Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 67(24): 4249-56, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20593215

ABSTRACT

Epstein-Barr virus (EBV) is a human gamma herpes virus that infects B cells and induces their transformation into immortalized lymphoblasts that can grow as cell lines (LCLs) in vitro. EBNA-3 is a member of the EBNA-3-protein family that can regulate transcription of cellular and viral genes. The identification of EBNA-3 cellular partners and a study of its influence on cellular pathways are important for understanding the transforming action of the virus. In this work, we have identified the vitamin D receptor (VDR) protein as a binding partner of EBNA-3. We found that EBNA3 blocks the activation of VDR-dependent genes and protects LCLs against vitamin-D3-induced growth arrest and/or apoptosis. The presented data shed some light on the anti-apoptotic EBV program and the role of the EBNA-3-VDR interaction in the viral strategy.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation , Herpesvirus 4, Human/metabolism , Receptors, Calcitriol/metabolism , Animals , Calcifediol/metabolism , Cell Line , Cell Line, Tumor , Epstein-Barr Virus Nuclear Antigens/genetics , Herpesvirus 4, Human/genetics , Humans , Lymphocytes/cytology , Lymphocytes/physiology , Mass Spectrometry , Receptors, Calcitriol/genetics
2.
Cell Signal ; 19(6): 1279-89, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17303383

ABSTRACT

Human Sin1 (SAPK-interacting protein 1) is a member of a conserved family of orthologous proteins that have an essential role in signal transduction in yeast and Dictyostelium. This study demonstrates that most Sin1 orthologues contain both a Raf-like Ras-binding domain (RBD) and a pleckstrin homology (PH) domain. These domains are functional in the human Sin1 protein, with the PH domain involved in lipid and membrane binding by Sin1, and the RBD able to bind activated H-and K-Ras. Sin1 and Ras co-immunoprecipitated and co-localised, showing that these proteins associate with each other in vivo. Overexpression of Sin1 inhibited the activation of ERK, Akt and JNK signalling pathways by Ras. In contrast, siRNA knockdown of endogenous Sin1 protein expression in HEK293 cells enhanced the activation of ERK1/2 by Ras. These data suggest that Sin1 is a mammalian Ras-inhibitor.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction , ras Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Cricetinae , Enzyme Activation , Humans , Lipids , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Protein Transport
3.
Breast Cancer Res Treat ; 104(3): 257-66, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17063265

ABSTRACT

BACKGROUND AND PURPOSE: Assays to determine the pathogenicity of unclassified sequence variants in disease-associated genes include the analysis of lymphoblastoid cell lines (LCLs). We assessed the ability of several assays of LCLs to distinguish carriers of germline BRCA1 and BRCA2 gene mutations from mutation-negative controls to determine their utility for use in a diagnostic setting. MATERIALS AND METHODS: Post-ionising radiation cell viability and micronucleus formation, and telomere length were assayed in LCLs carrying BRCA1 or BRCA2 mutations, and in unaffected mutation-negative controls. RESULTS: Post-irradiation cell viability and micronucleus induction assays of LCLs from individuals carrying pathogenic BRCA1 mutations, unclassified BRCA1 sequence variants or wildtype BRCA1 sequence showed significant phenotypic heterogeneity within each group. Responses were not consistent with predicted functional consequences of known pathogenic or normal sequences. Telomere length was also highly heterogeneous within groups of LCLs carrying pathogenic BRCA1 or BRCA2 mutations, and normal BRCA1 sequences, and was not predictive of mutation status. CONCLUSION: Given the significant degree of phenotypic heterogeneity of LCLs after gamma-irradiation, and the lack of association with BRCA1 or BRCA2 mutation status, we conclude that the assays evaluated in this study should not be used as a means of differentiating pathogenic and non-pathogenic sequence variants for clinical application. We suggest that a range of normal controls must be included in any functional assays of LCLs to ensure that any observed differences between samples reflect the genotype under investigation rather than generic inter-individual variation.


