GBM cell growth and EMT inhibition were induced by (125)I seeds with the involvement of a ROS-mediated signaling pathway. CONCLUSIONS: Radioactive (125)I seeds exhibit novel anticancer activity via a ROS-mediated signaling pathway. These findings have clinical implications for the treatment of patients with GBM by (125)I seeds.


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Iodine Radioisotopes/pharmacology , Radiopharmaceuticals/pharmacology , Animals , Apoptosis , Brain Neoplasms/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , G2 Phase Cell Cycle Checkpoints , Glioblastoma/pathology , Humans , Iodine Radioisotopes/therapeutic use , Mice, Inbred BALB C , Mice, Nude , Radiation Tolerance , Radiopharmaceuticals/therapeutic use , Reactive Oxygen Species/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
13.
Mol Cancer ; 13: 51, 2014 Mar 08.
Article in English | MEDLINE | ID: mdl-24606633

ABSTRACT

BACKGROUND: MiR-17-92 cluster and its paralogues have emerged as crucial regulators of many oncogenes and tumor suppressors. Transforming growth factor-ß receptor II (TGFßR2), as an important tumor suppressor, is involved in various cancer types. However, it is in cancer that only two miRNAs of this cluster and its paralogues have been reported so far to regulate TGFßR2. MiR-93 is oncogenic, but its targetome in cancer has not been fully defined. The role of miR-93 in nasopharyngeal carcinoma (NPC) still remains largely unknown. METHODS: We firstly evaluated the clinical signature of TGFßR2 down-regulation in clinical samples, and next used a miRNA expression profiling analysis followed by multi-validations, including Luciferase reporter assay, to identify miRNAs targeting TGFßR2 in NPC. In vitro and in vivo studies were performed to further investigate the effects of miRNA-mediated TGFßR2 down-regulation on NPC aggressiveness. Finally, mechanism studies were conducted to explore the associated pathway and genes influenced by this miRNA-mediated TGFßR2 down-regulation. RESULTS: TGFßR2 was down-regulated in more than 50% of NPC patients. It is an unfavorable prognosis factor contributing to clinical NPC aggressiveness. A cluster set of 4 TGFßR2-associated miRNAs was identified; they are all from miR-17-92 cluster and its paralogues, of which miR-93 was one of the most significant miRNAs, directly targeting TGFßR2, promoting cell proliferation, invasion and metastasis in vitro and in vivo. Moreover, miR-93 resulted in the attenuation of Smad-dependent TGF-ß signaling and the activation of PI3K/Akt pathway by suppressing TGFßR2, further promoting NPC cell uncontrolled growth, invasion, metastasis and EMT-like process. Impressively, the knockdown of TGFßR2 by siRNA displayed a consentaneous phenocopy with the effect of miR-93 in NPC cells, supporting TGFßR2 is a major target of miR-93. Our findings were also substantiated by investigation of the clinical signatures of miR-93 and TGFßR2 in NPC. CONCLUSION: The present study reports an involvement of miR-93-mediated TGFßR2 down-regulation in NPC aggressiveness, thus giving extended insights into molecular mechanisms underlying cancer aggressiveness. Approaches aimed at blocking miR-93 may serve as a promising therapeutic strategy for treating NPC patients.


Subject(s)
MicroRNAs/genetics , Nasopharyngeal Neoplasms/genetics , Neoplasm Invasiveness/genetics , Protein Serine-Threonine Kinases/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Adult , Aged , Carcinoma , Cell Movement/genetics , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , MicroRNAs/metabolism , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Neoplasm Metastasis , Prognosis , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Xenograft Model Antitumor Assays
14.
BMC Plant Biol ; 14: 226, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25181943

