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










Publication year range
1.
PLoS Pathog ; 18(4): e1010416, 2022 04.
Article in English | MEDLINE | ID: mdl-35377924

ABSTRACT

We investigated the impact of monocytes, NK cells, and CD8+ T-cells in primary HTLV-1 infection by depleting cell subsets and exposing macaques to either HTLV-1 wild type (HTLV-1WT) or to the HTLV-1p12KO mutant unable to infect replete animals due to a single point mutation in orf-I that inhibits its expression. The orf-I encoded p8/p12 proteins counteract cytotoxic NK and CD8+ T-cells and favor viral DNA persistence in monocytes. Double NK and CD8+ T-cells or CD8 depletion alone accelerated seroconversion in all animals exposed to HTLV-1WT. In contrast, HTLV-1p12KO infectivity was fully restored only when NK cells were also depleted, demonstrating a critical role of NK cells in primary infection. Monocyte/macrophage depletion resulted in accelerated seroconversion in all animals exposed to HTLV-1WT, but antibody titers to the virus were low and not sustained. Seroconversion did not occur in most animals exposed to HTLV-1p12KO. In vitro experiments in human primary monocytes or THP-1 cells comparing HTLV-1WT and HTLV-1p12KO demonstrated that orf-I expression is associated with inhibition of inflammasome activation in primary cells, with increased CD47 "don't-eat-me" signal surface expression in virus infected cells and decreased monocyte engulfment of infected cells. Collectively, our data demonstrate a critical role for innate NK cells in primary infection and suggest a dual role of monocytes in primary infection. On one hand, orf-I expression increases the chances of viral transmission by sparing infected cells from efferocytosis, and on the other may protect the engulfed infected cells by modulating inflammasome activation. These data also suggest that, once infection is established, the stoichiometry of orf-I expression may contribute to the chronic inflammation observed in HTLV-1 infection by modulating monocyte efferocytosis.


Subject(s)
HTLV-I Infections , Human T-lymphotropic virus 1 , Animals , Inflammasomes/metabolism , Killer Cells, Natural , Monocytes
2.
PLoS Pathog ; 17(9): e1009919, 2021 09.
Article in English | MEDLINE | ID: mdl-34543356

ABSTRACT

Viral infections are known to hijack the transcription and translation of the host cell. However, the extent to which viral proteins coordinate these perturbations remains unclear. Here we used a model system, the human T-cell leukemia virus type 1 (HTLV-1), and systematically analyzed the transcriptome and interactome of key effectors oncoviral proteins Tax and HBZ. We showed that Tax and HBZ target distinct but also common transcription factors. Unexpectedly, we also uncovered a large set of interactions with RNA-binding proteins, including the U2 auxiliary factor large subunit (U2AF2), a key cellular regulator of pre-mRNA splicing. We discovered that Tax and HBZ perturb the splicing landscape by altering cassette exons in opposing manners, with Tax inducing exon inclusion while HBZ induces exon exclusion. Among Tax- and HBZ-dependent splicing changes, we identify events that are also altered in Adult T cell leukemia/lymphoma (ATLL) samples from two independent patient cohorts, and in well-known cancer census genes. Our interactome mapping approach, applicable to other viral oncogenes, has identified spliceosome perturbation as a novel mechanism coordinated by Tax and HBZ to reprogram the transcriptome.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Gene Products, tax/metabolism , HTLV-I Infections/metabolism , Leukemia-Lymphoma, Adult T-Cell/virology , Retroviridae Proteins/metabolism , HEK293 Cells , HTLV-I Infections/etiology , Human T-lymphotropic virus 1 , Humans , Jurkat Cells , RNA Splicing , RNA, Messenger , Splicing Factor U2AF/metabolism
3.
Genome Biol ; 22(1): 97, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33823910

ABSTRACT

The integration of a viral genome into the host genome has a major impact on the trajectory of the infected cell. Integration location and variation within the associated viral genome can influence both clonal expansion and persistence of infected cells. Methods based on short-read sequencing can identify viral insertion sites, but the sequence of the viral genomes within remains unobserved. We develop PCIP-seq, a method that leverages long reads to identify insertion sites and sequence their associated viral genome. We apply the technique to exogenous retroviruses HTLV-1, BLV, and HIV-1, endogenous retroviruses, and human papillomavirus.


Subject(s)
Computational Biology/methods , Genome, Viral , Genomics/methods , Virus Integration , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , High-Throughput Nucleotide Sequencing , Humans , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Proviruses/genetics , Retroviridae/genetics
4.
Leukemia ; 35(3): 764-776, 2021 03.
Article in English | MEDLINE | ID: mdl-32555298

ABSTRACT

Adult T-cell leukemia/lymphoma (ATL) carries a poor prognosis even in indolent subtypes. We performed targeted deep sequencing combined with mapping of HTLV-1 proviral integration sites of 61 ATL patients of African and Caribbean origin. This revealed mutations mainly affecting TCR/NF-kB (74%), T-cell trafficking (46%), immune escape (29%), and cell cycle (26%) related pathways, consistent with the genomic landscape previously reported in a large Japanese cohort. To examine the evolution of mutational signatures upon disease progression while tracking the viral integration architecture of the malignant clone, we carried out a longitudinal study of patients who either relapsed or progressed from an indolent to an aggressive subtype. Serial analysis of relapsing patients identified several patterns of clonal evolution. In progressing patients, the longitudinal study revealed NF-kB/NFAT mutations at progression that were present at a subclonal level at diagnosis (allelic frequency < 5%). Moreover, the presence in indolent subtypes of mutations affecting the TCR/NF-kB pathway, whether clonal or subclonal, was associated with significantly shorter time to progression and overall survival. Our observations reveal the clonal dynamics of ATL mutational signatures at relapse and during progression. Our study defines a new subgroup of indolent ATLs characterized by a mutational signature at high risk of transformation.


Subject(s)
Biomarkers, Tumor/genetics , Clonal Evolution , High-Throughput Nucleotide Sequencing/methods , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Mutation , Adolescent , Adult , Aged , Disease Progression , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Young Adult
5.
Front Microbiol ; 11: 587306, 2020.
Article in English | MEDLINE | ID: mdl-33193242

ABSTRACT

The combined application of linear amplification-mediated PCR (LAM-PCR) protocols with next-generation sequencing (NGS) has had a large impact on our understanding of retroviral pathogenesis. Previously, considerable effort has been expended to optimize NGS methods to explore the genome-wide distribution of proviral integration sites and the clonal architecture of clinically important retroviruses like human T-cell leukemia virus type-1 (HTLV-1). Once sequencing data are generated, the application of rigorous bioinformatics analysis is central to the biological interpretation of the data. To better exploit the potential information available through these methods, we developed an optimized bioinformatics pipeline to analyze NGS clonality datasets. We found that short-read aligners, specifically designed to manage NGS datasets, provide increased speed, significantly reducing processing time and decreasing the computational burden. This is achieved while also accounting for sequencing base quality. We demonstrate the utility of an additional trimming step in the workflow, which adjusts for the number of reads supporting each insertion site. In addition, we developed a recall procedure to reduce bias associated with proviral integration within low complexity regions of the genome, providing a more accurate estimation of clone abundance. Finally, we recommend the application of a "clean-and-recover" step to clonality datasets generated from large cohorts and longitudinal studies. In summary, we report an optimized bioinformatics workflow for NGS clonality analysis and describe a new set of steps to guide the computational process. We demonstrate that the application of this protocol to the analysis of HTLV-1 and bovine leukemia virus (BLV) clonality datasets improves the quality of data processing and provides a more accurate definition of the clonal landscape in infected individuals. The optimized workflow and analysis recommendations can be implemented in the majority of bioinformatics pipelines developed to analyze LAM-PCR-based NGS clonality datasets.

6.
J Virol ; 93(19)2019 10 01.
Article in English | MEDLINE | ID: mdl-31315992

ABSTRACT

Human T cell leukemia virus type 1 (HTLV-1) is the ethological agent of adult T cell leukemia/lymphoma (ATLL) and a number of lymphocyte-mediated inflammatory conditions, including HTLV-1-associated myelopathy/tropical spastic paraparesis. HTLV-1 orf-I encodes two proteins, p8 and p12, whose functions in humans are to counteract innate and adaptive responses and to support viral transmission. However, the in vivo requirements for orf-I expression vary in different animal models. In macaques, the ablation of orf-I expression by mutation of its ATG initiation codon abolishes the infectivity of the molecular clone HTLV-1p12KO In rabbits, HTLV-1p12KO is infective and persists efficiently. We used humanized mouse models to assess the infectivity of both wild-type HTLV-1 (HTLV-1WT) and HTLV-1p12KO We found that NOD/SCID/γC-/- c-kit+ mice engrafted with human tissues 1 day after birth (designated NSG-1d mice) were highly susceptible to infection by HTLV-1WT, with a syndrome characterized by the rapid polyclonal proliferation and infiltration of CD4+ CD25+ T cells into vital organs, weight loss, and death. HTLV-1 clonality studies revealed the presence of multiple clones of low abundance, confirming the polyclonal expansion of HTLV-1-infected cells in vivo HTLV-1p12KO infection in a bone marrow-liver-thymus (BLT) mouse model prone to graft-versus-host disease occurred only following reversion of the orf-I initiation codon mutation within weeks after exposure and was associated with high levels of HTLV-1 DNA in blood and the expansion of CD4+ CD25+ T cells. Thus, the incomplete reconstitution of the human immune system in BLT mice may provide a window of opportunity for HTLV-1 replication and the selection of viral variants with greater fitness.IMPORTANCE Humanized mice constitute a useful model for studying the HTLV-1-associated polyclonal proliferation of CD4+ T cells and viral integration sites in the human genome. The rapid death of infected animals, however, appears to preclude the clonal selection typically observed in human ATLL, which normally develops in 2 to 5% of individuals infected with HTLV-1. Nevertheless, the expansion of multiple clones of low abundance in these humanized mice mirrors the early phase of HTLV-1 infection in humans, providing a useful model to investigate approaches to inhibit virus-induced CD4+ T cell proliferation.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Proliferation , HTLV-I Infections/pathology , HTLV-I Infections/virology , Host-Pathogen Interactions , Human T-lymphotropic virus 1/growth & development , Viral Regulatory and Accessory Proteins/metabolism , Animals , Disease Models, Animal , Disease Transmission, Infectious , Mice , Mice, Knockout , Mice, SCID , Viral Regulatory and Accessory Proteins/deficiency
7.
PLoS Pathog ; 14(3): e1006933, 2018 03.
Article in English | MEDLINE | ID: mdl-29566098

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia/lymphoma (ATLL), an aggressive malignant proliferation of activated CD4+ T lymphocytes. The viral Tax oncoprotein is critically involved in both HTLV-1-replication and T-cell proliferation, a prerequisite to the development of ATLL. In this study, we investigated the in vivo contribution of the Tax PDZ domain-binding motif (PBM) to the lymphoproliferative process. To that aim, we examined T-cell proliferation in humanized mice (hu-mice) carrying a human hemato-lymphoid system infected with either a wild type (WT) or a Tax PBM-deleted (ΔPBM) provirus. We observed that the frequency of CD4+ activated T-cells in the peripheral blood and in the spleen was significantly higher in WT than in ΔPBM hu-mice. Likewise, human T-cells collected from WT hu-mice and cultivated in vitro in presence of interleukin-2 were proliferating at a higher level than those from ΔPBM animals. We next examined the association of Tax with the Scribble PDZ protein, a prominent regulator of T-cell polarity, in human T-cells analyzed either after ex vivo isolation or after in vitro culture. We confirmed the interaction of Tax with Scribble only in T-cells from the WT hu-mice. This association correlated with the presence of both proteins in aggregates at the leading edge of the cells and with the formation of long actin filopods. Finally, data from a comparative genome-wide transcriptomic analysis suggested that the PBM-PDZ association is implicated in the expression of genes regulating proliferation, apoptosis and cytoskeletal organization. Collectively, our findings suggest that the Tax PBM is an auxiliary motif that contributes to the sustained growth of HTLV-1 infected T-cells in vivo and in vitro and is essential to T-cell immortalization.


Subject(s)
Cell Proliferation , Cell Transformation, Viral , Disease Models, Animal , Gene Products, tax/metabolism , HTLV-I Infections/pathology , Human T-lymphotropic virus 1/pathogenicity , T-Lymphocytes/pathology , Animals , Female , Gene Expression Profiling , Gene Products, tax/genetics , HEK293 Cells , HTLV-I Infections/metabolism , HTLV-I Infections/virology , Humans , Lymphocyte Activation , Male , Mice , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , PDZ Domains , Protein Binding , T-Lymphocytes/metabolism
8.
Nat Commun ; 8: 15890, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28639618

ABSTRACT

The human T-lymphotropic virus type 1 (HTLV-1) is efficiently transmitted through cellular contacts. While the molecular mechanisms of viral cell-to-cell propagation have been extensively studied in vitro, those facilitating the encounter between infected and target cells remain unknown. In this study, we demonstrate that HTLV-1-infected CD4 T cells secrete a potent chemoattractant, leukotriene B4 (LTB4). LTB4 secretion is dependent on Tax-induced transactivation of the pla2g4c gene, which encodes the cytosolic phospholipase A2 gamma. Inhibition of LTB4 secretion or LTB4 receptor knockdown on target cells reduces T-cell recruitment, cellular contact formation and virus propagation in vitro. Finally, blocking the synthesis of LTB4 in a humanized mouse model of HTLV-1 infection significantly reduces proviral load. This results from a decrease in the number of infected clones while their expansion is not impaired. This study shows the critical role of LTB4 secretion in HTLV-1 transmission both in vitro and in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Human T-lymphotropic virus 1/pathogenicity , Leukotriene B4/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Gene Products, tax/genetics , Gene Products, tax/metabolism , Group IV Phospholipases A2/genetics , Group IV Phospholipases A2/metabolism , HTLV-I Infections/drug therapy , HTLV-I Infections/metabolism , HTLV-I Infections/virology , Host-Pathogen Interactions , Humans , Indoles/pharmacology , Infant, Newborn , Jurkat Cells , Lipoxygenase Inhibitors/pharmacology , Male , Mice, Mutant Strains , NF-kappa B/genetics , NF-kappa B/metabolism
9.
Nat Commun ; 8: 15264, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28534499

ABSTRACT

Human T-cell leukaemia virus type-1 (HTLV-1) and bovine leukaemia virus (BLV) infect T- and B-lymphocytes, respectively, provoking a polyclonal expansion that will evolve into an aggressive monoclonal leukaemia in ∼5% of individuals following a protracted latency period. It is generally assumed that early oncogenic changes are largely dependent on virus-encoded products, especially TAX and HBZ, while progression to acute leukaemia/lymphoma involves somatic mutations, yet that both are independent of proviral integration site that has been found to be very variable between tumours. Here, we show that HTLV-1/BLV proviruses are integrated near cancer drivers which they affect either by provirus-dependent transcription termination or as a result of viral antisense RNA-dependent cis-perturbation. The same pattern is observed at polyclonal non-malignant stages, indicating that provirus-dependent host gene perturbation contributes to the initial selection of the multiple clones characterizing the asymptomatic stage, requiring additional alterations in the clone that will evolve into full-blown leukaemia/lymphoma.


Subject(s)
Carcinogenesis/genetics , Human T-lymphotropic virus 1/physiology , Leukemia Virus, Bovine/physiology , Leukemia/genetics , Leukemia/virology , Proviruses/physiology , Adult , Animals , Cattle , Female , Genome , Host-Pathogen Interactions/genetics , Human T-lymphotropic virus 1/genetics , Humans , Leukemia Virus, Bovine/genetics , Male , Models, Biological , Proviruses/genetics , RNA, Antisense/metabolism , Sheep , Transcription, Genetic , Virus Integration/genetics
10.
Retrovirology ; 13(1): 33, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27141823

ABSTRACT

BACKGROUND: Bovine Leukemia Virus (BLV) is a deltaretrovirus closely related to the Human T cell leukemia virus-1 (HTLV-1). Cattle are the natural host of BLV where it integrates into B-cells, producing a lifelong infection. Most infected animals remain asymptomatic but following a protracted latency period about 5 % develop an aggressive leukemia/lymphoma, mirroring the disease trajectory of HTLV-1. The mechanisms by which these viruses provoke cellular transformation remain opaque. In both viruses little or no transcription is observed from the 5'LTR in tumors, however the proviruses are not transcriptionally silent. In the case of BLV a cluster of RNA polymerase III transcribed microRNAs are highly expressed, while the HTLV-1 antisense transcript HBZ is consistently found in all tumors examined. RESULTS: Here, using RNA-seq, we demonstrate that the BLV provirus also constitutively expresses antisense transcripts in all leukemic and asymptomatic samples examined. The first transcript (AS1) can be alternately polyadenylated, generating a transcript of ~600 bp (AS1-S) and a less abundant transcript of ~2200 bp (AS1-L). Alternative splicing creates a second transcript of ~400 bp (AS2). The coding potential of AS1-S/L is ambiguous, with a small open reading frame of 264 bp, however the transcripts are primarily retained in the nucleus, hinting at a lncRNA-like role. The AS1-L transcript overlaps the BLV microRNAs and using high throughput sequencing of RNA-ligase-mediated (RLM) 5'RACE, we show that the RNA-induced silencing complex (RISC) cleaves AS1-L. Furthermore, experiments using altered BLV proviruses with the microRNAs either deleted or inverted point to additional transcriptional interference between the two viral RNA species. CONCLUSIONS: The identification of novel viral antisense transcripts shows the BLV provirus to be far from silent in tumors. Furthermore, the consistent expression of these transcripts in both leukemic and nonmalignant clones points to a vital role in the life cycle of the virus and its tumorigenic potential. Additionally, the cleavage of the AS1-L transcript by the BLV encoded microRNAs and the transcriptional interference between the two viral RNA species suggest a shared role in the regulation of BLV.


Subject(s)
Leukemia Virus, Bovine/genetics , Leukemia, B-Cell/virology , Lymphoma, B-Cell/virology , MicroRNAs/genetics , RNA, Antisense/genetics , RNA, Viral/genetics , Transcription, Genetic , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cattle , Enzootic Bovine Leukosis/virology , High-Throughput Nucleotide Sequencing , Humans , MicroRNAs/metabolism , RNA, Viral/metabolism , Retroviridae Proteins/genetics , Sheep , Terminal Repeat Sequences
11.
Proc Natl Acad Sci U S A ; 110(6): 2306-11, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23345446

ABSTRACT

Viral tumor models have significantly contributed to our understanding of oncogenic mechanisms. How transforming delta-retroviruses induce malignancy, however, remains poorly understood, especially as viral mRNA/protein are tightly silenced in tumors. Here, using deep sequencing of broad windows of small RNA sizes in the bovine leukemia virus ovine model of leukemia/lymphoma, we provide in vivo evidence of the production of noncanonical RNA polymerase III (Pol III)-transcribed viral microRNAs in leukemic B cells in the complete absence of Pol II 5'-LTR-driven transcriptional activity. Processed from a cluster of five independent self-sufficient transcriptional units located in a proviral region dispensable for in vivo infectivity, bovine leukemia virus microRNAs represent ∼40% of all microRNAs in both experimental and natural malignancy. They are subject to strong purifying selection and associate with Argonautes, consistent with a critical function in silencing of important cellular and/or viral targets. Bovine leukemia virus microRNAs are strongly expressed in preleukemic and malignant cells in which structural and regulatory gene expression is repressed, suggesting a key role in tumor onset and progression. Understanding how Pol III-dependent microRNAs subvert cellular and viral pathways will contribute to deciphering the intricate perturbations that underlie malignant transformation.


Subject(s)
Enzootic Bovine Leukosis/genetics , Enzootic Bovine Leukosis/virology , Leukemia Virus, Bovine/genetics , Leukemia, B-Cell/genetics , Leukemia, B-Cell/virology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/virology , MicroRNAs/genetics , RNA, Viral/genetics , Animals , Argonaute Proteins/metabolism , Base Sequence , Cattle , Cell Line, Tumor , Disease Models, Animal , Gene Expression , High Mobility Group Proteins/genetics , Human T-lymphotropic virus 1/genetics , Humans , Leukemia, B-Cell/veterinary , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/virology , Lymphoma, B-Cell/veterinary , MicroRNAs/chemistry , MicroRNAs/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , RNA Polymerase III/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Viral/chemistry , RNA, Viral/metabolism , Sequence Analysis, RNA , Sequence Homology, Nucleic Acid , Sheep , Sheep Diseases/genetics , Sheep Diseases/virology , Terminal Repeat Sequences
12.
Nucleic Acids Res ; 39(22): 9559-73, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21890901

ABSTRACT

Bovine leukemia virus expression relies on its chromatin organization after integration into the host cell genome. Proviral latency, which results from transcriptional repression in vivo, represents a viral strategy to escape the host immune system and likely allows for tumor progression. Here, we discriminated two types of latency: an easily reactivable latent state of the YR2 provirus and a 'locked' latent state of the L267 provirus. The defective YR2 provirus was characterized by the presence of nuclease hypersensitive sites at the U3/R junction and in the R/U5 region of the 5'-long terminal repeat (5'-LTR), whereas the L267 provirus displayed a closed chromatin configuration at the U3/R junction. Reactivation of viral expression in YR2 cells by the phorbol 12-myristate 13-acetate (PMA) plus ionomycin combination was accompanied by a rapid but transient chromatin remodeling in the 5'-LTR, leading to an increased PU.1 and USF-1/USF-2 recruitment in vivo sustained by PMA/ionomycin-mediated USF phosphorylation. In contrast, viral expression was not reactivated by PMA/ionomycin in L267 cells, because the 5'-LTR U3/R region remained inaccessible to nucleases and hypermethylated at CpG dinucleotides. Remarkably, we elucidated the BLV 5'-LTR chromatin organization in PBMCs isolated from BLV-infected cows, thereby depicting the virus hiding in vivo in its natural host.


Subject(s)
Chromatin/chemistry , Leukemia Virus, Bovine/genetics , Promoter Regions, Genetic , Transcriptional Activation , Animals , Binding Sites , Calcium Ionophores/pharmacology , Cattle , Cell Line , Chromatin/drug effects , Chromatin Assembly and Disassembly , Ionomycin/pharmacology , Nucleosomes/chemistry , Proto-Oncogene Proteins/metabolism , Sp1 Transcription Factor/metabolism , Terminal Repeat Sequences , Tetradecanoylphorbol Acetate/pharmacology , Trans-Activators/metabolism , Upstream Stimulatory Factors/metabolism
13.
Leuk Res ; 34(12): 1663-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20591480

ABSTRACT

Delta retrovirus-mediated leukemogenesis is dependent on the oncogenic potential of Tax. It is not clear, however, whether Tax-specific immune responses play a role in leukemia onset and progression. Using the BLV-associated leukemia model in sheep, we found that Tax-specific cytotoxic responses induced by DNA immunization or viral infection of naïve animals were not predictive of disease outcome and did not prevent tumor development. Furthermore, provirus and tax may be absent from blood for extended periods, emphasizing the relevance of surveying other compartments during chronic lymphoproliferative disorders. Our results support the conclusion that Tax-specific cytotoxic responses, even during the initial phase of infection, are not sufficient to prevent leukemogenesis.


Subject(s)
Gene Products, tax/immunology , Immunity, Cellular , Leukemia Virus, Bovine/immunology , Leukemia/immunology , Leukemia/virology , Animals , Disease Models, Animal , Female , Gene Products, tax/metabolism , Leukemia/metabolism , Leukemia/prevention & control , Leukemia Virus, Bovine/metabolism , Male , Sheep , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology , Viral Vaccines/genetics , Viral Vaccines/immunology , Viral Vaccines/pharmacology
14.
Front Biosci (Schol Ed) ; 1(1): 154-63, 2009 06 01.
Article in English | MEDLINE | ID: mdl-19482691

ABSTRACT

Bovine Leukemia Virus (BLV)-induced B-cell leukemia in sheep is a valuable large animal model for investigating oncogenic mechanisms, particularly those associated with human T-cell leukemia virus 1 (HTLV-1). Multiple factors including viral genes, genetic and epigenetic alterations, and the host immune system are likely to contribute and cooperate in the leukemogenesis of adult T-cell leukemia (ATL) in human and B-cell leukemia in sheep. While considerable effort has been made to explore the role of viral determinants in the transformation process, the participation of host-related mechanisms has been poorly addressed. We discuss recent evidence from sheep studies in the context of the growing knowledge that has accumulated in the field of epigenetics in human cancer. These results support the hypothesis that epigenetic events, which were initially identified as a causative mechanism of virus silencing, are also major players in host gene regulation. Future studies in sheep will increase the number of genes identified that are aberrantly regulated by epigenetic processes and identify potential biomarkers which may be used as therapeutic targets in leukemia.


Subject(s)
Epigenesis, Genetic , Leukemia Virus, Bovine/pathogenicity , Leukemia/virology , Oncogenes , Animals , Gene Silencing , Genes, Viral , Leukemia Virus, Bovine/genetics , Sheep
15.
Retrovirology ; 4: 51, 2007 Jul 23.
Article in English | MEDLINE | ID: mdl-17645797

ABSTRACT

BACKGROUND: During malignant progression, tumor cells need to acquire novel characteristics that lead to uncontrolled growth and reduced immunogenicity. In the Bovine Leukemia Virus-induced ovine leukemia model, silencing of viral gene expression has been proposed as a mechanism leading to immune evasion. However, whether proviral expression in tumors is completely suppressed in vivo was not conclusively demonstrated. Therefore, we studied viral expression in two selected experimentally-infected sheep, the virus or the disease of which had features that made it possible to distinguish tumor cells from their nontransformed counterparts. RESULTS: In the first animal, we observed the emergence of a genetically modified provirus simultaneously with leukemia onset. We found a Tax-mutated (TaxK303) replication-deficient provirus in the malignant B-cell clone while functional provirus (TaxE303) had been consistently monitored over the 17-month aleukemic period. In the second case, both non-transformed and transformed BLV-infected cells were present at the same time, but at distinct sites. While there was potentially-active provirus in the non-leukemic blood B-cell population, as demonstrated by ex-vivo culture and injection into naïve sheep, virus expression was completely suppressed in the malignant B-cells isolated from the lymphoid tumors despite the absence of genetic alterations in the proviral genome. These observations suggest that silencing of viral genes, including the oncoprotein Tax, is associated with tumor onset. CONCLUSION: Our findings suggest that silencing is critical for tumor progression and identify two distinct mechanisms-genetic and epigenetic-involved in the complete suppression of virus and Tax expression. We demonstrate that, in contrast to systems that require sustained oncogene expression, the major viral transforming protein Tax can be turned-off without reversing the transformed phenotype. We propose that suppression of viral gene expression is a contributory factor in the impairment of immune surveillance and the uncontrolled proliferation of the BLV-infected tumor cell.


Subject(s)
Gene Expression Regulation, Viral , Leukemia Virus, Bovine/genetics , Leukemia/veterinary , Animals , Cattle , Enzootic Bovine Leukosis/genetics , Leukemia/genetics , Leukemia Virus, Bovine/pathogenicity , Sheep/virology , Sheep Diseases/genetics
16.
J Virol ; 81(11): 5929-39, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17392371

ABSTRACT

Ovine leukemia/lymphoma resulting from bovine leukemia virus infection of sheep offers a large animal model for studying mechanisms underlying leukemogenesis. Silencing of viral information including Tax, the major contributor to the oncogenic potential of the virus, is critical if not mandatory for tumor progression. In this study, we have identified epigenetic mechanisms that govern the complete suppression of viral expression, using a lymphoma-derived B-cell clone carrying a silent provirus. Silencing was not relieved by injection of the malignant B cells into sheep. However, exogenous expression of Tax or treatment with either the DNA methyltransferase inhibitor 5'azacytidine or the histone deacetylase (HDAC) inhibitor trichostatin A rescued viral expression, as demonstrated by in vivo infectivity trials. Comparing silent and reactivated provirus, we found mechanistic connections between chromatin conformation and tumor-associated transcriptional repression. Silencing is associated with DNA methylation and decreased accessibility of promoter sequences. HDAC1 and the transcriptional corepressor mSin3A are associated with the inactive but not the reactivated promoter. Silencing correlates with a repressed chromatin structure marked by histone H3 and H4 hypoacetylation, a loss of methylation at H3 lysine 4, and an increase of H3 lysine 9 methylation. These observations point to the critical role of epigenetic mechanisms in tumor-specific virus/oncogene silencing, a potential strategy to evade immune response and favor the propagation of the transformed cell.


Subject(s)
Chromatin/pathology , Epigenesis, Genetic , Gene Expression Regulation, Viral/physiology , Histones/physiology , Leukemia Virus, Bovine/genetics , Leukemia, B-Cell/virology , Animals , Cell Line, Tumor , Chromatin/genetics , Histones/genetics , Leukemia, B-Cell/pathology , Leukemia, B-Cell/veterinary , Proviruses/genetics , Sheep/virology
17.
J Virol ; 80(4): 1922-38, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16439548

ABSTRACT

Large-animal models for leukemia have the potential to aid in the understanding of networks that contribute to oncogenesis. Infection of cattle and sheep with bovine leukemia virus (BLV), a complex retrovirus related to human T-cell leukemia virus type 1 (HTLV-1), is associated with the development of B-cell leukemia. Whereas the natural disease in cattle is characterized by a low tumor incidence, experimental infection of sheep leads to overt leukemia in the majority of infected animals, providing a model for studying the pathogenesis associated with BLV and HTLV-1. Tax(BLV), the major oncoprotein, initiates a cascade of events leading toward malignancy, although the basis of transformation is not fully understood. We have taken a cross-species ovine-to-human microarray approach to identify Tax(BLV)-responsive transcriptional changes in two sets of cultured ovine B cells following retroviral vector-mediated delivery of Tax(BLV). Using cDNA-spotted microarrays comprising 10,336 human genes/expressed sequence tags, we identified a cohort of differentially expressed genes, including genes related to apoptosis, DNA transcription, and repair; proto-oncogenes; cell cycle regulators; transcription factors; small Rho GTPases/GTPase-binding proteins; and previously reported Tax(HTLV-1)-responsive genes. Interestingly, genes known to be associated with human neoplasia, especially B-cell malignancies, were extensively represented. Others were novel or unexpected. The results suggest that Tax(BLV) deregulates a broad network of interrelated pathways rather than a single B-lineage-specific regulatory process. Although cross-species approaches do not permit a comprehensive analysis of gene expression patterns, they can provide initial clues for the functional roles of genes that participate in B-cell transformation and pinpoint molecular targets not identified using other methods in animal models.


Subject(s)
B-Lymphocytes/physiology , B-Lymphocytes/virology , Cell Transformation, Viral/genetics , Gene Expression Profiling , Gene Expression Regulation , Gene Products, tax/physiology , Leukemia Virus, Bovine/physiology , Animals , Blotting, Northern , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Sheep
18.
J Virol ; 78(24): 13848-64, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564493

ABSTRACT

Bovine leukemia virus (BLV) infection is characterized by viral latency in a large proportion of cells containing an integrated provirus. In this study, we postulated that mechanisms directing the recruitment of deacetylases to the BLV 5' long terminal repeat (LTR) could explain the transcriptional repression of viral expression in vivo. Accordingly, we showed that BLV promoter activity was induced by several deacetylase inhibitors (such as trichostatin A [TSA]) in the context of episomal LTR constructs and in the context of an integrated BLV provirus. Moreover, treatment of BLV-infected cells with TSA increased H4 acetylation at the viral promoter, showing a close correlation between the level of histone acetylation and transcriptional activation of the BLV LTR. Among the known cis-regulatory DNA elements located in the 5' LTR, three E box motifs overlapping cyclic AMP responsive elements (CREs) in U3 were shown to be involved in transcriptional repression of BLV basal gene expression. Importantly, the combined mutations of these three E box motifs markedly reduced the inducibility of the BLV promoter by TSA. E boxes are susceptible to recognition by transcriptional repressors such as Max-Mad-mSin3 complexes that repress transcription by recruiting deacetylases. However, our in vitro binding studies failed to reveal the presence of Mad-Max proteins in the BLV LTR E box-specific complexes. Remarkably, TSA increased the occupancy of the CREs by CREB/ATF. Therefore, we postulated that the E box-specific complexes exerted their negative cooperative effect on BLV transcription by steric hindrance with the activators CREB/ATF and/or their transcriptional coactivators possessing acetyltransferase activities. Our results thus suggest that the overlapping CRE and E box elements in the BLV LTR were selected during evolution as a novel strategy for BLV to allow better silencing of viral transcription and to escape from the host immune response.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Viral , Leukemia Virus, Bovine/metabolism , Promoter Regions, Genetic/genetics , Terminal Repeat Sequences/genetics , Transcription, Genetic , 5' Untranslated Regions/genetics , Acetylation , Activating Transcription Factors , Animals , Base Sequence , Blood Proteins/metabolism , Cattle , Cell Line , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Leukemia Virus, Bovine/genetics , Leukocytes, Mononuclear , Molecular Sequence Data , Sheep , Transcription Factors/metabolism , Virus Latency
19.
Oncogene ; 22(29): 4531-42, 2003 Jul 17.
Article in English | MEDLINE | ID: mdl-12881710

ABSTRACT

Transactivating proteins associated with complex onco-retroviruses including human T-cell leukemia virus-1 (HTLV-1) and bovine leukemia virus (BLV) mediate transformation using poorly understood mechanisms. To gain insight into the processes that govern tumor onset and progression, we have examined the impact of BLV-Tax expression on ovine B-cells, the targets of BLV in experimentally infected sheep, using B-cell clones that are dependent on CD154 and gammac-common cytokines. Tax was capable of mediating progression of B-cells from cytokine dependence to cytokine independence, indicating that the transactivator can over-ride signaling pathways typically controlled by cytokine receptor activation in B-cells. When examined in the presence of both CD154 and interleukin-4, Tax had a clear supportive role on B-cell growth, with an impact on B-cell proliferation, cell cycle phase distribution, and survival. Apoptotic B-cell death mediated by growth factor withdrawal, physical insult, and NF-kappaB inhibition was dramatically reduced in the presence of Tax. Furthermore, the expression of Tax was associated with higher Bcl-2 protein levels, providing rationale for the rescue signals mediated by the transactivator. Finally, Tax expression in B-cells led to a dramatic increase of nuclear RelB/p50 and p50/p50 NF-kappaB dimers, indicating that cellular signaling through NF-kappaB is a major contributory mechanism in the disruption of B-cell homeostasis. Although Tax is involved in aspects of pathogenesis that are unique to complex retroviruses, the viral strategies associated with this transactivating oncoprotein may have wide-ranging effects that are relevant to other B-cell malignancies.


Subject(s)
B-Lymphocytes/metabolism , Gene Products, tax/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Animals , Apoptosis/physiology , B-Lymphocytes/cytology , B-Lymphocytes/virology , CD40 Ligand/pharmacology , CD40 Ligand/physiology , Cell Cycle/genetics , Cell Division/genetics , Cells, Cultured , Clone Cells/cytology , Clone Cells/metabolism , Clone Cells/virology , Cytokines/metabolism , Cytokines/pharmacology , Gene Products, tax/genetics , Gene Transfer Techniques , Interleukin-4/pharmacology , Interleukin-4/physiology , Leukemia Virus, Bovine/chemistry , Retroviridae/genetics , Sheep , Up-Regulation
20.
Cancer Gene Ther ; 9(9): 715-24, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12189521

ABSTRACT

Gene delivery to dendritic cells (DCs) could represent a powerful method of inducing potent, long-lasting immunity. Although recent studies underline the intense interest in lentiviral vector-mediated monocyte-derived DC transduction, efficient gene transfer methods currently require high multiplicities of infection and are not compatible with clinical constraints. We have designed a strategy to optimize the efficiency and clinical relevance of this approach. Initially, using a third generation lentiviral vector expressing green fluorescent protein, we found that modifying the vector design, the DC precursor cell type, and the DC differentiation stage for transduction results in sustained transgene expression in 75-85% of immature DCs (transduction at a multiplicity of infection of 8). This high efficiency was reproducible among different donors irrespective of whether DCs were expanded from fresh or cryopreserved CD14(+) precursors. We then developed procedures that bypass the need for highly concentrated lentiviral preparations and the addition of polybrene to achieve efficient transduction. DCs transduced under these conditions retain their immature phenotype and immunostimulatory potential in both autologous and allogeneic settings. Furthermore, genetically modified DCs maintain their ability to respond to maturation signals and secrete bioactive IL-12, indicating that they are fully functional. Finally, the level of transgene expression is preserved in the therapeutically relevant mature DCs, demonstrating that there is neither promoter-silencing nor loss of transduced cells during maturation. The novel approach described should advance lentiviral-mediated monocyte-derived DC transduction towards a clinical reality.


Subject(s)
Dendritic Cells/metabolism , Gene Transfer Techniques , Genetic Vectors , HIV-1/genetics , Luminescent Proteins/metabolism , DNA Primers/chemistry , Dendritic Cells/virology , Flow Cytometry , Genetic Therapy , Green Fluorescent Proteins , Humans , Immunomagnetic Separation , Immunophenotyping , Interleukin-12/metabolism , Lipopolysaccharide Receptors/metabolism , Luminescent Proteins/genetics , Lymphocyte Activation , Monocytes/cytology , Monocytes/metabolism , Polymerase Chain Reaction , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...