Subject(s)
Genes, BRCA1 , Genes, BRCA2 , Lymphocytes/radiation effects , Mutation , Cell Line, Tumor , Cell Survival , DNA Mutational Analysis , Dose-Response Relationship, Radiation , Genetic Variation , Genotype , Humans , Micronucleus Tests , Phenotype , Radiation, Ionizing , Telomere/metabolism
4.
J Gen Virol ; 87(Pt 10): 2879-2884, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16963745

ABSTRACT

The Epstein-Barr nuclear antigen 3A (EBNA3A) is one of only six viral proteins essential for Epstein-Barr virus-induced transformation of primary human B cells in vitro. Viral proteins such as EBNA3A are able to interact with cellular proteins, manipulating various biochemical and signalling pathways to initiate and maintain the transformed state of infected cells. EBNA3A has been reported to have one nuclear-localization signal and is targeted to the nucleus during transformation, where it associates with components of the nuclear matrix. By using enhanced green fluorescent protein-tagged deletion mutants of EBNA3A in combination with site-directed mutagenesis, an additional five functional nuclear-localization signals have been identified in the EBNA3A protein. Two of these (aa 63-66 and 375-381) were computer-predicted, whilst the remaining three (aa 394-398, 573-578 and 598-603) were defined functionally in this study.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/metabolism , Nuclear Localization Signals/metabolism , HeLa Cells , Humans , Protein Transport
5.
Curr Biol ; 16(18): 1865-70, 2006 Sep 19.
Article in English | MEDLINE | ID: mdl-16919458

ABSTRACT

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that participates in at least two distinct multiprotein complexes, mTORC1 and mTORC2 . These complexes play important roles in the regulation of cell growth, proliferation, survival, and metabolism. mTORC2 is a hydrophobic motif kinase for the cell-survival protein Akt/PKB and, here, we identify mSin1 as a component of mTORC2 but not mTORC1. mSin1 is necessary for the assembly of mTORC2 and for its capacity to phosphorylate Akt/PKB. Alternative splicing generates at least five isoforms of the mSin1 protein , three of which assemble into mTORC2 to generate three distinct mTORC2s. Even though all mTORC2s can phosphorylate Akt/PKB in vitro, insulin regulates the activity of only two of them. Thus, we propose that cells contain several mTORC2 flavors that may phosphorylate Akt/PKB in response to different signals.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/physiology , Cell Line, Tumor , Humans , Insulin/metabolism , Phosphorylation , Protein Binding/drug effects , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Kinases/chemistry , Proto-Oncogene Proteins c-akt/chemistry , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
6.
J Gen Virol ; 87(Pt 4): 789-793, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16528026

ABSTRACT

The Epstein-Barr virus nuclear antigen (EBNA) 3B is a hydrophilic, proline-rich, charged protein that is thought to be involved in transcriptional regulation and is targeted exclusively to the cell nucleus, where it localizes to discrete subnuclear granules. Co-localization studies utilizing a fusion protein between enhanced green fluorescent protein (EGFP) and EBNA3B with FLAG-tagged EBNA3A and EBNA3C proteins demonstrated that EBNA3B co-localized with both EBNA3A and EBNA3C in the nuclei of cells when overexpressed. Computer analyses identified four potential nuclear-localization signals (NLSs) in the EBNA3B amino acid sequence. By utilizing fusion proteins with EGFP, deletion constructs of EBNA3B and site-directed mutagenesis, three of the four NLSs (aa 160-166, 430-434 and 867-873) were shown to be functional in truncated forms of EBNA3B, whilst an additional NLS (aa 243-246) was identified within the N-terminal region of EBNA3B. Only two of the NLSs were found to be functional in the context of the full-length EBNA3B protein.


Subject(s)
Cell Nucleus/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Nuclear Localization Signals/chemistry , Amino Acid Sequence , Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion
7.
Cell Signal ; 17(6): 761-7, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15722200

ABSTRACT

The orthologous proteins of the stress-activated protein kinase-interacting 1 (Sin1) family have been implicated in several different signal transduction pathways. In this study, we have investigated the function of the full-length human Sin1 protein and a C-terminally truncated isoform, Sin1alpha, which is produced by alternative splicing. Immunoblot analysis using an anti-Sin1 polyclonal antibody showed that full-length Sin1 and several smaller isoforms are widely expressed. Sin1 was demonstrated to bind to c-Jun N-terminal kinase (JNK) in vitro and in vivo, while no interaction with p38- or ERK1/2-family MAPKs was observed. The Sin1alpha isoform could also form a complex with JNK in vivo. Despite localizing in distinct compartments within the cell, both Sin1 and Sin1alpha co-localized with JNK, suggesting that the Sin1 proteins could recruit JNK. Over-expression of full-length Sin1 inhibited the activation of JNK by UV-C in DG75 cells, as well as basal JNK-activity in HEK293 cells. These data suggest that the human Sin1 proteins may act as scaffold molecules in the regulation of signaling by JNK.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Carrier Proteins/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/analysis , Carrier Proteins/genetics , Cell Line , Humans , JNK Mitogen-Activated Protein Kinases/analysis , Protein Isoforms/metabolism
8.
Gene ; 339: 17-23, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15363842

ABSTRACT

Limited but significant sequence similarity has been observed between an uncharacterized human protein, SIN1, and the S. pombe SIN1, Dictyostelium RIP3 and S. cerevisiae AVO1 proteins. The human Sin1 gene has been automatically predicted (MAPKAP1; GenBank accession number ); however, this sequence appears to be incomplete. In this study, we have cloned and characterized the full-length human Sin1 mRNA and identified a highly conserved domain that defines the family of SIN1 orthologues, members of which are widely distributed in the fungal and metazoan kingdoms. We demonstrate that Sin1 transcripts can use alternative polyadenylation signals and describe a number of Sin1 splice variants that potentially encode functionally different isoforms.


Subject(s)
Alternative Splicing , Carrier Proteins/genetics , Poly A/genetics , RNA, Messenger/genetics , 3' Untranslated Regions/genetics , 3' Untranslated Regions/metabolism , 5' Untranslated Regions/genetics , 5' Untranslated Regions/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/genetics , Gene Expression Profiling , Humans , Molecular Sequence Data , Poly A/metabolism , Polyadenylation , Protein Isoforms/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid
9.
Virology ; 318(1): 280-94, 2004 Jan 05.
Article in English | MEDLINE | ID: mdl-14972554

ABSTRACT

The Epstein-Barr virus nuclear antigen (EBNA)-6 protein is essential for Epstein-Barr virus (EBV)-induced immortalization of primary human B-lymphocytes in vitro. In this study, fusion proteins of EBNA-6 with green fluorescent protein (GFP) have been used to characterize its nuclear localization and organization within the nucleus. EBNA-6 associates with nuclear structures and in immunofluorescence demonstrate a punctate staining pattern. Herein, we show that the association of EBNA-6 with these nuclear structures was maintained throughout the cell cycle and with the use of GFP-E6 deletion mutants, that the region amino acids 733-808 of EBNA-6 contains a domain that can influence the association of EBNA-6 with these nuclear structures. Co-immunofluorescence and confocal analyses demonstrated that EBNA-6 and EBNA-3 co-localize in the nucleus of cells. Expression of EBNA-6, but not EBNA-3, caused a redistribution of nuclear survival of motor neurons protein (SMN) to the EBNA-6 containing nuclear structures resulting in co-localization of SMN with EBNA-6.


Subject(s)
Cell Nucleus/virology , Cell Transformation, Viral , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/physiology , Nerve Tissue Proteins/metabolism , Cell Line, Tumor , Cell Nucleus Structures , Cyclic AMP Response Element-Binding Protein , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Deletion , Green Fluorescent Proteins , HeLa Cells , Herpesvirus 4, Human/genetics , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Nerve Tissue Proteins/genetics , RNA-Binding Proteins , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SMN Complex Proteins
10.
Oncogene ; 23(7): 1342-53, 2004 Feb 19.
Article in English | MEDLINE | ID: mdl-14716295

ABSTRACT

The Epstein-Barr nuclear antigens (EBNA), EBNA-3, -4 and -6, have previously been shown to act as transcriptional regulators, however, this study identifies another function for these proteins, disruption of the G2/M checkpoint. Lymphoblastoid cell lines (LCLs) treated with a G2/M initiating drug azelaic bishydroxamine (ABHA) did not show a G2/M checkpoint response, but rather they display an increase in cell death, a characteristic of sensitivity to the cytotoxic effects of the drug. Cell cycle analysis demonstrated that the individual expression of EBNA-3, -4 or -6 are capable of disrupting the G2/M checkpoint response induced by ABHA resulting in increased toxicity, whereas EBNA-2, and -5 were not. EBNA-3 gene family protein expression also disrupted the G2/M checkpoint initiated in response to the genotoxin etoposide and the S phase inhibitor hydroxyurea. The G2 arrest in response to these drugs were sensitive to caffeine, suggesting that ATM/ATR signalling in these checkpoint responses may be blocked by the EBNA-3 family proteins. The function of EBNA-3, -4 and -6 proteins appears to be more complex than anticipated and these data suggest a role for these proteins in disrupting the host cell cycle machinery.


Subject(s)
Cell Cycle Proteins , Epstein-Barr Virus Nuclear Antigens/metabolism , G2 Phase/physiology , Mitosis/physiology , Ataxia Telangiectasia Mutated Proteins , Checkpoint Kinase 2 , DNA Damage/physiology , DNA-Binding Proteins , Epstein-Barr Virus Nuclear Antigens/immunology , G2 Phase/drug effects , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Mitosis/drug effects , Precipitin Tests , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins
11.
J Gen Virol ; 85(Pt 1): 165-172, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14718631

ABSTRACT

Epstein-Barr virus nuclear antigen (EBNA)-6 is essential for EBV-induced immortalization of primary human B-lymphocytes in vitro. Previous studies have shown that EBNA-6 acts as a transcriptional regulator of viral and cellular genes; however at present, few functional domains of the 140 kDa EBNA-6 protein have been completely characterized. There are five computer-predicted nuclear localization signals (NLS), four monopartite and one bipartite, present in the EBNA-6 amino acid sequence. To identify which of these NLS are functional, fusion proteins between green fluorescent protein and deletion constructs of EBNA-6 were expressed in HeLa cells. Each of the constructs containing at least one of the NLS was targeted to the nucleus of cells whereas a construct lacking all of the NLS was cytoplasmic. Site-directed mutation of these NLS demonstrated that only three of the NLS were functional, one at the N-terminal end (aa 72-80), one in the middle (aa 412-418) and one at the C-terminal end (aa 939-945) of the EBNA-6 protein.


Subject(s)
Cell Nucleus/metabolism , Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/genetics , Nuclear Localization Signals , Amino Acid Sequence , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Deletion , Green Fluorescent Proteins , HeLa Cells , Herpesvirus 4, Human/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Fusion Proteins
12.
J Gen Virol ; 84(Pt 7): 1871-1879, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12810882

ABSTRACT

The Epstein-Barr virus latent membrane protein (LMP 1) functions as a constitutively active signalling molecule and associates in lipid rafts clustered with other signalling molecules. Using immunofluorescent confocal microscopy, LMP 1 was shown to have an heterogeneous distribution among individual cells which was not related to the cell cycle stage. LMP 1 was shown to localize to intracellular compartments in cells other than the plasma membrane. Co-labelling of cells with both an LMP 1 antibody and an antibody to the Golgi protein GS15 revealed that the intracellular LMP 1 partly co-localized with the Golgi apparatus. Further confirmation of intracellular LMP 1 localization was obtained by immunoelectron microscopy with rabbit polyclonal LMP 1 antibodies and cryosectioning. As well as being present in intracellular foci, LMP 1 co-localized in part with MHC-II and was present on exosomes derived from a lymphoblastoid cell line. Preparations of LMP 1 containing exosomes were shown to inhibit the proliferation of peripheral blood mononuclear cells, suggesting that LMP 1 could be involved in immune regulation. This may be of particular relevance in EBV-associated tumours such as nasopharyngeal carcinoma and Hodgkin's disease, as LMP 1-containing exosomes may be taken up by infiltrating T-lymphocytes, where LMP 1 could exert an anti-proliferative effect, allowing the tumour cells to evade the immune system.


Subject(s)
Herpesvirus 4, Human/metabolism , Organelles/metabolism , Viral Matrix Proteins/metabolism , B-Lymphocytes , Cell Line, Transformed , Fluorescent Antibody Technique , Golgi Apparatus/metabolism , Herpesvirus 4, Human/physiology , Humans , Microscopy, Immunoelectron , Tumor Cells, Cultured
13.
Transplantation ; 73(2): 271-9, 2002 Jan 27.
Article in English | MEDLINE | ID: mdl-11821743

ABSTRACT

BACKGROUND: Posttransplant lymphoproliferative disease (PTLD), driven by the presence of Epstein-Barr virus (EBV), is becoming an increasingly important clinical problem after solid organ transplantation. The use of immunosuppressive therapy leads to the inhibition of the cytotoxic T cells that normally control the EBV latently infected B cells. The prognosis for many patients with PTLD is poor, and the optimal treatment strategy is not well defined. METHOD: This study investigates the use of a histone deacetylase inhibitor, azelaic bishydroxamic acid (ABHA), for its ability to effectively kill EBV-transformed lymphoblastoid cell lines. RESULTS: In vitro treatment of lymphoblastoid cell lines with ABHA showed that they were effectively killed by low doses of the drug (ID50 2-5 microg/ml) within 48 hr. As well as being effective against polyclonal B-cell lines, ABHA was also shown to be toxic to seven of eight clonal Burkitt's lymphoma cell lines, indicating that the drug may also be useful in the treatment of late-occurring clonal PTLD. In addition, ABHA treatment did not induce EBV replication or affect EBV latent gene expression. CONCLUSION: These studies suggest that ABHA effectively kills both polyclonal and clonal B-cell lines and has potential in the treatment of PTLD.


Subject(s)
B-Lymphocytes/drug effects , Enzyme Inhibitors/therapeutic use , Herpesvirus 4, Human/drug effects , Histone Deacetylase Inhibitors , Hydroxamic Acids/therapeutic use , Lymphoproliferative Disorders/drug therapy , Organ Transplantation/adverse effects , Cell Death/drug effects , Cell Line , Cell Transformation, Viral , G2 Phase/drug effects , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Lymphocyte Activation/drug effects , Mitosis/drug effects , Virus Replication/drug effects
14.
J Gen Virol ; 80 ( Pt 12): 3217-3226, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10567654

ABSTRACT

RBP, a transcriptional repressor, is intricately involved in Epstein-Barr virus (EBV) transformation of human B cells. The EBV nuclear proteins EBNA-2, -3, -4 and -6 all utilize RBP to regulate the transcription of both cellular and viral genes. This study investigates the isoforms of the RBP protein in Burkitt's lymphoma (BL) cells and in EBV-transformed lymphoblastoid cell lines (LCLs). Two-dimensional gel electrophoresis showed the presence of two different cellular isoforms of RBP; the molecular masses and isoelectric points of these two isoforms corresponded to RBP-Jkappa and RBP-2N. Fractionation studies and green fluorescent protein (GFP)-tagged expression studies demonstrated that both RBP isoforms were located predominantly in the cell nucleus. Interestingly, GFP-tagged RBP-Jkappa showed diffuse, uniform nuclear staining, whereas GFP-tagged RBP-2N showed a discrete nuclear pattern, demonstrating differences between the two isoforms. Within the nuclear fraction of EBV-negative BL cells, RBP existed both in a free form and bound to chromatin, whereas in LCLs the intranuclear RBP was predominantly chromatin-bound. Expression of the EBV latent proteins was found to lead to the sequestering of RBP from the cytoplasm into the cell nucleus and to an increase in the chromatin-bound forms of RBP.


Subject(s)
B-Lymphocytes/virology , Cell Transformation, Viral , DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/genetics , Nuclear Proteins , Repressor Proteins/metabolism , Transcription, Genetic , B-Lymphocytes/metabolism , Cell Line, Transformed , Cell Nucleus/metabolism , Chromatin/metabolism , Consensus Sequence , Cytoplasm/metabolism , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Electrophoresis, Gel, Two-Dimensional , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation, Viral , Green Fluorescent Proteins , Herpesvirus 4, Human/metabolism , Luminescent Proteins/metabolism , Protein Isoforms , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Tumor Cells, Cultured , Virus Latency/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...