Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Rev Neurol (Paris) ; 168(3): 257-69, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22405461

ABSTRACT

In 1980, Human T cell leukemia/lymphoma virus type 1 (HTLV-1) was the first oncogenic human retrovirus to be discovered. HTLV-1 belongs to the Retroviridae family, the Orthoretrovirinae subfamily and to the deltaretrovirus genus. HTLV-1 preferentially infects CD4(+) lymphoid cells in vivo. Three molecules have been identified for binding and/or entry of HTLV-1: heparan sulfate proteoglycans, neuropilin-1, and glucose transporter 1. An efficient transfer of the virus from an infected cell to a target cell can occur through the formation of a viral synapse and/or by virofilm structure. As for all retroviruses, HTLV-1 genome possesses three major ORFs (gag, pol and env) encoding the structural and enzymatic proteins. HTLV-1 encodes also some regulatory and auxillary proteins including the tax protein with transforming activities and the HBZ protein which plays a role in the proliferation and maintenance of the leukemic cells. HTLV-1 is present throughout the world with clusters of high endemicity including mainly Southern Japan, the Caribbean region, areas in South America and in intertropical Africa. The worldwide HTLV-1 infected population is estimated to be around 10-20 million. HTLV-1 has three modes of transmission: (1): mother to child, mainly linked to prolonged breast-feeding; (2): sexual, mainly occurring from male to female and (3): contaminated blood products. HTLV-1 possesses a remarkable genetic stability. HTLV-1 is the etiological agent of mainly two severe diseases: a malignant T CD4(+) cell lymphoproliferation, of very poor prognosis, named Adult T cell Leukemia/Lymphoma (ATLL), and a chronic neuro-myelopathy named Tropical spastic paraparesis/HTLV-1 Associated Myelopathy (TSP/HAM). The lifetime risk among HTLV-1 carriers is estimated to be around 0.25 to 3%. TSP/HAM mainly occurs in adults, with a mean age at onset of 40-50 years and it is more common in women than in men. Blood transfusion is a major risk factor for TSP/HAM development. Clinically, TSP/HAM is mainly defined as a chronic spastic paraparesis and minor sensory signs. The onset is insidious with often gait disturbance and urinary symptoms. In more than 90% of the cases, the neurological features involve: spasticity and/or hyperreflexia of the lower extremities, urinary bladder disturbance, lower extremity muscle weakness, and in around 50% of the cases, sensory disturbances with low back pain. Central functions and cranial nerves are usually spared. The clinical course is generally progressive without remission. High levels of antibodies titers directed against HTLV-1 antigens are present in blood and cerebrospinal fluid (CSF). A high HTLV-1 proviral load is frequently observed in the blood. Mild to moderate increase of proteins may be present in the CSF. However, intrathecal production of specific HTLV-1 antibody index provides additional data to support the diagnosis. Brain white matter lesions on magnetic resonance imaging are frequent. A mild atrophy of the thoracic spinal cord can also be observed. Pathologically, it is characterized by a chronic inflammation with perivascular lymphocytic cuffing and mild parenchymal lymphocytic infiltrates. The cells are mostly CD4(+) in early disease and mostly CD8(+) in latter disease. Pyramidal tract damage with myelin and axonal loss, mainly in the lower thoracic spinal cord are observed. TSP/HAM pathogenesis is still poorly understood and viral and host factors as the proviral load and the cellular immune response play a major role in disease progression. TSP/HAM can be associated with other HTLV-1 associated symptoms (uveitis, myositis, infective dermatitis). Therapy of TSP/HAM remains disappointing and symptomatic treatment remains still the mainstay of therapy.


Subject(s)
Deltaretrovirus , HTLV-I Infections/therapy , Paraparesis, Tropical Spastic/therapy , Adult , Female , HTLV-I Infections/epidemiology , HTLV-I Infections/pathology , HTLV-I Infections/virology , Human T-lymphotropic virus 1 , Humans , Magnetic Resonance Imaging , Male , Paraparesis, Tropical Spastic/epidemiology , Paraparesis, Tropical Spastic/pathology , Paraparesis, Tropical Spastic/virology , Tropical Medicine
2.
Bull Soc Pathol Exot ; 104(3): 181-7, 2011 Aug.
Article in French | MEDLINE | ID: mdl-21607661

ABSTRACT

HTLV-1 was the first human oncogenic retrovirus to be discovered. It is the etiological agent of adult T leukemia/lymphoma (ATLL) and of tropical spastic paraparesis/HTLV-1 associated myelopathy (TSP/HAM), two diseases that develop after a long latency period. Importantly, HTLV-1 does not cause ATLL through insertional mutagenesis. Apart from the gag, pro, pol and env genes, which are common to all retroviruses, HTLV-1 genome also encodes regulatory and auxiliary viral proteins. Among the former, Tax promotes cell transformation and HBZ is involved in the leukemic cells proliferation and in the maintenance of the transformed phenotype. Anti-ATLL therapies have lately made significant progress with an efficient antiviral treatment against the chronic and smoldering forms of this leukemia, but an efficient treatment of TSP/HAM patients is still lacking. Results from a recent study associating histone acetylase inhibitor with an anti-viral drug will be discussed here. While an increase in proviral load is considered a marker for disease progression, this treatment allows a significant drop of the proviral load in asymptomatic carriers.


Subject(s)
HTLV-I Infections/virology , Human T-lymphotropic virus 1/physiology , Animals , Anti-Retroviral Agents/therapeutic use , Clinical Trials as Topic , Drug Evaluation, Preclinical , Genome, Viral , HTLV-I Infections/drug therapy , HTLV-I Infections/prevention & control , Histone Deacetylase Inhibitors/therapeutic use , Human T-lymphotropic virus 1/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/virology , Paraparesis, Tropical Spastic/drug therapy , Paraparesis, Tropical Spastic/virology , Primates , Proviruses/drug effects , Retroviridae Proteins/genetics , Retroviridae Proteins/physiology , Valproic Acid/therapeutic use , Viral Load , Virus Replication
3.
Pathol Biol (Paris) ; 57(2): 161-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18456423

ABSTRACT

Human T cell leukemia/lymphoma virus Type 1 and 2 (HTLV-1 and HTLV-2), together with their simian counterparts (STLV-1, STLV-2), belong to the Primate T lymphotropic viruses group (PTLV). HTLV-1 infects 15 to 20million people worldwide, while STLV-1 is endemic in a number of simian or ape species living in Africa or Asia. The high percentage of homologies between HTLV-1 and STLV-1 strains, led to the demonstration that most HTLV-1 subtypes arose from interspecies transmission between monkeys and humans. STLV-3 viruses belong to the third PTLV type and are equally divergent from HTLV-1 than from HTLV-2. They are endemic in several monkey species that live in West, Central, and East Africa. In 2005, we and others reported the discovery of the human homolog (HTLV-3) of STLV-3 in two asymptomatic inhabitants from South Cameroon whose sera exhibited HTLV indeterminate serologies. More recently, we reported a third case of HTLV-3 infection in Cameroon suggesting that this virus is not rare in the human population living in Central Africa. Together with STLV-3, these three human viral strains belong therefore to the PTLV-3 type. A fourth HTLV type (HTLV-4) was also discovered in the same geographical area. Current studies are aimed at determining the prevalence, distribution and modes of transmission of these viruses as well as their possible association with human diseases. Furthermore, molecular characterization of their viral transactivator Tax is ongoing in order to look for possible oncogenic properties.


Subject(s)
Deltaretrovirus/classification , Africa/epidemiology , Animals , Deltaretrovirus Infections/epidemiology , Deltaretrovirus Infections/veterinary , Human T-lymphotropic virus 1/classification , Human T-lymphotropic virus 2/classification , Humans , Primate Diseases/epidemiology , Primate Diseases/virology
4.
Leukemia ; 22(4): 826-34, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18094712

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of primary effusion lymphoma (PEL) and of Kaposi's sarcoma. PEL is an aggressive proliferation of B cells with poor prognosis. We evaluated both in vitro and in vivo the potential role of angiogenic factors secreted by PEL cells, that is, their interaction with endothelial cells and their implication in the invasive behavior of tumoral cells. In vitro, PEL-induced angiogenesis is dependent on vascular endothelial growth factor (VEGF) and VEGF receptors. However, although PEL cells produce VEGF and basic fibroblast growth factor (b-FGF) transcripts, they only secrete VEGF in vitro. In vivo, very high levels of both VEGF and b-FGF were found in the ascitic fluid of NOD/SCID mice injected with PEL cells. We then show evidence of cell adhesion and gap junction-mediated heterocellular communication between PEL cells and endothelial cells. Finally, we show that PEL cells extravasate through the endothelial barrier and that the specific tyrosine kinase inhibitor of VEGF receptors, PTK-787/ZK-222584, the anti-VEGF antibody, bevacizumab or the gap junction inhibitor 18-alpha-glycyrrhetinic acid, partially attenuate PEL cell extravasation. Angiogenesis, cell adhesion and communication likely contribute to the development of PEL and represent potential therapeutic targets.


Subject(s)
Fibroblast Growth Factor 2/physiology , Herpesvirus 8, Human , Lymphoma, Primary Effusion/pathology , Lymphoma, Primary Effusion/virology , Neovascularization, Pathologic/etiology , Vascular Endothelial Growth Factor A/physiology , Animals , Cell Transformation, Viral , Coculture Techniques , Disease Models, Animal , Endothelial Cells/pathology , Fibroblast Growth Factor 2/metabolism , Gap Junctions/pathology , Humans , Mice , Neoplasms, Experimental , Neovascularization, Pathologic/pathology , Paracrine Communication , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
5.
Leukemia ; 21(8): 1792-801, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17568816

ABSTRACT

Kaposi's sarcoma (KS)-associated herpes virus (KSHV) is the causative agent of primary effusion lymphoma and of KS. Primary effusion lymphoma (PEL) is an aggressive proliferation of B cells. Conventional chemotherapy has limited benefits in PEL patients, and the prognosis is very poor. We previously reported that treatment of human T-cell leukemia virus type 1 (HTLV-1)-associated adult T-cell leukemia/lymphoma cells either with arsenic trioxide (As) combined to interferon-alpha (IFN-alpha) or with the bortezomib (PS-341) proteasome inhibitor induces cell cycle arrest and apoptosis, partly due to the reversal of the constitutive nuclear factor-kappaB (NF-kappaB) activation. PEL cells also display an activated NF-kappaB pathway that is necessary for their survival. This prompted us to investigate the effects of PS-341, or of the As/IFN-alpha combination on PEL cells. A dramatic inhibition of cell proliferation and induction of apoptosis was observed in PS-341 and in As/IFN-alpha treated cells. This was associated with the dissipation of the mitochondrial membrane potential, cytosolic release of cytochrome c, caspase activation and was reversed by the z-VAD caspase inhibitor. PS-341 and As/IFN-alpha treatment abrogated NF-kappaB translocation to the nucleus and decreased the levels of the anti-apoptotic protein Bcl-X(L). Altogether, these results provide a rational basis for a future therapeutic use of PS-341 or combined As and IFN-alpha in PEL patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Caspases/metabolism , Herpesvirus 8, Human/physiology , Lymphoma/pathology , Lymphoma/virology , Pyrazines/pharmacology , Arsenic Trioxide , Arsenicals/administration & dosage , Bortezomib , Cell Proliferation/drug effects , Humans , Interferon-alpha/administration & dosage , Lymphoma/enzymology , Membrane Potential, Mitochondrial/drug effects , NF-kappa B/metabolism , Oxides/administration & dosage , Protease Inhibitors/pharmacology , bcl-X Protein/metabolism
6.
Oncogene ; 25(32): 4470-82, 2006 Jul 27.
Article in English | MEDLINE | ID: mdl-16532031

ABSTRACT

Human T-cell leukemia virus and simian T-cell leukemia virus (STLV) form the primate T-cell lymphotropic viruses group. Human T-cell leukemia virus type 1 and type 2 (HTLV-1 and HTLV-2) encode the Tax viral transactivator (Tax1 and Tax2, respectively). Tax1 possesses an oncogenic potential and is responsible for cell transformation both in vivo and in vitro. We and others have recently discovered the existence of human T-cell lymphotropic virus type 3. However, there is currently no evidence for the presence of a Tax protein in HTLV-3-infected individuals. We show that the serum of an HTLV-3 asymptomatic carrier and the sera of two STLV-3-infected monkeys contain specific anti-Tax3 antibodies. We also show that tax3 mRNA is present in the PBMCs obtained from an STLV-3-infected monkey, demonstrating that Tax3 is expressed in vivo. We further demonstrate that Tax3 intracellular localization is very similar to that of Tax1 and that Tax3 binds to both CBP and p300 coactivators. Using purified Tax3, we show that the protein increases transcription from a 4TxRE G-free cassette plasmid in an in vitro transcription assay. In all cell types tested, including transiently transfected lymphocytes, Tax3 activates its own promoter STLV-3 long terminal repeat (LTR), which contains only two Tax Responsive Elements (TREs), and activates also HTLV-1 and HTLV-2 LTRs. In addition, Tax3 also activates the NF-kappaB pathway. We also show that Tax3 possesses a PDZ-binding sequence at its C-terminal end. Our results demonstrate that Tax3 is a transactivator, and that its properties are more similar to that of Tax1, rather than of Tax2. This suggests the possible occurrence of lymphoproliferative disorders among HTLV-3-infected populations.


Subject(s)
Gene Products, tax/genetics , Gene Products, tax/physiology , Human T-lymphotropic virus 1/chemistry , Human T-lymphotropic virus 2/physiology , Primate T-lymphotropic virus 3/chemistry , Amino Acid Sequence , Animals , Cell Line , Cercopithecinae , Gene Products, tax/biosynthesis , Gene Products, tax/chemistry , HeLa Cells , Human T-lymphotropic virus 1/physiology , Humans , Jurkat Cells , Molecular Sequence Data , Primate T-lymphotropic virus 3/physiology , Sequence Homology, Amino Acid
7.
Med Trop (Mars) ; 65(6): 525-8, 2005 Nov.
Article in French | MEDLINE | ID: mdl-16555510

ABSTRACT

Human T cell leukemia/lymphoma virus Type 1 and 2 (HTLV-1 and HTLV-2), together with their simian counterparts (STLV-1, STLV-2 and STLV-3), belong to the Primate T lymphotropic viruses group (PTLV). HTLV-1 infects 15 to 20 million people worldwide, while STLV-1 is endemic in a number of simian species living in the Old World. Due to the high percentage of homologies between HTLV-1 and STLV-1 strains, it has now been widely accepted that most HTLV-1 subtypes arose from interspecies transmission between monkeys and humans. On the opposite, there is no close human homolog of the two STLV-2 strains that have been discovered in African bonobos chimpanzees. These results suggest that the interspecies transmission that lead to the present day HTLV-2 must have occurred in a distant past. STLV-3 viruses are very divergent, both from HTLV-1 and from HTLV-2. They are endemic in several monkey species that live in west, central and east Africa. Recently, two laboratories independently reported the discovery of the human homolog (HTLV-3) of STLV-3 in two inhabitants from south Cameroon whose sera exhibited HTLV indeterminate serologies. Together with STLV-3, these two viruses belong therefore to the PTLV-3 group. In addition, a fourth HTLV type (HTLV-4) was also discovered in the same geographical area. Current studies are aimed at determining the molecular characterization of these viruses. In particular, the possible oncogenic properties of their viral transactivator Tax is being investigated, as well as their modes of transmission and their possible association with human diseases.


Subject(s)
Deltaretrovirus/classification , Animals , Deltaretrovirus Infections/virology , Humans
8.
Blood ; 98(13): 3762-9, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739184

ABSTRACT

Treatment of patients with adult T-cell leukemia-lymphoma (ATLL) using conventional chemotherapy has limited benefit because human T-cell leukemia virus type 1 (HTLV-1) cells are resistant to most apoptosis-inducing agents. The recent report that arsenic trioxide induces apoptosis in HTLV-1-transformed cells prompted investigation of the mechanism of action of this drug in HTLV-1 and HTLV-2 interleukin-2-independent T cells and in HTLV-1-immortalized cells or in ex vivo ATLL samples. Fluorescence-activated cell sorter analysis, fluorescence microscopy, and measures of mitochondrial membrane potential (Delta Psi m) demonstrated that arsenic trioxide alone was sufficient to induce programmed cell death in all HTLV-1 and -2 cells tested and in ATLL patient samples. I kappa B-alpha phosphorylation strongly decreased, and NF-kappa B translocation to the nucleus was abrogated. Expression of the antiapoptotic protein Bcl-X(L), whose promoter is NF-kappa B dependent, was down-regulated. The collapse of Delta Psi m and the release of cytochrome c to the cytosol resulted in the activation of caspase-3, as demonstrated by the cleavage of PARP. A specific caspase-3 inhibitor (Ac-DEVD-CHO) could reverse this phenotype. The antiapoptotic factor Bcl-2 was then cleaved, converting it to a Bax-like death effector. These results demonstrated that arsenic trioxide induces apoptosis in HTLV-1- and -2-infected cells through activation of the caspase pathway.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Caspases/metabolism , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 2/physiology , I-kappa B Proteins , Leukemia-Lymphoma, Adult T-Cell/pathology , Oxides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Arsenic Trioxide , Caspase 3 , Cell Line, Transformed , Cell Nucleus/metabolism , Cytochrome c Group/metabolism , DNA-Binding Proteins/metabolism , Flow Cytometry , Humans , Interferon-alpha/pharmacology , Leukemia-Lymphoma, Adult T-Cell/virology , Membrane Potentials , Microscopy, Fluorescence , Mitochondria/ultrastructure , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-bcl-2/analysis , Tumor Suppressor Protein p53/analysis , bcl-2-Associated X Protein , bcl-X Protein
9.
Mol Cell Biol ; 21(21): 7391-402, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11585920

ABSTRACT

The proto-oncogene c-myb is essential for a controlled balance between cell growth and differentiation. Aberrant c-Myb activity has been reported for numerous human cancers, and enforced c-Myb transcription can transform cells of lymphoid origin by stimulating cellular proliferation and inhibiting apoptotic pathways. Here we demonstrate that activation of the NF-kappaB pathway by the HTLV-1 Tax protein leads to transcriptional inactivation of c-Myb. This conclusion was supported by the fact that Tax mutants unable to stimulate the NF-kappaB pathway could not inhibit c-Myb transactivating functions. In addition, inhibition of Tax-mediated NF-kappaB activation by coexpression of IkappaBalpha restored c-Myb transcription, and Tax was unable to block c-Myb transcription in a NEMO knockout cell line. Importantly, physiological stimuli, such as signaling with the cellular cytokines tumor necrosis factor alpha, interleukin 1 beta (IL-1beta), and lipopolysaccharide, also inhibited c-Myb transcription. These results uncover a new link between extracellular signaling and c-Myb-dependent transcription. The mechanism underlying NF-kappaB-mediated repression was identified as sequestration of the coactivators CBP/p300 by RelA. Interestingly, an amino-terminal deletion form of p300 lacking the C/H1 and KIX domains and unable to bind RelA retained the ability to stimulate c-Myb transcription and prevented NF-kappaB-mediated repression.


Subject(s)
Gene Products, tax/metabolism , I-kappa B Proteins , NF-kappa B/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Transcription, Genetic , Animals , Cell Differentiation , Cell Division , Cell Line , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein , Enzyme Activation , Gene Deletion , Immunoblotting , Interleukin-1/metabolism , Ligases/metabolism , Luciferases/metabolism , Mice , Mice, Knockout , Microscopy, Fluorescence , Mutation , NF-KappaB Inhibitor alpha , Nuclear Proteins/metabolism , Phenotype , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Mas , Rabbits , Reticulocytes/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcriptional Activation , Transfection
10.
J Virol ; 75(4): 1736-43, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160671

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) Tat protein has been reported to transactivate several cellular genes, including the potent chemotactic factor interleukin-8 (IL-8). Consistent with these in vitro assays, elevated levels of IL-8 protein are found in the serum of HIV-infected individuals. We now extend these observations by demonstrating that Tat induction of IL-8 is linked to the cell cycle. Cells that constitutively express the Tat(1-86) protein (eTat) and control cells (pCEP) were reversibly blocked at the G(1)/S border with hydroxyurea or thymidine. The cells were subsequently released, and IL-8 expression was monitored by RNase protection assays and enzyme-linked immunosorbent assay (ELISA). RNase protection assays demonstrated that IL-8 mRNA expression is transiently induced, approximately fourfold, as the Tat-expressing cells enter S phase. Consistent with the RNase protection assay, an increase in IL-8 protein was observed in the cell supernatant using an IL-8 ELISA. Similar experiments were performed following a reversible block at the G(2)/M border with nocodazole and release into G(1). Using the RNase protection assay and ELISA, little or no increase in IL-8 expression was observed during G(1). Using gel shift as well as an immobilized DNA binding assay, we demonstrate that the increase in IL-8 gene expression correlates with a specific increase in p65 NF-kappa B binding activity only in the nucleus of the Tat-expressing cells. Moreover, the CREB-binding protein coactivator is present in the complex in the Tat cell line. Finally, we demonstrate that the presence of the proteasome inhibitor MG-132 inhibits the induction of NF-kappa B binding, as well as IL-8 expression, supporting the role of NF-kappa B.


Subject(s)
Cell Cycle/physiology , Gene Expression Regulation , Gene Products, tat/metabolism , HIV-1/metabolism , Interleukin-8/biosynthesis , Interleukin-8/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Gene Products, tat/genetics , HIV-1/genetics , HeLa Cells , Humans , Hydroxyurea/pharmacology , Leupeptins/pharmacology , NF-kappa B/metabolism , Promoter Regions, Genetic , S Phase , Transcription, Genetic , Transfection , tat Gene Products, Human Immunodeficiency Virus
11.
J Biol Chem ; 276(1): 200-5, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11036071

ABSTRACT

p53 plays a pivotal role in transmitting signals from many forms of genotoxic stress to genes and factors that control the cell cycle and apoptosis. We have previously shown that the human T-lymphotropic virus type I Tax protein can inhibit p53 function. Recently we reported that Tax inhibits p53 function in Jurkat cells and mouse embryo fibroblasts through a mechanism involving the nuclear factor kappa B pathway and correlates with phosphorylation on serines 15 and 392 of p53. However, several groups have also observed a mechanism that correlates with p300 binding of Tax. To address this controversy and to determine the mechanism by which Tax inhibits p53 function, we examined the activation functions of Tax required for p53 inhibition. In HeLa and H1299 cells the cAMP-response element-binding protein/activating transcription factor activation function is essential, as demonstrated by the Tax mutants M47 and K88A. In addition, expression of exogenous p300 in H1299 cells allows full recovery of p53 transactivation in the presence of Tax. Consistent with p300 being a limiting factor in H1299, Saos-2, and HeLa cells, we found that the level of endogenous p300 is relatively low in these cells compared with Jurkat cells or the human T-lymphotropic virus type I-infected C81 and MT2 cells. Thus our data suggests that Tax utilizes distinct mechanisms to inhibit p53 function that are cell type-dependent.


Subject(s)
DNA-Binding Proteins , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/physiology , Signal Transduction , Tumor Suppressor Protein p53/antagonists & inhibitors , Acetyltransferases/genetics , Acetyltransferases/metabolism , Activating Transcription Factor 1 , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Products, tax/genetics , Genes, Reporter , Histone Acetyltransferases , Humans , Lymphocytes/metabolism , Lymphocytes/virology , NF-kappa B/metabolism , Organ Specificity , Phosphorylation , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , p300-CBP Transcription Factors
12.
J Clin Microbiol ; 38(11): 4049-57, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11060067

ABSTRACT

To gain insight on the significance of human T-cell lymphotropic virus type 1 (HTLV-1) indeterminate serological reactivities, we studied villagers of South Cameroon, focusing on a frequent and specific HTLV-1 Gag indeterminate profile (HGIP) pattern (gag p19, p26, p28, and p30 without p24 or Env gp21 and gp46). Among the 102 sera studied, 29 from all age groups had a stable HGIP pattern over a period of 4 years. There was no epidemiological evidence for sexual or vertical transmission of HGIP. Seventy-five percent of HGIP sera reacted positively on MT2 HTLV-1-infected cells by immunofluorescence assay. However, we could not isolate any HTLV-1 virus or detect the presence of p19 Gag protein in cultures of peripheral blood mononuclear cells obtained from individuals with strong HGIP reactivity. PCR experiments conducted with primers for HTLV-1 and HTLV-2 (HTLV-1/2 primers) encompassing different regions of the virus did not yield HTLV-1/2 proviral sequences from individuals with HGIP. Using 11 peptides corresponding to HTLV-1 or HTLV-2 immunodominant B epitopes in an enzyme-linked immunosorbent assay, one epitope corresponding to the Gag p19 carboxyl terminus was identified in 75% of HGIP sera, while it was recognized by only 41% of confirmed HTLV-1-positive sera. A positive correlation between HTLV-1 optical density values and titers of antibody to Plasmodium falciparum was also demonstrated. Finally, passage of sera through a P. falciparum-infected erythrocyte-coupled column was shown to specifically abrogate HGIP reactivity but not the HTLV-1 pattern, suggesting the existence of cross-reactivity between HTLV-1 Gag proteins and malaria-derived antigens. These data suggest that in Central Africa, this frequent and specific Western blot is not caused by HTLV-1 infection but could instead be associated with P. falciparum infection.


Subject(s)
Deltaretrovirus Antibodies/blood , Gene Products, gag/immunology , HTLV-I Infections/immunology , Human T-lymphotropic virus 1/immunology , Malaria, Falciparum/immunology , Adult , Amino Acid Sequence , Animals , Antibodies, Protozoan/blood , Blotting, Western , Cameroon/epidemiology , Child , Cross Reactions , DNA, Viral/blood , Deltaretrovirus Antigens/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Gene Products, gag/chemistry , HTLV-I Infections/epidemiology , HTLV-I Infections/virology , HTLV-II Infections/immunology , HTLV-II Infections/virology , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/isolation & purification , Human T-lymphotropic virus 1/metabolism , Human T-lymphotropic virus 2/genetics , Human T-lymphotropic virus 2/immunology , Human T-lymphotropic virus 2/isolation & purification , Human T-lymphotropic virus 2/metabolism , Humans , Malaria, Falciparum/parasitology , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/immunology , Plasmodium falciparum/immunology
13.
AIDS Res Hum Retroviruses ; 16(16): 1629-32, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11080802

ABSTRACT

The B-myb gene was identified on the basis of its homology with the protooncogene c-myb, homolog of the avian myeloblastosis virus (AMV) and avian leukemia virus (E26) transforming genes. Several studies using antisense constructs or antisense oligonucleotides as well as overexpression experiments suggest that B-Myb plays an important role in the transition from G(1) to S phase of the cell cycle and that B-Myb expression is cell cycle regulated. We have previously demonstrated that the human T cell lymphotropic virus type 1 (HTLV1) trans-activator Tax is able to repress transcription from c-myb promoter reporter constructs as well as from the endogenous c-myb promoter in human T cells and that this effect is mediated through inhibition of the c-Myb trans-activating functions. Here we report that both HTLV-1 as well as HTLV-2 Tax proteins inhibit c-Myb trans-activation in mouse embryo fibroblasts (MEFs). In addition to c-Myb, B-Myb expression is also markedly downregulated in HTLV-1-transformed cells at both RNA and protein levels. Furthermore, by using a Jurkat T cell line stably transfected with a tax gene driven by a cadmium-inducible promoter (JPX9), we were able to demonstrate that Tax directly represses the endogenous B-myb promoter in T cells. Because c-Myb and B-Myb have been involved in cell cycle progression, our results suggest that Tax, by repressing both c-Myb and B-Myb endogenous promoters, may bypass their requirement for cell cycle progression in HTLV-1-transformed T cells.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/genetics , Gene Products, tax/pharmacology , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 2/physiology , T-Lymphocytes/virology , Trans-Activators/genetics , Transcriptional Activation , Animals , Cell Line, Transformed , Cell Transformation, Viral , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Fibroblasts , Humans , Jurkat Cells , Mice , Promoter Regions, Genetic , Trans-Activators/metabolism
14.
AIDS Res Hum Retroviruses ; 16(16): 1669-75, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11080808

ABSTRACT

The p53 protein plays a pivotal role in transmitting signals from many forms of genotoxic stress to genes and factors that control aspects of the cell cycle and death. Although mutated in approximately 60% of all human cancers, only a minority of human T-lymphotropic virus type 1 (HTLV-1)-transformed cells carry p53 mutations. Nevertheless, the p53 protein in HTLV-1-transformed cells is functionally inactive. We have previously demonstrated that the HTLV-1 Tax protein can inhibit p53 trans-activation function. Tax does not accomplish this by directly binding to p53, but rather by a unique mechanism that includes constitutive phosphorylation of p53 at Ser-15 and Ser-392. Analysis of Tax mutants in lymphocytes demonstrates that Tax-induced p53 inhibition correlates with the ability of Tax to activate NF-kappaB, but not p300 binding or CREB trans-activation. Consistent with these results, expression of the I-kappaBalpha(S32,36A) mutant that blocks NF-kappaB activation blocks Tax-mediated p53 inhibition. We further demonstrate the importance of Tax activation of NF-kappaB in p53 inhibition, using p65 knockout (KO) mouse embryo fibroblasts (MEFs). In the absence of p65 Tax could not inhibit p53. Tax does activate IKKbeta in the p65 KO MEFs, indicating that prenuclear events of NF-kappaB activation are not sufficient for Tax-mediated p53 inhibition, but rather NF-kappaB transcriptional activation is critical. Importantly, using phosphospecific antibodies, we demonstrate that phosphorylation of p53 at Ser-15 and Ser-392 correlates with Tax-mediated inhibition. In addition, mutation of p53 at Ser-15 and Ser-392 to alanines renders p53 resistant to Tax inhibition. This report reviews p53 inhibition by Tax and presents our current model.


Subject(s)
Gene Products, tax/metabolism , Human T-lymphotropic virus 1/metabolism , Lymphocytes/virology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Transformed , Gene Products, tax/genetics , Human T-lymphotropic virus 1/genetics , Humans , Mice , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
15.
AIDS Res Hum Retroviruses ; 16(16): 1677-81, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11080809

ABSTRACT

Human T cell lymphotropic virus type II (HTLV-2) was originally isolated from a patient with a hairy T cell leukemia. It has been associated with rare cases of CD8(+) T lymphoproliferative disorders, and has a controversial role as a pathogen. The loss of p53 function, as a consequence of mutation or inactivation, increases the chances of genetic damage. Indeed, the importance of p53 as a tumor suppressor is evident from the fact that over 60% of all human cancers have a mutant or inactive p53. p53 status has been extensively studied in HTLV-1-infected cell lines. Interestingly, despite the fact that p53 mutations have been found in only a minority of cells, the p53 functions were found to be impaired. We have analyzed the functional activity of the p53 tumor suppressor in cells transformed with HTLV-2 subtypes A and B. As with HTLV-1-infected cells, abundant levels of the p53 protein are detected in HTLV-2 virus-infected cell lines. Using p53 reporter plasmid or induction of p53-responsive genes in response to gamma-irradiation, the p53 was found to be transcriptionally inhibited in HTLV-2-infected cells. Interestingly, although Tax-2A and-2B inactivate p53, the Tax-2A protein appears to inhibit p53 function less efficiently than either Tax-1 or Tax-2B in T cells, but not in fibroblasts.


Subject(s)
Gene Products, tax/metabolism , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 2/physiology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Transformed , HTLV-I Infections/virology , HTLV-II Infections/virology , Humans , Mice , T-Lymphocytes/virology , Transcription, Genetic , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
16.
Bull Soc Pathol Exot ; 93(3): 163-71, 2000 Jul.
Article in French | MEDLINE | ID: mdl-11030050

ABSTRACT

Human T Cell leukemia/lymphoma virus type I, the first human oncogenic retrovirus, is the aetiological factor of Adult T cell leukemia (ATL), a CD4+ malignant lymphoproliferative disease and of a chronic neuromyelopathy, the tropical spastic paraparesis or HTLV-1 associated myelopathy (TSP/HAM). HTLV-1, which infects from 15 to 25 million individuals world-wide, is highly endemic in certain areas such as south-western Japan, Central Africa, the Caribbean basin and some regions of South America, Melanesia and of the middle East (for example the Mashhad area of Iran). The three major modes of transmission for HTLV-1 infection are perinatal, sexual and by blood transfusion. Recent molecular studies on HTLV-1 have shown the existence of several molecular subtypes (genotypes). These are related to the geographical origin of the infected populations and not to the associated diseases. The virus has a very high genetic stability. Viral amplification via clonal expansion of infected cells, rather than by use of reverse transcription could explain this remarkable phenomenon which can be used as a molecular tool for gaining new insights into the origin, evolution and modes of dissemination of HTLV-1. Analyses of HTLV-1 and STLV-1 (the simian counterpart) viral strains from throughout the world suggest that four events are responsible for this pattern of dissemination: 1) the transmission in the wild of STLV-1 between simian species, 2) the transmission of STLV-1 to humans as exemplified by the high percentage of identity between STLV-1 strains from chimpanzees or from mandrills with some HTLV-1 strains present in inhabitants of Central Africa, 3) persistence of HTLV-1 over a long period of time (by sexual and perinatal transmissions) in remote populations, as seen in the Australo-Melanesian region and 4) a global distribution of HTLV-1 via large scale human migrations, e.g., the slave trade from Africa to the New World.


Subject(s)
Deltaretrovirus Infections/epidemiology , Genetic Variation , HTLV-I Infections/epidemiology , Human T-lymphotropic virus 1/genetics , Simian T-lymphotropic virus 1/genetics , Animals , Biological Evolution , Deltaretrovirus Infections/transmission , Deltaretrovirus Infections/virology , HTLV-I Infections/transmission , HTLV-I Infections/virology , Human T-lymphotropic virus 1/classification , Humans , Pan troglodytes/virology , Papio/virology , Simian T-lymphotropic virus 1/classification
17.
Blood ; 96(1): 275-81, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10891462

ABSTRACT

Human T lymphotropic virus type I (HTLV-I) is the etiological agent of adult T-cell lymphocytic leukemia (ATLL), whereas HTLV-II has not been associated with hematopoietic malignancies. The control of apoptotic pathways has emerged as a critical step in the development of many cancer types. As a result, the underlying mechanism of long-term survival of HTLV-I and HTLV-II was studied in infected T cells in vitro and in ex vivo ATLL samples. Results indicate that HTLV-I- and HTLV-II-infected T cells in vitro express high levels of the antiapoptotic protein Bcl compared with other human leukemic T cell lines or uninfected peripheral blood mononuclear cells. The levels of proapoptotic proteins Bax, BAD, and Bak were not significantly altered. HTLV-I and HTLV-II viral transactivators, Tax1 and Tax2, are known to increase expression of cellular genes. These proteins were tested for increased transcription from the human Bcl2 and Bcl-X(L) promoters. Whereas no effect was observed on the Bcl2 promoter, both Tax1 and Tax2 increased transcription of the Bcl-X(L) promoter in T cells, although Tax1 appeared to be more efficient than Tax2. The biological significance of these observations was validated by the finding of an increased expression of Bcl-X(L) in ex vivo ATLL cells, especially from patients unresponsive to various chemotherapy regimens. Altogether, these data suggest that overexpression of Bcl-X(L )in vivo( )may be in part responsible for the resistance of ATLL cells to chemotherapy. In addition, inefficient activation of the Bcl-X(L) promoter by Tax2 may result in a shorter survival time of HTLV-II-infected cells in vivo and a diminished risk of leukemia development.


Subject(s)
Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 2/physiology , Leukemia-Lymphoma, Adult T-Cell/blood , Proto-Oncogene Proteins c-bcl-2/genetics , T-Lymphocytes/physiology , T-Lymphocytes/virology , Adult , Cell Line, Transformed , Gene Products, tax/genetics , Genes, bcl-2 , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 2/genetics , Humans , Jurkat Cells , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukocyte Count , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/blood , Recombinant Proteins/biosynthesis , Reference Values , T-Lymphocytes/immunology , Transcriptional Activation , Transfection , Tumor Cells, Cultured , bcl-X Protein
18.
J Virol ; 74(15): 6866-74, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10888626

ABSTRACT

We have analyzed the functional activity of the p53 tumor suppressor in human T-cell lymphotropic virus type 2 (HTLV-2)-transformed cells. Abundant levels of the p53 protein were detected in both HTLV-2A and -2B virus-infected cell lines. The p53 was functionally inactive, however, both in transient-transfection assays using a p53 reporter plasmid and in induction of p53-responsive genes in response to gamma irradiation. We further investigated HTLV-2A Tax and HTLV-2B Tax effects on p53 activity. Interestingly, although Tax-2A and -2B inactivate p53, the Tax-2A protein appears to inhibit p53 function less efficiently than either Tax-1 or Tax-2B. In transient-cotransfection assays, Tax-1 and Tax-2B inactivated p53 by 80%, while Tax2A reduced p53 activity by 20%. In addition, Tax-2A does not increase the steady-state level of cellular p53 as well as Tax-1 or -2B does in the same assays. Cotransfection assays demonstrated that Tax-2A could efficiently transactivate CREB-responsive promoters to the same level as Tax-1 and Tax-2B, indicating that the protein was functional. This report provides evidence of the first functional difference between the HTLV-2A and -2B subtypes. This comparison of the action of HTLV-1 and HTLV-2 Tax proteins on p53 function will provide important insights into the mechanism of HTLV transformation.


Subject(s)
Cell Transformation, Viral , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 2/physiology , Tumor Suppressor Protein p53/metabolism , Cell Line, Transformed , Gamma Rays , Gene Expression Regulation , Gene Products, tax/genetics , HTLV-II Infections/virology , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 2/genetics , Humans , Jurkat Cells , Phosphorylation , T-Lymphocytes/virology , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/radiation effects
19.
J Virol ; 74(16): 7270-83, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10906181

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1) is associated with adult T-cell leukemia (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). T-cell transformation is mainly due to the actions of the viral phosphoprotein Tax. Tax interacts with multiple transcriptional factors, aiding the transcription of many cellular genes. Here, we report that the cyclin-dependent kinase inhibitor p21/waf1 is overexpressed in all HTLV-1-infected cell lines tested as well as in ATL and HAM/TSP patient samples. Tax was found to be able to transactivate the endogenous p21/waf1 promoter, as detected by RNase protection, as well as activate a series of wild-type and 5'-deletion constructs linked to a luciferase reporter cassette. Wild-type but not a mutant form of Tax (M47) transactivated the p21/waf1 promoter in a p53-independent manner and utilized a minimal promoter that contained E2A and TATA box sequences. The p21/waf1 protein was reproducibly observed to be complexed with cyclin A/cdk2 and not with any other known G(1), S, or G(2)/M cyclins. Functionally, the association of p21/cyclin A/cdk2 decreased histone H1 phosphorylation in vitro, as observed in immunoprecipitations followed by kinase assays, and affected other substrates, such as the C terminus of Rb protein involved in c-Abl and histone deacetylase-1 (HDAC1) regulation. Interestingly, upon the use of a stress signal, such as gamma-irradiation, we found that the p21/cyclin A/cdk2 complex was able to block all known phosphorylation sites on the Rb molecule. Finally, using elutriated cell cycle fractions and a stress signal, we observed that the HTLV-1-infected T cells containing wild-type Tax, which had been in early or mid-G(1) phase prior to gamma-irradiation, arrested in G(1) and did not undergo apoptosis. This may be an important mechanism for an oncogenic virus such as HTLV-1 to stop the host at the G(1)/S boundary and to repair the damaged DNA upon injury, prior to S-phase entry.


Subject(s)
CDC2-CDC28 Kinases , Cyclin A/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Human T-lymphotropic virus 1/metabolism , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/virology , Animals , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Gamma Rays , Gene Products, tax/metabolism , HTLV-I Infections/virology , Humans , Leukemia-Lymphoma, Adult T-Cell/virology , Mice , Paraparesis, Tropical Spastic/virology , Phosphorylation , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured
20.
Oncogene ; 19(17): 2155-64, 2000 Apr 20.
Article in English | MEDLINE | ID: mdl-10815807

ABSTRACT

The c-Myb proto-oncogene is preferentially expressed in hematopoietic lineages, and highly expressed in several leukemia types. The Human T-cell Leukemia Virus Type I (HTLV-I) is the etiological agent of Adult T-cell Leukemia/Lymphoma (ATLL). A previous report suggested that Tax, the viral transactivator, is able to suppress the transactivation potential of c-Myb protein by competing for recruitment of CBP. We tested whether such a competition could affect transcription from the c-Myb promoter in Tax expressing T-cells. Using several c-Myb promoter reporter constructs carrying mutations in various regions, we demonstrate that Tax suppression of c-Myb transactivation results in transrepression of the c-Myb promoter through the Myb responsive elements in Jurkat T-cells. The ability of Tax mutants M22, M47 and V89A to interact with the full-length CBP and p300 proteins in vitro, and their ability to repress the c-Myb promoter, was then evaluated. Although both M47 and M22 bind to CBP and p300 to a similar extent, only M47 was able to repress the c-Myb promoter, suggesting that competition for CBP/p300 binding was not the mechanism underlying Tax's effect. This concept was further supported by the fact that the Tax mutant V89A transrepresses the c-Myb promoter efficiently in spite of an impaired binding to CBP and p300. Therefore, Tax-mediated repression of the c-Myb promoter appears to be independent from a direct competition between c-Myb and Tax for recruitment of CBP/p300. Interestingly, a decreased transcription from the endogenous c-Myb promoter was observed in several HTLV-I transformed T-cell lines. Finally, the ability of Tax to directly repress the endogenous c-Myb promoter was demonstrated in a Jurkat cell line stably transfected with a tax gene driven by a cadmium-inducible promoter.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/chemistry , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myb/genetics , Trans-Activators/metabolism , CREB-Binding Protein , Cell Line, Transformed , Cell Transformation, Viral , Gene Products, tax/genetics , Humans , Jurkat Cells/pathology , Jurkat Cells/virology , Mutation , Promoter Regions, Genetic , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myb/metabolism , Response Elements , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...