Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Virol ; 89(4): 1986-2001, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25505066

ABSTRACT

UNLABELLED: The role of the accessory viral Nef protein as a multifunctional manipulator of the host cell that is required for effective replication of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) in vivo is well established. It is unknown, however, whether Nef manipulates all or just specific subsets of CD4(+) T cells, which are the main targets of virus infection and differ substantially in their state of activation and importance for a functional immune system. Here, we analyzed the effect of Nef proteins differing in their T cell receptor (TCR)-CD3 downmodulation function in HIV-infected human lymphoid aggregate cultures and peripheral blood mononuclear cells. We found that Nef efficiently downmodulates TCR-CD3 in naive and memory CD4(+) T cells and protects the latter against apoptosis. In contrast, highly proliferative CD45RA(+) CD45RO(+) CD4(+) T cells were main producers of infectious virus but largely refractory to TCR-CD3 downmodulation. Such T cell subset-specific differences were also observed for Nef-mediated modulation of CD4 but not for enhancement of virion infectivity. Our results indicate that Nef predominantly modulates surface receptors on CD4(+) T cell subsets that are not already fully permissive for viral replication. As a consequence, Nef-mediated downmodulation of TCR-CD3, which distinguishes most primate lentiviruses from HIV type 1 (HIV-1) and its vpu-containing simian precursors, may promote a selective preservation of central memory CD4(+) T cells, which are critical for the maintenance of a functional immune system. IMPORTANCE: The Nef proteins of human and simian immunodeficiency viruses manipulate infected CD4(+) T cells in multiple ways to promote viral replication and immune evasion in vivo. Here, we show that some effects of Nef are subset specific. Downmodulation of CD4 and TCR-CD3 is highly effective in central memory CD4(+) T cells, and the latter Nef function protects this T cell subset against apoptosis. In contrast, highly activated/proliferating CD4(+) T cells are largely refractory to receptor downmodulation but are main producers of infectious HIV-1. Nef-mediated enhancement of virion infectivity, however, was observed in all T cell subsets examined. Our results provide new insights into how primate lentiviruses manipulate their target cells and suggest that the TCR-CD3 downmodulation function of Nef may promote a selective preservation of memory CD4(+) T cells, which are critical for immune function, but has little effect on activated/proliferating CD4(+) T cells, which are the main targets for viral replication.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV-1/immunology , T-Lymphocyte Subsets/virology , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Apoptosis , CD3 Complex/analysis , CD4-Positive T-Lymphocytes/immunology , Cell Survival , Humans , Leukocyte Common Antigens/analysis , Receptors, Antigen, T-Cell/analysis , T-Lymphocyte Subsets/immunology
2.
PLoS Pathog ; 10(8): e1004345, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25167059

ABSTRACT

A rare subset of HIV-infected individuals, designated viremic non-progressors (VNP), remain asymptomatic and maintain normal levels of CD4+ T-cells despite persistently high viremia. To identify mechanisms potentially responsible for the VNP phenotype, we compared VNPs (average >9 years of HIV infection) to HIV-infected individuals who have similar CD4+ T-cell counts and viral load, but who are likely to progress if left untreated ("putative progressors", PP), thus avoiding the confounding effect of differences related to substantial CD4+ T cell depletion. We found that VNPs, compared to PPs, had preserved levels of CD4+ stem cell memory cells (TSCM (p<0.0001), which was associated with decreased HIV infection of these cells in VNPs (r = -0.649, p = 0.019). In addition, VNPs had decreased HIV infection in CD4+ central memory (TCM) cells (p = 0.035), and the total number of TCM cells was associated with increased proliferation of memory CD4+ T cells (r = 0.733, p = 0.01). Our results suggest that, in HIV-infected VNPs, decreased infection of CD4+ TCM and TSCM, cells are involved in preservation of CD4+ T cell homeostasis and lack of disease progression despite high viremia.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Immunologic Memory/immunology , T-Lymphocyte Subsets/immunology , Viremia/immunology , CD4-Positive T-Lymphocytes/virology , Cell Separation , DNA, Viral/analysis , Disease Progression , HIV Infections/virology , Humans , Immunophenotyping , Oligonucleotide Array Sequence Analysis , Stem Cells/immunology , Stem Cells/virology , T-Lymphocyte Subsets/virology , Viral Load , Viremia/virology
3.
Retrovirology ; 12: 86, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26452480

ABSTRACT

BACKGROUND: Elderly HIV-1 infected individuals progress to AIDS more frequently and rapidly than people becoming infected at a young age. To identify possible reasons for these differences in clinical progression, we performed comprehensive phenotypic analyses of CD4+ T cells from uninfected young and elderly individuals, and examined their susceptibility to HIV-1 infection and programmed death. RESULTS: Peripheral blood mononuclear cells (PBMCs) from older people contain an increased percentage of central memory and Th17 CD4+ T cells that are main target cells of HIV-1 and strongly reduced proportions of naïve T cells that are poorly susceptible to HIV-1. Unstimulated T cells from elderly individuals expressed higher levels of activation markers, death receptors, and the viral CXCR4 co-receptor than those from young individuals but responded poorly to stimulation. CD4+ T cells from older individuals were highly susceptible to CXCR4- and CCR5-tropic HIV-1 infection but produced significantly lower quantities of infectious virus than cells from young individuals because they were highly prone to apoptosis and thus presumably had a very short life span. The increased susceptibility of T cells from the elderly to HIV-1 infection correlated directly with CXCR4 and inversely with CD4 expression. The levels of apoptosis correlated with the cell surface expression of FAS but not with the expression of programmed death receptor 1 (PD1) or tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). CONCLUSIONS: Increased levels of activated and highly susceptible HIV-1 target cells, reduced CD4 and enhanced CXCR4 cell surface expression, together with the high susceptibility to FAS-induced programmed cell death may contribute to the rapid CD4+ T cell depletion and accelerated clinical course of infection in elderly HIV-1-infected individuals.


Subject(s)
Aging , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , Receptors, CXCR4/genetics , fas Receptor/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Apoptosis , Female , HIV Infections/immunology , HIV-1/physiology , Humans , Immunologic Memory , Leukocytes, Mononuclear/immunology , Male , Membrane Glycoproteins , Middle Aged , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , Young Adult
4.
Retrovirology ; 11: 13, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24495362

ABSTRACT

BACKGROUND: A small minority of HIV-1-infected individuals show low levels of immune activation and do not develop immunodeficiency despite high viral loads. Since the accessory viral Nef protein modulates T cell activation and plays a key role in the pathogenesis of AIDS, we investigated whether specific properties of Nef may be associated with this highly unusual clinical outcome of HIV-1 infection. FINDINGS: Comprehensive functional analyses of sequential HIV-1 strains from three viremic long-term non-progressors (VNP) showed that they encode full-length Nef proteins that are capable of modulating CD4, CD28, CD8ß, MHC-I and CD74 cell surface expression. Similar to Nef proteins from HIV-1-infected individuals with progressive infection (P-Nefs) and unlike Nefs from simian immunodeficiency viruses (SIVs) that do not cause chronic immune activation and disease in their natural simian hosts, VNP-Nefs were generally unable to down-modulate TCR-CD3 cell surface expression to block T cell activation and apoptosis. On average, VNP-Nefs suppressed NF-AT activation less effectively than P-Nefs and were slightly less active in enhancing NF-κB activity. Finally, we found that VNP-Nefs increased virion infectivity and enhanced HIV-1 replication and cytopathicity in primary human cells and in ex vivo infected lymphoid tissues. CONCLUSIONS: Our results show that nef alleles from VNPs and progressors of HIV-1 infection show only modest differences in established functions. Thus, the lack of chronic immune activation and disease progression in HIV-1-infected VNPs is apparently not associated with unusual functional properties of the accessory viral Nef protein.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , HIV Long-Term Survivors , HIV-1/physiology , Viremia/immunology , Viremia/virology , nef Gene Products, Human Immunodeficiency Virus/metabolism , Antigens, CD/analysis , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Histocompatibility Antigens Class I/analysis , Humans
5.
Retrovirology ; 11: 1, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24383984

ABSTRACT

BACKGROUND: Increased cellular iron levels are associated with high mortality in HIV-1 infection. Moreover iron is an important cofactor for viral replication, raising the question whether highly divergent lentiviruses actively modulate iron homeostasis. Here, we evaluated the effect on cellular iron uptake upon expression of the accessory protein Nef from different lentiviral strains. RESULTS: Surface Transferrin receptor (TfR) levels are unaffected by Nef proteins of HIV-1 and its simian precursors but elevated in cells expressing Nefs from most other primate lentiviruses due to reduced TfR internalization. The SIV Nef-mediated reduction of TfR endocytosis is dependent on an N-terminal AP2 binding motif that is not required for downmodulation of CD4, CD28, CD3 or MHCI. Importantly, SIV Nef-induced inhibition of TfR endocytosis leads to the reduction of Transferrin uptake and intracellular iron concentration and is accompanied by attenuated lentiviral replication in macrophages. CONCLUSION: Inhibition of Transferrin and thereby iron uptake by SIV Nef might limit viral replication in myeloid cells. Furthermore, this new SIV Nef function could represent a virus-host adaptation that evolved in natural SIV-infected monkeys.


Subject(s)
Endocytosis/drug effects , Gene Products, nef/metabolism , Iron/metabolism , Receptors, Transferrin/antagonists & inhibitors , Simian Immunodeficiency Virus/physiology , Transferrin/metabolism , Animals , Haplorhini
6.
PLoS Pathog ; 8(12): e1003093, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23308067

ABSTRACT

HIV-1 groups M and N emerged within the last century following two independent cross-species transmissions of SIVcpz from chimpanzees to humans. In contrast to pandemic group M strains, HIV-1 group N viruses are exceedingly rare, with only about a dozen infections identified, all but one in individuals from Cameroon. Poor adaptation to the human host may be responsible for this limited spread of HIV-1 group N in the human population. Here, we analyzed the function of Vpu proteins from seven group N strains from Cameroon, the place where this zoonosis originally emerged. We found that these N-Vpus acquired four amino acid substitutions (E15A, V19A and IV25/26LL) in their transmembrane domain (TMD) that allow efficient interaction with human tetherin. However, despite these adaptive changes, most N-Vpus still antagonize human tetherin only poorly and fail to down-modulate CD4, the natural killer (NK) cell ligand NTB-A as well as the lipid-antigen presenting protein CD1d. These functional deficiencies were mapped to amino acid changes in the cytoplasmic domain that disrupt putative adaptor protein binding sites and an otherwise highly conserved ßTrCP-binding DSGxxS motif. As a consequence, N-Vpus exhibited aberrant intracellular localization and/or failed to recruit the ubiquitin-ligase complex to induce tetherin degradation. The only exception was the Vpu of a group N strain recently discovered in France, but originally acquired in Togo, which contained intact cytoplasmic motifs and counteracted tetherin as effectively as the Vpus of pandemic HIV-1 M strains. These results indicate that HIV-1 group N Vpu is under strong host-specific selection pressure and that the acquisition of effective tetherin antagonism may lead to the emergence of viral variants with increased transmission fitness.


Subject(s)
Antigens, CD/metabolism , HIV Infections/virology , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/metabolism , Selection, Genetic , Viral Regulatory and Accessory Proteins/metabolism , Amino Acid Sequence , Binding Sites , Blotting, Western , Cells, Cultured , Flow Cytometry , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , HIV Infections/metabolism , HIV-1/pathogenicity , Humans , Molecular Sequence Data , Protein Interaction Domains and Motifs , Sequence Homology, Amino Acid , Virus Release
7.
J Virol ; 86(1): 36-48, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22013062

ABSTRACT

It is well established that the Nef proteins of human and simian immunodeficiency viruses (HIV and SIV) modulate major histocompatibility complex class I (MHC-I) cell surface expression to protect infected cells against lysis by cytotoxic T lymphocytes (CTLs). Recent data supported the observation that Nef also manipulates CTLs directly by down-modulating CD8αß (J. A. Leonard, T. Filzen, C. C. Carter, M. Schaefer, and K. L. Collins, J. Virol. 85:6867-6881, 2011), but it remained unknown whether this Nef activity is conserved between different lineages of HIV and SIV. In this study, we examined a total of 42 nef alleles from 16 different primate lentiviruses representing most major lineages of primate lentiviruses, as well as nonpandemic HIV-1 strains and the direct precursors of HIV-1 (SIVcpz and SIVgor). We found that the vast majority of these nef alleles strongly down-modulate CD8ß in human T cells. Primate lentiviral Nefs generally interacted specifically with the cytoplasmic tail of CD8ß, and down-modulation of this receptor was dependent on the conserved dileucine-based motif and two adjacent acidic residues (DD/E) in the C-terminal flexible loop of SIV Nef proteins. Both of these motifs are known to be important for the interaction of HIV-1 Nef with AP-2, and they were also shown to be critical for down-modulation of CD4 and CD28, but not MHC-I, by SIV Nefs. Our results show that down-modulation of CD4, CD8ß, and CD28 involves largely overlapping (but not identical) domains and is most likely dependent on conserved interactions of primate lentiviral Nefs with cellular adaptor proteins. Furthermore, our data demonstrate that Nef-mediated down-modulation of CD8αß is a fundamental property of primate lentiviruses and suggest that direct manipulation of CD8+ T cells plays a relevant role in viral immune evasion.


Subject(s)
CD8 Antigens/genetics , Down-Regulation , Gene Products, nef/metabolism , Lentivirus Infections/genetics , Lentiviruses, Primate/metabolism , Animals , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Line , Cells, Cultured , Gene Products, nef/genetics , Humans , Lentivirus Infections/immunology , Lentivirus Infections/virology , Lentiviruses, Primate/classification , Lentiviruses, Primate/genetics , Lentiviruses, Primate/isolation & purification
8.
Cell Rep ; 30(7): 2261-2274.e7, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075764

ABSTRACT

The inability of Nef to downmodulate the CD3-T cell receptor (TCR) complex distinguishes HIV-1 from other primate lentiviruses and may contribute to its high virulence. However, the role of this Nef function in virus-mediated immune activation and pathogenicity remains speculative. Here, we selectively disrupted this Nef activity in SIVmac239 and analyzed the consequences for the virological, immunological, and clinical outcome of infection in rhesus macaques. The inability to downmodulate CD3-TCR does not impair viral replication during acute infection but is associated with increased immune activation and antiviral gene expression. Subsequent early reversion in three of six animals suggests strong selective pressure for this Nef function and is associated with high viral loads and progression to simian AIDS. In the absence of reversions, however, viral replication and the clinical course of infection are attenuated. Thus, Nef-mediated downmodulation of CD3 dampens the inflammatory response to simian immunodeficiency virus (SIV) infection and seems critical for efficient viral immune evasion.


Subject(s)
Immune Evasion/immunology , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Viral Regulatory and Accessory Proteins/metabolism , Animals , Female , Gene Products, nef , Inflammation/immunology , Inflammation/pathology , Macaca mulatta , Male , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Simian Immunodeficiency Virus/immunology
9.
Cell Host Microbe ; 20(3): 381-391, 2016 Sep 14.
Article in English | MEDLINE | ID: mdl-27631701

ABSTRACT

The cellular factor serine incorporator 5 (SERINC5) impairs HIV-1 infectivity but is antagonized by the viral Nef protein. We analyzed the anti-SERINC5 activity of Nef proteins across primate lentiviruses and examined whether SERINC5 represents a barrier to cross-species transmissions and/or within-species viral spread. HIV-1, HIV-2, and SIV Nefs counteract human, ape, monkey, and murine SERINC5 orthologs with similar potency. However, HIV-1 Nefs are more active against SERINC5 than HIV-2 Nefs, and chimpanzee SIV (SIVcpz) Nefs are more potent than those of their monkey precursors. Additionally, Nefs of HIV and most SIVs rely on the dileucine motif in the C-terminal loop for anti-SERINC5 activity, while the Nef from colobus SIV (SIVcol) evolved different inhibitory mechanisms. We also found a significant correlation between anti-SERINC5 potency and the SIV prevalence in the respective ape and monkey species. Thus, Nef-mediated SERINC5 antagonism may determine the ability of primate lentiviruses to spread within natural hosts.


Subject(s)
HIV-1/pathogenicity , HIV-2/pathogenicity , Simian Acquired Immunodeficiency Syndrome/epidemiology , Simian Immunodeficiency Virus/pathogenicity , Viral Regulatory and Accessory Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Cells, Cultured , HIV-1/immunology , HIV-2/immunology , Host-Pathogen Interactions , Humans , Mice , Primates , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Virulence Factors/metabolism
10.
Cell Rep ; 11(5): 737-47, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25921529

ABSTRACT

CXCL12-CXCR4 signaling controls multiple physiological processes and its dysregulation is associated with cancers and inflammatory diseases. To discover as-yet-unknown endogenous ligands of CXCR4, we screened a blood-derived peptide library for inhibitors of CXCR4-tropic HIV-1 strains. This approach identified a 16 amino acid fragment of serum albumin as an effective and highly specific CXCR4 antagonist. The endogenous peptide, termed EPI-X4, is evolutionarily conserved and generated from the highly abundant albumin precursor by pH-regulated proteases. EPI-X4 forms an unusual lasso-like structure and antagonizes CXCL12-induced tumor cell migration, mobilizes stem cells, and suppresses inflammatory responses in mice. Furthermore, the peptide is abundant in the urine of patients with inflammatory kidney diseases and may serve as a biomarker. Our results identify EPI-X4 as a key regulator of CXCR4 signaling and introduce proteolysis of an abundant precursor protein as an alternative concept for chemokine receptor regulation.


Subject(s)
Peptide Fragments/metabolism , Peptides/metabolism , Receptors, CXCR4/antagonists & inhibitors , Serum Albumin/metabolism , Amino Acid Sequence , Animals , Biomarkers/urine , Cell Line , Cell Movement/drug effects , HEK293 Cells , HIV-1/physiology , Half-Life , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Library , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Protein Structure, Tertiary , Receptors, CXCR4/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Sequence Alignment , Serum Albumin/chemistry , Serum Albumin/pharmacology , Signal Transduction/drug effects , Virus Internalization/drug effects
11.
Methods Mol Biol ; 1087: 115-23, 2014.
Article in English | MEDLINE | ID: mdl-24158818

ABSTRACT

Nef is a multifunctional protein encoded by all primate lentiviruses that modulates cell surface expression of a variety of cellular receptors and increases the infectivity of progeny virons. Here, we describe the use of bicistronic HIV-1 constructs that coexpress Nef and fluorescent proteins via an internal ribosome entry site to quantify Nef-mediated receptor modulation in virally infected cells. We also report how such proviral constructs and indicator cell lines can be used to quantify the effect of Nef on virion infectivity.


Subject(s)
HIV-1/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , HEK293 Cells , HIV-1/physiology , Humans , Receptors, Cell Surface/metabolism , Virion/physiology
12.
Cell Host Microbe ; 16(5): 639-50, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25525794

ABSTRACT

Most simian immunodeficiency viruses use their Nef protein to antagonize the host restriction factor tetherin. A deletion in human tetherin confers Nef resistance, representing a hurdle to successful zoonotic transmission. HIV-1 group M evolved to utilize the viral protein U (Vpu) to counteract tetherin. Although HIV-1 group O has spread epidemically in humans, it has not evolved a Vpu-based tetherin antagonism. Here we show that HIV-1 group O Nef targets a region adjacent to this deletion to inhibit transport of human tetherin to the cell surface, enhances virion release, and increases viral resistance to inhibition by interferon-α. The Nef protein of the inferred common ancestor of group O viruses is also active against human tetherin. Thus, Nef-mediated antagonism of human tetherin evolved prior to the spread of HIV-1 group O and likely facilitated secondary virus transmission. Our results may explain the epidemic spread of HIV-1 group O.


Subject(s)
Antigens, CD/genetics , HIV-1/pathogenicity , nef Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line, Tumor , Endocytosis , Evolution, Molecular , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , HIV-1/classification , Humans , Molecular Sequence Data , Protein Conformation , Sequence Analysis , Sequence Deletion , Virion/genetics , Virion/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics
13.
J Clin Invest ; 122(5): 1644-52, 2012 May.
Article in English | MEDLINE | ID: mdl-22505456

ABSTRACT

SIVs infecting wild-living apes in west central Africa have crossed the species barrier to humans on at least four different occasions, one of which spawned the AIDS pandemic. Although the chimpanzee precursor of pandemic HIV-1 strains must have been able to infect humans, the capacity of SIVcpz strains to replicate in human lymphoid tissues (HLTs) is not known. Here, we show that SIVcpz strains from two chimpanzee subspecies are capable of replicating in human tonsillary explant cultures, albeit only at low titers. However, SIVcpz replication in HLT was significantly improved after introduction of a previously identified human-specific adaptation at position 30 in the viral Gag matrix protein. An Arg or Lys at this position significantly increased SIVcpz replication in HLT, while the same mutation reduced viral replication in chimpanzee-derived CD4(+) T cells. Thus, naturally occurring SIVcpz strains are capable of infecting HLTs, the major site of HIV-1 replication in vivo. However, efficient replication requires the acquisition of a host-specific adaptation in the viral matrix protein. These results identify Gag matrix as a major determinant of SIVcpz replication fitness in humans and suggest a critical role in the emergence of HIV/AIDS.


Subject(s)
Lentivirus Infections/virology , Palatine Tonsil/virology , Simian Immunodeficiency Virus/physiology , Viral Matrix Proteins/metabolism , Virus Replication , Adaptation, Biological , Amino Acid Substitution , Animals , Cells, Cultured , Gene Products, gag/genetics , Gene Products, gag/metabolism , HIV-1/genetics , HIV-1/pathogenicity , HIV-1/physiology , Host-Pathogen Interactions , Humans , Mutagenesis, Site-Directed , Pan troglodytes , Phylogeny , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , Tissue Culture Techniques
14.
Cell Host Microbe ; 12(3): 373-80, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22980333

ABSTRACT

The interferon-induced host restriction factor tetherin poses a barrier for SIV transmission from primates to humans. After cross-species transmission, the chimpanzee precursor of pandemic HIV-1 switched from the accessory protein Nef to Vpu to effectively counteract human tetherin. As we report here, the experimental reintroduction of HIV-1 into its original chimpanzee host resulted in a virus that can use both Vpu and Nef to antagonize chimpanzee tetherin. Functional analyses demonstrated that alterations in and near the highly conserved ExxxLL motif in the C-terminal loop of Nef were critical for the reacquisition of antitetherin activity. Strikingly, just two amino acid changes allowed HIV-1 Nef to counteract chimpanzee tetherin and promote virus release. Our data demonstrate that primate lentiviruses can reacquire lost accessory gene functions during a single in vivo passage and suggest that other functional constraints keep Nef ready to regain antitetherin activity.


Subject(s)
Antigens, CD/immunology , HIV-1/immunology , Human Immunodeficiency Virus Proteins/metabolism , Primate Diseases/immunology , Viral Regulatory and Accessory Proteins/metabolism , Virulence Factors/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Substitution , Animals , Disease Models, Animal , HIV-1/growth & development , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Pan troglodytes , Primate Diseases/virology , nef Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL