Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Oncol ; 11: 640207, 2021.
Article in English | MEDLINE | ID: mdl-33718235

ABSTRACT

Nasopharyngeal carcinoma (NPC) is closely associated with Epstein-Barr virus (EBV) infection. It is also characterized by heavy infiltration with non-malignant leucocytes. The EBV-encoded latent membrane protein 1 (LMP1) is believed to play an important role in NPC pathogenesis by virtue of its ability to activate multiple cell signaling pathways which collectively promote cell proliferation and survival, angiogenesis, invasiveness, and aerobic glycolysis. LMP1 also affects cell-cell interactions, antigen presentation, and cytokine and chemokine production. Here, we discuss how LMP1 modulates local immune responses that contribute to the establishment of the NPC tumor microenvironment. We also discuss strategies for targeting the LMP1 protein as a novel therapy for EBV-driven malignancies.

2.
Cancers (Basel) ; 12(7)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32708965

ABSTRACT

Epstein-Barr virus (EBV) is closely linked to the development of a number of human cancers. EBV-associated malignancies are characterized by a restricted pattern of viral latent protein expression which is sufficient for the virus to both initiate and sustain cell growth and to protect virus-infected cells from immune attack. Expression of these EBV proteins in malignant cells provides an attractive target for therapeutic intervention. Among the viral proteins expressed in the EBV-associated epithelial malignancies, the protein encoded by the BamHI-A rightward frame 1 (BARF1) is of particular interest. BARF1 is a viral oncoprotein selectively expressed in latently infected epithelial cancers, nasopharyngeal carcinoma (NPC) and EBV-positive gastric cancer (EBV-GC). Here, we review the roles of BARF1 in oncogenesis and immunomodulation. We also discuss potential strategies for targeting the BARF1 protein as a novel therapy for EBV-driven epithelial cancers.

3.
J Pathol ; 246(2): 180-190, 2018 10.
Article in English | MEDLINE | ID: mdl-29968360

ABSTRACT

Nasopharyngeal carcinoma (NPC) is closely associated with Epstein-Barr virus (EBV) infection. The EBV-encoded latent membrane protein 1 (LMP1), which is commonly expressed in NPC, engages multiple signaling pathways that promote cell growth, transformation, and metabolic reprogramming. Here, we report a novel function of LMP1 in promoting de novo lipogenesis. LMP1 increases the expression, maturation and activation of sterol regulatory element-binding protein 1 (SREBP1), a master regulator of lipogenesis, and its downstream target fatty acid synthase (FASN). LMP1 also induces de novo lipid synthesis and lipid droplet formation. In contrast, small interfering RNA (siRNA) knockdown of LMP1 in EBV-infected epithelial cells diminished SREBP1 activation and lipid biosynthesis. Furthermore, inhibition of the mammalian target of rapamycin (mTOR) pathway, through the use of either mTOR inhibitors or siRNAs, significantly reduced LMP1-mediated SREBP1 activity and lipogenesis, indicating that LMP1 activation of the mTOR pathway is required for SREBP1-mediated lipogenesis. In primary NPC tumors, FASN overexpression is common, with high levels correlating significantly with LMP1 expression. Moreover, elevated FASN expression was associated with aggressive disease and poor survival in NPC patients. Luteolin and fatostatin, two inhibitors of lipogenesis, suppressed lipogenesis and proliferation of nasopharyngeal epithelial cells, effects that were more profound in cells expressing LMP1. Luteolin and fatostatin also dramatically inhibited NPC tumor growth in vitro and in vivo. Our findings demonstrate that LMP1 activation of SREBP1-mediated lipogenesis promotes tumor cell growth and is involved in EBV-driven NPC pathogenesis. Our results also reveal the therapeutic potential of utilizing lipogenesis inhibitors in the treatment of locally advanced or metastatic NPC. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cell Proliferation , Herpesvirus 4, Human/metabolism , Lipogenesis , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Viral Matrix Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Female , Herpesvirus 4, Human/genetics , Humans , Lipid Droplets/metabolism , Lipogenesis/drug effects , Luteolin/pharmacology , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Naphthyridines/pharmacology , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Pyridines/pharmacology , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Thiazoles/pharmacology , Viral Matrix Proteins/genetics , Xenograft Model Antitumor Assays
4.
J Pathol ; 244(4): 394-407, 2018 04.
Article in English | MEDLINE | ID: mdl-29230817

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a highly invasive epithelial malignancy that is prevalent in southern China and Southeast Asia. It is consistently associated with latent Epstein-Barr virus (EBV) infection. In NPC, miR-BARTs, the EBV-encoded miRNAs derived from BamH1-A rightward transcripts, are abundantly expressed and contribute to cancer development by targeting various cellular and viral genes. In this study, we establish a comprehensive transcriptional profile of EBV-encoded miRNAs in a panel of NPC patient-derived xenografts and an EBV-positive NPC cell line by small RNA sequencing. Among the 40 miR-BARTs, predominant expression of 22 miRNAs was consistently detected in these tumors. Among the abundantly expressed EBV-miRNAs, BART5-5p, BART7-3p, BART9-3p, and BART14-3p could negatively regulate the expression of a key DNA double-strand break (DSB) repair gene, ataxia telangiectasia mutated (ATM), by binding to multiple sites on its 3'-UTR. Notably, the expression of these four miR-BARTs represented more than 10% of all EBV-encoded miRNAs in tumor cells, while downregulation of ATM expression was commonly detected in all of our tested sequenced samples. In addition, downregulation of ATM was also observed in primary NPC tissues in both qRT-PCR (16 NP and 45 NPC cases) and immunohistochemical staining (35 NP and 46 NPC cases) analysis. Modulation of ATM expression by BART5-5p, BART7-3p, BART9-3p, and BART14-3p was demonstrated in the transient transfection assays. These findings suggest that EBV uses miRNA machinery as a key mechanism to control the ATM signaling pathway in NPC cells. By suppressing these endogenous miR-BARTs in EBV-positive NPC cells, we further demonstrated the novel function of miR-BARTs in inhibiting Zta-induced lytic reactivation. These findings imply that the four viral miRNAs work co-operatively to modulate ATM activity in response to DNA damage and to maintain viral latency, contributing to the tumorigenesis of NPC. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , MicroRNAs/genetics , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , RNA, Viral/genetics , 3' Untranslated Regions , Animals , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Binding Sites , Cell Line, Tumor , DNA Damage , Enzyme Repression , Epstein-Barr Virus Infections/diagnosis , Female , Gene Expression Regulation, Neoplastic , Heterografts , Host-Pathogen Interactions , Humans , Male , Mice, Nude , Middle Aged , Nasopharyngeal Carcinoma/enzymology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/enzymology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Transcriptome , Virus Latency
5.
Int J Cancer ; 141(8): 1512-1521, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28542909

ABSTRACT

The γ-herpesviruses, EBV and KSHV, are closely associated with a number of human cancers. While the signal transduction pathways exploited by γ-herpesviruses to promote cell growth, survival and transformation have been reported, recent studies have uncovered the impact of γ-herpesvirus infection on host cell metabolism. Here, we review the mechanisms used by γ-herpesviruses to induce metabolic reprogramming in host cells, focusing on their ability to modulate the activity of metabolic regulators and manipulate metabolic pathways. While γ-herpesviruses alter metabolic phenotypes as a means to support viral infection and long-term persistence, this modulation can inadvertently contribute to cancer development. Strategies that target deregulated metabolic phenotypes induced by γ-herpesviruses provide new opportunities for therapeutic intervention.


Subject(s)
Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Neoplasms/metabolism , Neoplasms/virology , Carcinogenesis , Cellular Reprogramming/physiology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/pathology , Herpesviridae Infections/metabolism , Herpesviridae Infections/pathology , Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/metabolism , Humans
6.
J Pathol ; 237(2): 238-48, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26096068

ABSTRACT

Non-keratinizing nasopharyngeal carcinoma (NPC) is closely associated with Epstein-Barr virus (EBV) infection. The EBV-encoded latent membrane protein 1 (LMP1) is believed to play an important role in NPC pathogenesis by virtue of its ability to activate multiple cell signalling pathways which collectively promote cell proliferation, transformation, angiogenesis, and invasiveness, as well as modulation of energy metabolism. In this study, we report that LMP1 increases cellular uptake of glucose and glutamine, enhances LDHA activity and lactate production, but reduces pyruvate kinase activity and pyruvate concentrations. LMP1 also increases the phosphorylation of PKM2, LDHA, and FGFR1, as well as the expression of PDHK1, FGFR1, c-Myc, and HIF-1α, regardless of oxygen availability. Collectively, these findings suggest that LMP1 promotes aerobic glycolysis. With respect to FGFR1 signalling, LMP1 not only increases FGFR1 expression, but also up-regulates FGF2, leading to constitutive activation of the FGFR1 signalling pathway. Furthermore, two inhibitors of FGFR1 (PD161570 and SU5402) attenuate LMP1-mediated aerobic glycolysis, cellular transformation (proliferation and anchorage-independent growth), cell migration, and invasion in nasopharyngeal epithelial cells, identifying FGFR1 signalling as a key pathway in LMP1-mediated growth transformation. Immunohistochemical staining revealed that high levels of phosphorylated FGFR1 are common in primary NPC specimens and that this correlated with the expression of LMP1. In addition, FGFR1 inhibitors suppress cell proliferation and anchorage-independent growth of NPC cells. Our current findings demonstrate that LMP1-mediated FGFR1 activation contributes to aerobic glycolysis and transformation of epithelial cells, thereby implicating FGF2/FGFR1 signalling activation in the EBV-driven pathogenesis of NPC.


Subject(s)
Cell Transformation, Viral , Epithelial Cells/metabolism , Glycolysis , Herpesvirus 4, Human/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharynx/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Viral Matrix Proteins/metabolism , Carcinoma , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Viral/drug effects , Epithelial Cells/pathology , Epithelial Cells/virology , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic , Glycolysis/drug effects , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Nasopharynx/pathology , Nasopharynx/virology , Neoplasm Invasiveness , Phosphorylation , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Time Factors , Viral Matrix Proteins/genetics
7.
J Pathol ; 230(3): 336-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23592276

ABSTRACT

The association of Epstein-Barr virus (EBV) infection with the development of nasopharyngeal carcinoma (NPC) is well established. Latent membrane protein 1 (LMP1), the major oncogene encoded by EBV, is believed to play a crucial role in NPC pathogenesis by virtue of its ability to constitutively activate multiple cell signalling pathways. The LKB1-AMPK pathway is a master regulator of cellular metabolism that, via modulation of energy metabolism, has tumour suppressor activity. In this study we identify a novel ability of LMP1 to inhibit the LKB1-AMPK pathway through phosphorylation of LKB1 at serine 428 with subsequent suppression of the phosphorylation of AMPK and its substrates, ACC and Raptor. We show that MEK/ERK-MAPK signalling, activated by the CTAR1 domain of LMP1, is responsible for LKB1-AMPK inactivation. In addition, reactivation of AMPK signalling by AMPK activator, AICAR, abolished LMP1-induced cellular transformation (proliferation and anchorage-independent growth) in nasopharyngeal epithelial cells. Immunohistochemical staining revealed that a low level of phosphorylated AMPK is common in primary NPC specimens, and that this correlated significantly with the expression of LMP1. AICAR treatment inhibited the proliferation and anchorage-independent growth of NPC cells as well as potentiating the cytotoxic effect of the chemotherapeutic drug 5-fluorouracil. The current findings demonstrate that LMP1-mediated AMPK inactivation contributes to the proliferation and transformation of epithelial cells, thereby implicating the LKB1-AMPK pathway in the EBV-driven pathogenesis of NPC. Our findings also suggest that AMPK activators could be used to enhance the efficacy of conventional chemotherapeutic agents in the treatment of local and metastatic NPC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Herpesvirus 4, Human/physiology , MAP Kinase Signaling System/drug effects , Nasopharyngeal Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Viral Matrix Proteins/metabolism , AMP-Activated Protein Kinase Kinases , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Carcinoma , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Fluorouracil/pharmacology , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/etiology , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/virology , Nasopharynx/metabolism , Nasopharynx/pathology , Phosphorylation , Protein Structure, Tertiary , Ribonucleotides/pharmacology , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 104(41): 16164-9, 2007 Oct 09.
Article in English | MEDLINE | ID: mdl-17911266

ABSTRACT

Epstein-Barr virus (EBV) was the first human virus found to encode microRNAs (miRNAs), but the function of these miRNAs has been obscure. Nasopharyngeal carcinoma (NPC) is associated with EBV infection, and the EBV-encoded LMP1 is believed to be a key factor in NPC development. However, detection of LMP1 protein in NPC is variable. Here, we report that EBV-encoded BART miRNAs target the 3' UTR of the LMP1 gene and negatively regulate LMP1 protein expression. These miRNAs also modulate LMP1-induced NF-kappaB signaling and alleviate the cisplatin sensitivity of LMP1-expressing NPC cells. Consistent with a previous study on the NPC C666-1 cell line and C15 xenograft, we found abundant expression of BART miRNAs in NPC tissues. Furthermore, DNA sequencing revealed that the 3' UTR of LMP1 is highly conserved in NPC-derived EBV isolates. The data provide insight into the discrepancy between LMP1 transcript and protein detection in NPC and highlight the role of the EBV miRNAs in regulating LMP1 downstream signaling to promote cancer development.


Subject(s)
Herpesvirus 4, Human/genetics , MicroRNAs/genetics , Viral Matrix Proteins/genetics , 3' Untranslated Regions , Antineoplastic Agents/pharmacology , Base Sequence , Cell Line, Tumor , Cisplatin/pharmacology , Gene Expression , Genes, Viral , Humans , Molecular Sequence Data , NF-kappa B/metabolism , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/virology , RNA, Neoplasm/genetics , RNA, Viral/genetics , Signal Transduction
9.
Neoplasia ; 8(3): 173-80, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16611410

ABSTRACT

Epstein-Barr virus (EBV) latent infection is a critical event in nasopharyngeal carcinoma (NPC) tumorigenesis. EBV-encoded genes have been shown to be involved in immune evasion and in the regulation of various cellular signaling cascades. To elucidate the roles of EBV in NPC development, stable infection of EBV in nasopharyngeal epithelial cell lines was established. Similar to primary tumors of NPC, these infected cells exhibited a type II EBV latency expression pattern. In this study, multiple cellular signaling pathways in EBV-infected cells were investigated. We first demonstrated that in vitro EBV infection resulted in the activation of STAT3 and NFkappaB signal cascades in nasopharyngeal epithelial cells. Increased expression of their downstream targets (c-Myc, Bcl-xL, IL-6, LIF, SOCS-1, SOCS-3, VEGF, and COX-2) was also observed. Moreover, EBV latent infection induced the suppression of p38-MAPK activities, but did not activate PKR cascade. Our findings suggest that EBV latent infection is able to manipulate multiple cellular signal cascades to protect infected cells from immunologic attack and to facilitate cancer development.


Subject(s)
Carcinoma/pathology , Cell Transformation, Viral , Epithelial Cells/virology , Epstein-Barr Virus Infections/physiopathology , Herpesvirus 4, Human/physiology , Nasopharyngeal Neoplasms/pathology , Nasopharynx/cytology , Signal Transduction , Carcinoma/immunology , Carcinoma/virology , Cell Line/physiology , Cell Line/virology , Cell Line, Tumor , Cytokines/biosynthesis , Cytokines/genetics , Epithelial Cells/physiology , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Humans , Inflammation , MAP Kinase Signaling System , NF-kappa B/physiology , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/virology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phosphorylation , Protein Processing, Post-Translational , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/physiology , Virus Latency , eIF-2 Kinase/physiology
10.
Int J Cancer ; 109(6): 919-25, 2004 May 10.
Article in English | MEDLINE | ID: mdl-15027126

ABSTRACT

Epstein-Barr virus (EBV) is closely associated with nasopharyngeal carcinoma (NPC), a common cancer in Hong Kong. The EBV-encoded LMP1 protein is believed to play an important role in cell transformation. We have previously identified a prevalent LMP1 variant (2117-LMP1) that is expressed in 86% of primary NPC in Hong Kong. In this study, the biologic phenotypes induced by 2117-LMP1 were compared with those of the prototypic B95.8-LMP1 in an immortalized nasopharyngeal epithelial cell line, NP69. The 2117-LMP1 could induce cell proliferation and resistance to apoptosis induced by growth factor deprivation. Expression of 2117-LMP1 also suppressed expression of p16, p21 and Bax but induced expression of CDK2 and A20. Compared with B95.8-LMP1, 2117-LMP1 could induce a higher migration ability in NP69 cells but was less efficient in inducing morphologic changes, anchorage-independent growth and cell invasion. Relatively weaker ability of 2117-LMP1 than B95.8-LMP1 in upregulation of vimentin, VEGF and MMP9 as well as in downregulation of E-cadherin was observed. 2117-LMP1 could activate higher level of NF-kappaB activity in HEK 293 cells than B95.8-LMP1. The present study supports a role of 2117-LMP1 in NPC development by enhancing cell proliferation, cell death inhibition and migration in premalignant nasopharyngeal epithelial cells. Furthermore, our study reveals significant functional differences between 2117-LMP1 and the prototypic B95.8-LMP1. Our results provide insights into the pathologic significance of this prevalent LMP1 variant, 2117-LMP1, in the development of NPC in the Hong Kong population.


Subject(s)
Herpesvirus 4, Human/physiology , Nasopharyngeal Neoplasms/virology , Proto-Oncogene Proteins c-bcl-2 , Viral Matrix Proteins/physiology , CDC2-CDC28 Kinases/metabolism , Cadherins/metabolism , Cell Division , Cell Movement , Cell Survival , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA-Binding Proteins , Hong Kong , Humans , Intracellular Signaling Peptides and Proteins , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Nuclear Proteins , Phenotype , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor alpha-Induced Protein 3 , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , bcl-2-Associated X Protein
11.
Lab Invest ; 83(5): 697-709, 2003 May.
Article in English | MEDLINE | ID: mdl-12746479

ABSTRACT

Undifferentiated nasopharyngeal carcinoma (NPC) is closely associated with EBV infection, and the EBV-encoded latent membrane protein 1 (LMP1) is frequently detected in NPC. However, little is known about the pathologic roles of LMP1 in this disease. Recently, we reported the morphologic transformation and increased expression of the LAMC2 and ITGalpha6 genes in LMP1-expressing NPC cell lines. In this study, we further examine the effects of LMP1 in an immortalized nasopharyngeal epithelial cell line called NP69. This cell line was established from primary nonmalignant nasopharyngeal epithelial cells and may represent a model of premalignant nasopharyngeal epithelial cells. LMP1 induced many phenotypic changes in NP69 cells. These include morphologic transformation, increased cell proliferation, anchorage-independent growth, resistance to serum free-induced cell death, and enhanced cell migration and invasion. In addition, expression array analysis identified 28 genes that demonstrated a more than 2-fold difference in expression of NP69 cells expressing LMP1 when compared with a vector control. Two of the up-regulated genes (VEGF and vimentin) identified have been previously reported as LMP1 targets. The majority of the identified genes are associated with cell growth, differentiation, cell shape, and invasion. The present findings support the proposed roles of LMP1 in promoting cell transformation, migration, and invasion in premalignant nasopharyngeal epithelial cells. The present study also indicates the activation of the Ras/MAPK pathway in LMP1-expressing cells, which may be involved in mediating some of the transforming effects of LMP1 observed in nasopharyngeal epithelial cells.


Subject(s)
Nasopharyngeal Neoplasms/etiology , Nasopharynx/pathology , Viral Matrix Proteins/physiology , Blotting, Western , Cell Differentiation , Cell Division , Cell Line , Cell Movement , Cell Survival , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Profiling , Humans , Mitogen-Activated Protein Kinases/metabolism , Nasopharynx/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...