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are a new class of endogenous regulators of a broad range of physiological processes, which act by regulating gene expression post-transcriptionally. The brassica vegetable, broccoli (Brassica oleracea var. italica), is very popular with a wide range of consumers, but environmental stresses such as salinity are a problem worldwide in restricting its growth and yield. Little is known about the role of miRNAs in the response of broccoli to salt stress. In this study, broccoli subjected to salt stress and broccoli grown under control conditions were analyzed by high-throughput sequencing. Differential miRNA expression was confirmed by real-time reverse transcription polymerase chain reaction (RT-PCR). The prediction of miRNA targets was undertaken using the Kyoto Encyclopedia of Genes and Genomes (KEGG) Orthology (KO) database and Gene Ontology (GO)-enrichment analyses. RESULTS: Two libraries of small (or short) RNAs (sRNAs) were constructed and sequenced by high-throughput Solexa sequencing. A total of 24,511,963 and 21,034,728 clean reads, representing 9,861,236 (40.23%) and 8,574,665 (40.76%) unique reads, were obtained for control and salt-stressed broccoli, respectively. Furthermore, 42 putative known and 39 putative candidate miRNAs that were differentially expressed between control and salt-stressed broccoli were revealed by their read counts and confirmed by the use of stem-loop real-time RT-PCR. Amongst these, the putative conserved miRNAs, miR393 and miR855, and two putative candidate miRNAs, miR3 and miR34, were the most strongly down-regulated when broccoli was salt-stressed, whereas the putative conserved miRNA, miR396a, and the putative candidate miRNA, miR37, were the most up-regulated. Finally, analysis of the predicted gene targets of miRNAs using the GO and KO databases indicated that a range of metabolic and other cellular functions known to be associated with salt stress were up-regulated in broccoli treated with salt. CONCLUSION: A comprehensive study of broccoli miRNA in relation to salt stress has been performed. We report significant data on the miRNA profile of broccoli that will underpin further studies on stress responses in broccoli and related species. The differential regulation of miRNAs between control and salt-stressed broccoli indicates that miRNAs play an integral role in the regulation of responses to salt stress.


Subject(s)
Brassica/metabolism , MicroRNAs/metabolism , Salinity , Stress, Physiological , Base Sequence , Conserved Sequence , High-Throughput Nucleotide Sequencing , Salt Tolerance , Sequence Analysis, RNA
15.
Mol Carcinog ; 53(11): 858-70, 2014 Nov.
Article in English | MEDLINE | ID: mdl-23908159

ABSTRACT

Previous studies have shown that promoter hypermethylation plays a key role in DLC-1 inactivation in nasopharyngeal carcinoma (NPC). However, DLC-1 mutation in NPC has not been reported, and there remain some discrepancies in methods and results between different groups. Here, we examined the mRNA and protein expression of DLC-1 in chronic nasopharyngitis (CN) and NPC tissues by reverse transcription-polymerase chain reaction/qPCR and immunohistochemistry, respectively. DLC-1 mRNA was undetectable in all the seven widely used NPC cell lines and absent or significantly down-regulated in 70% of NPC tissues. DLC-1 protein level was reduced in 74.3% of NPCs when compared with CN tissues, and significantly lower in NPC samples at advanced clinical stages than that at early stages. Then, we purified the same batch of specimens by microdissection and analyzed the possible mechanisms of DLC-1 downregulation with mutation and allelic loss analysis, methylation-specific PCR and bisulfite genomic sequencing. Only one mutation was detected at codon 693 of exon 8 in 3.3% of NPCs and five single nucleotide polymorphisms (SNPs) were identified. Loss of DLC-1 was detected in 23.3% of NPC tissues. The 100% of NPC cell lines, 80% of primary NPC and 22.2% of CN tissues showed methylation in DLC-1 promoter, while DLC-1 expression was recovered in seven NPC cell lines after 5-aza-dC treatment. Patched methylation assay confirmed that promoter methylation could repress DLC-1 expression. This report demonstrates that DLC-1 is negatively associated with NPC carcinogenesis, and promoter hypermethylation along with loss of heterozygosity, but not mutation, contributes to inactivation of DLC-1 in NPC.


Subject(s)
DNA Methylation/genetics , GTPase-Activating Proteins/genetics , Loss of Heterozygosity , Nasopharyngeal Neoplasms/genetics , Promoter Regions, Genetic/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Aged , Base Sequence , Carcinoma , Down-Regulation , Female , GTPase-Activating Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Mutation , Nasopharyngeal Carcinoma , Nasopharyngitis/genetics , Polymorphism, Single Nucleotide , RNA, Messenger/biosynthesis , Sequence Analysis, DNA , Tumor Suppressor Proteins/biosynthesis , Young Adult
16.
Mol Cancer ; 12: 53, 2013 Jun 08.
Article in English | MEDLINE | ID: mdl-23758864

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) is a common malignant tumor in southern China and Southeast Asia, but its molecular mechanisms of pathogenesis are poorly understood. Our previous work has demonstrated that BCAT1 mRNA is over expressed in NPC and knocking down its expression in 5-8F NPC cell line can potently inhibit cell cycle progression and cell proliferation. However, the mechanism of BCAT1 up-regulation and its functional role in NPC development remain to be elucidated yet. METHODS: Immunohistochemistry (IHC) method was utilized to detect the expression of BCAT1 protein in NPC at different pathological stages. The roles of gene mutation, DNA amplification and transcription factor c-Myc in regulating BCAT1 expression were analyzed using PCR-sequencing, quantitative polymerase chain reaction (qPCR), IHC, ChIP and luciferase reporter system, respectively. The functions of BCAT1 in colony formation, cell migration and invasion properties were evaluated by RNA interference (RNAi). RESULTS: The positive rates of BCAT1 protein expression in normal epithelia, low-to-moderate grade atypical hyperplasia tissues, high-grade atypical hyperplasia tissues and NPC tissues were 23.6% (17/72), 75% (18/24), 88.9% (8/9) and 88.8% (71/80), respectively. Only one SNP site in exon1 was detected, and 42.4% (12/28) of the NPC tissues displayed the amplification of microsatellite loci in BCAT1. C-Myc could directly bind to the c-Myc binding site in promoter region of BCAT1 and up-regulate its expression. The mRNA and protein of c-Myc and BCAT1 were co-expressed in 53.6% (15/28) and 59.1% (13/22) of NPC tissues, respectively, and BCAT1 mRNA expression was also down-regulated in c-Myc knockdown cell lines. In addition, BCAT1 knockdown cells demonstrated reduced proliferation and decreased cell migration and invasion abilities. CONCLUSIONS: Our study indicates that gene amplification and c-Myc up-regulation are responsible for BCAT1 overexpression in primary NPC, and overexpression of BCAT1 induces cell proliferation, migration and invasion. The results suggest that BCAT1 may be a novel molecular target for the diagnosis and treatment of NPC.


Subject(s)
Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Transaminases/metabolism , Animals , Base Sequence , Carcinoma , Cell Line , Cell Movement/genetics , Cell Proliferation , Exons , Gene Amplification , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Mutation , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Neoplasm Invasiveness , Neoplasm Staging , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transaminases/genetics , Tumor Stem Cell Assay
17.
Biochem Biophys Res Commun ; 435(1): 160-4, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23628413

ABSTRACT

ESRG (embryonic stem cell related gene, also known as HESRG), is a novel human gene first cloned and identified by our group with microarray analysis. Interestingly, it is expressed specifically in undifferentiated human embryonic stem cells (hESCs), while its expression pattern and its role in hESCs remain unclear. Here, full-length 3151nt ESRG cDNA was further identified by RNA ligase mediated rapid amplification of cDNA ends (RLM-RACE) technique. Meanwhile, an alternatively splicing ESRG transcript (ESRG-B) of 2837nt in length was also found. Surprisingly, bioinformatics analyses showed that the open reading frames (ORFs) of ESRG and ESRG-B were identical. Both of them consist of 669nt and encode a 222aa protein with a predicted molecular size of 24 kDa. The ESRG protein was located in the nuclei of hESCs as demonstrated by immunocytochemical staining and Western blotting using ESRG specific antibody generated by us. In contrast, ESRG located in the cytoplasm of COS7 cells when it was forced to be expressed in these cells by gene transfection strategy, suggesting there may be some special proteins present only in hESCs which can help ESRG protein transport into the nuclei of hESCs. By spatial expression analysis, we further discovered that ESRG only expressed in the ovary tissue and hESCs instead of other tissues or cell lines. Our current data provide us with an important basis for conducting further studies on the functions and regulatory mechanisms underlying the role of ESRG in hESCs.


Subject(s)
Cell Nucleus/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Proteins/genetics , Proteins/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , COS Cells , Cell Line , Chlorocebus aethiops , Cytoplasm/metabolism , Embryonic Stem Cells/cytology , Female , Fetus/metabolism , Humans , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Ovary/embryology , Ovary/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Long Noncoding , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem Biophys Res Commun ; 436(1): 19-24, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23685147

ABSTRACT

EBV-miR-BART1 has been found to be highly expressed in some cancers including nasopharyngeal carcinoma (NPC), but its exact roles in the pathogenesis of NPC remain unclear. Here, we did RNA deep sequencing to compare the gene expression profile between EBV-miR-BART1-expressing CNE1 cells and the control cells to determine the possible effects of EBV-miR-BART1 in NPC. Gene expression profiling analysis unexpectedly showed a significant number of up- and down-modulated metabolism-associated genes, such as G6PD, SAT1, ASS1, PAST1, FUT1, SGPL1, DHRS3, B4GALT1, PHGDH, IDH2, PISD, UGT8, LDHB and GALNT1, in EBV-miR-BART1-expressing NPC cells, which were next confirmed by RT-qPCR. Moreover, of these metabolism-genes, PSAT1 and PHGDH expression levels were significantly upregulated and most of other genes were obviously up-expressed in NPC specimens compared with chronic nasopharyngitis (CNP) tissues. Collectively, we for the first time found the effects of EBV-miR-BART1 on the expression of mechanism-associated genes in NPC, suggesting a novel role of EBV-miR-BART1 in cancer metabolism, which remains to be fully elucidated.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/metabolism , MicroRNAs/metabolism , Nasopharyngeal Neoplasms/metabolism , Biopsy , Carcinoma , Cell Line, Tumor , DNA, Viral/analysis , Epstein-Barr Virus Infections/metabolism , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/virology , Nasopharyngitis/metabolism , Transcription Factors
19.
Biochem Biophys Res Commun ; 431(3): 610-6, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23291181

ABSTRACT

The functions of miR-9 in some cancers are recently implicated in regulating proliferation, epithelial-mesenchymal transition (EMT), invasion and metastasis, apoptosis, and tumor angiogenesis, etc. miR-9 is commonly down-regulated in nasopharyngeal carcinoma (NPC), but the exact roles of miR-9 dysregulation in the pathogenesis of NPC remains unclear. Therefore, we firstly used miR-9-expressing CNE2 cells to determine the effects of miR-9 overexpression on global gene expression profile by microarray analysis. Microarray-based gene expression data unexpectedly demonstrated a significant number of up- or down-regulated immune- and inflammation-related genes, including many well-known interferon (IFN)-induced genes (e.g., IFI44L, PSMB8, IRF5, PSMB10, IFI27, PSB9_HUMAN, IFIT2, TRAIL, IFIT1, PSB8_HUMAN, IRF1, B2M and GBP1), major histocompatibility complex (MHC) class I molecules (e.g., HLA-B, HLA-C, HLA-F and HLA-H) and interleukin (IL)-related genes (e.g., IL20RB, GALT, IL7, IL1B, IL11, IL1F8, IL1A, IL6 and IL7R), which was confirmed by qRT-PCR. Moreover, the overexpression of miR-9 with the miRNA mimics significantly up- or down-regulated the expression of above-mentioned IFN-inducible genes, MHC class I molecules and IL-related genes; on the contrary, miR-9 inhibition by anti-miR-9 inhibitor in CNE2 and 5-8F cells correspondingly decreased or increased the aforementioned immune- and inflammation-related genes. Taken together, these findings demonstrate, for the first time, that miR-9 can modulate the expression of IFN-induced genes and MHC class I molecules in human cancer cells, suggesting a novel role of miR-9 in linking inflammation and cancer, which remains to be fully characterized.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, MHC Class I , Interferons/metabolism , MicroRNAs/physiology , Nasopharyngeal Neoplasms/genetics , Carcinoma , Humans , Inflammation/genetics , Inflammation/immunology , MicroRNAs/genetics , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/immunology , Oligonucleotide Array Sequence Analysis
20.
J Transl Med ; 11: 114, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23648139

ABSTRACT

BACKGROUND: Although the prognostic roles of ß-catenin expression in non-small cell lung cancer (NSCLC) have been reported in several immunohistochemical (IHC) studies, the results were not consistent because some studies lack sufficient number of the positive cases or did not evaluate the subcellular localization features of the protein. METHOD: In this study, we have evaluated the expression levels and subcellular localization of ß-catenin and Nanog proteins IHC staining in tissue specimens from 309 patients with NSCLC, and explored their association with clinicopathological features and patient outcome. RESULTS: We showed that patients with negative expression of membranous beta-catenin had a trend towards shorter survival (p=0.064) than those with positive expression. In contrast to previous studies, we found that increased expression of either cytoplasmic or nuclear ß-catenin was strongly associated with poor prognosis and was an independent prognosticator for overall survival (p <0.01). We further found that NSCLC cells frequently exhibited an abundance of nuclear Nanog protein which was significantly correlated with nuclear ß-catenin expression (p <0.01) and poor prognosis (p <0.01). Interestingly, immunofluorescent staining results revealed that increased expression of Nanog and nuclear translocation of ß-catenin occurred concomitantly in response to epidermal growth factor receptor(EGFR) signaling in A549 and H23 cells. Furthermore, western blot analysis show that nuclear ß-catenin rather than cytoplasmic ß-catenin expression in the A549 and H23 cells can be enhanced by adding EGF, Nanog expression in the A549 and H23 cells with knockdown of ß-catenin can not be obviously enhanced by adding EGF. CONCLUSION: We propose that evaluation of subcellular localization of ß-catenin and Nanog expression is of clinical significance for patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Lung Neoplasms/metabolism , beta Catenin/metabolism , Active Transport, Cell Nucleus , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Cytoplasm/metabolism , Female , Humans , Immunohistochemistry , Lung Neoplasms/diagnosis , Lung Neoplasms/mortality , Male , Middle Aged , Nanog Homeobox Protein , Prognosis , Signal Transduction , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL