Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Retrovirology ; 17(1): 36, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33228686

ABSTRACT

BACKGROUND: A reservoir of replication-competent but latent virus is the main obstacle to a cure for HIV-1 infection. Much of this reservoir resides in memory CD4 T cells. We hypothesized that these cells can be reactivated with antigens from HIV-1 and other common pathogens to reverse latency. RESULTS: We obtained mononuclear cells from the peripheral blood of antiretroviral-treated patients with suppressed viremia. We tested pools of peptides and proteins derived from HIV-1 and from other pathogens including CMV for their ability to reverse latency ex vivo by activation of memory responses. We assessed activation of the CD4 T cells by measuring the up-regulation of cell-surface CD69. We assessed HIV-1 expression using two assays: a real-time PCR assay for virion-associated viral RNA and a droplet digital PCR assay for cell-associated, multiply spliced viral mRNA. Reversal of latency occurred in a minority of cells from some participants, but no single antigen induced HIV-1 expression ex vivo consistently. When reversal of latency was induced by a specific peptide pool or protein, the extent was proportionally greater than that of T cell activation. CONCLUSIONS: In this group of patients in whom antiretroviral therapy was started during chronic infection, the latent reservoir does not appear to consistently reside in CD4 T cells of a predominant antigen-specificity. Peptide-antigens reversed HIV-1 latency ex vivo with modest and variable activity. When latency was reversed by specific peptides or proteins, it was proportionally greater than the extent of T cell activation, suggesting partial enrichment of the latent reservoir in cells of specific antigen-reactivity.


Subject(s)
Antigens/immunology , HIV Infections/virology , HIV-1/physiology , Virus Latency/immunology , Adult , Aged , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Dendritic Cells/immunology , Female , HIV-1/immunology , Humans , Immunologic Memory , Interferon-gamma/metabolism , Male , Middle Aged , Muromegalovirus/immunology , RNA, Messenger/metabolism , RNA, Viral/metabolism , Virion/metabolism , Virus Activation/immunology
3.
Virology ; 548: 73-81, 2020 09.
Article in English | MEDLINE | ID: mdl-32838948

ABSTRACT

The host protein SERINC5 inhibits the infectivity of HIV-1 virions in an Env-dependent manner and is counteracted by Nef. The conformation of the Env trimer reportedly correlates with sensitivity to SERINC5. Here, we tested the hypothesis that the "open" conformation of the Env trimer revealed by sensitivity to the V3-loop specific antibody 447-52D directly correlates with sensitivity to SERINC5. Of five Envs tested, SF162 was the most sensitive to neutralization by 447-52D, but it was not the most sensitive to SERINC5; instead the Env of LAI was substantially more sensitive to SERINC5 than all the other Envs. Mutational opening of the trimer by substitution of two tyrosines that mediate interaction between the V2 and V3 loops sensitized the Envs of JRFL and LAI to 447-52D as previously reported, but only BaL was sensitized to SERINC5. These data suggest that trimer "openness" is not sufficient for sensitivity to SERINC5.


Subject(s)
Antibodies, Monoclonal/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Membrane Proteins/immunology , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/immunology , Antibodies, Neutralizing/immunology , HIV Infections/genetics , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , HIV-1/physiology , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutation , env Gene Products, Human Immunodeficiency Virus/genetics
4.
Clin Infect Dis ; 60(6): 900-9, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25416754

ABSTRACT

BACKGROUND: The Shingles Prevention Study (SPS) demonstrated zoster vaccine efficacy through 4 years postvaccination. A Short-Term Persistence Substudy (STPS) demonstrated persistence of vaccine efficacy for at least 5 years. A Long-Term Persistence Substudy (LTPS) was undertaken to further assess vaccine efficacy in SPS vaccine recipients followed for up to 11 years postvaccination. Study outcomes were assessed for the entire LTPS period and for each year from 7 to 11 years postvaccination. METHODS: Surveillance, case determination, and follow-up were comparable to those in SPS and STPS. Because SPS placebo recipients were offered zoster vaccine before the LTPS began, there were no unvaccinated controls. Instead, SPS and STPS placebo results were used to model reference placebo groups. RESULTS: The LTPS enrolled 6867 SPS vaccine recipients. Compared to SPS, estimated vaccine efficacy in LTPS decreased from 61.1% to 37.3% for the herpes zoster (HZ) burden of illness (BOI), from 66.5% to 35.4% for incidence of postherpetic neuralgia, and from 51.3% to 21.1% for incidence of HZ, and declined for all 3 outcome measures from 7 through 11 years postvaccination. Vaccine efficacy for the HZ BOI was significantly greater than zero through year 10 postvaccination, whereas vaccine efficacy for incidence of HZ was significantly greater than zero only through year 8. CONCLUSIONS: Estimates of vaccine efficacy decreased over time in the LTPS population compared with modeled control estimates. Statistically significant vaccine efficacy for HZ BOI persisted into year 10 postvaccination, whereas statistically significant vaccine efficacy for incidence of HZ persisted only through year 8.


Subject(s)
Herpes Zoster Vaccine , Herpes Zoster/prevention & control , Aged , Aged, 80 and over , Cost of Illness , Epidemiological Monitoring , Female , Follow-Up Studies , Herpes Zoster/complications , Herpes Zoster/epidemiology , Herpes Zoster Vaccine/adverse effects , Herpes Zoster Vaccine/immunology , Humans , Incidence , Male , Middle Aged , Neuralgia, Postherpetic/epidemiology , Neuralgia, Postherpetic/prevention & control , Vaccination , Vaccine Potency
5.
J Virol ; 88(1): 763-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24155399

ABSTRACT

Alpha interferon (IFN-α) suppresses human immunodeficiency virus type 1 (HIV-1) replication in vitro by inducing cell-intrinsic retroviral restriction mechanisms. We investigated the effects of IFN-α/ribavirin (IFN-α/riba) treatment on 34 anti-HIV-1 restriction factors in vivo. Expression of several anti-HIV-1 restriction factors was significantly induced by IFN-α/riba in HIV/hepatitis C virus (HCV)-coinfected individuals. Fold induction of cumulative restriction factor expression in CD4(+) T cells was significantly correlated with viral load reduction during IFN-α/riba treatment (r(2) = 0.649; P < 0.016). Exogenous IFN-α induces supraphysiologic restriction factor expression associated with a pronounced decrease in HIV-1 viremia.


Subject(s)
HIV/immunology , Interferon-alpha/therapeutic use , Ribavirin/immunology , Virus Replication/immunology
6.
J Infect Dis ; 208(4): 559-63, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23633406

ABSTRACT

BACKGROUND: After completion of the Shingles Prevention Study (SPS; Department of Veterans Affairs Cooperative Studies Program Number 403), SPS participants who had initially received placebo were offered investigational zoster vaccine without charge. This provided an opportunity to determine the relative safety of zoster vaccine in older adults following documented herpes zoster (HZ). METHODS: A total of 13 681 SPS placebo recipients who elected to receive zoster vaccine were followed for serious adverse events (SAE) for 28 days after vaccination. In contrast to the SPS, a prior episode of HZ was not a contraindication to receiving zoster vaccine. The SPS placebo recipients who received zoster vaccine included 420 who had developed documented HZ during the SPS. RESULTS: The mean interval between the onset of HZ and the receipt of zoster vaccine in the 420 recipients with prior HZ was 3.61 years (median interval, 3.77 years [range, 3-85 months]); the interval was <5 years for approximately 80% of recipients. The proportion of vaccinated SPS placebo recipients with prior HZ who developed ≥ 1 SAE (0.95%) was not significantly different from that of vaccinated SPS placebo recipients with no prior history of HZ (0.66%), and the distribution of SAEs in the 2 groups was comparable. CONCLUSIONS: These results demonstrate that the general safety of zoster vaccine in older persons is not altered by a recent history of documented HZ, supporting the safety aspect of the Centers for Disease Control and Prevention Advisory Committee on Immunization Practices recommendation to administer zoster vaccine to all persons ≥ 60 years of age with no contraindications, regardless of a prior history of HZ.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/epidemiology , Herpes Zoster Vaccine/administration & dosage , Herpes Zoster Vaccine/adverse effects , Herpes Zoster/immunology , Aged , Aged, 80 and over , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Male , Middle Aged
7.
J Virol ; 85(1): 51-63, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980512

ABSTRACT

The HIV-1 protein Vpu counteracts the antiviral activity of the innate restriction factor BST-2/tetherin by a mechanism that partly depends on its interaction with ß-TrCP, a substrate adaptor for an SCF (Skp-Cullin 1-F box) E3 ubiquitin ligase complex. This suggests that Vpu stimulates the ubiquitination of BST-2 and that this underlies the relief of restriction. Here, we show that Vpu stimulates ubiquitination of BST-2. Mutation of all potential ubiquitination sites in the cytoplasmic domain of BST-2, including lysines, cysteines, serines, and threonines, abrogates Vpu-mediated ubiquitination. However, a serine-threonine-serine sequence specifically mediates the downregulation of BST-2 from the cell surface and the optimal relief of restricted virion release. Serine-threonine ubiquitination of BST-2 is likely part of the mechanism by which Vpu counteracts innate defenses.


Subject(s)
Antigens, CD/metabolism , Down-Regulation , Human Immunodeficiency Virus Proteins/metabolism , Ubiquitin/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Virus Release/physiology , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , HIV-1/genetics , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Molecular Sequence Data , Serine/metabolism , Threonine/metabolism , Virion/metabolism , beta-Transducin Repeat-Containing Proteins/genetics , beta-Transducin Repeat-Containing Proteins/metabolism
8.
Trends Microbiol ; 18(9): 388-96, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20688520

ABSTRACT

The interferon-inducible, transmembrane protein BST-2 (CD317, tetherin) directly holds fully formed enveloped virus particles to the cells that produce them, inhibiting their spread. BST-2 inhibits members of the retrovirus, filovirus, arenavirus and herpesvirus families. These viruses encode a variety of proteins to degrade BST-2 and/or direct it away from its site of action at the cell surface. Viral antagonism has subjected BST-2 to positive selection, leading to species-specific differences that presented a barrier to the transmission of simian immunodeficiency viruses (SIVs) to humans. This barrier was crossed by HIV-1 when its Vpu protein acquired activity as a BST-2 antagonist. Here, we review this new host-pathogen relationship and discuss its impact on the evolution of primate lentiviruses and the origins of the HIV pandemic.


Subject(s)
Antigens, CD/metabolism , HIV-1/immunology , HIV-1/physiology , Human Immunodeficiency Virus Proteins/metabolism , Immunity, Innate , Lentiviruses, Primate/physiology , Membrane Glycoproteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Acquired Immunodeficiency Syndrome/epidemiology , Acquired Immunodeficiency Syndrome/virology , Animals , Antigens, CD/chemistry , Antigens, CD/immunology , Cell Membrane/immunology , Cell Membrane/virology , Evolution, Molecular , GPI-Linked Proteins , HIV-1/metabolism , Host-Pathogen Interactions , Humans , Lentiviruses, Primate/genetics , Lentiviruses, Primate/immunology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/chemistry , Primates , Simian Immunodeficiency Virus/physiology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism
9.
Virology ; 406(2): 312-21, 2010 Oct 25.
Article in English | MEDLINE | ID: mdl-20708210

ABSTRACT

Pathogenic simian-human immunodeficiency viruses (SHIV) contain HIV-1 Vpu and SIV Nef, both shown to counteract BST-2 (HM1.24; CD317; tetherin) inhibition of virus release in a species-specific manner. We show that human and pig-tailed BST-2 (ptBST-2) restrict SHIV. We found that sequential "humanization" of the transmembrane domain (TMD) of the pig-tailed BST-2 (ptBST-2) protein resulted in a fluctuation in sensitivity to HIV-1 Vpu. Our results also show that the length of the TMD in human and ptBST-2 proteins is important for BST-2 restriction and susceptibility to Vpu. Taken together, our results emphasize the importance of tertiary structure in BST-2 antagonism and suggests that the HIV-1 Vpu transmembrane domain may have additional functions in vivo unrelated to BST-2 antagonism.


Subject(s)
Antigens, CD/metabolism , HIV Infections/metabolism , HIV-1/physiology , Membrane Glycoproteins/metabolism , Reassortant Viruses/physiology , Simian Immunodeficiency Virus/physiology , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Cell Line , GPI-Linked Proteins , Gene Products, nef/genetics , Gene Products, nef/metabolism , HIV Infections/virology , HIV-1/genetics , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Macaca mulatta , Macaca nemestrina , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Molecular Sequence Data , Protein Structure, Tertiary , Reassortant Viruses/genetics , Sequence Alignment , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
10.
Retrovirology ; 7: 51, 2010 Jun 07.
Article in English | MEDLINE | ID: mdl-20529266

ABSTRACT

BACKGROUND: In the absence of the Vpu protein, newly formed HIV-1 particles can remain attached to the surface of human cells due to the action of an interferon-inducible cellular restriction factor, BST-2/tetherin. Tetherin also restricts the release of other enveloped viral particles and is counteracted by a several viral anti-tetherin factors including the HIV-2 Env, SIV Nef and KSHV K5 proteins. RESULTS: We observed that a fraction of tetherin is located at the surface of restricting cells, and that co-expression of both HIV-1 Vpu and HIV-2 Env reduced this population. In addition, Vpu, but not the HIV-2 Env, reduced total cellular levels of tetherin. An additional effect observed for both Vpu and the HIV-2 Env was to redirect tetherin to an intracellular perinuclear compartment that overlapped with markers for the TGN (trans-Golgi network). Sequestration of tetherin in this compartment was independent of tetherin's normal endocytosis trafficking pathway. CONCLUSIONS: Both HIV-1 Vpu and HIV-2 Env redirect tetherin away from the cell surface and sequester the protein in a perinuclear compartment, which likely blocks the action of this cellular restriction factor. Vpu also promotes the degradation of tetherin, suggesting that it uses more than one mechanism to counteract tetherin restriction.


Subject(s)
Antigens, CD/metabolism , HIV Envelope Protein gp160/metabolism , HIV-1/pathogenicity , HIV-2/pathogenicity , Host-Pathogen Interactions , Human Immunodeficiency Virus Proteins/metabolism , Membrane Glycoproteins/metabolism , Protein Interaction Mapping , Viral Regulatory and Accessory Proteins/metabolism , Cell Line , Cell Membrane/chemistry , Endoplasmic Reticulum/chemistry , GPI-Linked Proteins , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Protein Binding
11.
Curr HIV Res ; 8(3): 240-52, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20201792

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) and several simian immunodeficiency viruses (SIV) encode for a transmembrane protein known as Vpu (viral protein U). While one of the smallest of the HIV-1 proteins, it has two important functions within virus-infected cells. The first of these functions is the down-regulation of the CD4 receptor to prevent its interaction with the HIV-1 envelope glycoprotein. Vpu interacts with the CD4 receptor in the rough endoplasmic reticulum (RER), resulting in its re-translocation across the RER and subsequent degradation via the proteasomal pathway. The second major function of the Vpu protein is to facilitate release of virus from infected cells. Previous studies have shown that virus release is cell type specific, suggesting that certain cells may express a restriction factor that inhibits virus release in the absence of Vpu. Recently, bone marrow stromal antigen 2 (BST-2/HM1.24/CD317/tetherin) has been identified as this factor. This review will focus on new findings within the last four years on the role of Vpu in CD4 down-regulation and the restriction of virus release from cells. We will relate these findings to virus pathogenesis and propose questions regarding BST-2 as a restriction factor.


Subject(s)
CD4 Antigens/biosynthesis , HIV-1/physiology , Human Immunodeficiency Virus Proteins/physiology , Simian Immunodeficiency Virus/physiology , Viral Regulatory and Accessory Proteins/physiology , Virulence Factors/physiology , Virus Release , Animals , Down-Regulation , HIV-1/pathogenicity , Humans , Simian Immunodeficiency Virus/pathogenicity
12.
Curr Top Microbiol Immunol ; 339: 27-45, 2009.
Article in English | MEDLINE | ID: mdl-20012522

ABSTRACT

Viral protein U (Vpu) is encoded by one of several accessory genes of HIV-1. Like the accessory gene products Vif and Vpr, Vpu targets host proteins such as CD4 for degradation via the recruitment of cellular multi-subunit ubiquitin ligases. Vpu also forms ion channels in cellular membranes. Through one or both of these attributes, Vpu antagonizes host cell factors that restrict the release of progeny virions from infected cells. A key target of Vpu has recently been identified as the interferon-induced transmembrane protein BST-2/CD317 (tetherin), which restricts viral replication by retaining nascent virions on the cell surface. The counteraction of this host defense allows Vpu to be considered an antagonist of the innate immune response to viral infection.


Subject(s)
Host-Pathogen Interactions , Human Immunodeficiency Virus Proteins/physiology , Viral Regulatory and Accessory Proteins/physiology , Adaptor Proteins, Signal Transducing/metabolism , Antigens, CD/metabolism , GPI-Linked Proteins , Human Immunodeficiency Virus Proteins/chemistry , Humans , Immune Evasion , Membrane Glycoproteins/metabolism , Protein Transport , Viral Regulatory and Accessory Proteins/chemistry , Virus Release , beta-Transducin Repeat-Containing Proteins/metabolism
13.
PLoS One ; 4(12): e8364, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-20020046

ABSTRACT

BACKGROUND: The down-regulation of the major histocompatibility complex class I (MHC-I) from the surface of infected cells by the Nef proteins of primate immunodeficiency viruses likely contributes to pathogenesis by providing evasion of cell-mediated immunity. HIV-1 Nef-induced down-regulation involves endosomal trafficking and a cooperative interaction between the cytoplasmic domain (CD) of MHC-I, Nef, and the clathrin adaptor protein complex-1 (AP-1). The CD of MHC-I contains a key tyrosine within the sequence YSQA that is required for down-regulation by Nef, but this sequence does not conform to the canonical AP-binding tyrosine-based motif Yxxphi, which mediates binding to the medium (micro) subunits of AP complexes. We previously proposed that Nef allows the MHC-I CD to bind the mu subunit of AP-1 (micro1) as if it contained a Yxxphimotif. METHODS AND FINDINGS: Here, we show that a direct interaction between the MHC-I CD/Nef and micro1 plays a primary role in the down-regulation of MHC-I: GST pulldown assays using recombinant proteins indicated that most of the MHC-I CD and Nef residues that are required for the down-regulation in human cells contribute to direct interactions with a truncated version of micro1. Specifically, the tyrosine residue of the YSQA sequence in the MHC-I CD as well as Nef residues E62-65 and P78 each contributed to the interaction between MHC-I CD/Nef and micro1 in vitro, whereas Nef M20 had little to no role. Conversely, residues F172/D174 and V392/L395 of the binding pocket on micro1 for Yxxphi motifs were required for a robust interaction. CONCLUSIONS: These data indicate that the MHC-I cytoplasmic domain, Nef, and the C-terminal two thirds of the mu subunit of AP-1 are sufficient to constitute a biologically relevant interaction. The data also reveal an unexpected role for a hydrophobic pocket in micro1 for interaction with MHC-I CD/Nef.


Subject(s)
Adaptor Protein Complex mu Subunits/metabolism , Endosomes/metabolism , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Adaptor Protein Complex mu Subunits/genetics , Amino Acids/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cell Membrane/metabolism , Cytoplasm/chemistry , Down-Regulation , Histocompatibility Antigens Class I/genetics , Humans , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics
14.
PLoS Pathog ; 5(5): e1000450, 2009 May.
Article in English | MEDLINE | ID: mdl-19478868

ABSTRACT

The interferon-induced transmembrane protein BST-2/CD317 (tetherin) restricts the release of diverse enveloped viruses from infected cells. The HIV-1 accessory protein Vpu antagonizes this restriction by an unknown mechanism that likely involves the down-regulation of BST-2 from the cell surface. Here, we show that the optimal removal of BST-2 from the plasma membrane by Vpu requires the cellular protein beta-TrCP, a substrate adaptor for a multi-subunit SCF E3 ubiquitin ligase complex and a known Vpu-interacting protein. beta-TrCP is also required for the optimal enhancement of virion-release by Vpu. Mutations in the DSGxxS beta-TrCP binding-motif of Vpu impair both the down-regulation of BST-2 and the enhancement of virion-release. Such mutations also confer dominant-negative activity, consistent with a model in which Vpu links BST-2 to beta-TrCP. Optimal down-regulation of BST-2 from the cell surface by Vpu also requires the endocytic clathrin adaptor AP-2, although the rate of endocytosis is not increased; these data suggest that Vpu induces post-endocytic membrane trafficking events whose net effect is the removal of BST-2 from the cell surface. In addition to its marked effect on cell-surface levels, Vpu modestly decreases the total cellular levels of BST-2. The decreases in cell-surface and intracellular BST-2 are inhibited by bafilomycin A1, an inhibitor of endosomal acidification; these data suggest that Vpu induces late endosomal targeting and partial degradation of BST-2 in lysosomes. The Vpu-mediated decrease in surface expression is associated with reduced co-localization of BST-2 and the virion protein Gag along the plasma membrane. Together, the data support a model in which Vpu co-opts the beta-TrCP/SCF E3 ubiquitin ligase complex to induce endosomal trafficking events that remove BST-2 from its site of action as a virion-tethering factor.


Subject(s)
HIV-1/pathogenicity , Human Immunodeficiency Virus Proteins/physiology , Lysosomes/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/physiology , beta-Transducin Repeat-Containing Proteins/metabolism , Adaptor Protein Complex 2 , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Line , Down-Regulation , Endosomes/metabolism , GPI-Linked Proteins , HIV-1/physiology , Human Immunodeficiency Virus Proteins/genetics , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mutation , Protein Transport , Viral Regulatory and Accessory Proteins/genetics , beta-Transducin Repeat-Containing Proteins/genetics
15.
J Virol ; 82(16): 7758-67, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18524831

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) Nef protein upregulates the expression of the invariant chain (Ii)/major histocompatibility complex class II (MHC-II) complex at the cell surface. This complex appears to reach the antigen-loading endosomal compartment at least in part via an indirect pathway in which it is internalized from the cell surface via the adaptor protein 2 (AP-2) complex. Here we provide evidence for a competition model to explain how Nef upregulates the expression of Ii at the cell surface. In this model, Nef and Ii compete for binding to AP-2. In support of this model, Nef decreased the rate of internalization of Ii from the cell surface. The AP-binding dileucine motif in Nef, ENTSLL(165), was necessary and sufficient for the upregulation of Ii. In addition, two leucine-based AP-binding motifs in the Ii cytoplasmic tail, DDQRDLI(8) and EQLPML(17), were critical for the efficient upregulation of Ii by Nef. Experiments using Nef variants in which the native dileucine-based sorting motif was replaced with similar motifs from cellular transmembrane proteins allowed modulation of AP-binding specificity. Analysis of these variants suggested that the binding of Nef to AP-2 is sufficient to upregulate Ii at the plasma membrane. Finally, interference with the expression of AP-2 caused an upregulation of Ii at the plasma membrane, and this decreased the effect of Nef. These data indicate that Nef usurps AP-2 complexes to dysregulate Ii trafficking and potentially interfere with antigen presentation in the context of MHC-II.


Subject(s)
Gene Expression Regulation, Viral , Gene Products, nef/biosynthesis , Gene Products, nef/physiology , Histocompatibility Antigens Class II/physiology , Adaptor Protein Complex 2/metabolism , Amino Acid Motifs , Antigen Presentation , Binding, Competitive , CD4-Positive T-Lymphocytes/metabolism , Cell Membrane/metabolism , Cytoplasm/metabolism , HeLa Cells , Histocompatibility Antigens Class II/chemistry , Humans , Leucine/chemistry , Leukocytes, Mononuclear/metabolism , Models, Biological
16.
J Virol ; 82(3): 1249-58, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18057255

ABSTRACT

Human immunodeficiency virus type 1 Nef provides immune evasion by decreasing the expression of major histocompatibility complex class I (MHC-I) at the surfaces of infected cells. The endosomal clathrin adaptor protein complex AP-1 is a key cellular cofactor for this activity, and it is recruited to the MHC-I cytoplasmic domain (CD) in the presence of Nef by an uncharacterized mechanism. To determine the molecular basis of this recruitment, we used an MHC-I CD-Nef fusion protein to represent the MHC-I CD/Nef complex during protein interaction assays. The MHC-I CD had no intrinsic ability to bind AP-1, but it conferred binding activity when fused to Nef. This activity was independent of the canonical leucine-based AP-binding motif in Nef; it required residue Y320 in the MHC-I CD and residues E62-65 and P78 in Nef, and it involved the mu but not the gamma/sigma subunits of AP-1. The impaired binding of mutants encoding substitutions of E62-65 or P78 in Nef was rescued by replacing the Y320SQA sequence in the MHC-I CD with YSQL, suggesting that Nef allows the YSQA sequence to act as if it were a canonical mu-binding motif. These data identify the mu subunit of AP-1 (mu1) as the key target of the MHC-I CD/Nef complex, and they indicate that both Y320 in the MHC-I CD and E62-65 in Nef interact directly with mu1. The data support a cooperative binding model in which Nef functions as a clathrin-associated sorting protein that allows recognition of an incomplete, tyrosine-based mu-binding signal in the MHC-I CD by AP-1.


Subject(s)
HIV-1/immunology , Histocompatibility Antigens Class I/metabolism , Transcription Factor AP-1/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Motifs , Amino Acid Substitution/genetics , Cell Line , Humans , Mutagenesis, Site-Directed , Mutant Proteins/metabolism , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary
17.
J Virol Methods ; 137(1): 125-33, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16876264

ABSTRACT

Rapid and precise quantitation of the infectivity of HIV is important for molecular virologic studies as well as for measuring the activities of antiviral drugs and neutralizing antibodies. In the present study, an indicator cell line and image-analysis software were used to quantify HIV infectivity. Cells of the P4R5 line, which express the receptors for HIV infection as well as beta-galactosidase under the control of the HIV-1 long terminal repeat, were infected either with CXCR4- or CCR5-using viruses, including primary isolates, then stained 2 days later with X-gal to turn infected cells blue. Digital images of monolayers of the infected cells were captured using a high resolution CCD video camera and a macro video zoom lens. A software program was developed to process the images and to count the blue-stained foci of infection. The assay was applied to assess the infectivity of site-directed viral mutants, and to measure the activity of antiviral drugs and neutralizing antibody. The results indicate that the described method allows for the rapid quantitation of infected cells over a wide range of viral inocula with reproducibility, accuracy and relatively low cost.


Subject(s)
HIV-1/growth & development , Image Processing, Computer-Assisted/methods , beta-Galactosidase/analysis , Anti-HIV Agents/pharmacology , Antibodies, Monoclonal/immunology , Genes, Reporter , HIV Antibodies/immunology , HIV Long Terminal Repeat/genetics , HIV-1/drug effects , HIV-1/genetics , HIV-1/immunology , HeLa Cells , Humans , Mutation , Neutralization Tests , Nevirapine/pharmacology , Receptors, CCR5/biosynthesis , Receptors, CXCR4/biosynthesis , Software , beta-Galactosidase/genetics
18.
Virology ; 354(2): 316-27, 2006 Oct 25.
Article in English | MEDLINE | ID: mdl-16905171

ABSTRACT

The cytoplasmic domain of the HIV-1 Env glycoprotein (gp41) contains sequences that affect the trafficking of Env within the host cell. We previously showed that the membrane-proximal tyrosine-based adaptor protein (AP)-binding signal of gp41 (Y712XXL) is required for optimal viral infectivity and entry into target cells. Because these effects were not attributable to an effect on the incorporation of Env into virions, we hypothesized that they involved targeting of viral assembly to specific endosomal membranes that conferred greater fusogenicity. To further elaborate this hypothesis, we mutated the C-terminal leucine-based AP-binding signal of gp41 (LL855/856). In contrast to Env Y712, the leucine signal was dispensable for viral infectivity in both single cycle assays and during spreading infections within cultures of peripheral blood mononuclear cells (PBMCs). To test the hypothesis that these AP-binding motifs target Env to endosomes during viral morphogenesis, we compared the subcellular localization of wild-type Env to mutants of the Y712 and LL855/856 signals. The results failed to support the hypothesis that these signals target viral assembly to specific endosomal membranes. Strikingly, in the context of a C2-V3 region that confers macrophage-tropism, mutation of Y712 no longer markedly affected viral infectivity in either single cycle assays or during spreading infection within PBMCs, and it did not impair viral entry. These data indicate that the importance of the tyrosine-based sorting signal in gp41 for optimal viral infectivity depends on sequences in gp120. This observation is consistent with the hypothesis that the Y712 residue is part of the ectodomain of gp41 in virion-associated Env. We speculate that as part of the ectodomain, Y712 could affect specifically the conformation of the more positively charged CXCR4-tropic V3 loop in a manner that augments viral fusogenicity and infectivity.


Subject(s)
Cell Membrane/metabolism , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp41/physiology , HIV-1/metabolism , Protein Sorting Signals/physiology , Amino Acid Motifs , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , HIV-1/pathogenicity , HeLa Cells , Humans , Protein Sorting Signals/genetics , Tyrosine/metabolism , Virion/metabolism
19.
J Virol ; 80(4): 1837-49, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16439540

ABSTRACT

The nef gene contributes to the replication of primate lentiviruses by altering the trafficking of cellular proteins involved in adaptive immunity (class I and II major histocompatibility complex [MHC]) and viral transmission (CD4 and DC-SIGN). A conserved acidic leucine-based sequence (E(160)xxxLL) within human immunodeficiency virus type 1 (HIV-1) Nef binds to the cellular adaptor protein (AP) complexes, which mediate protein sorting into endosomal vesicles. The leucine residues in this motif are required for the down-regulation of CD4 and for the up-regulation of DC-SIGN and the invariant chain of MHC class II, but the role of the acidic residue is unclear. Here, substitution of E160 with uncharged residues impaired the ability of Nef to up-regulate the expression of the invariant chain and DC-SIGN at the cell surface, whereas substitution with a basic residue was required for a similar effect on the down-regulation of CD4. All substitutions of E160 relieved the Nef-mediated block to transferrin uptake. E160 was required for the efficient interaction of Nef with AP-1 and AP-3 and for the stabilization of these complexes on endosomal membranes in living cells. Systematic mutation of the ExxxLL sequence together with correlation of binding and functional data leads to the hypotheses that AP-1 and AP-3 are major cofactors for the effect of Nef on the trafficking of transferrin, are less important but contribute to the modulation of the invariant chain and DC-SIGN, and are least critical for the modulation of CD4. The data suggest that the E160 residue plays a differential role in the modulation of leucine-dependent Nef-targets and support a model in which distinct AP complexes are used by Nef to modulate different cellular proteins.


Subject(s)
Gene Products, nef/metabolism , HIV-1/physiology , Protein Transport , Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex 3/metabolism , Amino Acid Motifs , Amino Acid Substitution , CD4 Antigens/analysis , Cell Adhesion Molecules/analysis , Cell Line , Cell Membrane/metabolism , Endosomes/chemistry , Gene Products, nef/chemistry , Histocompatibility Antigens Class I/analysis , Humans , Intracellular Membranes/metabolism , Lectins, C-Type/analysis , Mutagenesis, Site-Directed , Protein Binding , Receptors, Cell Surface/analysis , Transferrin/metabolism , nef Gene Products, Human Immunodeficiency Virus
20.
Virology ; 345(1): 148-55, 2006 Feb 05.
Article in English | MEDLINE | ID: mdl-16253302

ABSTRACT

HIV-1 Nef affects the trafficking of numerous cellular proteins to optimize viral replication and evade host defenses. The adaptor protein (AP) complexes, which form part of the cytoplasmic coat of endosomal vesicles, are key cellular co-factors for Nef. Nef binds these complexes and alters their physiologic cycle of attachment and release from membranes. Specifically, while AP-1 normally becomes cytosolic when attachment events are blocked by inhibition of the GTPase cycle of ADP-ribosylation factor-1 (ARF1), the complex remains membrane-associated in Nef-expressing cells. To investigate the mechanism of this effect, we used a permeabilized cell system to detect the de novo attachment of exogenous AP-1 to endosomal membranes. Nef did not mediate de novo attachment independently of ARF1, despite its ability to maintain the association of AP-1 with endosomal membranes when the activity of ARF1 was blocked. We conclude that Nef stabilizes AP complexes on endosomal membranes after ARF1-dependent attachment. This stabilization may facilitate coat formation and stimulate the trafficking of multiple cellular proteins.


Subject(s)
ADP-Ribosylation Factor 1/physiology , Adaptor Protein Complex 1/metabolism , Gene Products, nef/physiology , HIV-1/physiology , Intracellular Membranes/metabolism , ADP-Ribosylation Factor 1/antagonists & inhibitors , Animals , Cell Line , Cricetinae , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Intracellular Membranes/chemistry , Microscopy, Confocal , Models, Biological , Protein Binding , nef Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL