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










Database
Language
Publication year range
1.
Mol Carcinog ; 56(3): 1041-1054, 2017 03.
Article in English | MEDLINE | ID: mdl-27597634

ABSTRACT

In our previous whole-transcriptome sequencing analysis, downregulation of a long non-coding RNA, maternally expressed gene 3 (MEG3), was identified in NPC samples. This finding suggests the possible role of MEG3 as a tumor suppressor in this distinctive disease. In the present study, two MEG3 variants, AF119863 (MEG3-AF) and BX247998 (MEG3-BX), were found abundantly expressed in a normal nasopharyngeal epithelial cell line, NP69. Significant downregulation of MEG3-AF was further verified in a panel of NPC samples including xenografts and primary biopsies. MEG3 is an imprinted gene located within chromosome 14q32, a common deleted region in NPC. Both DNA copy number loss and aberrant promoter methylation contributed to MEG3 inactivation. Interestingly, MEG3 expression could successfully be rescued by the treatment of a demethylation agent. Besides, ectopic expression of MEG3 in NPC cell lines resulted in considerable repression of in vitro anchorage-independent growth and in vivo tumorigenicity, in addition to significant inhibition in cell proliferation, colony formation, and induction of cell cycle arrest. Finally, we revealed the association between MEG3 activity and the p53 signaling cascade. Our findings characterize MEG3 as a tumor suppressive long non-coding RNA in NPC and encourage the development of precise long non-coding RNA-targeted epigenetic therapy against this malignancy. © 2016 Wiley Periodicals, Inc.


Subject(s)
Carcinoma/genetics , DNA Copy Number Variations , DNA Methylation , Down-Regulation , Nasopharyngeal Neoplasms/genetics , RNA, Long Noncoding/genetics , Animals , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Chromosomes, Human, Pair 14/genetics , Gene Expression Regulation, Neoplastic , Genomic Imprinting , Humans , Mice , Nasopharyngeal Carcinoma , Neoplasm Transplantation , Promoter Regions, Genetic
2.
BMC Cancer ; 16: 495, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27431799

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV)-associated epithelial malignancy that exhibits distinct geographical and ethnic prevalence. Although the contemporary therapeutic approach of radio-/chemotherapy provides excellent results for patients with early-stage disease, it is far from satisfactory for those with disease remission and distant metastasis. Promising therapeutic strategies for advanced and relapsed NPC are still lacking. We recently identified and characterized a cancer stem-like cell (CSC) subpopulation in NPC that appeared to play an important role in tumor progression. Microarray analysis revealed downregulation of several stemness-inhibiting miRNAs in these CSC cells. Among these miRNAs, miR-96 and miR-183 showed the highest fold change and were selected to elucidate their role in repressing NPC CSC properties. METHODS: MiR-96 and miR-183 expression in NPC CSCs was detected by qRT-PCR. Transient and stable transfection was performed in EBV-positive NPC C666-1 cells to examine the effects of ectopic expression of miR-96 and miR-183 on repressing cell growth and CSC properties. Anchorage-dependent (colony formation) and anchorage-independent (tumor sphere formation) growths of these miR-96 and miR-183 expressing cells were determined. Expression of multiple CSC markers and related molecules were accessed by flow cytometry and Western blotting. The tumorigenicity of the stable miR-96- and miR-183-transfected NPC cells was examined in an in vivo nude mice model. RESULTS: Downregulation of miR-96 and miR-183 was confirmed in NPC spheroids. Using transient or stable transfection, we showed that ectopic expression of miR-96 and miR-183 suppressed cell growth and tumor sphere formation in NPC. Reduced NICD3 and NICD4 in miR-96- and miR-183-expressing NPC cells suggests the involvement of the NOTCH signaling pathway in their tumor suppressive function. Finally, we showed that the tumorigenicity of cells stably expressing miR-183 was significantly inhibited in the in vivo nude mice model. CONCLUSIONS: miR-183 is a tumor-suppressive miRNA in EBV-associated NPC. Its abilities to suppress CSC properties in vitro and effectively reduce tumor growth in vivo shed light on its role as a potential therapeutic target.


Subject(s)
Epstein-Barr Virus Infections/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Nasopharyngeal Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Female , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Humans , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/virology , Neoplastic Stem Cells/virology , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Receptor, Notch4 , Receptors, Notch/genetics , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular/metabolism , Spheroids, Cellular/virology , Transplantation, Heterologous/methods
3.
PLoS One ; 11(6): e0156833, 2016.
Article in English | MEDLINE | ID: mdl-27258148

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) is a peculiar Epstein Barr virus (EBV)-associated malignancy that is prevalent in South-East Asia. Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1) isomerizes specific phosphorylated amino acid residues, which makes it an important regulator in cell survival and apoptosis. In this study, we investigated the contribution made by PIN1 in NPC tumorigenesis and PIN1's potential role as a therapeutic target. METHODS: The expression of PIN1 was examined in a panel of NPC cell lines, xenografts and primary tumors. The functional roles of PIN1 in NPC cells were elucidated by the knockdown and overexpression of PIN1 in in vitro and in vivo nude mice models by siRNA and lenti-viral transfection, respectively. The antitumor effects of the PIN1 inhibitor Juglone in NPC cells were also evaluated. RESULTS: We revealed the consistent overexpression of PIN1 in almost all EBV-associated NPC cell lines, xenografts and primary tumors. PIN1 suppression was capable of inhibiting cyclin D1 expression and activating caspase-3 in NPC cells. It positively regulated NPC cell proliferation, colony formation and anchorage-independent growth. The inhibition of PIN1 suppressed tumor growth in vitro and in vivo. CONCLUSIONS: This study demonstrates the oncogenic role of PIN1 in NPC tumorigenesis, and shows that its overexpression can enhance tumor cell growth via the upregulation of cyclinD1. Our findings inform the development of novel treatments targeting PIN1 for NPC patients.


Subject(s)
Carcinoma/metabolism , Carcinoma/virology , Cell Proliferation/physiology , Cyclin D1/metabolism , Herpesvirus 4, Human/pathogenicity , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/virology , Animals , Carcinoma/drug therapy , Carcinoma/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclin D1/genetics , Female , HeLa Cells , Humans , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
4.
Chin J Cancer ; 33(11): 529-38, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25223912

ABSTRACT

Although the Epstein-Barr virus (EBV) has spread to all populations in the world, EBV-associated nasopharyngeal carcinoma (NPC) is prevalent only in South China and Southeast Asia. The role of EBV in the malignant transformation of nasopharyngeal epithelium is the main focus of current researches. Radiotherapy and chemoradiotherapy have been successful in treating early stage NPC, but the recurrence rates remain high. Unfortunately, local relapse and metastasis are commonly unresponsive to conventional treatments. These recurrent and metastatic lesions are believed to arise from residual or surviving cells that have the properties of cancer stem cells. These cancer stem-like cells (CSCs) have the ability to self-renew, differentiate, and sustain propagation. They are also chemo-resistant and can form spheres in anchorage-independent environments. This review summarizes recent researches on the CSCs in EBV-associated NPC, including the findings regarding cell surface markers, stem cell-related transcription factors, and various signaling pathways. In particular, the review focuses on the roles of EBV latent genes [latent membrane protein 1 (LMP1) and latent membrane protein 2A (LMP2A)], cellular microRNAs, and adenosine triphosphate (ATP)-binding cassette chemodrug transporters in contributing to the properties of CSCs, including the epithelial-mesenchymal transition, stem-like transition, and chemo-resistance. Novel therapeutics that enhance the efficacy of radiotherapy and chemoradiotherapy and inhibitors that suppress the properties of CSCs are also discussed.


Subject(s)
Cell Transformation, Neoplastic , Drug Resistance, Neoplasm/genetics , Herpesvirus 4, Human/genetics , Nasopharyngeal Neoplasms , Neoplastic Stem Cells , Carcinoma , China , Epithelial-Mesenchymal Transition , Humans , MicroRNAs , Multidrug Resistance-Associated Proteins , Nasopharyngeal Carcinoma , Nasopharynx , Neoplasm Metastasis , Neoplasm Recurrence, Local , Signal Transduction , Viral Matrix Proteins
5.
Mol Cancer ; 13: 184, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25098679

ABSTRACT

BACKGROUND: As a distinctive type of head and neck cancers, nasopharyngeal carcinoma (NPC) is genesis from the clonal Epstein-Barr virus (EBV)-infected nasopharyngeal epithelial cells accumulated with multiple genetic lesions. Among the recurrent genetic alterations defined, loss of 9p21.3 is the most frequent early event in the tumorigenesis of EBV-associated NPC. In addition to the reported CDKN2A/p16, herein, we elucidated the role of a miRNA, miR-31 within this 9p21.3 region as NPC-associated tumor suppressor. METHODS: The expression and promoter methylation of miR-31 were assessed in a panel of NPC tumor lines and primary tumors. Its in vitro and in vivo tumor suppression function was investigated through the ectopic expression of miR-31 in NPC cells. We also determined the miR-31 targeted genes and its involvement in the growth in NPC. RESULTS: Downregulation of miR-31 expression was detected in almost all NPC cell line, patient-derived xenografts (PDXs) and primary tumors. Both homozygous deletion and promoter hypermethylation were shown to be major mechanisms for miR-31 silencing in this cancer. Strikingly, loss of miR-31 was also obviously observed in the dysplastic lesions of nasopharynx. Restoration of miR-31 in C666-1 cells inhibited the cell proliferation, colony-forming and migratory capacities. Dramatic reduction of in vitro anchorage-independent growth and in vivo tumorigenic potential were demonstrated in the stable clones expressing miR-31. Furthermore, we proved that miR-31 suppressed the NPC cell growth via targeting FIH1 and MCM2. CONCLUSIONS: The findings provide strong evidence to support miR-31 as a new NPC-associated tumor suppressor on 9p21.3 region. The inactivation of miR-31 may contribute to the early development of NPC.


Subject(s)
Carcinogenesis/pathology , Herpesvirus 4, Human/physiology , MicroRNAs/metabolism , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/virology , Carcinogenesis/genetics , Carcinoma , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Comparative Genomic Hybridization , DNA Methylation/genetics , Down-Regulation/genetics , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Homozygote , Humans , MicroRNAs/genetics , Minichromosome Maintenance Complex Component 2/metabolism , Mixed Function Oxygenases/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Phosphorylation , Promoter Regions, Genetic , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
6.
PLoS One ; 9(8): e105163, 2014.
Article in English | MEDLINE | ID: mdl-25126743

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a common viral-associated neoplasm in which multiple signaling cascades are interfered with by Epstein-Bar virus (EBV) latent proteins and various genetic alterations. Aside from the previously reported PIK3CA amplification, we examined the role of INPP4B, a negative regulator of the PI3K/AKT signaling pathway in the development of NPC. By RT-PCR and Western blotting, we revealed that the expression of INPP4B was down-regulated in all five established EBV-positive tumor lines. While INPP4B was consistently expressed in normal nasopharyngeal epithelial cells, downregulation of INPP4B was found in 32/65 (49.2%) of primary tumors by immunohistochemistry. Furthermore, our study also demonstrated the hypermethylation of the 5'CpG island of INPP4B in the tumors in which INPP4B transcription was downregulated. Notably, the re-expression of INPP4B was detected in the NPC cells treated with the demethylation agent (5-aza-2'deoxycytidine). Our study showed that promoter hypermethylation was the major mechanism for transcriptional silencing of INPP4B in NPC. Furthermore, restoration of INPP4B expression significantly suppressed PI3K/AKT downstream signals in the NPC C666-1 cells. In vivo growth inhibition was clearly demonstrated in the tumor cells stably expressing INPP4B. The findings indicate that epigenetic inactivation of INPP4B is one of the key mechanisms in activating PI3K/AKT signaling cascade and playing a role in the tumorigenesis of NPC.


Subject(s)
Epstein-Barr Virus Infections/enzymology , Nasopharyngeal Neoplasms/enzymology , Phosphoric Monoester Hydrolases/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Epstein-Barr Virus Infections/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/virology , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
7.
PLoS One ; 7(12): e52426, 2012.
Article in English | MEDLINE | ID: mdl-23285037

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a unique EBV-associated epithelial malignancy, showing highly invasive and metastatic phenotype. Despite increasing evidence demonstrating the critical role of cancer stem-like cells (CSCs) in the maintenance and progression of tumors in a variety of malignancies, the existence and properties of CSC in EBV-associated NPC are largely unknown. Our study aims to elucidate the presence and role of CSCs in the pathogenesis of this malignant disease. Sphere-forming cells were isolated from an EBV-positive NPC cell line C666-1 and its tumor-initiating properties were confirmed by in vitro and in vivo assays. In these spheroids, up-regulation of multiple stem cell markers were found. By flow cytometry, we demonstrated that both CD44 and SOX2 were overexpressed in a majority of sphere-forming C666-1 cells. The CD44+SOX2+ cells was detected in a minor population in EBV-positive xenografts and primary tumors and considered as potential CSC in NPC. Notably, the isolated CD44+ NPC cells were resistant to chemotherapeutic agents and with higher spheroid formation efficiency, showing CSC properties. On the other hand, microarray analysis has revealed a number of differentially expressed genes involved in transcription regulation (e.g. FOXN4, GLI1), immune response (CCR7, IL8) and transmembrane transport (e.g. ABCC3, ABCC11) in the spheroids. Among these genes, increased expression of CCR7 in CD44+ CSCs was confirmed in NPC xenografts and primary tumors. Importantly, blocking of CCR7 abolished the sphere-forming ability of C666-1 in vitro. Expression of CCR7 was associated with recurrent disease and distant metastasis. The current study defined the specific properties of a CSC subpopulation in EBV-associated NPC. Our findings provided new insights into developing effective therapies targeting on CSCs, thereby potentiating treatment efficacy for NPC patients.


Subject(s)
Herpesvirus 4, Human/physiology , Hyaluronan Receptors/metabolism , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Neoplastic Stem Cells/pathology , Animals , Biomarkers, Tumor/metabolism , Carcinoma , Cell Membrane/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Clone Cells , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Neutralization Tests , Receptors, CCR7/metabolism , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...