Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Atherosclerosis ; 244: 22-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26581048

ABSTRACT

BACKGROUND: HIV protein Nef plays a key role in impairing cholesterol metabolism in both HIV infected and bystander cells. The existence of a small cohort of patients infected with Nef-deficient strain of HIV presented a unique opportunity to test the effect of Nef on lipid metabolism in a clinical setting. METHODS: Here we report the results of a study comparing six patients infected with Nef-deficient strain of HIV (ΔNefHIV) with six treatment-naïve patients infected with wild-type HIV (WT HIV). Lipoprotein profile, size and functionality of high density lipoprotein (HDL) particles as well as lipidomic and microRNA profiles of patient plasma were analyzed. RESULTS: We found that patients infected with ΔNefHIV had lower proportion of subjects with plasma HDL-C levels <1 mmol/l compared to patients infected with WT HIV. Furthermore, compared to a reference group of HIV-negative subjects, there was higher abundance of smaller under-lipidated HDL particles in plasma of patients infected with WT HIV, but not in those infected with ΔNefHIV. Lipidomic analysis of plasma revealed differences in abundance of phosphatidylserine and sphingolipids between patients infected with ΔNefHIV and WT HIV. MicroRNA profiling revealed that plasma abundance of 24 miRNAs, many of those involved in regulation of lipid metabolism, was differentially regulated by WT HIV and ΔNefHIV. CONCLUSION: Our findings are consistent with HIV protein Nef playing a significant role in pathogenesis of lipid-related metabolic complications of HIV disease.


Subject(s)
HIV Infections/blood , HIV-1/metabolism , Lipid Metabolism , Lipids/blood , RNA, Viral/genetics , nef Gene Products, Human Immunodeficiency Virus/deficiency , Adult , Aged , Female , HIV Infections/virology , HIV-1/genetics , Humans , Male , Middle Aged
2.
Nature ; 526(7572): 218-23, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26416733

ABSTRACT

HIV-1 Nef and the unrelated mouse leukaemia virus glycosylated Gag (glycoGag) strongly enhance the infectivity of HIV-1 virions produced in certain cell types in a clathrin-dependent manner. Here we show that Nef and glycoGag prevent the incorporation of the multipass transmembrane proteins serine incorporator 3 (SERINC3) and SERINC5 into HIV-1 virions to an extent that correlates with infectivity enhancement. Silencing of both SERINC3 and SERINC5 precisely phenocopied the effects of Nef and glycoGag on HIV-1 infectivity. The infectivity of nef-deficient virions increased more than 100-fold when produced in double-knockout human CD4(+) T cells that lack both SERINC3 and SERINC5, and re-expression experiments confirmed that the absence of SERINC3 and SERINC5 accounted for the infectivity enhancement. Furthermore, SERINC3 and SERINC5 together restricted HIV-1 replication, and this restriction was evaded by Nef. SERINC3 and SERINC5 are highly expressed in primary human HIV-1 target cells, and inhibiting their downregulation by Nef is a potential strategy to combat HIV/AIDS.


Subject(s)
HIV-1/chemistry , HIV-1/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Down-Regulation , Gene Deletion , Gene Products, gag/metabolism , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , HIV-1/growth & development , Host-Pathogen Interactions/drug effects , Humans , Leukemia Virus, Murine/chemistry , Membrane Glycoproteins , Membrane Proteins/deficiency , Membrane Proteins/pharmacology , Neoplasm Proteins/deficiency , Neoplasm Proteins/pharmacology , Protein Transport , Receptors, Cell Surface/deficiency , Virion/chemistry , Virion/drug effects , Virion/growth & development , Virion/physiology , Virus Replication/drug effects , nef Gene Products, Human Immunodeficiency Virus/deficiency
3.
J Virol ; 88(17): 10200-13, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965469

ABSTRACT

UNLABELLED: Host and viral factors influence the HIV-1 infection course. Reduced Nef function has been observed in HIV-1 controllers during the chronic phase, but the kinetics and mechanisms of Nef attenuation in such individuals remain unclear. We examined plasma RNA-derived Nef clones from 10 recently infected individuals who subsequently suppressed viremia to less than 2,000 RNA copies/ml within 1 year postinfection (acute controllers) and 50 recently infected individuals who did not control viremia (acute progressors). Nef clones from acute controllers displayed a lesser ability to downregulate CD4 and HLA class I from the cell surface and a reduced ability to enhance virion infectivity compared to those from acute progressors (all P<0.01). HLA class I downregulation activity correlated inversely with days postinfection (Spearman's R=-0.85, P=0.004) and positively with baseline plasma viral load (Spearman's R=0.81, P=0.007) in acute controllers but not in acute progressors. Nef polymorphisms associated with functional changes over time were identified in follow-up samples from six controllers. For one such individual, mutational analyses indicated that four polymorphisms selected by HLA-A*31 and B*37 acted in combination to reduce Nef steady-state protein levels and HLA class I downregulation activity. Our results demonstrate that relative control of initial HIV-1 viremia is associated with Nef clones that display reduced function, which in turn may influence the course of HIV-1 infection. Transmission of impaired Nef sequences likely contributed in part to this observation; however, accumulation of HLA-associated polymorphisms in Nef that impair function also suggests that CD8+ T-cell pressures play a role in this phenomenon. IMPORTANCE: Rare individuals can spontaneously control HIV-1 viremia in the absence of antiretroviral treatment. Understanding the host and viral factors that contribute to the controller phenotype may identify new strategies to design effective vaccines or therapeutics. The HIV-1 Nef protein enhances viral pathogenesis through multiple mechanisms. We examined the function of plasma HIV-1 RNA-derived Nef clones isolated from 10 recently infected individuals who subsequently controlled HIV viremia compared to the function of those from 50 individuals who failed to control viremia. Our results demonstrate that early Nef clones from HIV controllers displayed lower HLA class I and CD4 downregulation activity, as well as a reduced ability to enhance virion infectivity. The accumulation of HLA-associated polymorphisms in Nef during the first year postinfection was associated with impaired protein function in some controllers. This report highlights the potential for host immune responses to modulate HIV pathogenicity and disease outcome by targeting cytotoxic T lymphocyte (CTL) epitopes in Nef.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Viremia/immunology , nef Gene Products, Human Immunodeficiency Virus/deficiency , CD4 Antigens/analysis , Down-Regulation , Genotype , HIV-1/genetics , Histocompatibility Antigens Class I/analysis , Humans , Molecular Sequence Data , Plasma/virology , Polymorphism, Genetic , RNA, Viral/genetics , RNA, Viral/isolation & purification , Sequence Analysis, DNA , Viral Load , nef Gene Products, Human Immunodeficiency Virus/genetics
4.
J Virol ; 88(6): 3443-54, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24403584

ABSTRACT

UNLABELLED: Human immunodeficiency virus type 1 (HIV-1) Nef enhances the infectivity of progeny virions. However, Nef is dispensable for the production of HIV-1 virions of optimal infectivity if the producer cells are superinfected with certain gammaretroviruses. In the case of the ecotropic Moloney murine leukemia virus (M-MLV), the Nef-like effect is mediated by the glycosylated Gag (glycoGag) protein. We now show that the N-terminal intracellular domain of the type II transmembrane protein glycoGag is responsible for its effect on HIV-1 infectivity. In the context of a fully active minimal M-MLV glycoGag construct, truncations of the cytoplasmic domain led to a near total loss of activity. Furthermore, the cytoplasmic domain of M-MLV glycoGag was fully sufficient to transfer the activity to an unrelated type II transmembrane protein. Although the intracellular region of glycoGag is relatively poorly conserved even among ecotropic and xenotropic MLVs, it was also fully sufficient for the rescue of nef-deficient HIV-1 when derived from a xenotropic virus. A mutagenic analysis showed that only a core region of the intracellular domain that exhibits at least some conservation between murine and feline leukemia viruses is crucial for activity. In particular, a conserved YXXL motif in the center of this core region was critical. In addition, expression of the µ2 subunit of the AP-2 adaptor complex in virus producer cells was essential for activity. We conclude that the ability to enhance HIV-1 infectivity is a conserved property of the MLV glycoGag cytoplasmic domain and involves AP-2-mediated endocytosis. IMPORTANCE: The Nef protein of HIV-1 and the entirely unrelated glycosylated Gag (glycoGag) protein of a murine leukemia virus (MLV) similarly enhance the infectiousness of HIV-1 particles by an unknown mechanism. MLV glycoGag is an alternative version of the structural viral Gag protein with an extra upstream region that provides a cytosolic domain and a plasma membrane anchor. We now show for the first time that the cytosolic domain of MLV glycoGag contains all the information needed to enhance HIV-1 infectivity and that this function of the cytosolic domain is conserved despite limited sequence conservation. Within the cytosolic domain, a motif that resembles a cellular sorting signal is critical for activity. Furthermore, the enhancement of HIV-1 infectivity depends on an endocytic cellular protein that is known to interact with such sorting signals. Together, our findings implicate the endocytic machinery in the enhancement of HIV-1 infectivity by MLV glycoGag.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex mu Subunits/metabolism , Gene Products, gag/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Moloney murine leukemia virus/metabolism , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex mu Subunits/genetics , Animals , Gene Products, gag/chemistry , Gene Products, gag/genetics , Gene Products, nef/chemistry , Gene Products, nef/genetics , Gene Products, nef/metabolism , Glycosylation , HIV Infections/genetics , HIV-1/genetics , Humans , Mice , Moloney murine leukemia virus/chemistry , Moloney murine leukemia virus/genetics , Protein Structure, Tertiary , nef Gene Products, Human Immunodeficiency Virus/deficiency , nef Gene Products, Human Immunodeficiency Virus/genetics
5.
J Virol ; 87(7): 3729-40, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23325686

ABSTRACT

Nef is a human immunodeficiency virus type 1 (HIV-1) auxiliary protein that plays an important role in virus replication and the onset of acquired immunodeficiency. Although known functions of Nef might explain its contribution to HIV-1-associated pathogenesis, how Nef increases virus infectivity is still an open question. In vitro, Nef-deleted viruses have a defect that prevents efficient completion of early steps of replication. We have previously shown that this restriction is not due to the absence of Nef in viral particles. Rather, a loss of function in virus-producing cells accounts for the lower infectivity of nef-deleted viruses compared to wild-type (WT) viruses. Here we used DiGE and iTRAQ to identify differences between the proteomes of WT and nef-deleted viruses. We observe that glucosidase II is enriched in WT virions, whereas Ezrin, ALG-2, CD81, and EHD4 are enriched in nef-deleted virions. Functional analysis shows that glucosidase II, ALG-2, and CD81 have no or only Nef-independent effect on infectivity. In contrast, Ezrin and EHD4 are involved in the ability of Nef to increase virus infectivity (referred to thereafter as Nef potency). Indeed, simultaneous Ezrin and EHD4 depletion in SupT1 and 293T virus-producing cells result in an ∼30 and ∼70% decrease of Nef potency, respectively. Finally, while Ezrin behaves as an inhibitory factor counteracted by Nef, EHD4 should be considered as a cofactors required by Nef to increase virus infectivity.


Subject(s)
Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , HIV-1/genetics , HIV-1/pathogenicity , Nuclear Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus/deficiency , nef Gene Products, Human Immunodeficiency Virus/metabolism , Blotting, Western , Electrophoresis/methods , Gene Silencing , HEK293 Cells , HIV-1/metabolism , Humans , Proteomics , RNA, Small Interfering , Tetraspanin 28/metabolism , Ultracentrifugation , Virion/metabolism , alpha-Glucosidases/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics
6.
Virology ; 410(2): 316-26, 2011 Feb 20.
Article in English | MEDLINE | ID: mdl-21176845

ABSTRACT

The phenotypic changes that are induced by immune activation in CD4(+) T lymphocytes provide an optimal environment for efficient HIV-1 replication in these cells. The pathogenic Nef protein of HIV-1 modulates the T cell receptor (TCR) signaling, but whether this has a positive or negative effect on cellular activation is a matter of debate. Here we have investigated the response to TCR stimulation of primary CD4(+) T lymphocytes infected with wt or Nef-deficient HIV-1. Results show that, in freshly isolated quiescent T cells, Nef superinduces NFAT and IL-2 production bypassing early TCR effector molecules. Conversely, the early phosphorylation of PLC-γ1, the induction of NFAT, and the expression of IL-2 are impaired by Nef in sub-optimally activated/resting T cells. Our data indicate that Nef has a dual role in the modulation of TCR signaling aimed at favoring HIV-1 replication and spread in both quiescent and metabolically active CD4(+) T lymphocytes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV-1/pathogenicity , Receptors, Antigen, T-Cell/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , HIV-1/genetics , Interleukin-2/biosynthesis , Lymphocyte Activation , NFATC Transcription Factors/biosynthesis , nef Gene Products, Human Immunodeficiency Virus/deficiency
7.
Proc Natl Acad Sci U S A ; 107(20): 9364-9, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20439730

ABSTRACT

Optimal infectivity of HIV-1 virions requires synthesis of the HIV-1 regulatory protein Nef in some producer cells but not others. A survey of 18 lymphoid cell lines found that Nef was dispensable in three, each of which harbored gammaretroviruses. Nef-dependent cell lines were rendered Nef-independent by a cell-free supernatant from the independent lines or by transfection of cloned murine leukemia virus (MLV). Analysis of MLV deletion mutations identified glycosylated gag (glycogag) as the factor that rescues Nef-defective HIV-1 virions. Glycogag was also demonstrated to be required for the infectivity of MLV virions produced in lymphoid cells. Direct comparison of Nef and glycogag revealed identical dependence for activity on Env-pseudotype and producer cell type. The two proteins colocalize within cells, and both increase the yield of viral cDNA in target cells. The functional similarity of Nef and glycogag is a compelling example of convergent evolution in which two structurally unrelated proteins provide a function necessary for virion infectivity in lymphoid cells.


Subject(s)
Gene Products, gag/metabolism , HIV-1/pathogenicity , Leukemia Virus, Murine/metabolism , nef Gene Products, Human Immunodeficiency Virus/deficiency , Blotting, Western , DNA Primers/genetics , Electrophoresis, Polyacrylamide Gel , Evolution, Molecular , Glycosylation , HIV-1/metabolism , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/pathogenicity , Reverse Transcriptase Polymerase Chain Reaction
8.
J Immunol ; 181(12): 8460-77, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050264

ABSTRACT

The high mutation rate of HIV is linked to the generation of viruses expressing proteins with altered function whose impact on disease progression is unknown. We investigated how HIV-1 viruses lacking Env, Vpr, and Nef affect CD4(+) T cell survival. We found that in the absence of these proteins, HIV-1-infected CD4(+) primary T cells progress to the G(0) phase of the cell cycle and to cell death, indicating that viruses expressing inactive forms of these proteins can contribute to the CD4(+) T cell decline as the wild-type virus, suggesting that other HIV proteins are responsible for inducing apoptosis. Apoptosis in these cells is triggered by the alteration of the Egr1-PTEN-Akt (early growth response-1/phosphate and tensin homolog deleted on chromosome 10/Akt) and p53 pathways, which converge on the FOXO3a (Forkhead box transcription factor O class 3a) transcriptional activator. The FOXO3a target genes Fas ligand and TRAIL, involved in the extrinsic apoptotic pathway, and PUMA, Noxa, and Bim, which are part of the intrinsic apoptotic pathway, were also up-regulated, indicating that HIV infection leads to apoptosis by the engagement of multiple apoptotic pathways. RNAi-mediated knockdown of Egr1 and FOXO3a resulted in reduced apoptosis in HIV-infected HeLa and CD4(+) T cells, providing further evidence for their critical role in HIV-induced apoptosis and G(0) arrest. We tested the possibility that Tat is responsible for the T cell apoptosis observed with these mutant viruses. The induction of Egr1 and FOXO3a and its target genes was observed in Jurkat cells transduced by Tat alone. Tat-dependent activation of the Egr1-PTEN-FOXO3a pathway provides a mechanism for HIV-1-associated CD4(+) T cell death.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Forkhead Transcription Factors/physiology , HIV-1/immunology , tat Gene Products, Human Immunodeficiency Virus/physiology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , HIV-1/genetics , HeLa Cells , Humans , Jurkat Cells , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology , env Gene Products, Human Immunodeficiency Virus/deficiency , env Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/deficiency , nef Gene Products, Human Immunodeficiency Virus/genetics , vpr Gene Products, Human Immunodeficiency Virus/deficiency , vpr Gene Products, Human Immunodeficiency Virus/genetics
9.
Retrovirology ; 4: 66, 2007 Sep 23.
Article in English | MEDLINE | ID: mdl-17888184

ABSTRACT

In efforts to develop an effective vaccine, sterilizing immunity to primate lentiviruses has only been achieved by the use of live attenuated viruses carrying major deletions in nef and other accessory genes. Although live attenuated HIV vaccines are unlikely to be developed due to a myriad of safety concerns, opportunities exist to better understand the correlates of immune protection against HIV infection by studying rare cohorts of long-term survivors infected with attenuated, nef-deleted HIV strains such as the Sydney blood bank cohort (SBBC). Here, we review studies of viral evolution, pathogenicity, and immune responses to HIV infection in SBBC members. The studies show that potent, broadly neutralizing anti-HIV antibodies and robust CD8+ T-cell responses to HIV infection were not necessary for long-term control of HIV infection in a subset of SBBC members, and were not sufficient to prevent HIV sequence evolution, augmentation of pathogenicity and eventual progression of HIV infection in another subset. However, a persistent T-helper proliferative response to HIV p24 antigen was associated with long-term control of infection. Together, these results underscore the importance of the host in the eventual outcome of infection. Thus, whilst generating an effective antibody and CD8+ T-cell response are an essential component of vaccines aimed at preventing primary HIV infection, T-helper responses may be important in the generation of an effective therapeutic vaccine aimed at blunting chronic HIV infection.


Subject(s)
HIV Infections/immunology , HIV-1/pathogenicity , nef Gene Products, Human Immunodeficiency Virus/deficiency , Cohort Studies , HIV Infections/epidemiology , HIV Infections/virology , HIV Long Terminal Repeat , HIV Long-Term Survivors/statistics & numerical data , HIV-1/immunology , Sequence Deletion , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...