Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters











Publication year range
1.
Oncogene ; 29(30): 4287-96, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20498633

ABSTRACT

Epstein-Barr virus (EBV) is an oncogenic gamma-herpes virus associated with malignancies that develop in both lymphoid and epithelial cells including nasopharyngeal carcinoma (NPC). The EBV protein, latent membrane protein 2A (LMP2A), is expressed in NPC and can modulate epithelial proliferation, transformation and differentiation, and as such may promote malignancy. A key regulator of epithelial-cell differentiation is the transcription factor p63, a member of the p53 family. This study examines the potential contribution of p63 to LMP2A-mediated inhibition of epithelial-cell differentiation. Stable expression of LMP2A increased the protein level and stability of the DeltaNp63alpha isoform and in two epithelial cell lines, LMP2A interacted with DeltaNp63alpha under stable- and transient-expression systems. LMP2A and DeltaNp63alpha were localized to the cytoplasm and nuclear membrane and co-immunoprecipitated in the same fractions. Following induction of epithelial-cell differentiation by calcium, expression of differentiation markers was impaired in both DeltaNp63alpha- and LMP2A-expressing cells. Induction of p63alpha, association of p63alpha with LMP2A and impairment of differentiation required the PY and immunoreceptor tyrosine-based activation motif (ITAM) signaling motif of LMP2A. By associating with and being regulated by LMP2A, DeltaNp63alpha may function as a unique regulator of LMP2A effects on epithelial differentiation and contribute to EBV-associated epithelial cancers.


Subject(s)
Epithelial Cells/pathology , Trans-Activators/genetics , Tumor Suppressor Proteins/genetics , Viral Matrix Proteins/physiology , Calcium/physiology , Cell Differentiation , Cell Line , Humans , Trans-Activators/analysis , Trans-Activators/chemistry , Transcription Factors , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/chemistry , Viral Matrix Proteins/analysis
2.
Oncogene ; 25(2): 288-97, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16247482

ABSTRACT

Transgenic mice expressing Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) under the control of an immunoglobulin heavy-chain promoter and enhancer develop lymphoma at a threefold higher incidence than LMP1-negative mice. In vitro, LMP1 activates numerous signaling pathways including p38, c-Jun N terminal kinase (JNK), phosphatidylinositol 3 kinase (PI3K)/Akt, and NF-kappaB through interactions with tumor necrosis receptor-associated factors (TRAFs). These pathways are frequently activated in EBV-associated malignancies, although their activation cannot be definitively linked to LMP1 expression in vivo. In this study, interactions between LMP1 and TRAFs and the activation of PI3K/Akt, JNK, p38, and NF-kappaB were examined in LMP1 transgenic mice. LMP1 co-immunoprecipitated with TRAFs 1, 2, and 3. Akt, JNK, and p38 were activated in LMP1-positive and -negative splenocytes as well as LMP1-positive and -negative lymphomas. Multiple forms of NF-kappaB were activated in healthy splenocytes from LMP1 transgenic mice, in contrast to healthy splenocytes from LMP1-negative mice. However, in both LMP1-positive and -negative lymphomas, only the oncogenic NF-kappaB c-Rel, was specifically activated. Similarly to EBV-associated malignancies, p53 protein was detected at high levels in the transgenic lymphomas, although mutations were not detected in the p53 gene. These data indicate that NF-kappaB is activated in LMP1-positive healthy splenocytes; however, NF-kappaB c-Rel is specifically activated in both the transgenic lymphomas and in the rare lymphomas that develop in negative mice. The LMP1-mediated activation of NF-kappaB may contribute to the specific activation of c-Rel and lead to the increased development of lymphoma in the LMP1 transgenic mice.


Subject(s)
Genes, rel/physiology , Lymphoma , NF-kappa B/metabolism , Signal Transduction , Viral Matrix Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Class I Phosphatidylinositol 3-Kinases , Cytoskeletal Proteins , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases/metabolism , LIM Domain Proteins , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/metabolism , Spleen/metabolism , TNF Receptor-Associated Factor 1/metabolism , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 3/metabolism , Transcription Factor RelB/metabolism , Tumor Suppressor Protein p53/metabolism , Viral Matrix Proteins/genetics
3.
J Virol ; 77(22): 12276-84, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14581564

ABSTRACT

The Epstein-Barr virus (EBV) latent membrane protein 2A (LMP2A) functions to maintain latency in EBV-infected B lymphocytes. Although LMP2A is nonessential for the immortalization of B lymphocytes by EBV, its expression in B lymphocytes prevents viral reactivation by blocking B-cell receptor activation and signaling. LMP2A also provides an antiapoptotic signal in transgenic mice that express LMP2A in B lymphocytes. LMP2A activates phosphatidylinositol 3-kinase (PI3K) and the serine/threonine kinase Akt in lymphocytes and epithelial cells. Here we show that EBV LMP2A activates the PI3K and beta-catenin signaling pathways in telomerase-immortalized human foreskin keratinocytes (HFK). LMP2A activated Akt in a PI3K-dependent manner, and the downstream Akt targets glycogen synthase kinase 3beta (GSK3beta) and the Forkhead transcription factor FKHR were phosphorylated and inactivated in LMP2A-expressing HFK cells. GSK3beta is a negative regulator of the Wnt signaling pathway, and inactivation of GSK3beta by LMP2A signaling led to stabilization of beta-catenin, the central oncoprotein of Wnt signaling. In LMP2A-expressing cells, beta-catenin accumulated in the cytoplasm and translocated into the nucleus via a two-step mechanism. The cytoplasmic accumulation of beta-catenin downstream of LMP2A was independent of PI3K signaling, whereas its nuclear translocation was dependent on PI3K signaling. In the nucleus, beta-catenin activated a reporter responsive to T-cell factor, and this activation was augmented by LMP2A coexpression. The Wnt pathway is inappropriately activated in 90% of colon cancers and is dysregulated in several other cancers, and these data suggest that activation of this pathway by LMP2A may contribute to the generation of EBV-associated cancers.


Subject(s)
Cytoskeletal Proteins/metabolism , Protein Serine-Threonine Kinases , Signal Transduction/physiology , Trans-Activators/metabolism , Viral Matrix Proteins/physiology , Active Transport, Cell Nucleus , Cells, Cultured , DNA-Binding Proteins/physiology , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Herpesvirus 4, Human/pathogenicity , Humans , Keratinocytes/metabolism , Lymphoid Enhancer-Binding Factor 1 , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Telomerase/genetics , Transcription Factors/physiology , beta Catenin
4.
Oral Dis ; 8 Suppl 2: 161-8, 2002.
Article in English | MEDLINE | ID: mdl-12164651

ABSTRACT

Opportunistic DNA viruses, particularly members of the herpesvirus family, are frequently the aetiological agents of HIV-associated oral lesions. Oral lesions common to the early phase of the AIDS epidemic, including Kaposi's sarcoma (KS), oral aphthous ulceration, AIDS-associated oral lymphoma, and oral hairy leukoplakia (OHL), have been tested for the prevalence of Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV). While EBV DNA is detected by PCR in all of these lesions, abundant viral replication can only be detected in OHL. In OHL, a novel state of EBV infection has been discovered with concurrent expression of replicative and transforming proteins, with all of these proteins contributing to the development of the lesion. Activation of signalling pathways and up-regulation of the viral receptor, proliferative and antiapoptotic genes by these proteins induce several of the histological features common to OHL, such as acanthosis and hyperproliferation. In contrast to other permissive herpesvirus infections, expression of EBV transforming proteins within the permissively infected OHL tissue enables epithelial cell survival and may enhance viral replication. Detection of KSHV in these HIV-infected individuals has been localized only to their saliva. Replicative and latent KSHV gene products have been detected in association with the development of oral KS lesions. EBV, but not human cytomegalovirus (HCMV), has been detected by PCR in minor salivary gland biopsies of HIV-associated salivary gland disease. Human papillomaviruses (HPV) are associated with oral warts in HIV-positive individuals; a diagnosis that appears to be increasing in frequency in the era of highly active antiretroviral therapy. To date, there appears to be little increase in the incidence of HPV-associated oral cancer. The mechanisms of interaction between HIV and HPV are not fully understood. Expression of viral gene products is clearly important and necessary for the development of multiple AIDS-associated oral lesions.


Subject(s)
AIDS-Related Opportunistic Infections/virology , Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Mouth Diseases/virology , Papillomaviridae/physiology , Apoptosis/physiology , Basic-Leucine Zipper Transcription Factors , Carrier Proteins/analysis , Cell Division/physiology , Cell Survival , Cytomegalovirus/physiology , Cytomegalovirus Infections/complications , Epithelial Cells/virology , Epstein-Barr Virus Infections/complications , HIV Infections/complications , Humans , Leukoplakia, Hairy/virology , Lymphoma, AIDS-Related/virology , Mouth Mucosa/virology , Mouth Neoplasms/virology , Oncogene Proteins, Viral/analysis , Papillomavirus Infections/complications , Receptors, Virus/physiology , Repressor Proteins , Saliva/virology , Salivary Gland Diseases/virology , Sarcoma, Kaposi/virology , Signal Transduction/physiology , Stomatitis, Aphthous/virology , Up-Regulation/physiology , Viral Proteins/analysis , Virus Replication , Warts/virology
5.
Cancer Res ; 61(21): 7875-7, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691806

ABSTRACT

Nasopharyngeal carcinoma (NPC) is universally associated with EBV infection. We have shown that the phosphonated nucleoside analog, (S)-1-[3-hydroxy-2-(phosphonylmethoxy)-propyl]cytosine (HPMPC) strongly inhibits growth of NPC xenografts in nude mice by causing apoptosis (J. Neyts et al., Cancer Res., 58, 384-388, 1998). We, therefore, tested two additional members of this drug family that have different degrees of antiviral activity, 9-[2-(phosphonomethoxy)ethyl]adenine (PMEA) and 9-2-(R)-(phosphonomethoxy)propyladenine (PMPA). Intratumoral injection of PMEA (75 microl of 2% solution) in C15 NPC xenografts, which are latently infected with EBV, slowed tumor growth moderately, whereas PMPA (75 microl of 2% solution) slowed tumor growth only marginally. Compared with the previous results showing complete regression of tumor, PMEA had less antitumoral effect than HPMPC, and PMPA had the least. After 4 weeks of preventive treatment, tumors formed in 12.5, 50, and 100% of mice treated with HPMPC, PMEA, and PMPA, respectively, in contrast to the development of tumors in all of the PBS-treated control mice. We also investigated the effect of each drug on the EBV-positive epithelial cell line NPC-KT in vitro. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay showed inhibition of growth of NPC-KT cells by HPMPC and PMEA, but not by PMPA, which correlates with the results observed in tumor xenografts. Growth inhibition was attributable to induction of apoptosis in NPC-KT cells as indicated by a DNA fragmentation assay. Cleavage of poly(ADP-ribose) polymerase after treatment of NPC-KT cells with HPMPC was observed, which suggested that the apoptosis may be mediated by caspase(s). The apoptotic effects of the drugs are independent of any effects on EBV DNA polymerase, which is not expressed in these latently infected NPCs. These results suggest that HPMPC as well as PMEA could provide an adjunctive treatment for NPC.


Subject(s)
Adenine/analogs & derivatives , Adenine/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma/drug therapy , Cytosine/analogs & derivatives , Nasopharyngeal Neoplasms/drug therapy , Organophosphonates , Organophosphorus Compounds/pharmacology , Animals , Apoptosis/drug effects , Carcinoma/pathology , Carcinoma/prevention & control , Cell Division/drug effects , Cidofovir , Cytosine/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/virology , Growth Inhibitors/pharmacology , Herpesvirus 4, Human , Humans , Mice , Mice, Nude , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/prevention & control , Tenofovir , Xenograft Model Antitumor Assays
6.
J Virol ; 75(1): 384-95, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11119607

