Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Rev Mol Cell Biol ; 17(7): 426-38, 2016 07.
Article in English | MEDLINE | ID: mdl-27251421

ABSTRACT

RNA helicases comprise the largest family of enzymes involved in the metabolism of mRNAs, the processing and fate of which rely on their packaging into messenger ribonucleoprotein particles (mRNPs). In this Review, we describe how the capacity of some RNA helicases to either remodel or lock the composition of mRNP complexes underlies their pleiotropic functions at different steps of the gene expression process. We illustrate the roles of RNA helicases in coordinating gene expression steps and programmes, and propose that RNA helicases function as molecular drivers and guides of the progression of their mRNA substrates from one RNA-processing factory to another, to a productive mRNA pool that leads to protein synthesis or to unproductive mRNA pools that are stored or degraded.


Subject(s)
Gene Expression Regulation , RNA Helicases/physiology , Animals , Gene Expression , Humans , RNA Splicing , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
Nucleic Acids Res ; 52(4): 1527-1543, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38272542

ABSTRACT

The NF-κB protein p65/RelA plays a pivotal role in coordinating gene expression in response to diverse stimuli, including viral infections. At the chromatin level, p65/RelA regulates gene transcription and alternative splicing through promoter enrichment and genomic exon occupancy, respectively. The intricate ways in which p65/RelA simultaneously governs these functions across various genes remain to be fully elucidated. In this study, we employed the HTLV-1 Tax oncoprotein, a potent activator of NF-κB, to investigate its influence on the three-dimensional organization of the genome, a key factor in gene regulation. We discovered that Tax restructures the 3D genomic landscape, bringing together genes based on their regulation and splicing patterns. Notably, we found that the Tax-induced gene-gene contact between the two master genes NFKBIA and RELA is associated with their respective changes in gene expression and alternative splicing. Through dCas9-mediated approaches, we demonstrated that NFKBIA-RELA interaction is required for alternative splicing regulation and is caused by an intragenic enrichment of p65/RelA on RELA. Our findings shed light on new regulatory mechanisms upon HTLV-1 Tax and underscore the integral role of p65/RelA in coordinated regulation of NF-κB-responsive genes at both transcriptional and splicing levels in the context of the 3D genome.


The NF-κB pathway is essential for coordinating gene expression in response to various stimuli, including viral infections. Most studies have focused on the role of NF-κB in transcriptional regulation. In the present study, the impact of the potent NF-κB activator HTLV-1 Tax oncoprotein on the three-dimensional organization of the genome was investigated. Tax-mediated NF-κB activation was found to restructure the 3D genomic landscape in cells and to bring genes together in multigene complexes that are coordinately regulated either transcriptionally or through alternative splicing by NF-κB. Induced coordinate changes in transcription and alternative splicing included the two master genes of NF-κB pathway NFKBIA and RELA. The findings have significant implications for understanding cell fate determination and disease development associated with HTLV-1 infection, as well as chronic NF-κB activation in various human inflammatory diseases and cancer.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation , NF-kappa B p50 Subunit , Alternative Splicing/genetics , Chromatin Assembly and Disassembly/genetics , Gene Products, tax/genetics , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcriptional Activation , Humans , NF-kappa B p50 Subunit/metabolism
3.
Nucleic Acids Res ; 51(14): 7580-7601, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37254812

ABSTRACT

The selenocysteine (Sec) tRNA (tRNA[Ser]Sec) governs Sec insertion into selenoproteins by the recoding of a UGA codon, typically used as a stop codon. A homozygous point mutation (C65G) in the human tRNA[Ser]Sec acceptor arm has been reported by two independent groups and was associated with symptoms such as thyroid dysfunction and low blood selenium levels; however, the extent of altered selenoprotein synthesis resulting from this mutation has yet to be comprehensively investigated. In this study, we used CRISPR/Cas9 technology to engineer homozygous and heterozygous mutant human cells, which we then compared with the parental cell lines. This C65G mutation affected many aspects of tRNA[Ser]Sec integrity and activity. Firstly, the expression level of tRNA[Ser]Sec was significantly reduced due to an altered recruitment of RNA polymerase III at the promoter. Secondly, selenoprotein expression was strongly altered, but, more surprisingly, it was no longer sensitive to selenium supplementation. Mass spectrometry analyses revealed a tRNA isoform with unmodified wobble nucleotide U34 in mutant cells that correlated with reduced UGA recoding activities. Overall, this study demonstrates the pleiotropic effect of a single C65G mutation on both tRNA phenotype and selenoproteome expression.


Subject(s)
Selenium , Humans , Codon, Terminator , Mutation , Selenium/pharmacology , Selenium/metabolism , Selenocysteine/genetics , Selenocysteine/metabolism , Selenoproteins/genetics , Proteome
4.
Nucleic Acids Res ; 50(16): 9226-9246, 2022 09 09.
Article in English | MEDLINE | ID: mdl-36039747

ABSTRACT

DDX5 and DDX17 are DEAD-box RNA helicase paralogs which regulate several aspects of gene expression, especially transcription and splicing, through incompletely understood mechanisms. A transcriptome analysis of DDX5/DDX17-depleted human cells confirmed the large impact of these RNA helicases on splicing and revealed a widespread deregulation of 3' end processing. In silico analyses and experiments in cultured cells showed the binding and functional contribution of the genome organizing factor CTCF to chromatin sites at or near a subset of DDX5/DDX17-dependent exons that are characterized by a high GC content and a high density of RNA Polymerase II. We propose the existence of an RNA helicase-dependent relationship between CTCF and the dynamics of transcription across DNA and/or RNA structured regions, that contributes to the processing of internal and terminal exons. Moreover, local DDX5/DDX17-dependent chromatin loops spatially connect RNA helicase-regulated exons with their cognate promoter, and we provide the first direct evidence that de novo gene looping modifies alternative splicing and polyadenylation. Overall our findings uncover the impact of DDX5/DDX17-dependent chromatin folding on pre-messenger RNA processing.


Subject(s)
DEAD-box RNA Helicases , RNA , Humans , RNA/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , DEAD-box RNA Helicases/metabolism , Alternative Splicing , Chromatin/genetics
5.
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
6.
Genome Res ; 29(5): 711-722, 2019 05.
Article in English | MEDLINE | ID: mdl-30962178

ABSTRACT

The inclusion of exons during the splicing process depends on the binding of splicing factors to short low-complexity regulatory sequences. The relationship between exonic splicing regulatory sequences and coding sequences is still poorly understood. We demonstrate that exons that are coregulated by any given splicing factor share a similar nucleotide composition bias and preferentially code for amino acids with similar physicochemical properties because of the nonrandomness of the genetic code. Indeed, amino acids sharing similar physicochemical properties correspond to codons that have the same nucleotide composition bias. In particular, we uncover that the TRA2A and TRA2B splicing factors that bind to adenine-rich motifs promote the inclusion of adenine-rich exons coding preferentially for hydrophilic amino acids that correspond to adenine-rich codons. SRSF2 that binds guanine/cytosine-rich motifs promotes the inclusion of GC-rich exons coding preferentially for small amino acids, whereas SRSF3 that binds cytosine-rich motifs promotes the inclusion of exons coding preferentially for uncharged amino acids, like serine and threonine that can be phosphorylated. Finally, coregulated exons encoding amino acids with similar physicochemical properties correspond to specific protein features. In conclusion, the regulation of an exon by a splicing factor that relies on the affinity of this factor for specific nucleotide(s) is tightly interconnected with the exon-encoded physicochemical properties. We therefore uncover an unanticipated bidirectional interplay between the splicing regulatory process and its biological functional outcome.


Subject(s)
Alternative Splicing , Exons/genetics , RNA Splice Sites/genetics , RNA Splicing Factors/metabolism , Amino Acids/chemistry , Base Composition/genetics , Cell Line , Genetic Code , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Introns/genetics , Nucleotide Motifs/genetics , Sequence Analysis, Protein , Sequence Analysis, RNA , Serine-Arginine Splicing Factors/metabolism
7.
Genome Res ; 27(6): 1087-1097, 2017 06.
Article in English | MEDLINE | ID: mdl-28420690

ABSTRACT

Transcriptomic genome-wide analyses demonstrate massive variation of alternative splicing in many physiological and pathological situations. One major challenge is now to establish the biological contribution of alternative splicing variation in physiological- or pathological-associated cellular phenotypes. Toward this end, we developed a computational approach, named "Exon Ontology," based on terms corresponding to well-characterized protein features organized in an ontology tree. Exon Ontology is conceptually similar to Gene Ontology-based approaches but focuses on exon-encoded protein features instead of gene level functional annotations. Exon Ontology describes the protein features encoded by a selected list of exons and looks for potential Exon Ontology term enrichment. By applying this strategy to exons that are differentially spliced between epithelial and mesenchymal cells and after extensive experimental validation, we demonstrate that Exon Ontology provides support to discover specific protein features regulated by alternative splicing. We also show that Exon Ontology helps to unravel biological processes that depend on suites of coregulated alternative exons, as we uncovered a role of epithelial cell-enriched splicing factors in the AKT signaling pathway and of mesenchymal cell-enriched splicing factors in driving splicing events impacting on autophagy. Freely available on the web, Exon Ontology is the first computational resource that allows getting a quick insight into the protein features encoded by alternative exons and investigating whether coregulated exons contain the same biological information.


Subject(s)
Alternative Splicing , Exons , Gene Expression Profiling/methods , Genome, Human , Molecular Sequence Annotation/methods , Transcriptome , Autophagy , Cell Line, Tumor , Gene Ontology , Genome-Wide Association Study , Humans , MCF-7 Cells , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Signal Transduction , Software
8.
Nucleic Acids Res ; 46(15): 7686-7700, 2018 09 06.
Article in English | MEDLINE | ID: mdl-29931089

ABSTRACT

The Repressor Element 1-silencing transcription factor (REST) represses a number of neuronal genes in non-neuronal cells or in undifferentiated neural progenitors. Here, we report that the DEAD box RNA helicase DDX17 controls important REST-related processes that are critical during the early phases of neuronal differentiation. First, DDX17 associates with REST, promotes its binding to the promoter of a subset of REST-targeted genes and co-regulates REST transcriptional repression activity. During neuronal differentiation, we observed a downregulation of DDX17 along with that of the REST complex that contributes to the activation of neuronal genes. Second, DDX17 and its paralog DDX5 regulate the expression of several proneural microRNAs that are known to target the REST complex during neurogenesis, including miR-26a/b that are also direct regulators of DDX17 expression. In this context, we propose a new mechanism by which RNA helicases can control the biogenesis of intronic miRNAs. We show that the processing of the miR-26a2 precursor is dependent on RNA helicases, owing to an intronic regulatory region that negatively impacts on both miRNA processing and splicing of its host intron. Our work places DDX17 in the heart of a pathway involving REST and miRNAs that allows neuronal gene repression.


Subject(s)
DEAD-box RNA Helicases/genetics , Gene Expression Profiling , MicroRNAs/genetics , Repressor Proteins/genetics , Cell Line, Tumor , DEAD-box RNA Helicases/metabolism , Humans , MCF-7 Cells , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/metabolism , Repressor Proteins/metabolism
9.
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
10.
Retrovirology ; 11: 119, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25519886

ABSTRACT

BACKGROUND: Reprogramming cellular gene transcription sustains HTLV-1 viral persistence that ultimately leads to the development of adult T-cell leukemia/lymphoma (ATLL). We hypothesized that besides these quantitative transcriptional effects, HTLV-1 qualitatively modifies the pattern of cellular gene expression. RESULTS: Exon expression analysis shows that patients' untransformed and malignant HTLV-1(+) CD4(+) T-cells exhibit multiple alternate exon usage (AEU) events. These affect either transcriptionally modified or unmodified genes, culminate in ATLL, and unveil new functional pathways involved in cancer and cell cycle. Unsupervised hierarchical clustering of array data permitted to isolate exon expression patterns of 3977 exons that discriminate uninfected, infected, and transformed CD4(+) T-cells. Furthermore, untransformed infected CD4+ clones and ATLL samples shared 486 exon modifications distributed in 320 genes, thereby indicating a role of AEUs in HTLV-1 leukemogenesis. Exposing cells to splicing modulators revealed that Sudemycin E reduces cell viability of HTLV-1 transformed cells without affecting primary control CD4+ cells and HTLV-1 negative cell lines, suggesting that the huge excess of AEU might provide news targets for treating ATLL. CONCLUSIONS: Taken together, these data reveal that HTLV-1 significantly modifies the structure of cellular transcripts and unmask new putative leukemogenic pathways and possible therapeutic targets.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Exons , Gene Expression Regulation , Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , Leukemia-Lymphoma, Adult T-Cell/pathology , Human T-lymphotropic virus 1/growth & development , Humans , Transcription, Genetic
11.
J Gen Virol ; 94(Pt 4): 753-757, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23239567

ABSTRACT

Upon antiretroviral therapy (ART) human immunodeficiency virus (HIV)/human T-cell lymphotropic virus type 1 (HTLV-1) co-infected individuals frequently develop neurological disorders through hitherto unknown mechanisms. Here, we show that effective anti-HIV ART increases HTLV-1 proviral load through a polyclonal integration pattern of HTLV-1 in both CD4(+) and CD8(+) T-cell subsets that is reminiscent of that typically associated with HTLV-1-related inflammatory conditions. These data indicate that preventing ART-triggered clonal expansion of HTLV-1-infected cells in co-infected individuals deserves investigation.


Subject(s)
Anti-Retroviral Agents/administration & dosage , Antiretroviral Therapy, Highly Active/methods , HIV Infections/drug therapy , HTLV-I Infections/virology , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/pathogenicity , Virus Replication , Anti-Retroviral Agents/adverse effects , Antiretroviral Therapy, Highly Active/adverse effects , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Coinfection/drug therapy , HIV Infections/complications , Human T-lymphotropic virus 1/isolation & purification , Humans , Proviruses/drug effects , Proviruses/isolation & purification , Viral Load , Virus Integration/drug effects
12.
Database (Oxford) ; 20232023 Dec 21.
Article in English | MEDLINE | ID: mdl-38128543

ABSTRACT

One challenge faced by scientists from the alternative RNA splicing field is to decode the cooperative or antagonistic effects of splicing factors (SFs) to understand and eventually predict splicing outcomes on a genome-wide scale. In this manuscript, we introduce SplicingLore, an open-access database and web resource that help to fill this gap in a straightforward manner. The database contains a collection of RNA-sequencing-derived lists of alternative exons regulated by a total of 75 different SFs. All datasets were processed in a standardized manner, ensuring valid comparisons and correlation analyses. The user can easily retrieve a factor-specific set of differentially included exons from the database or provide a list of exons and search which SF(s) control(s) their inclusion. Our simple workflow is fast and easy to run, and it ensures a reliable calculation of correlation scores between the tested datasets. As a proof of concept, we predicted and experimentally validated a novel functional cooperation between the RNA helicases DDX17 and DDX5 and the heterogeneous nuclear ribonucleoprotein C (HNRNPC) protein. SplicingLore is available at https://splicinglore.ens-lyon.fr/. Database URL:  https://splicinglore.ens-lyon.fr/.


Subject(s)
Alternative Splicing , RNA Splicing , Humans , RNA Splicing Factors/genetics , RNA Splicing/genetics , Genome , Exons/genetics
13.
Int J Cancer ; 131(4): 821-33, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-21717459

ABSTRACT

Untransformed HTLV-1 positive CD4(+) cells from infected individuals are selected for expressing tax and displaying morphological features consistent with telomere dysfunctions. We show that in resting HTLV-1 positive CD4(+) cells cloned from patients, hTERT expression parallels tax expression and cell cycling. Upon activation, these cells dramatically augment tax expression, whereas their increase in telomerase activity is about 20 times lower than that of their uninfected counterpart. Activated HTLV-1 positive CD4(+) but not uninfected CD4(+) or CD8(+) clones also repress the transcription of TRF1, TPP1, TANK1, POT1, DNA-PKc and Ku80. Both infected and uninfected lymphocytes from infected individuals shared common telomere gene deregulations toward a pattern consistent with premature senescence. ATLL cells displayed the highest telomerase activity (TA) whereas recovered a telomere gene transcriptome close to that of normal CD4(+) cells. In conclusion HTLV-1-dependent telomere modulations seem involved in clonal expansion, immunosuppression, tumor initiation and progression.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HTLV-I Infections/enzymology , Human T-lymphotropic virus 1/isolation & purification , Telomerase/genetics , Telomere , Base Sequence , Clone Cells , DNA Primers , HTLV-I Infections/genetics , HTLV-I Infections/immunology , Homeostasis , Humans , Lymphocyte Activation , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Shelterin Complex , Telomere-Binding Proteins , Transcription, Genetic , Transcriptome
14.
Blood ; 116(19): 3802-8, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20587783

ABSTRACT

Approximately 3% of all human T-lymphotropic virus type 1 (HTLV-1)-infected persons will develop a disabling inflammatory disease of the central nervous system known as HTLV-1-associated myelopathy/tropical spastic paraparesis, against which there is currently no efficient treatment. As correlation exists between the proviral load (PVL) and the clinical status of the carrier, it is thought that diminishing the PVL could prevent later occurrence of the disease. We have conducted a study combining valproate, an inhibitor of histone deacetylases, and azidothymidine, an inhibitor of reverse transcriptase, in a series of baboons naturally infected with simian T-lymphotropic virus type 1 (STLV-1), whose PVL was equivalent to that of HTLV-1 asymptomatic carriers. We show that the combination of drugs caused a strong decrease in the PVL and prevented the transient rise in PVL that is seen after treatment with histone deacetylases alone. We then demonstrate that the PVL decline was associated with an increase in the STLV-1-specific cytotoxic T-cell population. We conclude that combined treatment with valproate to induce viral expression and azidothymidine to prevent viral propagation is a safe and effective means to decrease PVL in vivo. Such treatments may be useful to reduce the risk of HAM/TSP in asymptomatic carriers with a high PVL.


Subject(s)
Antiviral Agents/administration & dosage , Deltaretrovirus Infections/veterinary , Histone Deacetylase Inhibitors/administration & dosage , Monkey Diseases/drug therapy , Papio , Reverse Transcriptase Inhibitors/administration & dosage , Simian T-lymphotropic virus 1 , Animals , Antiretroviral Therapy, Highly Active , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Deltaretrovirus Infections/drug therapy , Deltaretrovirus Infections/immunology , Deltaretrovirus Infections/virology , Disease Models, Animal , Drug Therapy, Combination , Female , HTLV-I Infections/drug therapy , HTLV-I Infections/virology , Humans , Male , Monkey Diseases/immunology , Monkey Diseases/virology , Paraparesis, Tropical Spastic/drug therapy , Paraparesis, Tropical Spastic/virology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Valproic Acid/administration & dosage , Viral Load/drug effects , Zidovudine/administration & dosage
15.
Front Immunol ; 13: 939863, 2022.
Article in English | MEDLINE | ID: mdl-35979358

ABSTRACT

Adult T-cell leukemia/lymphoma (ATL) is a T-cell lymphoproliferative neoplasm caused by the human T-cell leukemia virus type 1 (HTLV-1). Two viral proteins, Tax-1 and HBZ play important roles in HTLV-1 infectivity and in HTLV-1-associated pathologies by altering key pathways of cell homeostasis. However, the molecular mechanisms through which the two viral proteins, particularly HBZ, induce and/or sustain the oncogenic process are still largely elusive. Previous results suggested that HBZ interaction with nuclear factors may alter cell cycle and cell proliferation. To have a more complete picture of the HBZ interactions, we investigated in detail the endogenous HBZ interactome in leukemic cells by immunoprecipitating the HBZ-interacting complexes of ATL-2 leukemic cells, followed by tandem mass spectrometry analyses. RNA seq analysis was performed to decipher the differential gene expression and splicing modifications related to HTLV-1. Here we compared ATL-2 with MOLT-4, a non HTLV-1 derived leukemic T cell line and further compared with HBZ-induced modifications in an isogenic system composed by Jurkat T cells and stably HBZ transfected Jurkat derivatives. The endogenous HBZ interactome of ATL-2 cells identified 249 interactors covering three main clusters corresponding to protein families mainly involved in mRNA splicing, nonsense-mediated RNA decay (NMD) and JAK-STAT signaling pathway. Here we analyzed in detail the cluster involved in RNA splicing. RNAseq analysis showed that HBZ specifically altered the transcription of many genes, including crucial oncogenes, by affecting different splicing events. Consistently, the two RNA helicases, members of the RNA splicing family, DDX5 and its paralog DDX17, recently shown to be involved in alternative splicing of cellular genes after NF-κB activation by HTLV-1 Tax-1, interacted and partially co-localized with HBZ. For the first time, a complete picture of the endogenous HBZ interactome was elucidated. The wide interaction of HBZ with molecules involved in RNA splicing and the subsequent transcriptome alteration strongly suggests an unprecedented complex role of the viral oncogene in the establishment of the leukemic state.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Human T-lymphotropic virus 1 , RNA Splicing , Retroviridae Proteins/metabolism , Adult , Alternative Splicing , DEAD-box RNA Helicases/metabolism , Human T-lymphotropic virus 1/physiology , Humans , Viral Proteins/metabolism
16.
Retrovirology ; 7: 17, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20222966

ABSTRACT

BACKGROUND: Adult T cell leukemia results from the malignant transformation of a CD4+ lymphoid clone carrying an integrated HTLV-1 provirus that has undergone several oncogenic events over a 30-60 year period of persistent clonal expansion. Both CD4+ and CD8+ lymphocytes are infected in vivo; their expansion relies on CD4+ cell cycling and on the prevention of CD8+ cell death. Cloned infected CD4+ but not CD8+ T cells from patients without malignancy also add up nuclear and mitotic defects typical of genetic instability related to the expression of the virus-encoded oncogene tax. HTLV-1 expression is cancer-prone in vitro, but in vivo numerous selection forces act to maintain T cell homeostasis and are possibly involved in clonal selection. RESULTS: Here we demonstrate that the HTLV-1 associated CD4+ preleukemic phenotype and the specific patterns of CD4+ and CD8+ clonal expansion are in vivo selected processes. By comparing the effects of recent (1 month) experimental infections performed in vitro and those observed in cloned T cells from patients infected for >6-26 years, we found that in chronically HTLV-1 infected individuals, HTLV-1 positive clones are selected for tax expression. In vivo, infected CD4+ cells are positively selected for cell cycling whereas infected CD8+ cells and uninfected CD4+ cells are negatively selected for the same processes. In contrast, the known HTLV-1-dependent prevention of CD8+ T cell death pertains to both in vivo and in vitro infected cells. CONCLUSIONS: Therefore, virus-cell interactions alone are not sufficient to initiate early leukemogenesis in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Cell Death , Cell Proliferation , Genes, pX , Human T-lymphotropic virus 1/pathogenicity , Leukemia-Lymphoma, Adult T-Cell/virology , Cell Transformation, Viral , Gene Expression , Host-Pathogen Interactions , Humans
17.
Nat Commun ; 11(1): 3045, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546717

ABSTRACT

Chronic NF-κB activation in inflammation and cancer has long been linked to persistent activation of NF-κB-responsive gene promoters. However, NF-κB factors also massively bind to gene bodies. Here, we demonstrate that recruitment of the NF-κB factor RELA to intragenic regions regulates alternative splicing upon NF-κB activation by the viral oncogene Tax of HTLV-1. Integrative analyses of RNA splicing and chromatin occupancy, combined with chromatin tethering assays, demonstrate that DNA-bound RELA interacts with and recruits the splicing regulator DDX17, in an NF-κB activation-dependent manner. This leads to alternative splicing of target exons due to the RNA helicase activity of DDX17. Similar results were obtained upon Tax-independent NF-κB activation, indicating that Tax likely exacerbates a physiological process where RELA provides splice target specificity. Collectively, our results demonstrate a physical and direct involvement of NF-κB in alternative splicing regulation, which significantly revisits our knowledge of HTLV-1 pathogenesis and other NF-κB-related diseases.


Subject(s)
Alternative Splicing/physiology , Gene Products, tax/metabolism , NF-kappa B/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation , Gene Products, tax/genetics , Human T-lymphotropic virus 1/pathogenicity , Humans , Leukocytes, Mononuclear/virology , NF-kappa B/genetics , Oncogenes , Transcription Factor RelA/metabolism
18.
Retrovirology ; 6: 30, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19344505

ABSTRACT

BACKGROUND: Retrovirus-induced tumors develop in a broad range of frequencies and after extremely variable periods of time, from only a few days to several decades, depending mainly on virus type. For hitherto unexplained reasons, deltaretroviruses cause hematological malignancies only in a minority of naturally infected organisms and after a very prolonged period of clinical latency. RESULTS: Here we demonstrate that the development of malignancies in sheep experimentally infected with the deltaretrovirus bovine leukemia virus (BLV) depends only on the level of BLV replication. Animals were experimentally infected with leukemogenic or attenuated, but infectious, BLV molecular clones and monitored prospectively through 8 months for viral replication. As early as 2 weeks after infection and subsequently at any time during follow-up, leukemogenic viruses produced significantly higher absolute levels of reverse transcription (RT), clonal expansion of infected cells, and circulating proviruses with RT- and somatic-dependent mutations than attenuated viruses. These differences were only quantitative, and both kinds of viruses triggered parallel temporal fluctuations of host lymphoid cells, viral loads, infected cell clonality and proliferation. CONCLUSION: Deltaretrovirus-associated leukemogenesis in sheep appears to be a two-hit process over time depending on the amounts of first horizontally and then vertically expanded viruses.


Subject(s)
Deltaretrovirus Infections , Leukemia Virus, Bovine/physiology , Leukemia Virus, Bovine/pathogenicity , Leukemia, Experimental , Sheep Diseases , Virus Replication , Animals , Cattle , DNA, Viral , Deltaretrovirus Infections/pathology , Deltaretrovirus Infections/virology , Leukemia Virus, Bovine/genetics , Leukemia, Experimental/pathology , Leukemia, Experimental/virology , Sheep , Sheep Diseases/pathology , Sheep Diseases/virology
19.
J Clin Invest ; 116(4): 974-83, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16585963

ABSTRACT

Human T cell leukemia virus type 1 (HTLV-1) infects both CD4+ and CD8+ lymphocytes, yet it induces adult T cell leukemia/lymphoma (ATLL) that is regularly of the CD4+ phenotype. Here we show that in vivo infected CD4+ and CD8+ T cells displayed similar patterns of clonal expansion in carriers without malignancy. Cloned infected cells from individuals without malignancy had a dramatic increase in spontaneous proliferation, which predominated in CD8+ lymphocytes and depended on the amount of tax mRNA. In fact, the clonal expansion of HTLV-1-positive CD8+ and CD4+ lymphocytes relied on 2 distinct mechanisms--infection prevented cell death in the former while recruiting the latter into the cell cycle. Cell cycling, but not apoptosis, depended on the level of viral-encoded tax expression. Infected tax-expressing CD4+ lymphocytes accumulated cellular defects characteristic of genetic instability. Therefore, HTLV-1 infection establishes a preleukemic phenotype that is restricted to CD4+ infected clones.


Subject(s)
CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Human T-lymphotropic virus 1/pathogenicity , Adult , Apoptosis , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Carrier State/virology , Cell Cycle , Cell Proliferation , Cells, Cultured , Female , Humans , Male , Middle Aged , Preleukemia/metabolism , Preleukemia/virology , Time Factors
20.
J Med Primatol ; 38(4): 279-89, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19457158

ABSTRACT

BACKGROUND: Although a wide variety of non-human primates are susceptible to simian T-cell leukaemia virus type 1 (STLV-1), little is known about the virological or molecular determinants of natural STLV-1 infection. METHODS: We determined STLV-1 virus tropism in vivo and its relation to the immune response by evaluating cytokine production and T-cell subsets in naturally infected and uninfected mandrills. RESULTS: With real-time PCR methods, we found that STLV-1 in mandrills infects both CD4(+) and CD8(+) T cells; however, proviral loads were significantly higher (P = 0.01) in CD4(+) than in CD8(+) cells (mean STLV-1 copies number per 100 cells (+/- SD) was 7.8 +/- 8 in CD4(+) T cells and 3.9 +/- 4.5 in CD8(+) T cells). After culture, STLV-1 provirus was detected in enriched CD4(+) but not in enriched CD8(+) T cells. After 6 months of culture, STLV-1-transformed cell lines expressing CD3(+), CD4(+) and HLADR(+) were established, and STLV-1 proteins and tax/rex mRNA were detected. In STLV-1 infected monkeys, there was a correlation between high proviral load and elevated levels of interleukin (IL)-2, IL-6, IL-10, interferon-gamma and tumour necrosis factor-alpha. The two monkeys with the highest STLV-1 proviral load had activated CD4(+)HLADR(+) and CD8(+)HLADR(+) T-cell subsets and a high percentage of CD25(+) in CD4(+) and CD8(+) T cells. CONCLUSIONS: Our study provides the first cellular, immunological and virological characterization of natural STLV-1 infection in mandrills and shows that they are an appropriate animal model for further physiopathological studies of the natural history of human T-cell leukaemia viruses.


Subject(s)
Cytokines/metabolism , Mandrillus , Simian T-lymphotropic virus 1/immunology , T-Lymphocytes/physiology , Animals , Antibodies , Cells, Cultured , Chronic Disease , Gene Expression Profiling , Gene Expression Regulation, Viral/physiology , Polymerase Chain Reaction/veterinary , T-Lymphocytes/cytology , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL