Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 13(6): e0199171, 2018.
Article in English | MEDLINE | ID: mdl-29944671

ABSTRACT

JC virus (JCV), a ubiquitous human polyomavirus, can cause fatal progressive multifocal leukoencephalopathy (PML) in immune compromised patients. The viral genome is composed of two conserved coding regions separated by a highly variable non-coding control region (NCCR). We analyzed the NCCR sequence from 10 PML JCV strains and found new mutations. Remarkably, the NCCR f section was mutated in most cases. We therefore explored the importance of this section in JCV expression in renal (HEK293H) and glioblastoma (U-87MG) cell lines, by adapting the emerging technology of DNA minicircles. Using bidirectional fluorescent reporters, we revealed that impaired NCCR-driven late expression in glioblastoma cells was restored by a short deletion overlapping e and f sections. This study evidenced a relevant link between JCV NCCR polymorphism and cell-type dependent expression. The use of DNA minicircles opens new insights for monitoring the impact of NCCR variation.


Subject(s)
Gene Expression Regulation, Viral , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/virology , Polyomavirus Infections/virology , Cell Line, Tumor , Genome, Viral , HEK293 Cells , Humans , Mutation , Polymorphism, Genetic , Untranslated Regions
2.
Retrovirology ; 12: 28, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25880753

ABSTRACT

BACKGROUND: The TRIM5α restriction factor interferes with retroviral infections by inhibiting an early step of viral replication. TRIM5α activity was recently proposed to be regulated by the SUMO machinery and one SUMO consensus conjugation site as well as three putative SUMO interacting motifs (SIMs) were identified within TRIM5α sequence. Whereas mutation of the SIM sequences was found to abolish TRIM5α antiviral activity, mutation of the consensus SUMO conjugation site did not affect its restriction capacity, although this putative site has never been shown to be actually a SUMO substrate. FINDINGS: Here we further demonstrate that TRIM5α relies on the SUMO machinery to promote restriction, since SUMO1 overexpression enhances TRIM5α-mediated retroviral inhibition whereas knockdown of SUMO1 or E2 SUMO conjugating enzyme Ubc9 prevents restriction. Furthermore, we show for the first time that TRIM5α is SUMOylated both in vitro and in cellulo and that Lysine 10 is the main SUMOylation site. Mutation of the consensus SUMO conjugation motif in position 10 abrogated SUMOylation at this position, but did not disrupt TRIM5α antiviral activity. CONCLUSIONS: Altogether, our results confirm that the SUMO machinery is involved in TRIM5α-mediated retroviral restriction, and demonstrate that TRIM5α is a SUMO 1 and SUMO 2 substrate. The inability to abrogate TRIM5α antiviral activity by mutating its main SUMO conjugation motif supports the notion that non-covalent interaction with SUMO or SUMOylated proteins rather than TRIM5α direct SUMOylation is required.


Subject(s)
HIV-1/immunology , Proteins/metabolism , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Cell Line , Humans , Macaca mulatta , Proteolysis , Sumoylation , Ubiquitin-Protein Ligases
3.
Biochimie ; 107 Pt B: 300-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25260582

ABSTRACT

Xenotropic Murine Leukemia Virus-related Virus (XMRV) is a new gammaretrovirus generated by genetic recombination between two murine endogenous retroviruses, PreXMRV1 and PreXMRV2, during passaging of human prostate cancer xenografts in laboratory mice. XMRV is representative of an early founder virus that jumps species from mouse to human cell lines. Relatively little information is available concerning the XMRV integrase (IN), an enzyme that catalyzes a key stage in the retroviral cycle, and whose sequence is conserved among replication competent retroviruses emerging from recombination between the murine endogenous PreXMRV-1 and PreXMRV-2 genomes. Previous studies have shown that IN inhibitors efficiently block XMRV multiplication in cells. We thus aimed at characterizing the biochemical properties and sensitivity of the XMRV IN to the raltegravir, dolutegravir, 118-D-24 and elvitegravir inhibitors in vitro. We report for the first time the purification and enzymatic characterization of recombinant XMRV IN. This IN, produced in Escherichia coli and purified under native conditions, is optimally active over a pH range of 7-8.5, in the presence of Mg(2+) (15 mM and 30 mM for 3'-processing and strand transfer, respectively) and is poorly sensitive to the addition of dithiothreitol. Raltegravir was shown to be a very potent inhibitor (IC50 âˆ¼ 30 nM) whereas dolutegravir and elvitegravir were less effective (IC50 âˆ¼ 230 nM and 650 nM, respectively). The 118-D-24 drug had no impact on XMRV IN activity. Interestingly, the substrate specificity of XMRV IN seems to be less marked compared to HIV-1 IN since XMRV IN is able to process various donor substrates that share little homology. Finally, our analysis revealed some original properties of the XMRV IN such as its relatively low sequence specificity.


Subject(s)
Integrase Inhibitors/pharmacology , Integrases/chemistry , Integrases/metabolism , Xenotropic murine leukemia virus-related virus/enzymology , Amino Acid Sequence , Dithiothreitol/pharmacology , HIV Integrase/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Hydrogen-Ion Concentration , Integrases/genetics , Integrases/isolation & purification , Molecular Sequence Data , Oxazines , Piperazines , Pyridones , Pyrrolidinones/pharmacology , Quinolones/pharmacology , Raltegravir Potassium , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sequence Homology, Amino Acid , Substrate Specificity , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
4.
Virology ; 456-457: 28-38, 2014 May.
Article in English | MEDLINE | ID: mdl-24889222

ABSTRACT

Xenotropic Murine leukemia virus-Related Virus (XMRV) directly arose from genetic recombinations between two endogenous murine retroviruses that occurred during human xenografts in laboratory mice. Studies on XMRV could thus bring clues on how a new retrovirus could circumvent barrier species. We observed that XMRV exhibits a weak promoter activity in human cells, similar to the transcription level of a Tat-defective HIV-1. Despite this low fitness, XMRV can efficiently propagate through the huge accumulation of viral copies (≈40 copies per cell) that compensates for the low expression level of individual proviruses. We further demonstrate that there is an inverse relationship between the maximum number of viral copies per infected cell and the level of viral expression, which is explained by viral envelope interference mechanisms. Low viral expression compensation by viral copy accumulation through delayed interference could a priori contribute to the propagation of others viruses following species jumps.


Subject(s)
Gammaretrovirus/physiology , Gene Expression , Proviruses/physiology , Transcription, Genetic , Virus Replication , Animals , Cell Line , Gammaretrovirus/genetics , Humans , Mice , Promoter Regions, Genetic , Proviruses/genetics , Transduction, Genetic
5.
PLoS One ; 8(7): e69691, 2013.
Article in English | MEDLINE | ID: mdl-23922776

ABSTRACT

Ku, a cellular complex required for human cell survival and involved in double strand break DNA repair and multiple other cellular processes, may modulate retroviral multiplication, although the precise mechanism through which it acts is still controversial. Recently, Ku was identified as a possible anti-human immunodeficiency virus type 1 (HIV-1) target in human cells, in two global approaches. Here we investigated the role of Ku on the HIV-1 replication cycle by analyzing the expression level of a panel of non-replicative lentiviral vectors expressing the green fluorescent protein in human colorectal carcinoma HCT 116 cells, stably or transiently depleted of Ku. We found that in this cellular model the depletion of Ku did not affect the efficiency of (pre-)integrative steps but decreased the early HIV-1 expression by acting at the transcriptional level. This negative effect was specific of the HIV-1 promoter, required the obligatory step of viral DNA integration and was reversed by transient depletion of p53. We also provided evidence on a direct binding of Ku to HIV-1 LTR in transduced cells. Ku not only promotes the early transcription from the HIV-1 promoter, but also limits the constitution of viral latency. Moreover, in the presence of a normal level of Ku, HIV-1 expression was gradually lost over time, likely due to the counter-selection of HIV-1-expressing cells. On the contrary, the reactivation of transgene expression from HIV-1 by means of trichostatin A- or tumor necrosis factor α-administration was enhanced under condition of Ku haplodepletion, suggesting a phenomenon of provirus latency. These observations plead in favor of the hypothesis that Ku has an impact on HIV-1 expression and latency at early- and mid-time after integration.


Subject(s)
DNA Helicases/metabolism , HIV-1/physiology , DNA Helicases/genetics , HCT116 Cells , Humans , Ku Autoantigen , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Virus Integration/physiology , Virus Latency/physiology
6.
PLoS One ; 4(3): e4894, 2009.
Article in English | MEDLINE | ID: mdl-19290053

ABSTRACT

BACKGROUND: Tripartite motif (TRIM) proteins constitute a family of proteins that share a conserved tripartite architecture. The recent discovery of the anti-HIV activity of TRIM5alpha in primate cells has stimulated much interest in the potential role of TRIM proteins in antiviral activities and innate immunity. PRINCIPAL FINDINGS: To test if TRIM genes are up-regulated during antiviral immune responses, we performed a systematic analysis of TRIM gene expression in human primary lymphocytes and monocyte-derived macrophages in response to interferons (IFNs, type I and II) or following FcgammaR-mediated activation of macrophages. We found that 27 of the 72 human TRIM genes are sensitive to IFN. Our analysis identifies 9 additional TRIM genes that are up-regulated by IFNs, among which only 3 have previously been found to display an antiviral activity. Also, we found 2 TRIM proteins, TRIM9 and 54, to be specifically up-regulated in FcgammaR-activated macrophages. CONCLUSIONS: Our results present the first comprehensive TRIM gene expression analysis in primary human immune cells, and suggest the involvement of additional TRIM proteins in regulating host antiviral activities.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation/drug effects , Interferons/pharmacology , Membrane Proteins/genetics , Humans , Macrophages/drug effects , Macrophages/metabolism , Phylogeny , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
7.
Retrovirology ; 5: 59, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18613956

ABSTRACT

BACKGROUND: TRIM5 alpha is a restriction factor that interferes with retroviral infections in a species-specific manner in primate cells. Although TRIM5 alpha is constitutively expressed, its expression has been shown to be up-regulated by type I interferon (IFN). Among primates, a particular case exists in owl monkey cells, which express a fusion protein between TRIM5 and cyclophilin A, TRIMCyp, specifically interfering with HIV-1 infection. No studies have been conducted so far concerning the possible induction of TRIMCyp by IFN. We investigated the consequences of IFN treatment on retroviral restriction in diverse primate cells and evaluated the implication of TRIM5 alpha or TRIMCyp in IFN-induced anti-retroviral activities. RESULTS: First, we show that human type I IFN can enhance TRIM5 alpha expression in human, African green monkey and macaque cells, as well as TRIMCyp expression in owl monkey cells. In TRIM5 alpha-expressing primate cell lines, type I IFN has little or no effect on HIV-1 infection, whereas it potentiates restriction activity against N-MLV in human and African green monkey cells. In contrast, type I IFN treatment of owl monkey cells induces a great enhancement of HIV-1 restriction, as well as a strain-tropism independent restriction of MLV. We were able to demonstrate that TRIM5 alpha is the main mediator of the IFN-induced activity against N-MLV in human and African green monkey cells, whereas TRIMCyp mediates the IFN-induced HIV-1 restriction enhancement in owl monkey cells. In contrast, the type I IFN-induced anti-MLV restriction in owl monkey cells is independent of TRIMCyp expression. CONCLUSION: Together, our observations indicate that both TRIM5 alpha and TRIMCyp are implicated in IFN-induced anti-retroviral response in primate cells. Furthermore, we found that type I IFN also induces a TRIMCyp-independent restriction activity specific to MLV in owl monkey cells.


Subject(s)
Antiviral Agents/pharmacology , Cyclophilin A/metabolism , HIV-1/drug effects , Interferons/pharmacology , Leukemia Virus, Murine/drug effects , Proteins/metabolism , Animals , Antiviral Restriction Factors , Aotus trivirgatus , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Chlorocebus aethiops , Cyclophilin A/genetics , Gene Expression Regulation , HIV-1/pathogenicity , HeLa Cells , Humans , Leukemia Virus, Murine/pathogenicity , Macaca mulatta , Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Vero Cells
8.
Retrovirology ; 3: 16, 2006 Mar 03.
Article in English | MEDLINE | ID: mdl-16515685

ABSTRACT

BACKGROUND: As it is the very first step of the HIV replication cycle, HIV entry represents an attractive target for the development of new antiviral drugs. In this context, fusion inhibitors are the third class of anti-HIV drugs to be used for treatment, in combination with nucleoside analogues and antiproteases. But the precise mechanism of HIV fusion mechanism is still unclear. Gp41 ectodomain-derived synthetic peptides represent ideal tools for clarifying this mechanism, in order to design more potent anti-HIV drugs. RESULTS: Two soluble trimeric recombinant gp41 proteins, termed Rgp41B and Rgp41A were designed. Both comprise the N- and C-terminal heptad repeat regions of the ectodomain of HIV-1 gp41, connected by a 7-residue hydrophilic linker, in order to mimic the trimeric fusogenic state of the transmembrane glycoprotein. Both recombinant proteins were found to inhibit HIV-1 entry into target cells in a dose-dependent manner. Rgp41A, the most potent inhibitor, was able to inhibit both X4 and R5 isolates into HeLa cells and primary T lymphocytes. X4 viruses were found to be more susceptible than R5 isolates to inhibition by Rgp41A. In order to elucidate how the trimeric recombinant gp41 protein can interfere with HIV-1 entry into target cells, we further investigated its mode of action. Rgp41A was able to bind gp120 but did not induce gp120-gp41 dissociation. Furthermore, this inhibitor could also interfere with a late step of the fusion process, following the mixing of lipids. CONCLUSION: Taken together, our results suggest that Rgp41A can bind to gp120 and also interfere with a late event of the fusion process. Interestingly, Rgp41A can block membrane fusion without preventing lipid mixing. Although further work will be required to fully understand its mode of action, our results already suggest that Rgp41A can interfere with multiple steps of the HIV entry process.


Subject(s)
Antiviral Agents/pharmacology , HIV Envelope Protein gp41/pharmacology , HIV/physiology , Recombinant Proteins/pharmacology , Antiviral Agents/chemistry , Cell Fusion , Cell Line , Gene Deletion , Gene Products, env/genetics , Genes, Reporter , HIV/isolation & purification , HIV Core Protein p24/analysis , HIV Core Protein p24/immunology , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/metabolism , HIV Infections/blood , HeLa Cells , Humans , Kidney , Lymphocyte Activation , Lymphocytes/immunology , Plasmids , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Virus Replication
9.
Retrovirology ; 2: 73, 2005 Nov 23.
Article in English | MEDLINE | ID: mdl-16305739

ABSTRACT

BACKGROUND: The persistence of latent HIV-1 reservoirs is the principal barrier preventing the eradication of HIV-1 infection in patients by current antiretroviral therapy. It is thus crucial to understand the molecular mechanisms involved in the establishment, maintenance and reactivation of HIV-1 latency. Since chromatin remodeling has been implicated in the transcriptional reactivation of the HIV-1 promoter, we assessed the role of the histone deacetylase inhibitor sodium butyrate (NaB) on two HIV-1 latently infected cell lines (U1 and ACH-2) gene expression. RESULTS: Analysis of microarrays data led us to select two candidate genes: NCoA3 (Nuclear Receptor Coactivator 3), a nuclear receptor coactivator and IRF8 (Interferon Regulatory Factor 8), an interferon regulatory factor. NCoA3 gene expression is upregulated following NaB treatment of latently infected cells whereas IRF8 gene expression is strongly downregulated in the promonocytic cell line following NaB treatment. Their differential expressions were confirmed at the transcriptional and translational levels. Moreover, NCoA3 gene expression was also upregulated after treatment of U1 and ACH-2 cells with phorbol myristyl acetate (PMA) but not trichostatin A (TSA) and after treatment with NaB of two others HIV-1 latently infected cell lines (OM10.1 and J1.1). IRF8 gene is only expressed in U1 cells and was also downregulated after treatment with PMA or TSA. Functional analyses confirmed that NCoA3 synergizes with Tat to enhance HIV-1 promoter transcription and that IRF8 represses the IRF1-mediated activation through the HIV-1 promoter Interferon-stimulated response element (ISRE). CONCLUSION: These results led us to postulate that NCoA3 could be involved in the transcriptional reactivation of the HIV-1 promoter from latency and that IRF8 may contribute to the maintenance of the latent state in the promonocytic cell line. Implication of these factors in the maintenance or reactivation of the viral latency may provide potential new targets to control HIV-1 replication in latent viral reservoirs.


Subject(s)
Acetyltransferases/genetics , HIV-1/physiology , Interferon Regulatory Factors/genetics , Trans-Activators/genetics , Virus Latency , Acetyltransferases/physiology , Butyrates/pharmacology , HIV Long Terminal Repeat , HIV-1/genetics , Histone Acetyltransferases , Humans , Interferon Regulatory Factors/physiology , Interferons/pharmacology , Jurkat Cells , Nuclear Receptor Coactivator 3 , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Protein Biosynthesis , RNA, Messenger/analysis , Response Elements/physiology , Trans-Activators/physiology , Transcription, Genetic , Virus Replication
10.
J Virol ; 79(3): 1930-3, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15650218

ABSTRACT

CXCR4 is one of two physiologically relevant human immunodeficiency type 1 (HIV-1) entry coreceptors. Studies of CXCR4 mutants have not clearly identified the determinants of coreceptor function and specificity. We therefore used a panel of monoclonal antibodies to further elucidate CXCR4 expression, structure, and function. Our findings show the existence of conformational subpopulations of CXCR4 that are in equilibrium on the cell surface but are not cell type specific as previously reported. HIV-1 X4 isolates can interact with multiple CXCR4 conformations in order to gain entry into target cells.


Subject(s)
Antibodies, Monoclonal/immunology , Epitope Mapping , HIV-1/pathogenicity , Receptors, CXCR4/chemistry , Receptors, CXCR4/immunology , Antibodies, Monoclonal/metabolism , Cell Line , HIV-1/metabolism , HeLa Cells , Humans , Point Mutation , Protein Conformation , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , T-Lymphocytes
11.
AIDS ; 17(17): 2537-9, 2003 Nov 21.
Article in English | MEDLINE | ID: mdl-14600528

ABSTRACT

We reassessed the infection ability of human primary preadipocytes. The use of X4, R5 or VSV-G-pseudotyped viral particles indicated that viral entry is the limiting step. However, transfection with HIV-1 receptors restored efficient infection. Analyses of CD4, CXCR4 and CCR5 expression on preadipocytes and adipocytes revealed that receptor co-expression levels did not permit HIV-1 entry into adipose cells from all biopsies tested. We concluded that adipose tissue cannot be infected with HIV-1 in vivo.


Subject(s)
Adipocytes/virology , HIV Infections/virology , HIV-1/pathogenicity , Transfection , Adipose Tissue/virology , CD4 Antigens/genetics , Gene Expression , HIV Infections/genetics , HIV-1/growth & development , HeLa Cells , Humans , Receptors, CCR5/genetics , Receptors, CXCR4/genetics
12.
FASEB J ; 16(10): 1254-6, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12153994

ABSTRACT

The concept that adipocytes belong to an essential endocrine system with some characteristics of immune cells has recently emerged. The aim of this paper is to present evidence of the expression of CD4, CXCR4, and CCR5 receptors by human adipocytes and to test whether adipose cells support HIV entry. Primary human preadipocytes were cultured and differentiated in vitro. Expression of the three receptors on preadipocytes and adipocytes was demonstrated by reverse transcriptase-polymerase chain reaction, immunocytochemical, and immunohistochemical analysis. Infection of adipose cells to HIV-1 was then investigated. The measurement of the viral p24 antigen in preadipocyte culture medium showed an increase of p24 levels between 24 and 72 h postexposure and then a progressive decrease to reach a low level at 10-15 days. Ten days after the infection test, supernatant of preadipocytes contained infectious particles able to infect the susceptible T-CD4 CEM cell line. The expression of viral proteins by adipocytes was confirmed using a fusion test. The presence of viral DNA was exhibited by gag-specific polymerase chain reaction, supporting the hypothesis of HIV-1 X4- and R5-virus entry in preadipocytes. Adipose cells represent the first cell type that does not belong to the immune system expressing all specific HIV receptors and may represent new HIV-1 target cells.


Subject(s)
Adipocytes/virology , CD4 Antigens/metabolism , HIV-1/metabolism , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , Receptors, Virus/metabolism , Adipocytes/metabolism , Biological Transport , Cells, Cultured , DNA, Viral/analysis , Gene Products, env/metabolism , HIV-1/genetics , Humans , Models, Biological
13.
J Gen Virol ; 80 ( Pt 8): 1975-1982, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10466793

ABSTRACT

A previous report from this laboratory described the isolation of the first CD4-independent human immunodeficiency virus type 1 isolate, m7NDK. This independence of CD4 is due to seven mutations located in the C2, V3 and C3 regions of the gp120 protein. The present report describes the entry features of the m5NDK virus, which contains five of the seven m7NDK mutations, located in the V3 loop and C3 region. The entry of this virus is strictly CD4-dependent but it can fuse with African green monkey (agm) COS-7 cells bearing human CD4 (h-CD4). This fusion is directly due to the five mutations in the envgene. It has also been shown that entry of m7NDK is CD4-independent in COS-7 cells. Since the wild-type NDK and m7NDK viruses use the human CXCR4 protein as co-receptor, agm-CXCR4 was cloned and used in transfection and fusion inhibition experiments to show that this receptor can be used by the m5 and m7NDK viruses. The wild-type NDK virus, which does not enter COS-7 cells, can use agm-CXCR4, but only when the receptor is transfected into target cells. Although co-receptor nature and expression levels are still major determinants of virus entry, this is the first case where a few mutations in the env gene can overcome this restriction.


Subject(s)
Genes, env , HIV Envelope Protein gp120/metabolism , HIV-1/genetics , HIV-1/metabolism , Peptide Fragments/metabolism , Receptors, CXCR4/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Line , Chemokine CXCL12 , Chemokines, CXC/metabolism , Chlorocebus aethiops , Cloning, Molecular , HIV Envelope Protein gp120/genetics , HIV-1/isolation & purification , HIV-1/physiology , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis , Peptide Fragments/genetics , Phenotype , Receptors, CXCR4/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...