ABSTRACT

The Epstein-Barr virus (EBV) BamHI A mRNAs were originally identified in cDNA libraries from nasopharyngeal carcinoma, where they are expressed at high levels. The RNAs are differentially spliced to form several open reading frames and also contain the BARF0 open reading frame at the 3' end. One cDNA, RK-BARF0, included a potential endoplasmic reticulum-targeting signal peptide sequence. The RK-BARF0 protein is shown here to interact with the Notch4 ligand binding domain, using yeast two-hybrid screening, coimmunoprecipitation, and confocal microscopy. This interaction induces translocation of a portion of the full-length unprocessed Notch4 to the nucleus by using the Notch nuclear localization signal. These effects of RK-BARF0 on Notch intracellular location indicate that EBV possibly modulates Notch signaling. Unprocessed Notch4 was also detected in immunoprecipitated complexes from EBV-infected cells by using a rabbit antiserum raised against a BARF0-specific peptide. This finding provides additional evidence for expression of RK-BARF0 and its interaction with Notch during EBV infection. In EBV-infected, EBNA2-negative cells, RK-BARF0 induced the expression of EBV latent membrane protein 1 (LMP1), and this induction was dependent on the RK-BARF0/Notch interaction domain. The activation of LMP1 expression by RK-BARF0 may be responsible for expression of LMP1 in EBV latent infections in the absence of EBNA2.


Subject(s)
Herpesvirus 4, Human/physiology , Membrane Proteins/physiology , Viral Proteins/physiology , Animals , Biological Transport , Cell Nucleus/metabolism , Endoplasmic Reticulum/metabolism , Humans , Open Reading Frames , Rabbits , Receptors, Notch , Tumor Cells, Cultured , Viral Matrix Proteins/biosynthesis , Viral Proteins/chemistry
7.
Virology ; 291(2): 208-14, 2001 Dec 20.
Article in English | MEDLINE | ID: mdl-11878890

ABSTRACT

LMP2A deletion and point mutants, with mutations in phosphotyrosine-containing protein-protein interaction motifs, were transiently expressed in 293 cells and their phosphorylation was examined in immune complex kinase assays as well as in vivo. In vitro LMP2A phosphorylation depended on tyrosine 112. In vivo, mutations of single tyrosines did not eliminate LMP2 phosphorylation, although mutation of the LMP2A ITAM decreased LMP2A phosphorylation. The relationship between LMP2A in vitro phosphorylation and that induced by cell-extracellular matrix (ECM) interactions was also investigated. While LMP2A was phosphorylated to higher levels in whole-cell extracts of stimulated cells, a difference in in vitro kinase assays with extracts from stimulated and unstimulated cells was not detected, indicating that the ECM-mediated regulation of LMP2A phosphorylation is lost in vitro. In the presence of LMP2A, several cellular proteins with molecular weights between 70 and 80 kDa were phosphorylated on tyrosine. This increase in cellular protein phosphorylation depended on the LMP2A ITAM motif and suggests that the ITAM may participate in signal-transduction events in epithelial cells.


Subject(s)
Herpesvirus 4, Human/metabolism , Viral Matrix Proteins/metabolism , Binding Sites , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Humans , Mutagenesis , Phosphorylation , Tyrosine/genetics , Tyrosine/metabolism , Viral Matrix Proteins/genetics , src-Family Kinases/metabolism
8.
J Virol ; 74(22): 10681-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11044112

ABSTRACT

The Epstein-Barr virus LMP2A protein was expressed in a human keratinocyte cell line, HaCaT, and effects on epithelial cell growth were detected in organotypic raft cultures and in vivo in nude mice. Raft cultures derived from LMP2A-expressing cells were hyperproliferative, and epithelial differentiation was inhibited. The LMP2A-expressing HaCaT cells were able to grow anchorage independently and formed colonies in soft agar. HaCaT cells expressing LMP2A were highly tumorigenic and formed aggressive tumors in nude mice. The LMP2A tumors were poorly differentiated and highly proliferative, in contrast to occasional tumors that arose from parental HaCaT cells and vector control cells, which grew slowly and remained highly differentiated. Animals injected with LMP2A-expressing cells developed frequent metastases, which predominantly involved lymphoid organs. Involucrin, a marker of epithelial differentiation, and E-cadherin, involved in the maintenance of intercellular contact, were downregulated in LMP2A tumors. Whereas activation of the mitogen-activated protein kinase pathway was not observed, phosphatidylinositol-3-kinase (PI3-kinase)-dependent activation of the serine-threonine kinase Akt was detected in LMP2A-expressing cells and LMP2A tumors. Inhibition of this pathway blocked growth in soft agar. These data indicate that LMP2A greatly affects cell growth and differentiation pathways in epithelial cells, in part through activation of the PI3-kinase-Akt pathway.


Subject(s)
Cell Differentiation , Cell Transformation, Neoplastic , Cell Transformation, Viral , Herpesvirus 4, Human/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Viral Matrix Proteins/physiology , Animals , Cell Division , Cell Line , Cell Transplantation , Collagen , Epithelial Cells/virology , Fibroblasts , Humans , Keratinocytes/metabolism , MAP Kinase Signaling System , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Organ Culture Techniques , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt , Retroviridae/genetics , Signal Transduction , Viral Matrix Proteins/metabolism
9.
J Virol ; 74(16): 7610-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10906215

ABSTRACT

Human herpesviruses are characterized by distinct states of infection. Typically in permissive herpesvirus infection, abundant virus production results in cell lysis. In latent transforming Epstein-Barr virus (EBV) infection, viral proteins that induce cell growth are expressed. The immunodeficiency-associated hairy leukoplakia (HLP) lesion is the only pathologic manifestation of permissive EBV infection; however, within HLP, viral proteins characteristic of latent infection have also been detected. In this study, we further analyzed expression of EBV latent genes and investigated their contribution to the unique histologic phenotype of HLP. Coexpression of lytic and transforming viral proteins was detected simultaneously within individual HLP keratinocytes. LMP1 has now been shown to be uniformly expressed in the affected tissue, and it is associated and colocalizes with tumor necrosis factor receptor-associated factor (TRAF) signaling molecules. Effects induced by activated TRAF signaling that were detected in HLP included activation of NF-kappaB and c-Jun terminal kinase 1 (JNK1) and upregulated expression of epidermal growth factor receptor (EGFR), CD40, A20, and TRAFs. This study identifies a novel state of EBV infection with concurrent expression of replicative and transforming proteins. It is probable that both replicative and latent proteins contribute to HLP development and induce many of the histologic features of HLP, such as acanthosis and hyperproliferation. In contrast to other permissive herpesvirus infections, expression of EBV transforming proteins within the permissively infected HLP tissue enables epithelial cell survival and may enhance viral replication.


Subject(s)
Cell Transformation, Viral , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/physiology , Leukoplakia, Hairy/pathology , Leukoplakia, Hairy/virology , AIDS-Related Opportunistic Infections/virology , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation, Viral , Humans , Reverse Transcriptase Polymerase Chain Reaction , Tongue/metabolism , Tongue/virology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Activation , Virus Latency/genetics , Virus Replication
10.
Int J Cancer ; 86(2): 244-50, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10738252

ABSTRACT

Nasopharyngeal carcinoma (NPC) is endemic in parts of southern China and is etiologically associated with Epstein-Barr virus (EBV) infection as well as other dietary and environmental factors. Loss of heterozygosity (LOH) on chromosome 3p has been described in NPC from the endemic region. In this study, tumors originating from both the NPC nonendemic area of northern China and the endemic area in southern China were analyzed for LOH at 8 microsatellite markers on chromosome 3. Allele loss was detected at D3S1300 in 3p14.2 in more than 50% of tumors from both the endemic and nonendemic areas, suggesting that LOH at this locus probably does not account for the endemicity of NPC in southern China. The 3p14.2 region encompasses FHIT, a candidate tumor suppressor gene previously shown to be rearranged in several NPC cell lines. In this study, analysis of FHIT gene structure and transcription in primary tumors did not support a role for FHIT in NPC. However, the high frequency of allele loss at 3p14.2 in NPC from endemic and nonendemic regions supports the possibility that an important tumor suppressor gene other than FHIT complements EBV transformation and resides in this region.


Subject(s)
Acid Anhydride Hydrolases , Chromosomes, Human, Pair 3 , Endemic Diseases , Nasopharyngeal Neoplasms/epidemiology , Nasopharyngeal Neoplasms/genetics , Neoplasm Proteins , Alleles , China , Epstein-Barr Virus Infections , Gene Deletion , Genes, Tumor Suppressor , Humans , Loss of Heterozygosity , Microsatellite Repeats , Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL