Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Eur J Immunol ; 47(1): 168-179, 2017 01.
Article in English | MEDLINE | ID: mdl-27861791

ABSTRACT

The forkhead box P1 (FOXP1) transcription factor has been shown to regulate the generation and maintenance of quiescent naïve murine T cells. In humans, FOXP1 expression has been correlated with overall survival in patients with peripheral T-cell lymphoma (PTCL), although its regulatory role in T-cell function is currently unknown. We found that FOXP1 is normally expressed in all human leukocyte subpopulations. Focusing on primary human CD4+ T cells, we show that nuclear expression of FOXP1 predominates in naïve cells with significant downregulation detected in memory cells from blood and tonsils. FOXP1 is repressed following in vitro T-cell activation of naïve T cells, and later re-established in memory CD4+ T cells, albeit at lower levels. DNA methylation analysis revealed that epigenetic mechanisms participate in regulating the human FOXP1 gene. ShRNA-mediated FOXP1 repression induces CD4+ T cells to enter the cell cycle, acquire memory-like markers and upregulate helper T-cell differentiation genes. In patients with lymphoproliferative disorders, FOXP1 expression is constitutionally repressed in the clonal T cells in parallel with overexpression of helper T-cell differentiation genes. Collectively, these data identify FOXP1 as an essential transcriptional regulator for primary human CD4+ T cells and suggest its potential important role in the development of PTCL.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/metabolism , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Repressor Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Biomarkers , Cell Cycle/genetics , Cell Line , DNA Methylation , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Gene Expression , Gene Expression Regulation , Humans , Immunophenotyping , Leukocytes/immunology , Leukocytes/metabolism , Lymphocyte Activation/immunology , Lymphoproliferative Disorders/genetics , Phenotype , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/metabolism , Repressor Proteins/genetics
2.
Retrovirology ; 10: 75, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23880157

ABSTRACT

BACKGROUND: Transformation by the Tax oncoprotein of the human T cell leukemia virus type 1 (HTLV-1) is governed by actions on cellular regulatory signals, including modulation of specific cellular gene expression via activation of signaling pathways, acceleration of cell cycle progression via stimulation of cyclin-dependent kinase activity leading to retinoblastoma protein (pRb) hyperphosphorylation and perturbation of survival signals. These actions control early steps in T cell transformation and development of Adult T cell leukemia (ATL), an aggressive malignancy of HTLV-1 infected T lymphocytes. Post-translational modifications of Tax by phosphorylation, ubiquitination, sumoylation and acetylation have been implicated in Tax-mediated activation of the NF-κB pathway, a key function associated with Tax transforming potential. RESULTS: In this study, we demonstrate that acetylation at lysine K(346) in the carboxy-terminal domain of Tax is modulated in the Tax nuclear bodies by the acetyltransferase p300 and the deacetylases HDAC5/7 and controls phosphorylation of the tumor suppressor pRb by Tax-cyclin D3-CDK4-p21(CIP) complexes. This property correlates with the inability of the acetylation deficient K(346)R mutant, but not the acetylation mimetic K(346)Q mutant, to promote anchorage-independent growth of Rat-1 fibroblasts. By contrast, acetylation at lysine K(346) had no effects on the ability of Tax carboxy-terminal PDZ-binding domain to interact with the tumor suppressor hDLG. CONCLUSIONS: The identification of the acetyltransferase p300 and the deacetylase HDAC7 as enzymes modulating Tax acetylation points to new therapeutic targets for the treatment of HTLV-1 infected patients at risk of developing ATL.


Subject(s)
Cell Transformation, Viral , Gene Products, tax/metabolism , Histone Deacetylases/metabolism , Human T-lymphotropic virus 1/pathogenicity , Lysine/metabolism , Protein Processing, Post-Translational , p300-CBP Transcription Factors/metabolism , Acetylation , Animals , Cell Line , Fibroblasts/virology , Humans , Rats
3.
Retrovirology ; 9: 102, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23217160

ABSTRACT

BACKGROUND: Retroviruses HTLV-1 and HTLV-2 have homologous genomic structures but differ significantly in pathogenicity. HTLV-1 is associated with Adult T cell Leukemia (ATL), whereas infection by HTLV-2 has no association with neoplasia. Transformation of T lymphocytes by HTLV-1 is linked to the capacity of its oncoprotein Tax-1 to alter cell survival and cell cycle control mechanisms. Among these functions, Tax-1-mediated activation of cellular gene expression via the NF-κB pathway depends on Tax-1 post-translational modifications by ubiquitination and sumoylation. The Tax-2 protein of HTLV-2B (Tax-2B) is also modified by ubiquitination and sumoylation and activates the NF-κB pathway to a level similar to that of Tax-1. The present study aims to understand whether ubiquitination and sumoylation modifications are involved in Tax-2B-mediated activation of the NF-κB pathway. RESULTS: The comparison of Tax-1 and Tax-2B lysine to arginine substitution mutants revealed conserved patterns and levels of ubiquitination with notable difference in the lysine usage for sumoylation. Neither Tax-1 nor Tax-2B ubiquitination and sumoylation deficient mutants could activate the NF-κB pathway and fusion of ubiquitin or SUMO-1 to the C-terminus of the ubiquitination and sumoylation deficient Tax-2B mutant strikingly restored transcriptional activity. In addition, ubiquitinated forms of Tax-2B colocalized with RelA and IKKγ in prominent cytoplasmic structures associated with the Golgi apparatus, whereas colocalization of Tax-2B with the RelA subunit of NF-κB and the transcriptional coactivator p300 in punctate nuclear structures was dependent on Tax-2B sumoylation, as previously observed for Tax-1. CONCLUSIONS: Both Tax-1 and Tax-2 activate the NF-κB pathway via similar mechanisms involving ubiquitination and sumoylation. Therefore, the different transforming potential of HTLV-1 and HTLV-2 is unlikely to be related to different modes of activation of the canonical NF-κB pathway.


Subject(s)
Gene Products, tax/metabolism , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 2/physiology , NF-kappa B/metabolism , Sumoylation , Amino Acid Sequence , Amino Acid Substitution , Cell Nucleus/metabolism , Conserved Sequence , E1A-Associated p300 Protein/metabolism , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , I-kappa B Kinase/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Transport , Transcriptional Activation , Ubiquitination
4.
J Biol Chem ; 285(33): 25831-40, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20558726

ABSTRACT

The oncogenic protein BCL-3 activates or represses gene transcription through binding with the NF-kappaB proteins p50 and p52 and is degraded through a phospho- and GSK3-dependent pathway. However, the mechanisms underlying its degradation remain poorly understood. Yeast two-hybrid analysis led to the identification of the proteasome subunit PSMB1 as a BCL-3-associated protein. The binding of BCL-3 to PSMB1 is required for its degradation through the proteasome. Indeed, PSMB1-depleted cells are defective in degrading polyubiquitinated BCL-3. The N-terminal part of BCL-3 includes lysines 13 and 26 required for the Lys(48)-linked polyubiquitination of BCL-3. Moreover, the E3 ligase FBW7, known to polyubiquitinate a variety of substrates phosphorylated by GSK3, is dispensable for BCL-3 degradation. Thus, our data defined a unique motif of BCL-3 that is needed for its recruitment to the proteasome and identified PSMB1 as a key protein required for the proteasome-mediated degradation of a nuclear and oncogenic IkappaB protein.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , B-Cell Lymphoma 3 Protein , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoprecipitation , Lysine/metabolism , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Phosphorylation/genetics , Phosphorylation/physiology , Proteasome Endopeptidase Complex/genetics , Protein Binding/genetics , Protein Binding/physiology , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/physiology
5.
Carcinogenesis ; 31(3): 473-80, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19917629

ABSTRACT

Among high-risk human papillomaviruses (HPV), HPV-16 infection is the most prevalent causative factor for cervical cancer. Beside other mucosal targets, HPV-16 was reported to infect the placenta and to replicate in trophoblastic cells. Since these cells share invasive properties of tumoral cells, they represent an ideal model to investigate several oncogenic processes. In the present work, we analyzed the impacts of HPV-16 E5, E6 and E7 oncoproteins on the trophoblastic model. Our results showed that E5 impaired the viability of trophoblastic and cervical cell lines but E6 and E7, favoring cell growth, neutralized the E5 cytotoxic effect. In addition, E5 decreased the adhesiveness of trophoblastic cells to the tissue culture plastic and to endometrial cells similarly as described previously for E6 and E7. E5 and E6 plus E7 increased also their migration and their invasive properties. Cells expressing HPV-16 early proteins under the control of the long control region endogenous promoter displayed growth advantage and were also more motile and invasive compared with control cells. Interestingly, the E-cadherin was downregulated in trophoblastic cells expressing E5, E6 and E7. Nuclear factor-kappaB and activator protein-1 activities were also enhanced. In conclusion, HPV-16 early proteins enhanced trophoblastic growth and intensify the malignant phenotype by impairing cell adhesion leading to increased cellular motile and invasive properties. HPV-16 E5 participated, with E6 and E7, in these changes by impairing E-cadherin expression, a hallmark of malignant progression.


Subject(s)
Choriocarcinoma/pathology , Endometrial Neoplasms/pathology , Human papillomavirus 16/physiology , Oncogene Proteins, Viral/physiology , Papillomavirus E7 Proteins/physiology , Repressor Proteins/physiology , Uterine Cervical Neoplasms/pathology , Uterine Neoplasms/pathology , Cadherins/biosynthesis , Cadherins/genetics , Cell Adhesion , Cell Line, Tumor/cytology , Cell Line, Tumor/virology , Chemotaxis/physiology , Cytopathogenic Effect, Viral , Female , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Human papillomavirus 16/genetics , Human papillomavirus 16/pathogenicity , Humans , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phenotype , Transfection
6.
J Virol ; 82(16): 7913-22, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18495761

ABSTRACT

The human T-cell leukemia virus type 1 (HTLV-1) Tax transactivator is known to induce or repress various cellular genes, several of them encoding transcription factors. As Tax is known to deregulate various basic bHLH factors, we looked more specifically at its effect on TAL1 (T-cell acute lymphoblastic leukemia 1), also known as SCL (stem cell leukemia). Indeed, TAL1 is deregulated in a high percentage of T-cell acute lymphoblastic leukemia cells, and its oncogenic properties are well-established. Here we show that Tax induces transcription of this proto-oncogene by stimulating the activity of the TAL1 gene promoter 1b, through both the CREB and NF-kappaB pathways. It was also observed that TAL1 upregulates HTLV-1 promoter activity, in either the presence or the absence of Tax. The viral promoter is inhibited in trans by expression of the E2A protein E47, and TAL1 is able to abrogate this inhibition. These data show the existence of a positive feedback loop between Tax and TAL1 expression and support the notion that this proto-oncogene participates in generation of adult T-cell leukemia/lymphoma by increasing the amount of the Tax oncoprotein but also possibly by its own transforming activities.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Viral , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/metabolism , Leukemia-Lymphoma, Adult T-Cell/virology , Proto-Oncogene Proteins/metabolism , Binding Sites , Cell Line , Feedback, Physiological , HeLa Cells , Humans , Models, Biological , NF-kappa B/metabolism , Promoter Regions, Genetic , Proto-Oncogene Mas , T-Cell Acute Lymphocytic Leukemia Protein 1 , Thymus Gland/cytology
7.
Mol Cell Biol ; 25(23): 10391-406, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16287853

ABSTRACT

The transcription factor NF-kappaB is critical for the induction of cancer, including adult T-cell leukemia, which is linked to infection by human T-cell leukemia virus type 1 and the expression of its regulatory protein Tax. Although activation of the NF-kappaB pathway by Tax involves its interaction with the regulatory subunit of the IkappaB kinase (IKK) complex, NEMO/IKKgamma, the mechanism by which Tax activates specific cellular genes in the nucleus remains unknown. Here, we demonstrate that the attachment of SUMO-1 to Tax regulates its localization in nuclear bodies and the recruitment of both the RelA subunit of NF-kappaB and free IKKgamma in these nuclear structures. However, this sumoylation step is not sufficient for the activation of the NF-kappaB pathway by Tax. This activity requires the prior ubiquitination and colocalization of ubiquitinated Tax with IKK complexes in the cytoplasm and the subsequent migration of the RelA subunit of NF-kappaB to the nucleus. Thus, the ubiquitination and sumoylation of Tax function in concert to result in the migration of RelA to the nucleus and its accumulation with IKKgamma in nuclear bodies for activation of gene expression. These modifications may result in targets for the treatment of adult T-cell leukemia.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/metabolism , Lysine/metabolism , NF-kappa B/chemistry , NF-kappa B/metabolism , SUMO-1 Protein/metabolism , Adaptor Proteins, Signal Transducing , Autophagy-Related Proteins , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Expression Regulation , Gene Products, tax/genetics , Human T-lymphotropic virus 1/genetics , Humans , I-kappa B Kinase/metabolism , Lysine/genetics , Mutation/genetics , NF-kappa B/genetics , Phenotype , Protein Binding , Protein Transport , SUMO-1 Protein/genetics , Signal Transduction , Transcription Factor RelA/metabolism , Transcription, Genetic/genetics
8.
Gene ; 386(1-2): 191-201, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17071021

ABSTRACT

The human T-cell leukemia virus HTLV-1 encodes regulatory proteins, Tax, Rex and p30(II), which are involved in the control of viral gene expression at the transcriptional and post-transcriptional levels. Tax localizes in unique nuclear bodies that contain components of the transcription and splicing complexes. In this work, we studied the relative intracellular localizations of Tax, Rex and p30(II). Run-on transcription assays and immunocytochemistry at light and electron microscopy levels indicated that the Tax nuclear bodies included both de novo transcribed RNA and the RNA polymerase II form that is phosphorylated on its carboxy-terminal domain whereas contacts with chromatin were observed at the periphery of these nuclear bodies. Rex first accumulated in nucleolar foci and then spread across the whole nucleus to display a diffuse and punctuate nucleoplasmic distribution. This distribution of Rex was observed in HTLV-1 transformed lymphocytes and in COS cells expressing the HTLV-1 provirus. Rex colocalized with the cellular export factor CRM-1 in the nucleolar foci as well as in the nucleoplasmic foci that did not overlap with Tax nuclear bodies but were found at the boundaries of the Tax bodies. In addition, we demonstrate that p30(II) interacts with Rex and colocalizes with the Rex/CRM-1 complexes in the nucleoli leading to their clearance from the nucleoplasm. Our results suggest that transcripts originating from Tax-induced activation of gene expression at the boundaries of the Tax bodies are transported out of the nucleus by nucleoplasmic Rex/CRM-1 complexes that are first assembled in nucleolar foci. In addition, p30(II) might exert its negative effect on viral RNA transport by preventing the release of the Rex/CRM-1 complexes from sequestration in nucleolar foci. These data support the idea that the transcriptional and post-transcriptional regulation of HTLV-1 gene expression depends on the concentration of select regulatory complexes at specific area of the nucleus.


Subject(s)
Cell Nucleolus/metabolism , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral/genetics , Gene Products, rex/metabolism , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/genetics , Retroviridae Proteins/metabolism , Animals , COS Cells , Cell Line , Cell Nucleolus/virology , Chlorocebus aethiops , Cricetinae , Humans , Phosphorylation , RNA Polymerase II/physiology , RNA, Viral/metabolism
9.
Mol Cell Biol ; 23(17): 6200-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12917341

ABSTRACT

Previous studies have implicated acetylases and deacetylases in regulating the transcriptional activity of NF-kappa B. Here, we show that inhibitors of deacetylases such as trichostatin A (TSA) and sodium butyrate (NaBut) potentiated TNF-induced expression of several natural NF-kappa B-driven promoters. This transcriptional synergism observed between TNF and TSA (or NaBut) required intact kappa B sites in all promoters tested and was biologically relevant as demonstrated by RNase protection on two instances of endogenous NF-kappa B-regulated gene transcription. Importantly, TSA prolonged both TNF-induced DNA-binding activity and the presence of NF-kappa B in the nucleus. We showed that the p65 subunit of NF-kappa B was acetylated in vivo. However, this acetylation was weak, suggesting that other mechanisms could be implicated in the potentiated binding and transactivation activities of NF-kappa B after TNF plus TSA versus TNF treatment. Western blot and immunofluorescence confocal microscopy experiments revealed a delay in the cytoplasmic reappearance of the I kappa B alpha inhibitor that correlated temporally with the prolonged intranuclear binding and presence of NF-kappa B. This delay was due neither to a defect in I kappa B alpha mRNA production nor to a nuclear retention of I kappa B alpha but was rather due to a persistent proteasome-mediated degradation of I kappa B alpha. A prolongation of I kappa B kinase activity could explain, at least partially, the delayed I kappa B alpha cytoplasmic reappearance observed in presence of TNF plus TSA.


Subject(s)
Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Butyric Acid/pharmacology , Cysteine Endopeptidases , HeLa Cells , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , I-kappa B Kinase , I-kappa B Proteins/drug effects , I-kappa B Proteins/genetics , Leupeptins/pharmacology , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Multienzyme Complexes/antagonists & inhibitors , NF-KappaB Inhibitor alpha , NF-kappa B/drug effects , NF-kappa B/genetics , Proteasome Endopeptidase Complex , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Transcription Factor RelA
10.
Mol Cancer Ther ; 5(12): 3042-51, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17148762

ABSTRACT

BACKGROUND: Preclinical and clinical studies have shown that the proteasome inhibitor bortezomib (PS341, Velcade) is highly effective when combined with chemotherapeutic agents. The value of trastuzumab (Herceptin) in HER-2-positive (3+ score by immunohistochemistry or fluorescence in situ hybridization positive) breast cancer is also known; however, the response rate is <40% for metastatic breast cancer. These two pharmacologic agents prevent nuclear factor-kappaB (NF-kappaB) activation and induce nuclear accumulation of the cyclin-dependent kinase inhibitor p27(kip1), suggesting that combining bortezomib with trastuzumab could increase trastuzumab efficacy. METHODS: Drug cytotoxicity, both individually and together, and drug effects on p27 localization and NF-kappaB activation were investigated on four breast cancer cell lines: SKBR-3 (HER-2+++), MDA-MB-453 (HER-2++), HER-2-transfected MCF-7 (HER-2+++), and MCF-7 (HER-2-). RESULTS: Bortezomib induced apoptosis in HER-2-positive and HER-2-negative breast cancer cells in a dose- and time-dependent manner. Together, these drugs induced apoptosis of HER-2++/+++ cells at low concentrations, which had no effect when used alone, indicating there was a synergistic effect. Sequential treatment (trastuzumab then bortezomib) induced either necrosis or apoptosis, depending on the trastuzumab preincubation time. Susceptibility to bortezomib alone and the drug combination correlated with NF-kappaB activity and p27 localization. CONCLUSIONS: The addition of bortezomib to trastuzumab increases the effect of trastuzumab in HER-2+++/++ cell lines in a synergistic way. This effect likely results from the ability of these two drugs to target the NF-kappaB and p27 pathways. The potential clinical application of this drug combination is under current evaluation by our group in a phase 1 clinical trial.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/pharmacology , Breast Neoplasms/drug therapy , Pyrazines/pharmacology , Receptor, ErbB-2/biosynthesis , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Boronic Acids/administration & dosage , Bortezomib , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Drug Resistance, Neoplasm , Drug Synergism , Humans , NF-kappa B/biosynthesis , NF-kappa B/metabolism , Pyrazines/administration & dosage , Trastuzumab
11.
Oncogene ; 24(3): 419-30, 2005 Jan 13.
Article in English | MEDLINE | ID: mdl-15543232

ABSTRACT

HTLV-I associated adult T-cell leukemia (ATL) and HTLV-I-negative peripheral T-cell lymphomas are associated with poor prognosis. Using pharmacological concentrations of the proteasome inhibitor PS-341, we demonstrate inhibition of cell proliferation and induction of apoptosis in fresh ATL cells, HTLV-I transformed and HTLV-I-negative malignant T cells, while normal resting or activated T lymphocytes were resistant. Combination of PS-341 and doxorubicin or etoposide resulted in an additive growth inhibition. In HTLV-I-negative malignant cells, PS-341 treatment significantly downregulated the antiapoptotic protein X-IAP and to a lesser extent c-IAP-1 and bcl-X(L) and resulted in caspase-dependent apoptosis. In HTLV-I transformed cells, the inhibition of the proteasomal degradation of Tax by PS-341 likely explains the relative protection of HTLV-I infected cells against caspase-dependent apoptosis. PS-341 treatment of these cells stabilized IkappaBalpha, IkappaBbeta, IkappaBvarepsilon, p21, p27 and p53 proteins and selectively inhibited Rel-A DNA binding NF-kappaB complexes. In both HTLV-I-positive and -negative cells, PS-341 treatment induced ceramide accumulation that correlated with apoptosis. We conclude that PS-341 affects multiple pathways critical for the survival of HTLV-I-positive and -negative malignant T cells supporting a potential therapeutic role for PS-341 in both ATL and HTLV-I-negative T-cell lymphomas, whether alone or in combination with chemotherapy.


Subject(s)
Antineoplastic Agents/toxicity , Boronic Acids/toxicity , Leukemia-Lymphoma, Adult T-Cell/pathology , Lymphoma, T-Cell/pathology , Protease Inhibitors/toxicity , Proteasome Inhibitors , Pyrazines/toxicity , Adult , Apoptosis/drug effects , Bortezomib , Cell Division/drug effects , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Ceramides/metabolism , Humans , Jurkat Cells
12.
Retrovirology ; 2: 8, 2005 Feb 09.
Article in English | MEDLINE | ID: mdl-15703079

ABSTRACT

BACKGROUND: In this study, we have examined the role of heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) in viral gene expression in T lymphocytes transformed by HTLV-1. RESULTS: We have previously observed that hnRNP A1 (A1) down-modulates the post transcriptional activity of Rex protein of HTLV-1. Here, we tested whether the ectopic expression of a dominant negative mutant (NLS-A1-HA) defective in shuttling activity or knockdown of the hnRNPA1 gene using RNA interference could inhibit Rex-mediated export of viral mRNAs in HTLV-1 producing C91PL T-cells. We show that the expression of NLS-A1-HA does not modify the export of Rex-dependent viral mRNAs. Conversely, inhibiting A1 expression in C91PL cells by RNA interference provoked an increase in the Rex-dependent export of unspliced and singly spliced mRNAs. Surprisingly, we also observed a significant increase in proviral transcription and an accumulation of unspliced mRNAs, suggesting that the splicing process was affected. Finally, A1 knockdown in C91PL cells increased viral production by these cells. Thus, hnRNP A1 is implicated in the modulation of the level of HTLV-1 gene expression in T cells transformed by this human retrovirus. CONCLUSIONS: These observations provide an insight into a new cellular control of HTLV-1 replication and suggest that hnRNP A1 is likely part of the regulatory mechanisms of the life cycle of this human retrovirus in T cells.


Subject(s)
Cell Transformation, Viral , Gene Expression Regulation, Viral , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Human T-lymphotropic virus 1/physiology , T-Lymphocytes/virology , Virus Replication/drug effects , Cell Line, Transformed , Gene Deletion , Gene Products, rex/genetics , Gene Products, rex/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/pharmacology , Human T-lymphotropic virus 1/drug effects , Humans , Jurkat Cells , RNA Interference , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism
13.
Cloning Stem Cells ; 6(2): 140-9, 2004.
Article in English | MEDLINE | ID: mdl-15268788

ABSTRACT

The genetic make-up of a cell resides entirely in its DNA. Now that the nucleotide sequence of several genomes has been determined, the major challenging problem is to understand how cell differentiation, proliferation or death are controlled. Major steps include analysis of the determinants of the cell cycle, the unravelling of RNAs and proteins involved in the control of gene expression and the dissection of the protein-destruction machinery. The successive steps to be considered are transcription of RNA on the DNA template, mRNA stabilization or degradation, and mRNA translation and protein localization in the right cell compartment. Gene expression or gene silencing is the result of many DNA-RNA-protein interactions and chromatin is among the key regulators of gene expression. Open chromatin (euchromatin) allows expression of the DNA message. This chromatin structure is generally characterized by the presence on the gene promoters of transcription complexes associated with histone acetyltransferases (HATs). On the contrary, closed chromatin (heterochromatin) is poorly acetylated and more condensed. It contains histone deacetylases (HDACs), potentially associated with DNA methyltransferases (DNMTs). DNMT activity leads to methylation and silencing of the DNA. Thus, a major problem in the field of gene regulation resides in understanding chromatin structure at each promoter, a formidable task for the years to come.


Subject(s)
Gene Expression Regulation , Gene Silencing , Heterochromatin/genetics , Acetylation , Animals , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Chromatin Assembly and Disassembly , Heterochromatin/metabolism , Histones/metabolism , Humans , Promoter Regions, Genetic , Retroviridae/genetics , Transcriptional Activation
14.
Front Microbiol ; 4: 271, 2013 Sep 09.
Article in English | MEDLINE | ID: mdl-24058363

ABSTRACT

Human T cell leukemia viruses (HTLVs) are complex human retroviruses of the Deltaretrovirus genus. Four types have been identified thus far, with HTLV-1 and HTLV-2 much more prevalent than HTLV-3 or HTLV-4. HTLV-1 and HTLV-2 possess strictly related genomic structures, but differ significantly in pathogenicity, as HTLV-1 is the causative agent of adult T cell leukemia and of HTLV-associated myelopathy/tropical spastic paraparesis, whereas HTLV-2 is not associated with neoplasia. HTLVs code for a protein named Tax that is responsible for enhancing viral expression and drives cell transformation. Much effort has been invested to dissect the impact of Tax on signal transduction pathways and to identify functional differences between the HTLV Tax proteins that may explain the distinct oncogenic potential of HTLV-1 and HTLV-2. This review summarizes our current knowledge of Tax-1 and Tax-2 with emphasis on their structure, role in activation of the NF-κB (nuclear factor kappa-B) pathway, and interactions with host factors.

15.
Viruses ; 3(6): 829-57, 2011 06.
Article in English | MEDLINE | ID: mdl-21994756

ABSTRACT

The HTLV-1 Tax protein both activates viral replication and is involved in HTLV-1-mediated transformation of T lymphocytes. The transforming properties of Tax include altering the expression of select cellular genes via activation of cellular pathways and perturbation of both cell cycle control mechanisms and apoptotic signals. The recent discovery that Tax undergoes a hierarchical sequence of posttranslational modifications that control its intracellular localization provides provocative insights into the mechanisms regulating Tax transcriptional and transforming activities.


Subject(s)
Gene Products, tax/metabolism , HTLV-I Infections/virology , Human T-lymphotropic virus 1/metabolism , Animals , Cell Transformation, Viral , Gene Products, tax/genetics , HTLV-I Infections/genetics , HTLV-I Infections/metabolism , Human T-lymphotropic virus 1/genetics , Humans , Signal Transduction , Transcriptional Activation
16.
Virology ; 408(1): 39-48, 2010 Dec 05.
Article in English | MEDLINE | ID: mdl-20875659

ABSTRACT

HTLV-1 is more pathogenic than HTLV-2 despite having a similar genome and closely related transactivating oncoproteins. Both Tax-1 protein from HTLV-1 and Tax-2 from HTLV-2 activate the NF-κB pathway. The mechanisms involved in Tax-1 deregulation of this signalling pathway have been thoroughly investigated, but little is known about regulation by Tax-2. We have compared the interaction of Tax-1 and Tax-2 with two key NF-κB signalling factors: TAK1-binding protein 2 (TAB2), an adaptor involved in the activation of TAK1 kinase, and RelA, the active subunit of the canonical RelA/p50 NF-κB transcription factor. Tax-2 formed stable complexes with both RelA and TAB2. These two NF-κB factors colocalized with Tax proteins in dotted cytoplasmic structures targeted by calreticulin, a multi-process calcium-buffering chaperone. Co-expression of RelA and/or TAB2 markedly increased Tax-mediated NF-κB activation. These findings provide new insights into the role of RelA, TAB2 and Tax in the deregulation of the NF-κB pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calreticulin/analysis , Cytoplasmic Structures/virology , Gene Products, tax/metabolism , NF-kappa B/immunology , Transcription Factor RelA/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , Cytoplasmic Structures/chemistry , Gene Products, tax/immunology , Human T-lymphotropic virus 1/immunology , Human T-lymphotropic virus 2/immunology , Humans , NF-kappa B/metabolism , Protein Binding
17.
Virology ; 386(1): 68-78, 2009 Mar 30.
Article in English | MEDLINE | ID: mdl-19200568

ABSTRACT

The oncogenic potential of the HTLV-1 Tax protein involves activation of the NF-kappaB pathway, which depends on Tax phosphorylation, ubiquitination and sumoylation. We demonstrate that the nuclei of Tax-expressing cells, including HTLV-1 transformed T-lymphocytes, contain a pool of Tax molecules acetylated on lysine residue at amino acid position 346 by the transcriptional coactivator p300. Phosphorylation of Tax on serine residues 300/301 was a prerequisite for Tax localization in the nucleus and correlated with its subsequent acetylation by p300, whereas sumoylation, resulting in the formation of Tax nuclear bodies in which p300 was recruited, favored Tax acetylation. Overexpression of p300 markedly increased Tax acetylation and the ability of a wild type HTLV-1 provirus, but not of a mutant provirus carrying an acetylation deficient Tax gene, to activate gene expression from an integrated NF-kappaB-controlled promoter. Thus, Tax acetylation favors NF-kappaB activation and might play an important role in HTLV-1-induced cell transformation.


Subject(s)
E1A-Associated p300 Protein/metabolism , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/physiology , NF-kappa B/metabolism , Acetylation , Cell Nucleus/chemistry , HeLa Cells , Humans , Phosphorylation
18.
Virology ; 386(1): 6-11, 2009 Mar 30.
Article in English | MEDLINE | ID: mdl-19195675

ABSTRACT

HTLV-1 is more pathogenic than HTLV-2B. The difference is generally attributed to the properties of their individual transactivating Tax proteins. By using internal Flag-6His tagged Tax-1 and Tax-2B, which display transcriptional activities comparable to the untagged proteins and can be recognized by a single anti-Flag antibody, we demonstrate that Tax-2B is modified by ubiquitination and sumoylation. In addition, Tax2B is distributed in punctuate nuclear structures that include the RelA subunit of NF-kappaB, as has been previously demonstrated for Tax-1.


Subject(s)
Gene Products, tax/metabolism , Human T-lymphotropic virus 2/physiology , Cell Line , Cell Nucleus/chemistry , Humans , Transcription Factor RelA/metabolism , Ubiquitination
19.
J Biol Chem ; 282(29): 21308-18, 2007 Jul 20.
Article in English | MEDLINE | ID: mdl-17537731

ABSTRACT

IKKalpha regulates many chromatin events in the nuclear phase of the NF-kappaB program, including phosphorylation of histone H3 and removal of co-repressors from NF-kappaB-dependent promoters. However, all of the nuclear functions of IKKalpha are not understood. In this study, using mouse embryonic fibroblasts IKKalpha knock-out and reexpressing IKKalpha after retroviral transduction, we demonstrate that IKKalpha contributes to NF-kappaB/p65 DNA binding activity on an exogenous kappaB element and on some, but not all, endogenous NF-kappaB-target promoters. Indeed, p65 chromatin immunoprecipitation assays revealed that IKKalpha is crucial for p65 binding on kappaB sites of icam-1 and mcp-1 promoters but not on ikappabalpha promoter. The mutation of IKKalpha putative nuclear localization sequence, which prevents its nuclear translocation, or of crucial serines in the IKKalpha activation loop completely inhibits p65 binding on icam-1 and mcp-1 promoters and rather enhances p65 binding on the ikappabalpha promoter. Further molecular studies demonstrated that the removal of chromatin-bound HDAC3, a histone deacetylase inhibiting p65 DNA binding, is differentially regulated by IKKalpha in a promoter-specific manner. Indeed, whereas the absence of IKKalpha induces HDAC3 recruitment and repression on the icam-1 promoter, it has an opposite effect on the ikappabalpha promoter, where a better p65 binding occurs. We conclude that nuclear IKKalpha is required for p65 DNA binding in a gene-specific manner.


Subject(s)
I-kappa B Kinase/genetics , Promoter Regions, Genetic , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Animals , Cell Nucleus/metabolism , Chromatin/metabolism , Fibroblasts/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Mice , Models, Biological , Mutation , Phosphorylation , Protein Binding
20.
Eur J Immunol ; 37(2): 376-84, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17274002

ABSTRACT

Human papillomavirus type 16 is commonly implicated in cervical cancers. The viral genome encodes potential targets like the oncoprotein E7, expressed in transformed cells but thought to represent a poorly immunogenic antigen. We describe in this work a DNA-based vaccination protocol aimed at inducing an efficient anti-E7 immune response in vivo. Plasmids allowing the expression of the E7 protein in distinct cellular compartments were generated and assayed in an in vivo model of tumor growth. Our data demonstrate that mice vaccinated with a plasmid encoding for an E7 protein fused to a domain of the MHC class II-associated invariant chain (IiE7) were protected against tumor challenge. Mice immunized against an ubiquitinated form of E7 (Ub(Ala)E7) failed to control tumor growth. Protection induced by IiE7 was correlated with the development of CD8+ CTL and required the presence of CD4+ cells. In vitro studies confirmed that the IiE7 fusion protein was expressed at high levels in the endosomal compartment of transfected cells, while the natural and the ubiquitin-modified form of E7 were mainly nuclear. The present study suggests that an efficient anti-tumor response can be induced in vivo by DNA constructs encoding for E7 protein forms localizing at the endosomal compartment.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endosomes/metabolism , Oncogene Proteins, Viral/immunology , Papillomavirus Vaccines/immunology , Uterine Cervical Neoplasms/prevention & control , Vaccines, DNA/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , Cancer Vaccines/immunology , Endosomes/immunology , Female , Flow Cytometry , Genetic Vectors , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Human papillomavirus 16/immunology , Humans , Immunohistochemistry , Mice , Microscopy, Confocal , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins , Papillomavirus Infections/complications , Papillomavirus Infections/immunology , Plasmids/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Transfection , Ubiquitin/immunology , Ubiquitin/metabolism , Uterine Cervical Neoplasms/virology
SELECTION OF CITATIONS
SEARCH DETAIL