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










Publication year range
1.
Cell Rep Med ; 5(4): 101483, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38579727

ABSTRACT

Immune cell phenotyping frequently detects lineage-unrelated receptors. Here, we report that surface receptors can be transferred from primary macrophages to CD4 T cells and identify the Fcγ receptor CD32 as driver and cargo of this trogocytotic transfer. Filamentous CD32+ nanoprotrusions deposit distinct plasma membrane patches onto target T cells. Transferred receptors confer cell migration and adhesion properties, and macrophage-derived membrane patches render resting CD4 T cells susceptible to infection by serving as hotspots for HIV-1 binding. Antibodies that recognize T cell epitopes enhance CD32-mediated trogocytosis. Such autoreactive anti-HIV-1 envelope antibodies can be found in the blood of HIV-1 patients and, consistently, the percentage of CD32+ CD4 T cells is increased in their blood. This CD32-mediated, antigen-independent cell communication mode transiently expands the receptor repertoire and functionality of immune cells. HIV-1 hijacks this mechanism by triggering the generation of trogocytosis-promoting autoantibodies to gain access to immune cells critical to its persistence.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Humans , CD4-Positive T-Lymphocytes , Receptors, IgG/metabolism , Autoantibodies/metabolism , Trogocytosis
2.
Cell Rep ; 43(3): 113941, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38478523

ABSTRACT

Resting CD4 T cells resist productive HIV-1 infection. The HIV-2/simian immunodeficiency virus protein viral accessory protein X (Vpx) renders these cells permissive to infection, presumably by alleviating blocks at cytoplasmic reverse transcription and subsequent nuclear import of reverse-transcription/pre-integration complexes (RTC/PICs). Here, spatial analyses using quantitative virus imaging techniques reveal that HIV-1 capsids containing RTC/PICs are readily imported into the nucleus, recruit the host dependency factor CPSF6, and translocate to nuclear speckles in resting CD4 T cells. Reverse transcription, however, remains incomplete, impeding proviral integration and viral gene expression. Vpx or pharmacological inhibition of the deoxynucleotide triphosphohydrolase (dNTPase) activity of the restriction factor SAM domain and HD domain-containing protein 1 (SAMHD1) increases levels of nuclear reverse-transcribed cDNA and facilitates HIV-1 integration. Nuclear import and intranuclear transport of viral complexes therefore do not pose important blocks to HIV-1 in resting CD4 T cells, and the limitation to reverse transcription by SAMHD1's dNTPase activity constitutes the main pre-integration block to infection.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Monomeric GTP-Binding Proteins , Animals , Humans , HIV-1/genetics , CD4-Positive T-Lymphocytes/metabolism , SAM Domain and HD Domain-Containing Protein 1/metabolism , HIV-2/genetics , Viral Regulatory and Accessory Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , HEK293 Cells
3.
mBio ; 13(2): e0370521, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35229634

ABSTRACT

Combinations of direct-acting antivirals are needed to minimize drug resistance mutations and stably suppress replication of RNA viruses. Currently, there are limited therapeutic options against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and testing of a number of drug regimens has led to conflicting results. Here, we show that cobicistat, which is an FDA-approved drug booster that blocks the activity of the drug-metabolizing proteins cytochrome P450-3As (CYP3As) and P-glycoprotein (P-gp), inhibits SARS-CoV-2 replication. Two independent cell-to-cell membrane fusion assays showed that the antiviral effect of cobicistat is exerted through inhibition of spike protein-mediated membrane fusion. In line with this, incubation with low-micromolar concentrations of cobicistat decreased viral replication in three different cell lines including cells of lung and gut origin. When cobicistat was used in combination with remdesivir, a synergistic effect on the inhibition of viral replication was observed in cell lines and in a primary human colon organoid. This was consistent with the effects of cobicistat on two of its known targets, CYP3A4 and P-gp, the silencing of which boosted the in vitro antiviral activity of remdesivir in a cobicistat-like manner. When administered in vivo to Syrian hamsters at a high dose, cobicistat decreased viral load and mitigated clinical progression. These data highlight cobicistat as a therapeutic candidate for treating SARS-CoV-2 infection and as a potential building block of combination therapies for COVID-19. IMPORTANCE The lack of effective antiviral treatments against SARS-CoV-2 is a significant limitation in the fight against the COVID-19 pandemic. Single-drug regimens have so far yielded limited results, indicating that combinations of antivirals might be required, as previously seen for other RNA viruses. Our work introduces the drug booster cobicistat, which is approved by the FDA and typically used to potentiate the effect of anti-HIV protease inhibitors, as a candidate inhibitor of SARS-CoV-2 replication. Beyond its direct activity as an antiviral, we show that cobicistat can enhance the effect of remdesivir, which was one of the first drugs proposed for treatment of SARS-CoV-2. Overall, the dual action of cobicistat as a direct antiviral and a drug booster can provide a new approach to design combination therapies and rescue the activity of compounds that are only partially effective in monotherapy.


Subject(s)
COVID-19 Drug Treatment , Hepatitis C, Chronic , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cobicistat , Cricetinae , Disease Progression , Humans , Mesocricetus , Pandemics , SARS-CoV-2 , Viral Load
4.
Cell Rep ; 38(7): 110387, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35134331

ABSTRACT

SARS-CoV-2 variants of concern (VOCs) display enhanced transmissibility and resistance to antibody neutralization. Comparing the early 2020 isolate EU-1 to the VOCs Alpha, Beta, and Gamma in mice transgenic for human ACE2 reveals that VOCs induce a broadened scope of symptoms, expand systemic infection to the gastrointestinal tract, elicit the depletion of natural killer cells, and trigger variant-specific cytokine production patterns. Gamma infections result in accelerated disease progression associated with increased immune activation and inflammation. All four SARS-CoV-2 variants induce pDC depletion in the lungs, paralleled by reduced interferon responses. Remarkably, VOCs also use the murine ACE2 receptor for infection to replicate in the lungs of wild-type animals, which induce cellular and innate immune responses that apparently curtail the spread of overt disease. VOCs thus display distinct intrinsic pathogenic properties with broadened tissue and host range. The enhanced pathogenicity of VOCs and their potential for reverse zoonotic transmission pose challenges to clinical and pandemic management.


Subject(s)
COVID-19/virology , Disease Models, Animal , SARS-CoV-2/physiology , SARS-CoV-2/pathogenicity , Animals , COVID-19/immunology , Cytokines/metabolism , Host Specificity , Immunity, Cellular , Immunity, Innate , Lung/immunology , Lung/virology , Mice , Species Specificity , Viral Load , Viral Tropism , Virulence , Virus Replication
5.
Mol Ther ; 30(5): 2005-2023, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35038579

ABSTRACT

Despite rapid development and deployment of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), clinically relevant modalities to curb the pandemic by directly attacking the virus on a genetic level remain highly desirable and are urgently needed. Here we comprehensively illustrate the capacity of adeno-associated virus (AAV) vectors co-expressing a cocktail of three short hairpin RNAs (shRNAs; RNAi triggers) directed against the SARS-CoV-2 RdRp and N genes as versatile and effective antiviral agents. In cultured monkey cells and human gut organoids, our most potent vector, SAVIOR (SARS virus repressor), suppressed SARS-CoV-2 infection to background levels. Strikingly, in control experiments using single shRNAs, multiple SARS-CoV-2 escape mutants quickly emerged from infected cells within 24-48 h. Importantly, such adverse viral adaptation was fully prevented with the triple-shRNA AAV vector even during long-term cultivation. In addition, AAV-SAVIOR efficiently purged SARS-CoV-2 in a new model of chronically infected human intestinal cells. Finally, intranasal AAV-SAVIOR delivery using an AAV9 capsid moderately diminished viral loads and/or alleviated disease symptoms in hACE2-transgenic or wild-type mice infected with human or mouse SARS-CoV-2 strains, respectively. Our combinatorial and customizable AAV/RNAi vector complements ongoing global efforts to control the coronavirus disease 2019 (COVID-19) pandemic and holds great potential for clinical translation as an original and flexible preventive or therapeutic antiviral measure.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antiviral Agents , COVID-19/prevention & control , Dependovirus , Mice , Pandemics , RNA Interference , RNA, Small Interfering/genetics , SARS-CoV-2/genetics
6.
Viruses ; 12(8)2020 08 07.
Article in English | MEDLINE | ID: mdl-32784757

ABSTRACT

Rapid large-scale testing is essential for controlling the ongoing pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The standard diagnostic pipeline for testing SARS-CoV-2 presence in patients with an ongoing infection is predominantly based on pharyngeal swabs, from which the viral RNA is extracted using commercial kits, followed by reverse transcription and quantitative PCR detection. As a result of the large demand for testing, commercial RNA extraction kits may be limited and, alternatively, non-commercial protocols are needed. Here, we provide a magnetic bead RNA extraction protocol that is predominantly based on in-house made reagents and is performed in 96-well plates supporting large-scale testing. Magnetic bead RNA extraction was benchmarked against the commercial QIAcube extraction platform. Comparable viral RNA detection sensitivity and specificity were obtained by fluorescent and colorimetric reverse transcription loop-mediated isothermal amplification (RT-LAMP) using a primer set targeting the N gene, as well as RT-qPCR using a primer set targeting the E gene, showing that the RNA extraction protocol presented here can be combined with a variety of detection methods at high throughput. Importantly, the presented diagnostic workflow can be quickly set up in a laboratory without access to an automated pipetting robot.


Subject(s)
Betacoronavirus/chemistry , Betacoronavirus/genetics , Clinical Laboratory Techniques/methods , Coronavirus Infections/virology , Pneumonia, Viral/virology , RNA, Viral/isolation & purification , Betacoronavirus/isolation & purification , COVID-19 , COVID-19 Testing , Coronavirus Infections/diagnosis , Humans , Magnetic Phenomena , Molecular Diagnostic Techniques/methods , Nucleic Acid Amplification Techniques/methods , Pandemics , Pneumonia, Viral/diagnosis , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction/methods , Reverse Transcription , SARS-CoV-2 , Sensitivity and Specificity
7.
J Immunol ; 201(9): 2731-2743, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30257886

ABSTRACT

HIV-1 Nef is a multifunctional protein that optimizes virus spread and promotes immune evasion of infected cells to accelerate disease progression in AIDS patients. As one of its activities, Nef reduces the motility of infected CD4+ T lymphocytes in confined space. In vivo, Nef restricts T lymphocyte homing to lymph nodes as it reduces the ability for extravasation at the diapedesis step. Effects of Nef on T lymphocyte motility are typically mediated by its ability to reduce actin remodeling. However, interference with diapedesis does not depend on residues in Nef required for inhibition of host cell actin dynamics. In search for an alternative mechanism by which Nef could alter T lymphocyte extravasation, we noted that the viral protein interferes with the polarization of primary human CD4+ T lymphocytes upon infection with HIV-1. Expression of Nef alone is sufficient to disrupt T cell polarization, and this effect is conserved among lentiviral Nef proteins. Nef acts by arresting the oscillation of CD4+ T cells between polarized and nonpolarized morphologies. Mapping studies identified the binding site for the Nef-associated kinase complex (NAKC) as critical determinant of this Nef activity and a NAKC-binding-deficient Nef variant fails to impair CD4+ T lymphocyte extravasation and homing to lymph nodes. These results thus imply the disruption of T lymphocyte polarity via its NAKC binding site as a novel mechanism by which lentiviral Nef proteins alter T lymphocyte migration in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Polarity/immunology , Chemotaxis, Leukocyte/immunology , Transendothelial and Transepithelial Migration/immunology , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Binding Sites , CD4-Positive T-Lymphocytes/immunology , Humans , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
8.
Proc Natl Acad Sci U S A ; 114(10): 2729-2734, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28228523

ABSTRACT

Early after entry into monocytes, macrophages, dendritic cells, and resting CD4 T cells, HIV encounters a block, limiting reverse transcription (RT) of the incoming viral RNA genome. In this context, dNTP triphosphohydrolase SAM domain and HD domain-containing protein 1 (SAMHD1) has been identified as a restriction factor, lowering the concentration of dNTP substrates to limit RT. The accessory lentiviral protein X (Vpx) proteins from the major simian immunodeficiency virus of rhesus macaque, sooty mangabey, and HIV-2 (SIVsmm/SIVmac/HIV-2) lineage packaged into virions target SAMHD1 for proteasomal degradation, increase intracellular dNTP pools, and facilitate HIV cDNA synthesis. We find that virion-packaged Vpx proteins from a second SIV lineage, SIV of red-capped mangabeys or mandrills (SIVrcm/mnd-2), increased HIV infection in resting CD4 T cells, but not in macrophages, and, unexpectedly, acted in the absence of SAMHD1 degradation, dNTP pool elevation, or changes in SAMHD1 phosphorylation. Vpx rcm/mnd-2 virion incorporation resulted in a dramatic increase of HIV-1 RT intermediates and viral cDNA in infected resting CD4 T cells. These analyses also revealed a barrier limiting HIV-1 infection of resting CD4 T cells at the level of nuclear import. Single amino acid changes in the SAMHD1-degrading Vpx mac239 allowed it to enhance early postentry steps in a Vpx rcm/mnd-2-like fashion. Moreover, Vpx enhanced HIV-1 infection of SAMHD1-deficient resting CD4 T cells of a patient with Aicardi-Goutières syndrome. These results indicate that Vpx, in addition to SAMHD1, overcomes a previously unappreciated restriction for lentiviruses at the level of RT that acts independently of dNTP concentrations and is specific to resting CD4 T cells.


Subject(s)
HIV Infections/genetics , Reverse Transcription/genetics , SAM Domain and HD Domain-Containing Protein 1/genetics , Viral Regulatory and Accessory Proteins/genetics , Animals , CD4-Positive T-Lymphocytes/virology , Genome, Viral/genetics , HIV Infections/virology , HIV-1/genetics , HIV-1/pathogenicity , HIV-2/genetics , HIV-2/pathogenicity , Host-Pathogen Interactions/genetics , Humans , Macaca mulatta/genetics , Macaca mulatta/virology , Monocytes/virology , Proteolysis , RNA, Viral/genetics , Virion/genetics , Virion/pathogenicity , Virus Replication/genetics
9.
J Biol Chem ; 290(45): 27345-27359, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26370074

ABSTRACT

Siglec-1 (sialoadhesin, CD169) is a surface receptor on human cells that mediates trans-enhancement of HIV-1 infection through recognition of sialic acid moieties in virus membrane gangliosides. Here, we demonstrate that mouse Siglec-1, expressed on the surface of primary macrophages in an interferon-α-responsive manner, captures murine leukemia virus (MLV) particles and mediates their transfer to proliferating lymphocytes. The MLV infection of primary B-cells was markedly more efficient than that of primary T-cells. The major structural protein of MLV particles, Gag, frequently co-localized with Siglec-1, and trans-infection, primarily of surface-bound MLV particles, efficiently occurred. To explore the role of sialic acid for MLV trans-infection at a submolecular level, we analyzed the potential of six sialic acid precursor analogs to modulate the sialylated ganglioside-dependent interaction of MLV particles with Siglec-1. Biosynthetically engineered sialic acids were detected in both the glycolipid and glycoprotein fractions of MLV producer cells. MLV released from cells carrying N-acyl-modified sialic acids displayed strikingly different capacities for Siglec-1-mediated capture and trans-infection; N-butanoyl, N-isobutanoyl, N-glycolyl, or N-pentanoyl side chain modifications resulted in up to 92 and 80% reduction of virus particle capture and trans-infection, respectively, whereas N-propanoyl or N-cyclopropylcarbamyl side chains had no effect. In agreement with these functional analyses, molecular modeling indicated reduced binding affinities for non-functional N-acyl modifications. Thus, Siglec-1 is a key receptor for macrophage/lymphocyte trans-infection of surface-bound virions, and the N-acyl side chain of sialic acid is a critical determinant for the Siglec-1/MLV interaction.


Subject(s)
Moloney murine leukemia virus/pathogenicity , Sialic Acid Binding Ig-like Lectin 1/chemistry , Sialic Acid Binding Ig-like Lectin 1/physiology , Animals , Binding Sites , Cell Line , Gangliosides/chemistry , Gangliosides/metabolism , Host-Pathogen Interactions/physiology , Humans , Interferon-alpha/physiology , Leukemia, Experimental/physiopathology , Leukemia, Experimental/virology , Lymphocytes/physiology , Lymphocytes/virology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/physiology , N-Acetylneuraminic Acid/chemistry , Receptors, Virus/chemistry , Receptors, Virus/physiology , Retroviridae Infections/physiopathology , Retroviridae Infections/virology , Sialic Acid Binding Ig-like Lectin 1/genetics , Tumor Virus Infections/physiopathology , Tumor Virus Infections/virology
10.
Nat Med ; 18(11): 1682-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22972397

ABSTRACT

Unlike activated CD4(+) T cells, resting CD4(+) T cells are highly resistant to productive HIV-1 infection. Early after HIV-1 entry, a major block limits reverse transcription of incoming viral genomes. Here we show that the deoxynucleoside triphosphate triphosphohydrolase SAMHD1 prevents reverse transcription of HIV-1 RNA in resting CD4(+) T cells. SAMHD1 is abundantly expressed in resting CD4(+) T cells circulating in peripheral blood and residing in lymphoid organs. The early restriction to infection in unstimulated CD4(+) T cells is overcome by HIV-1 or HIV-2 virions into which viral Vpx is artificially or naturally packaged, respectively, or by addition of exogenous deoxynucleosides. Vpx-mediated proteasomal degradation of SAMHD1 and elevation of intracellular deoxynucleotide pools precede successful infection by Vpx-carrying HIV. Resting CD4(+) T cells from healthy donors following SAMHD1 silencing or from a patient with Aicardi-Goutières syndrome homozygous for a nonsense mutation in SAMHD1 were permissive for HIV-1 infection. Thus, SAMHD1 imposes an effective restriction to HIV-1 infection in the large pool of noncycling CD4(+) T cells in vivo. Bypassing SAMHD1 was insufficient for the release of viral progeny, implicating other barriers at later stages of HIV replication. Together, these findings may unveil new ways to interfere with the immune evasion and T cell immunopathology of pandemic HIV-1.


Subject(s)
CD4-Positive T-Lymphocytes , HIV-1 , Monomeric GTP-Binding Proteins , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , HIV Infections , HIV-1/genetics , HIV-1/metabolism , HIV-2/genetics , HIV-2/metabolism , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Nervous System Malformations/genetics , Nervous System Malformations/virology , Reverse Transcription/genetics , SAM Domain and HD Domain-Containing Protein 1 , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Virion/genetics , Virion/growth & development , Virus Replication
11.
Chem Biol Drug Des ; 80(3): 349-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22650354

ABSTRACT

The self-assembling protein nanoparticle (SAPN) is an antigen-presenting system that has been shown to be suitable for use as a vaccine platform. The SAPN scaffold is based on the principles of icosahedral symmetry, beginning from a monomeric chain that self-assembles into an ordered oligomeric state. The monomeric chain contains two covalently linked α-helical coiled-coil domains, an N-terminal de novo-designed pentameric tryptophan zipper and a C-terminal de novo-designed trimeric leucine zipper, which assemble along the internal symmetry axes of an icosahedron. In this study, we incorporated the membrane proximal external region (MPER) of HIV-1 gp41 from HXB2 into the N-terminal pentamer, referred to as MPER-SAPN, attempting to reproduce the α-helical state of the 4E10 epitope while maintaining a structurally less-constrained 2F5 epitope. Sprague-Dawley rats were immunized with MPER-SAPNs, and their sera were analyzed for induced humoral anti-HIV-1 responses. We show that high membrane proximal external region-specific titers can be raised via the repetitive antigen display of MPER on the SAPN without the need for adjuvant. However, none of the sera displayed a detectable neutralizing activity against HIV-1. Thus, 4E10- and 2F5-like neutralizing antibodies could not be elicited by MPER conformationally restrained in the SAPN context.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/immunology , Epitopes/immunology , HIV Envelope Protein gp41/immunology , HIV Infections/prevention & control , HIV-1/immunology , AIDS Vaccines/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Epitopes/chemistry , Female , HIV Envelope Protein gp41/chemistry , HIV Infections/immunology , Humans , Immunity, Humoral , Immunization , Models, Molecular , Molecular Sequence Data , Nanoparticles/chemistry , Neutralization Tests , Protein Structure, Secondary , Rats , Rats, Sprague-Dawley
12.
Retrovirology ; 8: 9, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21310048

ABSTRACT

BACKGROUND: The cellular transmembrane protein CD317/BST-2/HM1.24/Tetherin restricts HIV-1 infection by physically tethering mature virions to the surface of infected cells. HIV-1 counteracts this restriction by expressing the accessory protein Vpu, yet the mechanism of this antagonism is incompletely understood. ß-TrCP is the substrate recognition domain of an E3 ubiquitin ligase complex that interacts with the di-serine motif S52/S56 in the cytoplasmic tail of Vpu to target the CD4 receptor for proteasomal degradation. Recently, it has been suggested that ß-TrCP is also critically involved in Vpu's ability to overcome the CD317-mediated virion release block. RESULTS: To test this model, we analyzed the consequences of several experimental strategies to interfere with the Vpu-ß-TrCP protein-protein interaction. Under these conditions, we studied effects of Vpu on expression and localization of CD317 and CD4, as well as on its ability to promote HIV-1 release. Our results demonstrate a strict requirement for Vpu's di-serine motif for degradation of CD4 and also CD317, reduction of cell surface exposure of CD317, and HIV-1 release enhancement. We further show a critical role of ß-TrCP2, but not of the structurally related ß-TrCP1 isoform, for Vpu-mediated degradation of both receptors. Most importantly, Vpu remained active in downregulating CD317 from the cell surface and in overcoming the HIV-1 release restriction in ß-TrCP-depleted cells. CONCLUSIONS: These results demonstrate that ß-TrCP is not strictly required for Vpu's ability to counteract the CD317-imposed virion release block and support the relevance of cell surface down-modulation of the restriction factor as a central mechanism of Vpu antagonism. Moreover, we propose the existence of a critical, yet to be identified cellular factor that interacts with Vpu via its di-serine motif to alter the trafficking of the restriction factor.


Subject(s)
Antigens, CD/metabolism , HIV-1/physiology , Human Immunodeficiency Virus Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Antigens, CD/genetics , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Down-Regulation , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HIV-1/genetics , HIV-1/metabolism , HeLa Cells , Human Immunodeficiency Virus Proteins/genetics , Humans , Viral Regulatory and Accessory Proteins/genetics , Virion/metabolism , Virion/physiology , Virus Release/physiology , beta-Transducin Repeat-Containing Proteins/genetics
13.
J Virol ; 84(8): 4089-94, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20147395

ABSTRACT

Vpu antagonizes human immunodeficiency virus type 1 (HIV-1) particle release inhibition by CD317/BST-2/Tetherin. Whether this Vpu activity strictly requires cellular depletion of the restriction factor is unclear. Here, we characterized CD317 variants with mutations in putative sorting or ubiquitination motifs. All mutants still potently impaired release of Vpu-defective HIV-1 and remained sensitive to Vpu-mediated release enhancement. Importantly, this virological antagonism correlated with surface downregulation of CD317 mutants by Vpu, while intracellular pools of these mutants, which were consistently depleted of the wild-type protein, were highly variable or even enhanced. Thus, Vpu can efficiently antagonize virion tethering in the absence of CD317 degradation.


Subject(s)
HIV-1/pathogenicity , Human Immunodeficiency Virus Proteins/physiology , Membrane Glycoproteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/physiology , Virulence Factors/physiology , Antigens, CD/genetics , GPI-Linked Proteins , Human Immunodeficiency Virus Proteins/genetics , Humans , Membrane Glycoproteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL