Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
2.
PLoS Pathog ; 15(10): e1008093, 2019 10.
Article in English | MEDLINE | ID: mdl-31600344

ABSTRACT

ISG20 is a broad spectrum antiviral protein thought to directly degrade viral RNA. However, this mechanism of inhibition remains controversial. Using the Vesicular Stomatitis Virus (VSV) as a model RNA virus, we show here that ISG20 interferes with viral replication by decreasing protein synthesis in the absence of RNA degradation. Importantly, we demonstrate that ISG20 exerts a translational control over a large panel of non-self RNA substrates including those originating from transfected DNA, while sparing endogenous transcripts. This activity correlates with the protein's ability to localize in cytoplasmic processing bodies. Finally, these functions are conserved in the ISG20 murine ortholog, whose genetic ablation results in mice with increased susceptibility to viral infection. Overall, our results posit ISG20 as an important defense factor able to discriminate the self/non-self origins of the RNA through translation modulation.


Subject(s)
Antiviral Agents/pharmacology , Exoribonucleases/pharmacology , Protein Biosynthesis , RNA, Viral/metabolism , Vesicular Stomatitis/immunology , Vesiculovirus/immunology , Virus Replication/drug effects , Animals , Exoribonucleases/physiology , HeLa Cells , Humans , Mice , Mice, Knockout , RNA Stability , RNA, Viral/genetics , Vesicular Stomatitis/drug therapy , Vesicular Stomatitis/virology , Vesiculovirus/drug effects
3.
PLoS Pathog ; 13(4): e1006353, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28426803

ABSTRACT

Human T lymphotropic Virus type 1 (HTLV-1) is the etiological agent of Adult T cell Leukemia/Lymphoma (ATLL) and HTLV-1-Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). Both CD4+ T-cells and dendritic cells (DCs) infected with HTLV-1 are found in peripheral blood from HTLV-1 carriers. We previously demonstrated that monocyte-derived IL-4 DCs are more susceptible to HTLV-1 infection than autologous primary T-cells, suggesting that DC infection precedes T-cell infection. However, during blood transmission, breast-feeding or sexual transmission, HTLV-1 may encounter different DC subsets present in the blood, the intestinal or genital mucosa respectively. These different contacts may impact HTLV-1 ability to infect DCs and its subsequent transfer to T-cells. Using in vitro monocyte-derived IL-4 DCs, TGF-ß DCs and IFN-α DCs that mimic DCs contacting HTLV-1 in vivo, we show here that despite their increased ability to capture HTLV-1 virions, IFN-α DCs restrict HTLV-1 productive infection. Surprisingly, we then demonstrate that it is not due to the antiviral activity of type-I interferon produced by IFN-α DCs, but that it is likely to be linked to a distinct trafficking route of HTLV-1 in IL-4 DCs vs. IFN-α DCs. Finally, we demonstrate that, in contrast to IL-4 DCs, IFN-α DCs are impaired in their capacity to transfer HTLV-1 to CD4 T-cells, both after viral capture and trans-infection and after their productive infection. In conclusion, the nature of the DCs encountered by HTLV-1 upon primo-infection and the viral trafficking route through the vesicular pathway of these cells determine the efficiency of viral transmission to T-cells, which may condition the fate of infection.


Subject(s)
Antiviral Agents/pharmacology , Cytokines/immunology , Dendritic Cells/immunology , HTLV-I Infections/immunology , Human T-lymphotropic virus 1/immunology , Paraparesis, Tropical Spastic/immunology , Adult , Dendritic Cells/virology , HTLV-I Infections/transmission , HTLV-I Infections/virology , Human T-lymphotropic virus 1/pathogenicity , Human T-lymphotropic virus 1/physiology , Humans , Interferon Type I/immunology , Models, Biological , Paraparesis, Tropical Spastic/pathology , Paraparesis, Tropical Spastic/virology , T-Lymphocytes/immunology , T-Lymphocytes/virology
5.
Virologie (Montrouge) ; 23(5): 16-31, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31826846

ABSTRACT

Currently, more than 10% of human cancers are associated with viral infection. Studies on oncoviruses led to the development of clinical intervention strategies and elucidated fundamental cellular events altered upon cell transformation. Cancer cells exhibit several hallmarks including genomic instability, defined as a high frequency of mutations including gain or loss of chromosomes. The centrosome is an organelle that governs mitotic chromosome segregation and that functions as a signaling platform downstream of the DNA damage response. Here, we review the current literature to highlight how oncoviruses induce genomic instability via the deregulation of the centrosome. Viral interference with the centrosome duplication cycle, leading to centrosome amplification, is illustrated, with a special emphasis on mechanisms shared by several viral families. In addition, we discuss how oncoviruses could alter the signaling functions of the centrosome, and we comment on the bibliographic gaps that could be addressed by future research.


Subject(s)
Aneuploidy , Genomic Instability , Mitosis , Cell Transformation, Neoplastic/genetics , Centrosome , Genomic Instability/genetics , Humans , Mitosis/genetics
6.
Virologie (Montrouge) ; 23(5): 304-320, 2019 10 01.
Article in French | MEDLINE | ID: mdl-31826851

ABSTRACT

Currently, more than 10% of human cancers are associated with viral infection. Studies on oncoviruses led to the development of clinical intervention strategies and elucidated fundamental cellular events altered upon cell transformation. Cancer cells exhibit several hallmarks including genomic instability, defined as a high frequency of mutations including gain or loss of chromosomes. The centrosome is an organelle that governs mitotic chromosome segregation and that functions as a signaling platform downstream of the DNA damage response. Here, we review the current literature to highlight how oncoviruses induce genomic instability via the deregulation of the centrosome. Viral interference with the centrosome duplication cycle, leading to centrosome amplification, is illustrated, with a special emphasis on mechanisms shared by several viral families. In addition, we discuss how oncoviruses could alter the signaling functions of the centrosome, and we comment on the bibliographic gaps that could be addressed by future research.


Subject(s)
Aneuploidy , Genomic Instability , Mitosis , Cell Transformation, Neoplastic/genetics , Centrosome , Genomic Instability/genetics , Humans , Mitosis/genetics
7.
J Virol ; 90(17): 7607-17, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27334587

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) and human T-lymphotropic virus type 1 (HTLV-1) are complex retroviruses mainly infecting CD4(+) T lymphocytes. In addition, antigen-presenting cells such as dendritic cells (DCs) are targeted in vivo by both viruses, although to a lesser extent. Interaction of HIV-1 with DCs plays a key role in viral dissemination from the mucosa to CD4(+) T lymphocytes present in lymphoid organs. While similar mechanisms may occur for HTLV-1 as well, most HTLV-1 data were obtained from T-cell studies, and little is known regarding the trafficking of this virus in DCs. We first compared the efficiency of cell-free versus cell-associated viral sources of both retroviruses at infecting DCs. We showed that both HIV-1 and HTLV-1 cell-free particles are poorly efficient at productively infecting DCs, except when DC-SIGN has been engaged. Furthermore, while SAMHD-1 accounts for restriction of cell-free HIV-1 infection, it is not involved in HTLV-1 restriction. In addition, cell-free viruses lead mainly to a nonproductive DC infection, leading to trans-infection of T-cells, a process important for HIV-1 spread but not for that of HTLV-1. Finally, we show that T-DC cell-to-cell transfer implies viral trafficking in vesicles that may both increase productive infection of DCs ("cis-infection") and allow viral escape from immune surveillance. Altogether, these observations allowed us to draw a model of HTLV-1 and HIV-1 trafficking in DCs.


Subject(s)
HIV-1/physiology , Human T-lymphotropic virus 1/physiology , Virus Internalization , Virus Release , Biological Transport , CD4-Positive T-Lymphocytes/virology , Dendritic Cells/virology , Humans , Models, Biological , Virus Replication
8.
J Virol ; 89(15): 8092-5, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25995252

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1)-induced adult T-cell leukemia/lymphoma is an aggressive malignancy. HTLV-2 is genetically related to HTLV-1 but does not cause any malignant disease. HTLV-1 Tax transactivator (Tax-1) contributes to leukemogenesis via NF-κB. We describe transgenic Drosophila models expressing Tax in the compound eye and plasmatocytes. We demonstrate that Tax-1 but not Tax-2 induces ommatidial perturbation and increased plasmatocyte proliferation and that the eye phenotype is dependent on Kenny (IKKγ/NEMO), thus validating this new in vivo model.


Subject(s)
Cell Transformation, Viral , Drosophila melanogaster/virology , Gene Products, tax/metabolism , HTLV-I Infections/virology , Human T-lymphotropic virus 1/physiology , Animals , Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eye/pathology , Eye/virology , Gene Products, tax/genetics , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 2/genetics , Human T-lymphotropic virus 2/metabolism , Humans
9.
J Virol ; 89(2): 931-51, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25355890

ABSTRACT

UNLABELLED: Human T-cell lymphotropic virus type 1 (HTLV-1) and HTLV-2 encode auxiliary proteins that play important roles in viral replication, viral latency, and immune escape. The presence of auxiliary protein-encoding open reading frames (ORFs) in HTLV-3, the latest HTLV to be discovered, is unknown. Simian T-cell lymphotropic virus type 3 (STLV-3) is almost identical to HTLV-3. Given the lack of HTLV-3-infected cell lines, we took advantage of STLV-3-infected cells and of an STLV-3 molecular clone to search for the presence of auxiliary transcripts. Using reverse transcriptase PCR (RT-PCR), we first uncovered the presence of three unknown viral mRNAs encoding putative proteins of 5, 8, and 9 kDa and confirmed the presence of the previously reported RorfII transcript. The existence of these viral mRNAs was confirmed by using splice site-specific RT-PCR with ex vivo samples. We showed that p5 is distributed throughout the cell and does not colocalize with a specific organelle. The p9 localization is similar to that of HTLV-1 p12 and induced a strong decrease in the calreticulin signal, similarly to HTLV-1 p12. Although p8, RorfII, and Rex-3 share an N-terminal sequence that is predicted to contain a nucleolar localization signal (NoLS), only p8 is found in the nucleolus. The p8 location in the nucleolus is linked to a bipartite NoLS. p8 and, to a lesser extent, p9 repressed viral expression but did not alter Rex-3-dependent mRNA export. Using a transformation assay, we finally showed that none of the STLV-3 auxiliary proteins had the ability to induce colony formation, while both Tax-3 and antisense protein of HTLV-3 (APH-3) promoted cellular transformation. Altogether, these results complete the characterization of the newly described primate T-lymphotropic virus type 3 (PTLV-3). IMPORTANCE: Together with their simian counterparts, HTLVs form the primate T-lymphotropic viruses. HTLVs arose from interspecies transmission between nonhuman primates and humans. HTLV-1 and HTLV-2 encode auxiliary proteins that play important roles in viral replication, viral latency, and immune escape. The presence of ORFs encoding auxiliary proteins in HTLV-3 or STLV-3 genomes was unknown. Using in silico analyses, ex vivo samples, or in vitro experiments, we have uncovered the presence of 3 previously unknown viral mRNAs encoding putative proteins and confirmed the presence of a previously reported viral transcript. We characterized the intracellular localization of the four proteins. We showed that two of these proteins repress viral expression but that none of them have the ability to induce colony formation. However, both Tax and the antisense protein APH-3 promote cell transformation. Our results allowed us to characterize 4 new retroviral proteins for the first time.


Subject(s)
Gene Expression Profiling , Simian T-lymphotropic virus 3/genetics , Simian T-lymphotropic virus 3/physiology , Viral Proteins/analysis , Viral Proteins/genetics , Animals , Cell Line , Cell Nucleus/chemistry , Cytosol/chemistry , Humans , Molecular Weight , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/chemistry
10.
Retrovirology ; 11: 93, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25389016

ABSTRACT

BACKGROUND: The role of innate immunity in general and of type I interferon (IFN-I) in particular in HTLV-1 pathogenesis is still a matter of debate. ADAR1-p150 is an Interferon Stimulated Gene (ISG) induced by IFN-I that can edit viral RNAs. We therefore investigated whether it could play the role of an anti-HTLV factor. RESULTS: We demonstrate here that ADAR1 is also expressed in the absence of IFN stimulation in activated primary T-lymphocytes that are the natural target of this virus and in HTLV-1 or HTLV-2 chronically infected T-cells. ADAR1 expression is also increased in primary lymphocytes obtained from HTLV-1 infected individuals. We show that ADAR1 enhances HTLV-1 and HTLV-2 infection in T-lymphocytes and that this proviral effect is independent from its editing activity. ADAR1 expression suppresses IFN-α inhibitory effect on HTLV-1 and HTLV-2 and acts through the repression of PKR phosphorylation. DISCUSSION: This study demonstrates that two interferon stimulated genes, i.e. PKR and ADAR1 have opposite effects on HTLV replication in vivo. The balanced expression of those proteins could determine the fate of the viral cycle in the course of infection.


Subject(s)
Adenosine Deaminase/metabolism , Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 2/physiology , RNA-Binding Proteins/metabolism , Virus Replication , eIF-2 Kinase/antagonists & inhibitors , Cells, Cultured , Humans , Inhibition, Psychological , Molecular Sequence Data , Sequence Analysis, DNA , T-Lymphocytes/immunology , T-Lymphocytes/virology
11.
J Virol ; 87(2): 1123-36, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23135727

ABSTRACT

Permanent activation of the NF-κB pathway by the human T cell leukemia virus type 1 (HTLV-1) Tax (Tax1) viral transactivator is a key event in the process of HTLV-1-induced T lymphocyte immortalization and leukemogenesis. Although encoding a Tax transactivator (Tax2) that activates the canonical NF-κB pathway, HTLV-2 does not cause leukemia. These distinct pathological outcomes might be related, at least in part, to distinct NF-κB activation mechanisms. Tax1 has been shown to be both ubiquitinated and SUMOylated, and these two modifications were originally proposed to be required for Tax1-mediated NF-κB activation. Tax1 ubiquitination allows recruitment of the IKK-γ/NEMO regulatory subunit of the IKK complex together with Tax1 into centrosome/Golgi-associated cytoplasmic structures, followed by activation of the IKK complex and RelA/p65 nuclear translocation. Herein, we compared the ubiquitination, SUMOylation, and acetylation patterns of Tax2 and Tax1. We show that, in contrast to Tax1, Tax2 conjugation to endogenous ubiquitin and SUMO is barely detectable while both proteins are acetylated. Importantly, Tax2 is neither polyubiquitinated on lysine residues nor ubiquitinated on its N-terminal residue. Consistent with these observations, Tax2 conjugation to ubiquitin and Tax2-mediated NF-κB activation is not affected by overexpression of the E2 conjugating enzyme Ubc13. We further demonstrate that a nonubiquitinable, non-SUMOylable, and nonacetylable Tax2 mutant retains a significant ability to activate transcription from a NF-κB-dependent promoter after partial activation of the IKK complex and induction of RelA/p65 nuclear translocation. Finally, we also show that Tax2 does not interact with TRAF6, a protein that was shown to positively regulate Tax1-mediated activation of the NF-κB pathway.


Subject(s)
Gene Products, tax/metabolism , Human T-lymphotropic virus 2/pathogenicity , NF-kappa B/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Ubiquitin/metabolism , Acetylation , HeLa Cells , Humans , Jurkat Cells , Protein Processing, Post-Translational
12.
J Virol ; 87(24): 13386-96, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089560

ABSTRACT

Type I interferon (IFN-I) inhibits the replication of different viruses. However, the effect of IFN-I on the human T-lymphotropic virus type 1 (HTLV-1) viral cycle is controversial. Here, we investigated the consequences of IFN-α addition for different steps of HTLV-1 and HTLV-2 infection. We first show that alpha interferon (IFN-α) efficiently impairs HTLV-1 and HTLV-2 de novo infection in a T cell line and in primary lymphocytes. Using pseudotyped viruses expressing HTLV-1 envelope, we then show that cell-free infection is insensitive to IFN-α, demonstrating that the cytokine does not affect the early stages of the viral cycle. In contrast, intracellular levels of Gag, Env, or Tax protein are affected by IFN-α treatment in T cells, primary lymphocytes, or 293T cells transfected with HTLV-1 or HTLV-2 molecular clones, demonstrating that IFN-α acts during the late stages of infection. We show that IFN-α does not affect Tax-mediated transcription and acts at a posttranscriptional level. Using either small interfering RNA (siRNA) directed against PKR or a PKR inhibitor, we demonstrate that PKR, whose expression is induced by interferon, plays a major role in IFN-α-induced HTLV-1/2 inhibition. These results indicate that IFN-α has a strong repressive effect on the HTLV-1 and HTLV-2 viral cycle during de novo infection of cells that are natural targets of the viruses.


Subject(s)
HTLV-I Infections/enzymology , HTLV-II Infections/enzymology , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 2/physiology , Interferon-alpha/metabolism , eIF-2 Kinase/metabolism , Cell Line , Enzyme Activation , HTLV-I Infections/genetics , HTLV-I Infections/virology , HTLV-II Infections/genetics , HTLV-II Infections/virology , Host-Pathogen Interactions , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 2/genetics , Humans , Interferon alpha-2 , Recombinant Proteins/metabolism , eIF-2 Kinase/genetics
13.
Virologie (Montrouge) ; 16(6): 356-370, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-31910554

ABSTRACT

Innate immunity plays a critical role in the host response to a viral infection. In particular, type I interferons (IFN-I) are major effectors of antiviral innate immunity. Herein, interplays between HTLV-1 and the IFN-I response are reviewed. Particular emphasis is put on virus sensing by innate immunity receptors and on anti-HTLV-1 effects of IFN-I. We also discuss HTLV-1-induced alteration of IFN-I function and how IFN-I/AZT treatment of adult T-cell leukemia/lymphoma patients can lead to complete remission despite virus-induced escape mechanisms.

14.
Viruses ; 14(8)2022 07 23.
Article in English | MEDLINE | ID: mdl-35893677

ABSTRACT

Human T cell leukemia virus type 1 (HTLV-1), the etiological agent of adult T cell leukemia/lymphoma (ATLL) and of HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), was identified a few years before Human Immunodeficiency Virus (HIV). However, forty years later, our comprehension of HTLV-1 immune detection and the host immune responses to HTLV-1 is far more limited than for HIV. In addition to innate and adaptive immune responses that rely on specialized cells of the immune system, host cells may also express a range of antiviral factors that inhibit viral replication at different stages of the cycle, in a cell-autonomous manner. Multiple antiviral factors allowing such an intrinsic immunity have been primarily and extensively described in the context HIV infection. Here, we provide an overview of whether known HIV restriction factors might act on HTLV-1 replication. Interestingly, many of them do not exert any antiviral activity against HTLV-1, and we discuss viral replication cycle specificities that could account for these differences. Finally, we highlight future research directions that could help to identify antiviral factors specific to HTLV-1.


Subject(s)
HIV Infections , HTLV-I Infections , Human T-lymphotropic virus 1 , Leukemia-Lymphoma, Adult T-Cell , Paraparesis, Tropical Spastic , Adult , Antiviral Agents , Humans
15.
Life Sci Alliance ; 5(7)2022 07.
Article in English | MEDLINE | ID: mdl-35396335

ABSTRACT

The InterFeron-Induced TransMembrane proteins (IFITMs) are members of the dispanin/CD225 family that act as broad viral inhibitors by preventing viral-to-cellular membrane fusion. In this study, we uncover egress from the Golgi as an important step in the biology of IFITM3 by identifying the domain that regulates this process and that similarly controls the egress of the dispanins IFITM1 and PRRT2, protein linked to paroxysmal kinesigenic dyskinesia. In the case of IFITM3, high levels of expression of wild-type, or mutations in the Golgi egress domain, lead to accumulation of IFITM3 in the Golgi and drive generalized glycoprotein trafficking defects. These defects can be relieved upon incubation with Amphotericin B, compound known to relieve IFITM-driven membrane fusion defects, as well as by v-SNARE overexpression, suggesting that IFITM3 interferes with membrane fusion processes important for Golgi functionalities. The comparison of glycoprotein trafficking in WT versus IFITMs-KO cells indicates that the modulation of the secretory pathway is a novel feature of IFITM proteins. Overall, our study defines a novel domain that regulates the egress of several dispanin/CD225 members from the Golgi and identifies a novel modulatory function for IFITM3.


Subject(s)
Membrane Proteins , RNA-Binding Proteins , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , Secretory Pathway , Virus Internalization
16.
PLoS Pathog ; 5(7): e1000521, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19609363

ABSTRACT

Nuclear factor (NF)-kappaB is a major survival pathway engaged by the Human T-Lymphotropic Virus type 1 (HTLV-1) Tax protein. Tax1 activation of NF-kappaB occurs predominantly in the cytoplasm, where Tax1 binds NF-kappaB Essential Modulator (NEMO/IKKgamma) and triggers the activation of IkappaB kinases. Several independent studies have shown that Tax1-mediated NF-kappaB activation is dependent on Tax1 ubiquitination. Here, we identify by co-immunoprecipitation assays NEMO-Related Protein (NRP/Optineurin) as a binding partner for Tax1 in HTLV-1 infected and Tax1/NRP co-expressing cells. Immunofluorescence studies reveal that Tax1, NRP and NEMO colocalize in Golgi-associated structures. The interaction between Tax1 and NRP requires the ubiquitin-binding activity of NRP and the ubiquitination sites of Tax1. In addition, we observe that NRP increases the ubiquitination of Tax1 along with Tax1-dependent NF-kappaB signaling. Surprisingly, we find that in addition to Tax1, NRP interacts cooperatively with the Tax1 binding protein TAX1BP1, and that NRP and TAX1BP1 cooperate to modulate Tax1 ubiquitination and NF-kappaB activation. Our data strongly suggest for the first time that NRP is a critical adaptor that regulates the assembly of TAX1BP1 and post-translationally modified forms of Tax1, leading to sustained NF-kappaB activation.


Subject(s)
Gene Products, tax/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Transcription Factor TFIIIA/metabolism , Binding Sites , Cell Cycle Proteins , Cell Line, Tumor , Gene Products, tax/genetics , Golgi Apparatus , HeLa Cells , Humans , Immunoprecipitation , Intracellular Space/metabolism , Membrane Transport Proteins , Protein Interaction Domains and Motifs , Two-Hybrid System Techniques , Ubiquitination
17.
Blood ; 114(12): 2427-38, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19602711

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) and type 2 (HTLV-2) retroviruses infect T lymphocytes. The minus strand of the HTLV-1 genome encodes HBZ, a protein that could play a role in the development of leukemia in infected patients. Herein, we demonstrate that the complementary strand of the HTLV-2 genome also encodes a protein that we named APH-2 for "antisense protein of HTLV-2." APH-2 mRNA is spliced, polyadenylated, and initiates in the 3'-long terminal repeat at different positions. This transcript was detected in all HTLV-2-infected cell lines and short-term culture of lymphocytes obtained from HTLV-2 African patients tested and in 4 of 15 HTLV-2-infected blood donors. The APH-2 protein is 183 amino acids long, is localized in the cell nucleus, and is detected in vivo. Despite the lack of a consensus basic leucine zipper domain, APH-2 interacts with cyclic adenosine monophosphate-response element binding protein (CREB) and represses Tax2-mediated transcription in Tax2-expressing cells and in cells transfected with an HTLV-2 molecular clone. Altogether, our results demonstrate the existence of an antisense strand-encoded protein in HTLV-2, which could represent an important player in the development of disorders, such as lymphocytosis, which is frequently observed in HTLV-2 patients.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Gene Products, tax/genetics , Human T-lymphotropic virus 2/physiology , RNA Splicing/genetics , RNA, Antisense/genetics , Transcription, Genetic , Viral Proteins/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , Blotting, Northern , Blotting, Western , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Viral , Gene Products, tax/metabolism , Humans , Immunoenzyme Techniques , Immunoprecipitation , Jurkat Cells , Luciferases/genetics , Luciferases/metabolism , Middle Aged , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Transfection , Viral Proteins/metabolism
18.
Virologie (Montrouge) ; 20(3): 141-142, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-33065891
19.
Virologie (Montrouge) ; 15(4): 222-234, 2011 Aug 01.
Article in French | MEDLINE | ID: mdl-36151672

ABSTRACT

Viruses represent an important cause of cancer in humans: infections are estimated to account for close to one cancer case out of five.With the ongoing discovery of new infectious agents, this number should be raising in the near future. In 2006, the discovery of a new _-retrovirus in prostate cancer biopsies launched an intense research activity: could this new xenotropic MLV-related virus (XMRV) be the cause of prostate cancer? Five years later, the initial enthusiasm of retrovirologists has dramatically diminished. One by one, arguments favouring the hypothesis of human infection with XMRV are being refuted. The aim of this review article is to present the discovery of XMRV and to analyze recent data arguing against its existence in humans. A synthetic interpretation of XMRV literature will then be suggested.

20.
Pathogens ; 10(8)2021 Aug 09.
Article in English | MEDLINE | ID: mdl-34451465

ABSTRACT

The years 2020 and 2021 will remain memorable years for many reasons [...].

SELECTION OF CITATIONS
SEARCH DETAIL