Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Virol ; 91(24)2017 12 15.
Article in English | MEDLINE | ID: mdl-28978713

ABSTRACT

HIV-1-infected macrophages participate in virus dissemination and establishment of virus reservoirs in host tissues, but the mechanisms for virus cell-to-cell transfer to macrophages remain unknown. Here, we reveal the mechanisms for cell-to-cell transfer from infected T cells to macrophages and virus spreading between macrophages. We show that contacts between infected T lymphocytes and macrophages lead to cell fusion for the fast and massive transfer of CCR5-tropic viruses to macrophages. Through the merge of viral material between T cells and macrophages, these newly formed lymphocyte-macrophage fused cells acquire the ability to fuse with neighboring noninfected macrophages. Together, these two-step envelope-dependent cell fusion processes lead to the formation of highly virus-productive multinucleated giant cells reminiscent of the infected multinucleated giant macrophages detected in HIV-1-infected patients and simian immunodeficiency virus-infected macaques. These mechanisms represent an original mode of virus transmission for viral spreading and a new model for the formation of macrophage virus reservoirs during infection.IMPORTANCE We reveal a very efficient mechanism involved in cell-to-cell transfer from infected T cells to macrophages and subsequent virus spreading between macrophages by a two-step cell fusion process. Infected T cells first establish contacts and fuse with macrophage targets. The newly formed lymphocyte-macrophage fused cells then acquire the ability to fuse with surrounding uninfected macrophages, leading to the formation of infected multinucleated giant cells that can survive for a long time, as evidenced in vivo in lymphoid organs and the central nervous system. This route of infection may be a major determinant for virus dissemination and the formation of macrophage virus reservoirs in host tissues during HIV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Giant Cells/virology , HIV Infections/immunology , HIV-1/physiology , Macrophages/cytology , Animals , CD4-Positive T-Lymphocytes/virology , Cell Fusion , Cell Line , Giant Cells/cytology , HEK293 Cells , HIV-1/pathogenicity , Humans , Jurkat Cells , Macaca mulatta , Macrophages/virology , Simian Immunodeficiency Virus/pathogenicity , Simian Immunodeficiency Virus/physiology
2.
Cell Rep ; 20(9): 2044-2056, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28854357

ABSTRACT

Oxidative damage to telomere DNA compromises telomere integrity. We recently reported that the DNA glycosylase NEIL3 preferentially repairs oxidative lesions in telomere sequences in vitro. Here, we show that loss of NEIL3 causes anaphase DNA bridging because of telomere dysfunction. NEIL3 expression increases during S phase and reaches maximal levels in late S/G2. NEIL3 co-localizes with TRF2 and associates with telomeres during S phase, and this association increases upon oxidative stress. Mechanistic studies reveal that NEIL3 binds to single-stranded DNA via its intrinsically disordered C terminus in a telomere-sequence-independent manner. Moreover, NEIL3 is recruited to telomeres through its interaction with TRF1, and this interaction enhances the enzymatic activity of purified NEIL3. Finally, we show that NEIL3 interacts with AP Endonuclease 1 (APE1) and the long-patch base excision repair proteins PCNA and FEN1. Taken together, we propose that NEIL3 protects genome stability through targeted repair of oxidative damage in telomeres during S/G2 phase.


Subject(s)
Chromosome Segregation , DNA Damage , DNA Repair , Mitosis , N-Glycosyl Hydrolases/metabolism , S Phase , Telomere/pathology , CD4-Positive T-Lymphocytes/metabolism , Cell Cycle Checkpoints , Cell Nucleus/metabolism , DNA/metabolism , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Humans , Microtubules/metabolism , N-Glycosyl Hydrolases/chemistry , Oxidative Stress , Protein Binding , Protein Domains , Spindle Apparatus/metabolism
3.
Retrovirology ; 13: 26, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27068393

ABSTRACT

BACKGROUND: Through incorporation into virus particles, the HIV-1 Vpr protein participates in the early steps of the virus life cycle by influencing the reverse transcription process. We previously showed that this positive impact on reverse transcription was related to Vpr binding to the uracil DNA glycosylase 2 enzyme (UNG2), leading to enhancement of virus infectivity in established CD4-positive cell lines via a nonenzymatic mechanism. RESULTS: We report here that Vpr can form a trimolecular complex with UNG2 and the p32 subunit (RPA32) of the replication protein A (RPA) complex and we explore how these cellular proteins can influence virus replication and dissemination in the primary target cells of HIV-1, which express low levels of both proteins. Virus infectivity and replication in peripheral blood mononuclear cells and monocyte-derived macrophages (MDMs), as well as the efficiency of the viral DNA synthesis, were significantly reduced when viruses were produced from cells depleted of endogenous UNG2 or RPA32. Moreover, viruses produced in macrophages failed to replicate efficiently in UNG2- and RPA32-depleted T lymphocytes. Reciprocally, viruses produced in UNG2-depleted T cells did not replicate efficiently in MDMs confirming the positive role of UNG2 for virus dissemination. CONCLUSIONS: Our data show the positive effect of UNG2 and RPA32 on the reverse transcription process leading to optimal virus replication and dissemination between the primary target cells of HIV-1.


Subject(s)
DNA Glycosylases/metabolism , HIV-1/physiology , Host-Pathogen Interactions , Replication Protein A/metabolism , Reverse Transcription , Virus Replication , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Cells, Cultured , Humans , Leukocytes, Mononuclear/virology
4.
J Virol ; 89(6): 3247-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25568205

ABSTRACT

UNLABELLED: Tetraspanins constitute a family of cellular proteins that organize various membrane-based processes. Several members of this family, including CD81, are actively recruited by HIV-1 Gag to viral assembly and release sites. Despite their enrichment at viral exit sites, the overall levels of tetraspanins are decreased in HIV-1-infected cells. Here, we identify Vpu as the main viral determinant for tetraspanin downregulation. We also show that reduction of CD81 levels by Vpu is not a by-product of CD4 or BST-2/tetherin elimination from the surfaces of infected cells and likely occurs through an interaction between Vpu and CD81. Finally, we document that Vpu-mediated downregulation of CD81 from the surfaces of infected T cells can contribute to preserving the infectiousness of viral particles, thus revealing a novel Vpu function that promotes virus propagation by modulating the host cell environment. IMPORTANCE: The HIV-1 accessory protein Vpu has previously been shown to downregulate various host cell factors, thus helping the virus to overcome restriction barriers, evade immune attack, and maintain the infectivity of viral particles. Our study identifies tetraspanins as an additional group of host factors whose expression at the surfaces of infected cells is lowered by Vpu. While the downregulation of these integral membrane proteins, including CD81 and CD82, likely affects more than one function of HIV-1-infected cells, we document that Vpu-mediated lowering of CD81 levels in viral particles can be critical to maintaining their infectiousness.


Subject(s)
Down-Regulation , HIV Infections/genetics , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/metabolism , Tetraspanin 28/genetics , Viral Regulatory and Accessory Proteins/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions , Human Immunodeficiency Virus Proteins/genetics , Humans , Protein Binding , Tetraspanin 28/metabolism , Viral Regulatory and Accessory Proteins/genetics
5.
J Virol ; 88(13): 7645-58, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24760896

ABSTRACT

UNLABELLED: During cell-to-cell transmission of HIV-1, viral and cellular proteins transiently accumulate at the contact zone between infected (producer) and uninfected (target) cells, forming the virological synapse. Rearrangements of the cytoskeleton in producer and target cells are required for proper targeting of viral and cellular components during synapse formation, yet little is known about how these processes are regulated, particularly within the producer cell. Since ezrin-radixin-moesin (ERM) proteins connect F-actin with integral and peripheral membrane proteins, are incorporated into virions, and interact with cellular components of the virological presynapse, we hypothesized that they play roles during the late stage of HIV-1 replication. Here we document that phosphorylated (i.e., active) ezrin specifically accumulates at the HIV-1 presynapse in T cell lines and primary CD4(+) lymphocytes. To investigate whether ezrin supports virus transmission, we sought to ablate ezrin expression in producer cells. While cells did not tolerate a complete knockdown of ezrin, even a modest reduction of ezrin expression (~50%) in HIV-1-producing cells led to the release of particles with impaired infectivity. Further, when cocultured with uninfected target cells, ezrin-knockdown producer cells displayed reduced accumulation of the tetraspanin CD81 at the synapse and fused more readily with target cells, thus forming syncytia. Such an outcome likely is not optimal for virus dissemination, as evidenced by the fact that, in vivo, only relatively few infected cells form syncytia. Thus, ezrin likely helps secure efficient virus spread not only by enhancing virion infectivity but also by preventing excessive membrane fusion at the virological synapse. IMPORTANCE: While viruses, in principal, can propagate through successions of syncytia, HIV-1-infected cells in the majority of cases do not fuse with potential target cells during viral transmission. This mode of spread is coresponsible for key features of HIV-1 pathogenesis, including killing of bystander cells and establishment of latently infected T lymphocytes. Here we identify the ERM protein family member ezrin as a cellular factor that contributes to the inhibition of cell-cell fusion and thus to suppressing excessive syncytium formation. Our analyses further suggest that ezrin, which connects integral membrane proteins with actin, functions in concert with CD81, a member of the tetraspanin family of proteins. Additional evidence, documented here and elsewhere, suggests that ezrin and CD81 cooperate to prevent cytoskeleton rearrangements that need to take place during the fusion of cellular membranes.


Subject(s)
Cell Communication , Cytoskeletal Proteins/metabolism , HIV Infections/virology , HIV-1/physiology , Virus Internalization , Blotting, Western , Cell Fusion , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , Flow Cytometry , HEK293 Cells , HIV Infections/immunology , HIV Infections/metabolism , HeLa Cells , Humans , Phosphorylation , RNA, Small Interfering/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology
6.
Viruses ; 6(3): 1078-90, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24608085

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) transmission takes place primarily through cell-cell contacts known as virological synapses. Formation of these transient adhesions between infected and uninfected cells can lead to transmission of viral particles followed by separation of the cells. Alternatively, the cells can fuse, thus forming a syncytium. Tetraspanins, small scaffolding proteins that are enriched in HIV-1 virions and actively recruited to viral assembly sites, have been found to negatively regulate HIV-1 Env-induced cell-cell fusion. How these transmembrane proteins inhibit membrane fusion, however, is currently not known. As a first step towards elucidating the mechanism of fusion repression by tetraspanins, e.g., CD9 and CD63, we sought to identify the stage of the fusion process during which they operate. Using a chemical epistasis approach, four fusion inhibitors were employed in tandem with CD9 overexpression. Cells overexpressing CD9 were found to be sensitized to inhibitors targeting the pre-hairpin and hemifusion intermediates, while they were desensitized to an inhibitor of the pore expansion stage. Together with the results of a microscopy-based dye transfer assay, which revealed CD9- and CD63-induced hemifusion arrest, our investigations strongly suggest that tetraspanins block HIV-1-induced cell-cell fusion at the transition from hemifusion to pore opening.


Subject(s)
Cell Fusion , HIV-1/physiology , Membrane Fusion , Tetraspanin 29/metabolism , Tetraspanin 30/metabolism , Tetraspanins/metabolism , Cell Line , Host-Pathogen Interactions , Humans
7.
J Virol ; 87(13): 7516-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23637402

ABSTRACT

HIV-1 Env mediates virus attachment to and fusion with target cell membranes, and yet, while Env is still situated at the plasma membrane of the producer cell and before its incorporation into newly formed particles, Env already interacts with the viral receptor CD4 on target cells, thus enabling the formation of transient cell contacts that facilitate the transmission of viral particles. During this first encounter with the receptor, Env must not induce membrane fusion, as this would prevent the producer cell and the target cell from separating upon virus transmission, but how Env's fusion activity is controlled remains unclear. To gain a better understanding of the Env regulation that precedes viral transmission, we examined the nanoscale organization of Env at the surface of producer cells. Utilizing superresolution microscopy (stochastic optical reconstruction microscopy [STORM]) and fluorescence recovery after photobleaching (FRAP), we quantitatively assessed the clustering and dynamics of Env upon its arrival at the plasma membrane. We found that Gag assembly induced the aggregation of small Env clusters into larger domains and that these domains were completely immobile. Truncation of the cytoplasmic tail (CT) of Env abrogated Gag's ability to induce Env clustering and restored Env mobility at assembly sites, both of which correlated with increased Env-induced fusion of infected and uninfected cells. Hence, while Env trapping by Gag secures Env incorporation into viral particles, Env clustering and its sequestration at assembly sites likely also leads to the repression of its fusion function, and thus, by preventing the formation of syncytia, Gag helps to secure efficient transfer of viral particles to target cells.


Subject(s)
HIV Envelope Protein gp120/metabolism , Virus Assembly/physiology , Virus Attachment , Virus Internalization , Analysis of Variance , Cell Fusion , Fluorescence Recovery After Photobleaching , Gene Products, gag/metabolism , HeLa Cells , Humans , Microscopy/methods
8.
Retrovirology ; 6: 64, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19602278

ABSTRACT

BACKGROUND: The presence of the tetraspanins CD9, CD63, CD81 and CD82 at HIV-1 budding sites, at the virological synapse (VS), and their enrichment in HIV-1 virions has been well-documented, but it remained unclear if these proteins play a role in the late phase of the viral replication cycle. Here we used overexpression and knockdown approaches to address this question. RESULTS: Neither ablation of CD9, CD63 and/or CD81, nor overexpression of these tetraspanins was found to affect the efficiency of virus release. However, confirming recently reported data, tetraspanin overexpression in virus-producing cells resulted in the release of virions with substantially reduced infectivity. We also investigated the roles of these tetraspanins in cell-to-cell transmission of HIV-1. Overexpression of CD9 and CD63 led to reduced cell-to-cell transmission of this virus. Interestingly, in knockdown experiments we found that ablation of CD63, CD9 and/or CD81 had no effect on cell-free infectivity. However, knockdown of CD81, but not CD9 and CD63, enhanced productive particle transmission to target cells, suggesting additional roles for tetraspanins in the transmission process. Finally, tetraspanins were found to be downregulated in HIV-1-infected T lymphocytes, suggesting that HIV-1 modulates the levels of these proteins in order to maximize the efficiency of its transmission within the host. CONCLUSION: Altogether, these results establish an active role of tetraspanins in HIV-1 producer cells.


Subject(s)
HIV Infections/drug therapy , HIV Infections/virology , HIV Integrase Inhibitors/therapeutic use , HIV Integrase/genetics , HIV-1/genetics , Polymorphism, Genetic , Amino Acid Substitution/genetics , Drug Resistance, Viral , HIV Integrase Inhibitors/pharmacology , Humans , Molecular Sequence Data , Mutation, Missense , Sequence Analysis, DNA
9.
Gene ; 390(1-2): 52-66, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17088026

ABSTRACT

Botmar1 elements are mariner-like elements (MLEs), class II transposable elements that occur in the genome of the bumble bee, Bombus terrestris. Each haploid B. terrestris genome contains about 230 Botmar1, consisting entirely of 1.3-kb and 0.85-kb elements. During their evolution in the B. terrestris genome, two Botmar1 lineages have been differentiated in terms of their nucleic acid sequences and the differences found in their 5' untranslated regions suggest that they could be transcribed differently in B. terrestris. Here, we show that small amounts of Botmar1 mRNA occur in RNA extracts purified from B. terrestris imagoes. This indicates that the Botmar1 transcription is either weak in imagoes, or is restricted to very few cells. The cloning of several mRNAs reveals that only lineage-2 Botmar1 elements are transcribed. This transcription is specific, and uses cardinal initiators and terminators of eukaryotic elements in the Botmar1 elements. The intrastrand stem-loop folds in the mRNA theoretically synthesized by elements of the first lineage suggest that mRNA maintenance in cells might be self-regulated by RNA interference.


Subject(s)
Bees/genetics , DNA Transposable Elements , 3' Untranslated Regions , 5' Untranslated Regions , Animals , Base Sequence , Bees/growth & development , Bees/metabolism , DNA Transposable Elements/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Molecular Sequence Data , Nucleic Acid Conformation , Phylogeny , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid , Transcription, Genetic
10.
J Virol ; 81(1): 125-40, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17050592

ABSTRACT

The motifs involved in the various functions of the human immunodeficiency virus type 1 (HIV-1) gp41 cytoplasmic tail (CT), particularly those related to the intracellular trafficking and assembly of envelope glycoproteins (Env) onto core particles, have generally been assessed with a restricted panel of T-cell laboratory-adapted virus strains. Here, we investigated gp41 CT sequences derived from individuals infected with HIV-1 viruses of various subtypes. We identified four patients harboring HIV variants with a natural polymorphism in the membrane-proximal tyrosine-based signal Y(712)SPL or the Y(802)W(803) diaromatic motif, which are two major determinants of Env intracellular trafficking. Confocal microscopy showed that the intracellular distribution of Env with a mutation in the tyrosine or diaromatic motif differed from that of Env with no mutation in these motifs. Surprisingly, the gp41 CTs of the primary viruses also had differential effects on the intracellular distribution of Env, independently of mutations in the tyrosine or diaromatic motifs, suggesting the involvement of additional determinants. Furthermore, analyses of virus replication kinetics indicated that the effects of mutations in the tyrosine or diaromatic motifs on viral replication depended on the gp41 CT context. These effects were at least partly due to differences in the efficiency of Env incorporation into virions. Thus, polymorphisms in primary HIV-1 gp41 CTs at the quasispecies or subtype level can influence the intracellular distribution of Env, its incorporation into virions, and viral replication capacity.


Subject(s)
Gene Products, env/analysis , HIV Envelope Protein gp41/genetics , HIV-1/genetics , Polymorphism, Genetic , Virus Assembly/genetics , Amino Acid Motifs , Amino Acid Sequence , Gene Products, gag/metabolism , HIV Envelope Protein gp41/analysis , HIV Envelope Protein gp41/metabolism , HIV Infections/virology , HIV-1/physiology , HIV-1/ultrastructure , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Protein Transport , Sequence Alignment , Virion/metabolism , Virus Replication
11.
J Clin Microbiol ; 43(2): 750-4, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15695675

ABSTRACT

The hepatitis C virus (HCV) envelope protein 2 (E2) interacts in vitro with the interferon alpha (IFN-alpha)-inducible double-stranded RNA-activated protein kinase, suggesting a possible mechanism by which HCV may evade the antiviral effects of IFN-alpha. Variability in the part of the HCV E2 gene encoding the carboxy-terminal part of the protein, which includes the interaction domain (E2-PePHD), was explored in 25 patients infected with HCV genotype 1b and receiving IFN-alpha therapy. PCR products were generated and sequenced for 15 patients with a sustained response and for 10 patients with no virological response after treatment with IFN-alpha and ribavirin. PePHD amino acid sequences were obtained for isolates from serum collected before and during treatment, after 2 months in responders, and after 6 months in nonresponders. Quasispecies analysis of the pretreatment PePHD region was performed for isolates from patients displaying amino acid substitutions in this domain on direct sequencing. The E2-PePHD sequence was highly conserved in both resistant and susceptible genotype 1b strains and was identical to the prototype HCV type J sequence. No significant emergence of PePHD mutants during therapy was observed in our clonal analysis, and sporadic mutations and treatment outcomes were not found to be correlated. The PePHD sequence before or during treatment cannot be used to predict reliably the outcome of treatment in HCV type 1b-infected patients.


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Hepatitis C/drug therapy , Interferon-alpha/therapeutic use , Mutation , Ribavirin/therapeutic use , Viral Envelope Proteins/genetics , Amino Acid Sequence , Drug Therapy, Combination , Genotype , Hepacivirus/classification , Hepacivirus/genetics , Hepatitis C/virology , Humans , Molecular Sequence Data , Sequence Analysis, DNA , Treatment Outcome , Viral Envelope Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...