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










Publication year range
1.
Pathogens ; 12(6)2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37375521

ABSTRACT

Infection with human T-cell leukemia virus type 1 (HTLV-1) can produce a spectrum of pathological effects ranging from inflammatory disorders to leukemia. In vivo, HTLV-1 predominantly infects CD4+ T-cells. Infectious spread within this population involves the transfer of HTLV-1 virus particles from infected cells to target cells only upon cell-to-cell contact. The viral protein, HBZ, was found to enhance HTLV-1 infection through transcriptional activation of ICAM1 and MYOF, two genes that facilitate viral infection. In this study, we found that HBZ upregulates the transcription of COL4A1, GEM, and NRP1. COL4A1 and GEM are genes involved in viral infection, while NRP1, which encodes neuropilin 1 (Nrp1), serves as an HTLV-1 receptor on target cells but has no reported function on HTLV-1-infected cells. With a focus on Nrp1, cumulative results from chromatin immunoprecipitation assays and analyses of HBZ mutants support a model in which HBZ upregulates NRP1 transcription by augmenting recruitment of Jun proteins to an enhancer downstream of the gene. Results from in vitro infection assays demonstrate that Nrp1 expressed on HTLV-1-infected cells inhibits viral infection. Nrp1 was found to be incorporated into HTLV-1 virions, and deletion of its ectodomain removed the inhibitory effect. These results suggest that inhibition of HTLV-1 infection by Nrp1 is caused by the ectodomain of Nrp1 extended from virus particles, which may inhibit the binding of virus particles to target cells. While HBZ has been found to enhance HTLV-1 infection using cell-based models, there may be certain circumstances in which activation of Nrp1 expression negatively impacts viral infection, which is discussed.

2.
PLoS Pathog ; 19(2): e1011202, 2023 02.
Article in English | MEDLINE | ID: mdl-36827461

ABSTRACT

The complex retrovirus, human T-cell leukemia virus type 1 (HTLV-1), primarily infects CD4+ T-cells in vivo. Infectious spread within this cell population requires direct contact between virally-infected and target cells. The HTLV-1 accessory protein, HBZ, was recently shown to enhance HTLV-1 infection by activating intracellular adhesion molecule 1 (ICAM-1) expression, which promotes binding of infected cells to target cells and facilitates formation of a virological synapse. In this study we show that HBZ additionally enhances HTLV-1 infection by activating expression of myoferlin (MyoF), which functions in membrane fusion and repair and vesicle transport. Results from ChIP assays and quantitative reverse transcriptase PCR indicate that HBZ forms a complex with c-Jun or JunB at two enhancer sites within the MYOF gene and activates transcription through recruitment of the coactivator p300/CBP. In HTLV-1-infected T-cells, specific inhibition of MyoF using the drug, WJ460, or shRNA-mediated knockdown of MyoF reduced infection efficiency. This effect was associated with a decrease in cell adhesion and an intracellular reduction in the abundance of HTLV-1 envelope (Env) surface unit (SU) and transmembrane domain (TM). Lysosomal protease inhibitors partially restored SU levels in WJ460-treated cells, and SU localization to LAMP-2 sites was increased by MyoF knockdown, suggesting that MyoF restricts SU trafficking to lysosomes for degradation. Consistent with these effects, less SU was associated with cell-free virus particles. Together, these data suggest that MyoF contributes to HTLV-1 infection through modulation of Env trafficking and cell adhesion.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Human T-lymphotropic virus 1 , Retroviridae Proteins , Humans , Basic-Leucine Zipper Transcription Factors/genetics , CD4-Positive T-Lymphocytes/metabolism , Human T-lymphotropic virus 1/pathogenicity , Human T-lymphotropic virus 1/physiology , Retroviridae Proteins/metabolism
3.
Virology ; 549: 51-58, 2020 10.
Article in English | MEDLINE | ID: mdl-32841759

ABSTRACT

HBZ is expressed by the complex retrovirus, Human T-cell Leukemia Virus type 1, and implicated in pathological effects associated with viral infection. From the nucleus, HBZ alters gene expression by interacting with a variety of transcriptional regulatory proteins, among which is c-Jun. Previously, one of the three HBZ variants, HBZUS, was reported to decrease c-Jun expression by promoting its degradation. Here we show that another variant, HBZS1, produces the opposite effect. In the presence of HBZS1, c-Jun expression increases due to its stabilization. Our data suggest that this effect requires the ability of HBZS1 to interact with c-Jun. We provide evidence that HBZS1 inhibits the proteosomal degradation of c-Jun initiated by the Cop1-containing ubiquitin ligase complex. HBZS1 is the most abundant variant in HTLV-1-infected T-cells, and our data indicate that levels of c-Jun expression in infected cells are consistent with effects of HBZS1.


Subject(s)
Alternative Splicing , Basic-Leucine Zipper Transcription Factors/genetics , Host-Pathogen Interactions/genetics , Human T-lymphotropic virus 1/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Retroviridae Proteins/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/virology , Cullin Proteins/genetics , Cullin Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Human T-lymphotropic virus 1/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Jurkat Cells , Primary Cell Culture , Protein Binding , Protein Stability , Proteolysis , Retroviridae Proteins/metabolism , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
4.
J Virol ; 93(19)2019 10 01.
Article in English | MEDLINE | ID: mdl-31315993

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) causes multiple pathological effects, ranging from a form of leukemia to a spectrum of inflammation-mediated diseases. These diseases arise from one or several infected CD4+ T cells among thousands acquiring proliferation and survival advantages and ultimately becoming pathogenic. Given the low incidence of HTLV-1-associated diseases among carriers, such cellular evolutionary processes appear to occur rarely. Therefore, infectious spread of HTLV-1 within the T-cell population may be one underlying factor influencing disease development. Free HTLV-1 virions are poorly infectious, so infection of T cells relies on direct contact between infected and target cells. Following contact, virions pass to target cells through a virological synapse or cellular conduits or are transferred to target cells within an extracellular matrix. Lymphocyte functioning antigen 1 (LFA-1) on the surface of the target cell engaging with its ligand, ICAM-1, on the surface of the infected cell (effector cell) initiates and stabilizes cell-cell contact for infection. We found that stable expression of an HTLV-1 accessory protein, HTLV-1 bZIP factor (HBZ), in Jurkat T cells increases homotypic aggregation. This phenotype was attributed to elevated ICAM-1 expression in the presence of HBZ. Using a single-cycle replication-dependent luciferase assay, we found that HBZ expression in Jurkat cells (used as effector cells) increases HTLV-1 infection. Despite this effect, HBZ could not replace the critical infection-related functions of the HTLV-1 regulatory protein Tax. However, in HTLV-1-infected T cells, knockdown of HBZ expression did lead to a decrease in infection efficiency. These overall results suggest that HBZ contributes to HTLV-1 infectivity.IMPORTANCE Human T-cell leukemia virus type 1 (HTLV-1) causes a variety of diseases, ranging from a fatal form of leukemia to immune-mediated inflammatory diseases. These diseases occur rarely, arising from one or a small subset of virally infected cells infrequently evolving into a pathogenic state. Thus, the process of HTLV-1 cell-to-cell transmission within the host helps influence the probability of disease development. HTLV-1 primarily infects T cells and initially spreads within this cell population when virally infected T cells dock to uninfected target T cells and then transfer HTLV-1 virus particles to the target cells. Here we found that the viral protein HTLV-1 bZIP factor (HBZ) promotes infectivity. HBZ accomplishes this task by increasing the surface abundance of a cellular adhesion protein known as intercellular adhesion molecule 1 (ICAM-1), which helps initiate and stabilize contact (docking) between infected and target T cells. These results define a novel and unexpected function of HBZ, diverging from its defined functions in cellular survival and proliferation.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Host-Pathogen Interactions , Human T-lymphotropic virus 1/growth & development , Intercellular Adhesion Molecule-1/biosynthesis , Retroviridae Proteins/metabolism , T-Lymphocytes/virology , Up-Regulation , Cell Aggregation , Humans , Jurkat Cells
5.
PLoS Pathog ; 15(6): e1007922, 2019 06.
Article in English | MEDLINE | ID: mdl-31251786

ABSTRACT

Adult T-cell Leukemia (ATL) is a lymphoproliferative disease of CD4+ T-cells infected with Human T-cell Leukemia Virus type I (HTLV-1). With the exception of allogeneic hematopoietic stem cell transplantation, there are no effective treatments to cure ATL, and ATL cells often acquire resistance to conventional chemotherapeutic agents. Accumulating evidence shows that development and maintenance of ATL requires key contributions from the viral protein, HTLV-1 basic leucine zipper factor (HBZ). In this study we found that HBZ activates expression of Heme Oxygenase 1 (HMOX-1), a component of the oxidative stress response that functions to detoxify free heme. Transcription of HMOX1 and other antioxidant genes is regulated by the small Mafs. These cellular basic leucine zipper (bZIP) factors control transcription by forming homo- or heterodimers among themselves or with other cellular bZIP factors that then bind Maf responsive elements (MAREs) in promoters or enhancers of antioxidant genes. Our data support a model in which HBZ activates HMOX1 transcription by forming heterodimers with the small Mafs that bind MAREs located in an upstream enhancer region. Consistent with this model, we found that HMOX-1 is upregulated in HTLV-1-transformed T-cell lines and confers these cells with resistance to heme-induced cytotoxicity. In this context, HBZ-mediated activation of HMOX-1 expression may contribute to resistance of ATL cells to certain chemotherapeutic agents. We also provide evidence that HBZ counteracts oxidative stress caused by two other HTLV-1-encoded proteins, Tax and p13. Tax induces oxidative stress as a byproduct of driving mitotic expansion of infected cells, and p13 is believed to induce oxidative stress to eliminate infected cells that have become transformed. Therefore, in this context, HBZ-mediated activation of HMOX-1 expression may facilitate transformation. Overall, this study characterizes a novel function of HBZ that may support the development and maintenance of ATL.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Transformation, Viral , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/biosynthesis , Human T-lymphotropic virus 1/metabolism , Leukemia-Lymphoma, Adult T-Cell/metabolism , Oxidative Stress , Retroviridae Proteins/metabolism , Up-Regulation , Basic-Leucine Zipper Transcription Factors/genetics , Female , Gene Products, tax/genetics , Gene Products, tax/metabolism , HEK293 Cells , HeLa Cells , Heme Oxygenase-1/genetics , Human T-lymphotropic virus 1/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Male , Retroviridae Proteins/genetics , Transcription, Genetic
6.
J Virol ; 92(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29769340

ABSTRACT

Adult T-cell leukemia (ATL) is a fatal malignancy of CD4+ T cells infected with human T-cell leukemia virus type 1 (HTLV-1). ATL cells often exhibit random gross chromosomal rearrangements that are associated with the induction and improper repair of double-stranded DNA breaks (DSBs). The viral oncoprotein Tax has been reported to impair DSB repair but has not been shown to be consistently expressed throughout all phases of infection. The viral oncoprotein HTLV-1 basic leucine zipper (bZIP) factor (HBZ) is consistently expressed prior to and throughout disease progression, but it is unclear whether it also influences DSB repair. We report that HBZ attenuates DSB repair by nonhomologous end joining (NHEJ), in a manner dependent upon the bZIP domain. HBZ was found to interact with two vital members of the NHEJ core machinery, Ku70 and Ku80, and to be recruited to DSBs in a bZIP-dependent manner in vitro We observed that HBZ expression also resulted in a bZIP-dependent delay in DNA protein kinase (DNA-PK) activation following treatment with etoposide. Although Tax is reported to interact with Ku70, we did not find Tax expression to interfere with HBZ:Ku complex formation. However, as Tax was reported to saturate NHEJ, we found that this effect masked the attenuation of NHEJ by HBZ. Overall, these data suggest that DSB repair mechanisms are impaired not only by Tax but also by HBZ and show that HBZ expression may significantly contribute to the accumulation of chromosomal abnormalities during HTLV-1-mediated oncogenesis.IMPORTANCE Human T-cell leukemia virus type 1 (HTLV-1) infects 15 million to 20 million people worldwide. Approximately 90% of infected individuals are asymptomatic and may remain undiagnosed, increasing the risk that they will unknowingly transmit the virus. About 5% of the HTLV-1-positive population develop adult T-cell leukemia (ATL), a fatal disease that is not highly responsive to treatment. Although ATL development remains poorly understood, two viral proteins, Tax and HBZ, have been implicated in driving disease progression by manipulating host cell signaling and transcriptional pathways. Unlike Tax, HBZ expression is consistently observed in all infected individuals, making it important to elucidate the specific role of HBZ in disease progression. Here, we present evidence that HBZ could promote the accumulation of double-stranded DNA breaks (DSBs) through the attenuation of the nonhomologous end joining (NHEJ) repair pathway. This effect may lead to genome instability, ultimately contributing to the development of ATL.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Transformation, Viral , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/metabolism , Retroviridae Proteins/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Gene Products, tax/genetics , HEK293 Cells , HeLa Cells , Human T-lymphotropic virus 1/genetics , Humans , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Retroviridae Proteins/genetics
7.
J Virol ; 90(7): 3600-10, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26792732

ABSTRACT

UNLABELLED: Human T-cell leukemia virus type 1 (HTLV-1) is a retrovirus, and, as such, its genome becomes chromosomally integrated following infection. The resulting provirus contains identical 5' and 3' peripheral long terminal repeats (LTRs) containing bidirectional promoters. Antisense transcription from the 3' LTR regulates expression of a single gene, hbz, while sense transcription from the 5' LTR controls expression of all other viral genes, including tax. Both the HBZ and Tax proteins are implicated in the development of adult T-cell leukemia (ATL), a T-cell malignancy caused by HTLV-1 infection. However, these proteins appear to harbor opposing molecular functions, indicating that they may act independently and at different time points prior to leukemogenesis. Here, we used bidirectional reporter constructs to test whether transcriptional interference serves as a mechanism that inhibits simultaneous expression of Tax and HBZ. We found that sense transcription did not interfere with antisense transcription from the 3' LTR and vice versa, even with strong transcription emanating from the opposing direction. Therefore, bidirectional transcription across the provirus might not restrict hbz or tax expression. Single-cell analyses revealed that antisense transcription predominates in the absence of Tax, which transactivates viral sense transcription. Interestingly, a population of Tax-expressing cells exhibited antisense but not activated sense transcription. Consistent with the ability of Tax to induce cell cycle arrest, this population was arrested in G(0)/G(1) phase. These results imply that cell cycle arrest inhibits Tax-mediated activation of sense transcription without affecting antisense transcription, which may be important for long-term viral latency. IMPORTANCE: The chromosomally integrated form of the retrovirus human T-cell leukemia virus type 1 (HTLV-1) contains identical DNA sequences, known as long terminal repeats (LTRs), at its 5' and 3' ends. The LTRs modulate transcription in both forward (sense) and reverse (antisense) directions. We found that sense transcription from the 5' LTR does not interfere with antisense transcription from the 3' LTR, allowing viral genes encoded on opposite DNA strands to be simultaneously transcribed. Two such genes are tax and hbz, and while they are thought to function at different times during the course of infection to promote leukemogenesis of infected T cells, our results indicate that they can be simultaneously transcribed. We also found that the ability of Tax to induce cell cycle arrest inhibits its fundamental function of activating viral sense transcription but does not affect antisense transcription. This regulatory mechanism may be important for long-term HTLV-1 infection.


Subject(s)
Human T-lymphotropic virus 1/genetics , Terminal Repeat Sequences , Transcription, Genetic , Cell Cycle , Cell Line , Gene Expression Profiling , Genes, Reporter , Humans , Promoter Regions, Genetic , Virus Replication
8.
Oncotarget ; 7(2): 1687-706, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26625199

ABSTRACT

Adult T-cell leukemia (ATL) is an often fatal malignancy caused by infection with the complex retrovirus, human T-cell Leukemia Virus, type 1 (HTLV-1). In ATL patient samples, the tumor suppressor, p53, is infrequently mutated; however, it has been shown to be inactivated by the viral protein, Tax. Here, we show that another HTLV-1 protein, HBZ, represses p53 activity. In HCT116 p53+/+ cells treated with the DNA-damaging agent, etoposide, HBZ reduced p53-mediated activation of p21/CDKN1A and GADD45A expression, which was associated with a delay in G2 phase-arrest. These effects were attributed to direct inhibition of the histone acetyltransferase (HAT) activity of p300/CBP by HBZ, causing a reduction in p53 acetylation, which has be linked to decreased p53 activity. In addition, HBZ bound to, and inhibited the HAT activity of HBO1. Although HBO1 did not acetylate p53, it acted as a coactivator for p53 at the p21/CDKN1A promoter. Therefore, through interactions with two separate HAT proteins, HBZ impairs the ability of p53 to activate transcription. This mechanism may explain how p53 activity is restricted in ATL cells that do not express Tax due to modifications of the HTLV-1 provirus, which accounts for a majority of patient samples.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Histone Acetyltransferases/metabolism , Retroviridae Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation/drug effects , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Etoposide/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Retroviridae Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics
9.
J Virol ; 88(22): 13482-94, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25210182

ABSTRACT

UNLABELLED: Brain-derived neurotrophic factor (BDNF) is a neurotrophin that promotes neuronal proliferation, survival, and plasticity. These effects occur through autocrine and paracrine signaling events initiated by interactions between secreted BDNF and its high-affinity receptor, TrkB. A BDNF/TrkB autocrine/paracrine signaling loop has additionally been implicated in augmenting the survival of cells representing several human cancers and is associated with poor patient prognosis. Adult T-cell leukemia (ATL) is a fatal malignancy caused by infection with the complex retrovirus human T-cell leukemia virus type 1 (HTLV-1). In this study, we found that the HTLV-1-encoded protein HBZ activates expression of BDNF, and consistent with this effect, BDNF expression is elevated in HTLV-1-infected T-cell lines compared to uninfected T cells. Expression of TrkB is also higher in HTLV-1-infected T-cell lines than in uninfected T cells. Furthermore, levels of both BDNF and TrkB mRNAs are elevated in peripheral blood mononuclear cells (PBMCs) from ATL patients, and ATL patient sera contain higher concentrations of BDNF than sera from noninfected individuals. Finally, chemical inhibition of TrkB signaling increases apoptosis in HTLV-1-infected T cells and reduces phosphorylation of glycogen synthase kinase 3ß (GSK-3ß), a downstream target in the signaling pathway. These results suggest that HBZ contributes to an active BDNF/TrkB autocrine/paracrine signaling loop in HTLV-1-infected T cells that enhances the survival of these cells. IMPORTANCE: Infection with human T-cell leukemia virus type 1 (HTLV-1) can cause a rare form of leukemia designated adult T-cell leukemia (ATL). Because ATL patients are unresponsive to chemotherapy, this malignancy is fatal. As a retrovirus, HTLV-1 integrates its genome into a host cell chromosome in order to utilize host factors for replication and expression of viral proteins. However, in infected cells from ATL patients, the viral genome is frequently modified to block expression of all but a single viral protein. This protein, known as HBZ, is therefore believed to modulate cellular pathways necessary for the leukemic state and the chemotherapeutic resistance of the cell. Here we provide evidence to support this hypothesis. We found that HBZ promotes a BDNF/TrkB autocrine/paracrine signaling pathway that is known to enhance the survival and chemotherapeutic resistance of other types of cancer cells. It is possible that inhibition of this pathway may improve treatments for ATL.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cell Transformation, Viral , Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/virology , Viral Proteins/metabolism , Cell Survival , Humans , Receptor, trkB , Retroviridae Proteins , T-Lymphocytes/physiology
10.
J Virol ; 86(17): 9070-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22696638

ABSTRACT

Infection with the human T-cell leukemia virus type 1 (HTLV-1) results in a variety of diseases including adult T-cell leukemia (ATL), a fatal malignancy characterized by the uncontrolled proliferation of virally infected CD4(+) T cells. The HTLV-1 basic leucine zipper factor (HBZ) is believed to contribute to development and maintenance of ATL. Unlike the other HTLV-1 genes, the hbz gene is encoded on the complementary strand of the provirus and therefore is not under direct control of the promoter within the 5' long terminal repeat (LTR) of the provirus. This promoter can undergo inactivating genetic or epigenetic changes during the course of ATL that eliminates expression of all viral genes except that of hbz. In contrast, repressive modifications are not known to occur on the hbz promoter located in the 3' LTR, and hbz expression has been consistently detected in all ATL patient samples. Although Sp1 regulates basal transcription from the HBZ promoter, other factors that activate transcription remain undefined. In this study, we used a proviral reporter construct deleted of the 5' LTR to show that HBZ upregulates its own expression through cooperation with JunD. Activation of antisense transcription was apparent in serum-deprived cells in which the level of JunD was elevated, and elimination of JunD expression by gene knockout or shRNA-mediated knockdown abrogated this effect. Activation through HBZ and JunD additionally required Sp1 binding at the hbz promoter. These data favor a model in which JunD is recruited to the promoter through Sp1, where it heterodimerizes with HBZ thereby enhancing its activity. Separately, hbz gene expression led to an increase in JunD abundance, and this effect correlated with emergence of features of transformed cells in immortalized fibroblasts. Overall, our results suggest that JunD represents a novel therapeutic target for the treatment of ATL.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation, Viral , Human T-lymphotropic virus 1/genetics , Leukemia-Lymphoma, Adult T-Cell/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA, Antisense/genetics , Terminal Repeat Sequences , Viral Proteins/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cell Line , Human T-lymphotropic virus 1/physiology , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/virology , Mice , Protein Binding , Proto-Oncogene Proteins c-jun/genetics , RNA, Antisense/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Up-Regulation , Viral Proteins/genetics
11.
Nucleic Acids Res ; 40(13): 5910-25, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22434882

ABSTRACT

The homologous cellular coactivators p300 and CBP contain intrinsic lysine acetyl transferase (termed HAT) activity. This activity is responsible for acetylation of several sites on the histones as well as modification of transcription factors. In a previous study, we found that HBZ, encoded by the Human T-cell Leukemia Virus type 1 (HTLV-1), binds to multiple domains of p300/CBP, including the HAT domain. In this study, we found that HBZ inhibits the HAT activity of p300/CBP through the bZIP domain of the viral protein. This effect correlated with a reduction of H3K18 acetylation, a specific target of p300/CBP, in cells expressing HBZ. Interestingly, lower levels of H3K18 acetylation were detected in HTLV-1 infected cells compared to non-infected cells. The inhibitory effect of HBZ was not limited to histones, as HBZ also inhibited acetylation of the NF-κB subunit, p65, and the tumor suppressor, p53. Recent studies reported that mutations in the HAT domain of p300/CBP that cause a defect in acetylation are found in certain types of leukemia. These observations suggest that inhibition of the HAT activity by HBZ is important for the development of adult T-cell leukemia associated with HTLV-1 infection.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Viral Proteins/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , Acetylation , Basic-Leucine Zipper Transcription Factors/chemistry , Cell Line , Curcumin/pharmacology , Enzyme Inhibitors/pharmacology , HeLa Cells , Histones/metabolism , Humans , Protein Interaction Domains and Motifs , Retroviridae Proteins , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Transcription Factors/metabolism , Viral Proteins/chemistry , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/metabolism
12.
Viruses ; 3(8): 1485-500, 2011 08.
Article in English | MEDLINE | ID: mdl-21994792

ABSTRACT

Adult T-cell leukemia (ATL) is one of the primary diseases caused by Human T-cell Leukemia Virus type 1 (HTLV-1) infection. The virally-encoded Tax protein is believed to initiate early events in the development of this disease, as it is able to promote immortalization of T-cells and transformation of other cell types. These processes may be aided by the ability of the viral protein to directly deregulate expression of specific cellular genes through interactions with numerous transcriptional regulators. To identify gene promoters where Tax is localized, we isolated Tax-DNA complexes from an HTLV-1-infected T-cell line through a chromatin immunoprecipitation (ChIP) assay and used the DNA to probe a CpG island microarray. A site within the RNASET2 gene was found to be occupied by Tax. Real-time PCR analysis confirmed this result, and transient expression of Tax in uninfected cells led to the recruitment of the viral protein to the promoter. This event correlated with a decrease in the level of RNase T2 mRNA and protein, suggesting that Tax represses expression of this gene. Loss of RNase T2 expression occurs in certain hematological malignancies and other forms of cancer, and RNase T2 was recently reported to function as a tumor suppressor. Consequently, a reduction in the level of RNase T2 by Tax may play a role in ATL development.


Subject(s)
Endoribonucleases/metabolism , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/genetics , Ribonucleases/metabolism , Tumor Suppressor Proteins/metabolism , Binding Sites , Cell Transformation, Viral , Chromatin Immunoprecipitation , CpG Islands , Endoribonucleases/genetics , Gene Expression Regulation, Leukemic , Gene Expression Regulation, Viral , Gene Products, tax/genetics , HeLa Cells , Host-Pathogen Interactions , Human T-lymphotropic virus 1/metabolism , Humans , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/virology , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribonucleases/genetics , Tumor Suppressor Proteins/genetics
13.
J Mol Biol ; 409(3): 384-98, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21497608

ABSTRACT

The complex retrovirus human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia. Deregulation of cellular transcription is thought to be an important step for T-cell transformation caused by viral infection. HTLV-1 basic leucine zipper factor (HBZ) is one of the viral proteins believed to be involved in this process, as it deregulates the expression of numerous cellular genes. In the context of the provirus, HBZ represses HTLV-1 transcription, in part, by binding to the homologous cellular coactivators p300 and CBP. These coactivators play a central role in transcriptional regulation. In this study, we determined that HBZ binds with high affinity to the KIX domain of p300/CBP. This domain contains two binding surfaces that are differentially targeted by multiple cellular factors. We show that two φXXφφ motifs in the activation domain of HBZ mediate binding to a single surface of the KIX domain, the mixed-lineage leukemia (MLL) binding surface. Formation of this interaction inhibits binding of MLL to the KIX domain while enhancing the binding of the transcription factor c-Myb to the opposite surface of KIX. Consequently, HBZ inhibits transcriptional activation mediated by MLL and enhances activation mediated by c-Myb. CREB, which binds the same surface of KIX as c-Myb, also exhibited an increase in activity through HBZ. These results indicate that HBZ is able to alter gene expression by competing with transcription factors for the occupancy of one surface of KIX while enhancing the binding of factors to the other surface.


Subject(s)
Basic-Leucine Zipper Transcription Factors/chemistry , Host-Derived Cellular Factors/chemistry , Protein Interaction Domains and Motifs , Viral Proteins/chemistry , p300-CBP Transcription Factors/chemistry , Amino Acid Sequence , Binding Sites , Gene Expression Regulation, Viral , Human T-lymphotropic virus 1/genetics , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myb/chemistry , Retroviridae Proteins , Transcriptional Activation
14.
Retrovirology ; 7: 61, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20653953

ABSTRACT

BACKGROUND: Human T-cell leukemia virus type 1 (HTLV-1) is the etiologic agent of adult T-cell leukemia, a malignancy characterized by uncontrolled proliferation of virally-infected CD4+ T-cells. Hypercalcemia and bone lesions due to osteoclast-mediated bone resorption are frequently associated with more aggressive forms of the disease. The HTLV-1 provirus contains a unique antisense gene that expresses HTLV-1 basic leucine zipper (bZIP) factor (HBZ). HBZ is localized to the nucleus where it regulates levels of transcription by binding to certain cellular transcriptional regulators. Among its protein targets, HBZ forms a stable complex with the homologous cellular coactivators, p300 and CBP, which is modulated through two N-terminal LXXLL motifs in the viral protein and the conserved KIX domain in the coactivators. RESULTS: To determine the effects of these interactions on transcription, we performed a preliminary microarray analysis, comparing levels of gene expression in cells with wild-type HBZ versus cells with HBZ mutated in its LXXLL motifs. DKK1, which encodes the secreted Wnt signaling inhibitor, Dickkopf-1 (Dkk1), was confirmed to be transcriptionally activated by HBZ, but not its mutant. Dkk1 plays a major role in the development of bone lesions caused by multiple myeloma. In parallel with the initial findings, activation of Dkk1 expression by HBZ was abrogated by siRNA-mediated knockdown of p300/CBP or by a truncated form of p300 containing the KIX domain. Among HTLV-1-infected T-cell lines tested, the detection of Dkk1 mRNA partially correlated with a threshold level of HBZ mRNA. In addition, an uninfected and an HTLV-1-infected T-cell line transfected with an HBZ expression vector exhibited de novo and increased DKK1 transcription, respectively. In contrast to HBZ, The HTLV-1 Tax protein repressed Dkk1 expression. CONCLUSIONS: These data indicate that HBZ activates Dkk1 expression through its interaction with p300/CBP. However, this effect is limited in HTLV-1-infected T-cell lines, which in part, may be due to suppression of Dkk1 expression by Tax. Consequently, the ability of HBZ to regulate expression of Dkk1 and possibly other cellular genes may only be significant during late stages of ATL, when Tax expression is repressed.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Human T-lymphotropic virus 1/pathogenicity , Intercellular Signaling Peptides and Proteins/biosynthesis , Viral Proteins/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Bone Resorption , Cell Line , Gene Products, tax/metabolism , Gene Silencing , Humans , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Retroviridae Proteins , T-Lymphocytes/virology , Viral Proteins/genetics , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/metabolism
15.
J Biol Chem ; 283(35): 23903-13, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18599479

ABSTRACT

Activation of human T cell leukemia virus type 1 (HTLV-1) transcription is established through the formation of protein complexes on the viral promoter that are essentially composed of the cellular basic leucine zipper (bZIP) transcription factor cAMP-response element-binding protein (CREB (or certain other members of the ATF/CREB family), the HTLV-1-encoded transactivator Tax, and the pleiotropic cellular coactivators p300/CBP. HTLV-1 bZIP factor (HBZ) is a protein encoded by HTLV-1 that contains a bZIP domain and functions to repress HTLV-1 transcription. HBZ has been shown to repress viral transcription by dimerizing with CREB, which occurs specifically through the bZIP domain in each protein, and preventing CREB from binding to the DNA. However, we previously found that HBZ causes only partial removal of CREB from a chromosomally integrated viral promoter, and more importantly, an HBZ mutant lacking the COOH-terminal bZIP domain retains the ability to repress viral transcription. These results suggest that an additional mechanism contributes to HBZ-mediated repression of HTLV-1 transcription. In this study, we show that HBZ binds directly to the p300 and CBP coactivators. Two LXXLL-like motifs located within the NH(2)-terminal region of HBZ are important for this interaction and specifically mediate binding to the KIX domain of p300/CBP. We provide evidence that this interaction interferes with the ability of Tax to bind p300/CBP and thereby inhibits the association of the coactivators with the viral promoter. Our findings demonstrate that HBZ utilizes a bipartite mechanism to repress viral transcription.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Down-Regulation/physiology , Gene Expression Regulation, Viral/physiology , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/physiology , Transcription, Genetic/physiology , Viral Proteins/metabolism , p300-CBP Transcription Factors/metabolism , Amino Acid Motifs/physiology , Basic-Leucine Zipper Transcription Factors/genetics , Cell Line , Dimerization , Gene Products, tax/genetics , Humans , Mutation , Promoter Regions, Genetic/physiology , Protein Structure, Tertiary/physiology , Retroviridae Proteins , Viral Proteins/genetics , Virus Activation/physiology , p300-CBP Transcription Factors/genetics
16.
J Virol ; 81(4): 1543-53, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17151132

ABSTRACT

The complex human T-cell leukemia virus type 1 (HTLV-1) retrovirus encodes several proteins that are unique to the virus within its 3'-end region. Among them, the viral transactivator Tax and posttranscriptional regulator Rex are well characterized, and both positively regulate HTLV-1 viral expression. Less is known about the other regulatory proteins encoded in this region of the provirus, including the recently discovered HBZ protein. HBZ has been shown to negatively regulate basal and Tax-dependent HTLV-1 transcription through its ability to interact with specific basic-leucine zipper (bZIP) proteins. In the present study, we found that HBZ reduces HTLV-1 transcription and virion production. We then characterized the interaction between HBZ and the cellular transcription factor CREB. CREB plays a critical role in Tax-mediated HTLV-1 transcription by forming a complex with Tax that binds to viral cyclic AMP-response elements (CREs) located within the viral promoter. We found that HBZ and CREB interact in vivo and directly in vitro, and this interaction occurs through the bZIP domain of each protein. We also found that CREM-Ia and ATF-1, which share significant homology in their bZIP domains with the bZIP domain of CREB, interact with HBZ-bZIP. The interaction between CREB and HBZ prevents CREB binding to the viral CRE elements in vitro and in vivo, suggesting that the reduction in HTLV-1 transcription by HBZ is partly due to the loss of CREB at the promoter. We also found that HBZ displaces CREB from a cellular CRE, suggesting that HBZ may deregulate CREB-dependent cellular gene expression.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Human T-lymphotropic virus 1/genetics , Viral Proteins/genetics , Animals , Basic-Leucine Zipper Transcription Factors/chemistry , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Line , Down-Regulation , Gene Expression Regulation, Viral , Gene Products, tax/metabolism , Humans , Promoter Regions, Genetic , Protein Structure, Tertiary , Transcription, Genetic , Viral Proteins/metabolism
17.
J Biol Chem ; 281(19): 13075-13082, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16547351

ABSTRACT

The human T-cell leukemia virus type 1 (HTLV-1) is integrated into the host cell DNA and assembled into nucleosomes. Within the repressive chromatin environment, the virally encoded Tax protein mediates the recruitment of the coactivators CREB-binding protein/p300 to the HTLV-1 promoter, located within the long terminal repeats (LTRs) of the provirus. These proteins carry acetyltransferase activity that is essential for strong transcriptional activation of the virus in the context of chromatin. Consistent with this, the amino-terminal tails of nucleosomal histones at the viral promoter are acetylated in Tax-expressing cells. We have developed a system in which we transfect Tax into cells carrying integrated copies of the HTLV-1 LTR driving the luciferase gene to analyze changes in "activating" histone modifications at the LTR. Unexpectedly, Tax transactivation led to an apparent reduction of these modifications at the HTLV-1 promoter and downstream region that correlates with a similar reduction in histone H3 and linker histone H1. Micrococcal nuclease protection analysis showed that less LTR-luciferase DNA is nucleosomal in Tax-expressing cells. Furthermore, nucleosome depletion correlated with RNA polymerase II recruitment and loss of SWI/SNF. The M47 Tax mutant, deficient in HTLV-1 transcriptional activation, was also defective for nucleosome depletion. Although this mutant formed complexes with CREB and p300 at the HTLV-1 promoter in vivo, it was unable to mediate RNA polymerase II recruitment or SWI/SNF displacement. These results support a model in which nucleosomes are depleted from the LTR and transcribed region during Tax-mediated transcriptional activation and correlate RNA polymerase II recruitment with nucleosome depletion.


Subject(s)
Gene Expression Regulation, Viral , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Nucleosomes/metabolism , Transcriptional Activation , Animals , CHO Cells , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cricetinae , Gene Products, tax/genetics , Histones/metabolism , Humans , Mutation , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Transcription Factors/metabolism
18.
Mol Cell Biol ; 24(14): 6117-26, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15226416

ABSTRACT

The human T-cell leukemia virus type 1 (HTLV-1) is a retrovirus that integrates randomly into the T-cell genome. Two long terminal repeats (LTRs) flank the integrated provirus. The upstream and downstream LTRs carry identical promoter sequences. Studies with other retroviruses suggest that the downstream promoter is silent and that RNA polymerases initiating at the upstream promoter proceed through the 3' LTR. In this study, we used the chromatin immunoprecipitation assay to compare the binding of transcription regulatory proteins at both the upstream and downstream promoters in HTLV-1-infected cell lines and adult T-cell leukemia-lymphoma cells. Unexpectedly, we detected a nearly equal distribution of activator (Tax, CREB, ATF-1, ATF-2, c-Fos, and c-Jun) and regulatory protein (CBP, p300, TAF(II)250, and polymerase II) binding at both the upstream and downstream promoters. Consistent with this observation, we found that the downstream promoter was transcriptionally active, suggesting that the two promoters are functionally equivalent. We also detected asymmetrical binding of histone deacetylases (HDAC-1, -2, and -3) at both promoters. All three HDACs strongly repressed Tax transactivation, and this repression correlated with displacement of Tax from the HTLV-1 promoter. These effects were reciprocal, as Tax expression reversed HDAC repression and displaced HDACs from the HTLV-1 promoter. These data suggest that HTLV-1 transcriptional regulation at both the 5' and 3' LTRs is mediated, in part, through the mutually exclusive binding of Tax and HDACs at the proviral promoters.


Subject(s)
3' Flanking Region/genetics , 5' Flanking Region/genetics , Gene Expression Regulation, Viral , Human T-lymphotropic virus 1/genetics , Terminal Repeat Sequences , Transcription Factors/metabolism , Transcription, Genetic , Adult , Animals , Cell Line , Gene Products, tax/genetics , Gene Products, tax/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Human T-lymphotropic virus 1/metabolism , Humans , Macromolecular Substances , Promoter Regions, Genetic , Protein Binding , T-Lymphocytes/cytology , T-Lymphocytes/physiology
19.
J Biol Chem ; 277(51): 49459-65, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12386157

ABSTRACT

The human T-cell leukemia virus (HTLV-I)-encoded Tax protein is a potent transcriptional activator that stimulates expression of the integrated provirus. Biochemical studies indicate that Tax, together with cellular transcription factors, interacts with viral cAMP-response element enhancer elements to recruit the pleiotropic coactivators CREB-binding protein and p300. Histone acetylation by these coactivators has been shown to play a major role in activating HTLV-I transcription from chromatin templates in vitro. However, the extent of histone modification and the precise identity of the cellular regulatory proteins bound at the HTLV-I promoter in vivo is not known. Chromatin immunoprecipitation analysis was used to investigate factor binding and histone modification at the integrated HTLV-I provirus in infected T-cells (SLB-1). These studies reveal the presence of Tax, a variety of ATF/CREB and AP-1 family members (CREB, CREB-2, ATF-1, ATF-2, c-Fos, and c-Jun), and both p300 and CREB-binding protein at the HTLV-I promoter. Consistent with the binding of these coactivators, we observed histone H3 and H4 acetylation at three regions within the proviral genome. Histone deacetylases were also present at the viral promoter and, following their inhibition, we observe an increase in histone H4 acetylation on the HTLV-I promoter and a concomitant increase in viral RNA. Together, these results suggest that a variety of transcriptional activators, coactivators, and histone deacetylases participate in the regulation of HTLV-I transcription in infected T-cells.


Subject(s)
Genes, Viral , Human T-lymphotropic virus 1/metabolism , T-Lymphocytes/virology , Transcription Factors/metabolism , Acetylation , Chromatin/metabolism , Cyclic AMP/metabolism , Histones/metabolism , Humans , Models, Genetic , Nuclear Proteins/metabolism , Plasmids/metabolism , Polymerase Chain Reaction , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic
20.
Nucleic Acids Res ; 30(9): 1977-84, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11972335

ABSTRACT

TFIIIA is required to activate RNA polymerase III transcription from 5S RNA genes. Although all known TFIIIA homologs harbor nine zinc fingers that mediate DNA binding, very limited sequence homology is found among these proteins, which reflects unique properties of some TFIIIA homologs. For example, the Acanthamoeba castellanii homolog directly regulates 5S RNA transcription. We have purified and characterized A.castellanii TFIIIA (AcTFIIIA) as a step toward obtaining a clearer understanding of these differences and of the regulatory process. AcTFIIIA is 59 kDa, significantly larger than all other TFIIIA homologs isolated to date. Nevertheless, it exhibits a DNase I footprint very similar to those produced by the smaller vertebrate TFIIIA homologs, but distinct from the smaller footprint of the 51 kDa TFIIIA from Saccharomyces cerevisiae. Similar footprinting is not reflected in greater sequence similarity between the A.castellanii and vertebrate promoters.


Subject(s)
Acanthamoeba/metabolism , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Transcription Factors/isolation & purification , Transcription Factors/metabolism , Acanthamoeba/genetics , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Cells, Cultured , DNA Footprinting , DNA, Protozoan/metabolism , DNA-Binding Proteins/chemistry , Deoxyribonuclease I/chemistry , Peptides/chemistry , Promoter Regions, Genetic , RNA, Ribosomal, 5S/genetics , Transcription Factor TFIIIA , Transcription Factors/chemistry , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...