Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 162
Filter
Add more filters










Publication year range
1.
J Virol ; 79(15): 9579-87, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014920

ABSTRACT

The human cytidine deaminase Apobec3F (h-A3F), a protein related to the previously recognized antiviral factor Apobec3G (h-A3G), has antiviral activity against human immunodeficiency virus type 1 (HIV-1) that is suppressed by the viral protein Vif. The mechanism of HIV-1 Vif-mediated suppression of h-A3F is not fully understood. Here, we demonstrate that while h-A3F, like h-A3G, was able to suppress primate lentiviruses other than HIV-1 (simian immunodeficiency virus from African green monkeys [SIVagm] and Rhesus macaques [SIVmac]), the interaction between Vif proteins and h-A3F appeared to differ from that with h-A3G. H-A3F showed no change in its species specificity against HIV-1 or SIVagm Vif when a negatively charged amino acid was replaced with a lysine at position 128, a residue critical for h-A3G recognition by HIV-1 Vif. However, HIV-1 Vif, but not SIVagm Vif, was able to bind h-A3F and induce its polyubiquitination and degradation through the Cul5-containing E3 ubiquitin ligase. Interference with Cul5-E3 ligase function by depletion of Cul5, through RNA interference or overexpression of Cul5 mutants, blocked the ability of HIV-1 Vif to suppress h-A3F. A BC-box mutant of HIV-1 Vif that failed to recruit Cul5-E3 ligase but was still able to interact with h-A3F failed to suppress h-A3F. Interestingly, interference with Cul5-E3 ligase function or overexpression of h-A3F or h-A3G also increased the stability of HIV-1 Vif, suggesting that like the substrate molecules h-A3F and h-A3G, the substrate receptor protein Vif is itself also regulated by Cul5-E3 ligase. Our results indicate that Cul5-E3 ligase appears to be a common pathway hijacked by HIV-1 Vif to defeat both h-A3F and h-A3G. Developing inhibitors to disrupt the interaction between Vif and Cul5-E3 ligase could be therapeutically useful, allowing multiple host antiviral factors to suppress HIV-1.


Subject(s)
Antiviral Agents/physiology , Cullin Proteins/metabolism , Cytosine Deaminase/physiology , Gene Products, vif/metabolism , HIV-1/physiology , Ubiquitin-Protein Ligases/metabolism , Antiviral Agents/metabolism , Cell Line , Cullin Proteins/genetics , Cytosine Deaminase/metabolism , Gene Deletion , Humans , Lentiviruses, Primate/physiology , Ubiquitin-Protein Ligase Complexes/metabolism , Virus Replication , vif Gene Products, Human Immunodeficiency Virus
2.
Hepatology ; 42(2): 301-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16025511

ABSTRACT

APOBEC3G is a cellular cytidine deaminase displaying broad antiretroviral activity. Recently, it was shown that APOBEC3G can also suppress hepatitis B virus (HBV) production in human hepatoma cells. In the present study, we characterized the mechanisms of APOBEC-mediated antiviral activity against HBV and related hepadnaviruses. We show that human APOBEC3G blocks HBV production in mammalian and nonmammalian cells and is active against duck HBV as well. Early steps of viral morphogenesis, including RNA and protein synthesis, binding of pregenomic RNA to core protein, and self-assembly of viral core protein, were unaffected. However, APOBEC3G rendered HBV core protein-associated full-length pregenomic RNA nuclease-sensitive. Ongoing reverse-transcription in capsids that had escaped the block in morphogenesis was not significantly inhibited. The antiviral effect was not modulated by abrogating or enhancing expression of the accessory HBV X protein, suggesting that HBV X protein does not represent a functional homologue to the HIV vif protein. Furthermore, human APOBEC3F but not rat APOBEC1 inhibited HBV DNA production. Viral RNA and low-level DNA produced in the presence of APOBEC3F or rat APOBEC1 occasionally displayed mutations, but the majority of clones were wild-type. In conclusion, APOBEC3G and APOBEC3F but not rat APOBEC1 can downregulate the production of replication-competent hepadnaviral nucleocapsids. In contrast to HIV and other retroviruses, however, APOBEC3G/3F-mediated editing of nucleic acids does not seem to represent an effective innate defense mechanism for hepadnaviruses.


Subject(s)
Antiviral Agents/physiology , Cytosine Deaminase/physiology , Hepatitis B Virus, Duck/physiology , Hepatitis B virus/physiology , Proteins/physiology , APOBEC-1 Deaminase , APOBEC-3G Deaminase , Animals , Chickens , Cytidine Deaminase/physiology , DNA, Viral/biosynthesis , Hepatitis B virus/genetics , Nucleoside Deaminases , RNA-Directed DNA Polymerase/metabolism , Rats , Repressor Proteins , Viral Core Proteins/biosynthesis , Virus Assembly
3.
J Virol ; 79(14): 8969-78, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15994791

ABSTRACT

The tripartite motif 5alpha protein (TRIM5alpha) is one of several factors expressed by mammalian cells that inhibit retrovirus replication. Human TRIM5alpha (huTRIM5alpha) inhibits infection by N-tropic murine leukemia virus (N-MLV) but is inactive against human immunodeficiency virus type 1 (HIV-1). However, we show that replacement of a small segment in the carboxy-terminal B30.2/SPRY domain of huTRIM5alpha with its rhesus macaque counterpart (rhTRIM5alpha) endows it with the ability to potently inhibit HIV-1 infection. The B30.2/SPRY domain and an additional domain in huTRIM5alpha, comprising the amino-terminal RING and B-box components of the TRIM motif, are required for N-MLV restriction activity, while the intervening coiled-coil domain is necessary and sufficient for huTRIM5alpha multimerization. Truncated huTRIM5alpha proteins that lack either or both the N-terminal RING/B-Box or the C-terminal B30.2/SPRY domain form heteromultimers with full-length huTRIM5alpha and are dominant inhibitors of its N-MLV restricting activity, suggesting that homomultimerization of intact huTRIM5alpha monomers is necessary for N-MLV restriction. However, localization in large cytoplasmic bodies is not required for inhibition of N-MLV by huTRIM5alpha or for inhibition of HIV-1 by chimeric or rhTRIM5alpha.


Subject(s)
Antiviral Agents/physiology , Carrier Proteins/chemistry , Carrier Proteins/physiology , Retroviridae Infections/prevention & control , Amino Acid Sequence , Antiviral Restriction Factors , HIV-1/physiology , HeLa Cells , Humans , Leukemia Virus, Murine/physiology , Molecular Sequence Data , Structure-Activity Relationship , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
4.
Biochem Biophys Res Commun ; 331(4): 1358-64, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15883025

ABSTRACT

Human respiratory syncytial virus (hRSV) membrane fusion is promoted by the formation of a trimer-of-hairpins structure that brings the amino- and carboxyl-terminal regions of fusion (F) protein into close proximity. Two heptad-repeat (HR1 and HR2) regions in F protein play an important role in this process. Our previous study demonstrated that peptides derived from HR1 and HR2 regions of F protein were potent inhibitors of hRSV entry. Here we showed that HR1 peptide and its analog denoted 5-Helix which contained a central coiled-coil formed by three HR1s could induce highly potent antibody response in the immunized rabbits. Both antibodies could recognize F1 domain of the F protein and inhibited hRSV entry with the neutralizing antibody titers of 1:61 and 1:115, respectively. These suggested that 5-Helix could induce potent neutralizing antibody response and the central coiled-coil might be a highly conserved neutralization site for hRSV F protein.


Subject(s)
Antibodies/immunology , Antiviral Agents/physiology , Peptide Fragments/immunology , Respiratory Syncytial Viruses/metabolism , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Cell Line , Electrophoresis, Polyacrylamide Gel , Immune Sera , Molecular Sequence Data , Neutralization Tests
6.
Science ; 308(5721): 557-60, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15845854

ABSTRACT

In eukaryotes, 21- to 24-nucleotide-long RNAs engage in sequence-specific interactions that inhibit gene expression by RNA silencing. This process has regulatory roles involving microRNAs and, in plants and insects, it also forms the basis of a defense mechanism directed by small interfering RNAs that derive from replicative or integrated viral genomes. We show that a cellular microRNA effectively restricts the accumulation of the retrovirus primate foamy virus type 1 (PFV-1) in human cells. PFV-1 also encodes a protein, Tas, that suppresses microRNA-directed functions in mammalian cells and displays cross-kingdom antisilencing activities. Therefore, through fortuitous recognition of foreign nucleic acids, cellular microRNAs have direct antiviral effects in addition to their regulatory functions.


Subject(s)
Antiviral Agents/physiology , MicroRNAs/physiology , RNA Interference , Spumavirus/genetics , Spumavirus/physiology , Animals , Arabidopsis/genetics , Cell Line , Cricetinae , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Oligonucleotides, Antisense , Plants, Genetically Modified , Protein Biosynthesis , RNA, Viral , Retroviridae Proteins/genetics , Retroviridae Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection , Virus Replication
7.
Eur J Med Res ; 10(3): 117-20, 2005 Mar 29.
Article in English | MEDLINE | ID: mdl-15851378

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome (SARS) is a novel infectious disease which is characterized by an overaggressive immune response. Chemokines are important inflammatory mediators and regulate disease due to viral infection. In previous study, we found that SARS-CoV has the ability to replicate in mononuclear cells. In present work, we sought to characterize the replication of SARS-CoV at the presence of RANTES in THP-1 cells. METHODS: To determine whether RANTES play an role in the process of SARS, THP-1 cells were incubated with heat-inactivated SARS-CoV and ELISA was used to test RANTES levels in the supernatants; Then the effect of dexamethasone on the induced secretion was evaluated. Real-time PCR was used to investigate the effort of RANTES on the replication of SARS-CoV in vitro. Macrophages, induced by THP-1 cells, were used as cell model. FINDINGS: Inactive SARS-CoV could induce THP-1 cells secret RANTES and this increase effect could not be suppressed by DXM. RANTES itself could inhibit the replication of SARS-CoV in THP-1 cells when it was added into the culture before or at the same time with the virus; No inhibition effect was shown when RANTES were added into the culture after SARS-CoV infected the cells.


Subject(s)
Antiviral Agents/physiology , Chemokine CCL5/physiology , Severe acute respiratory syndrome-related coronavirus/physiology , Virus Replication/physiology , Animals , CHO Cells , Cell Line, Tumor , Chlorocebus aethiops , Cricetinae , Humans , Macrophages/virology , Vero Cells
8.
J Immunol ; 174(4): 1932-7, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699120

ABSTRACT

Toll-like receptors (TLRs) mediate host cell activation by various microbial components. TLR2, TLR3, TLR4, TLR7, TLR8, and TLR9 are the receptors that have been associated with virus-induced immune response. We have previously reported that all these TLRs, except TLR9, are expressed at mRNA levels in human monocyte-derived macrophages. Here we have studied TLR2, TLR3, TLR4, and TLR7/8 ligand-induced IFN-alpha, IFN-beta, IL-28, and IL-29 expression in human macrophages. IFN-alpha pretreatment of macrophages was required for efficient TLR3 and TLR4 agonist-induced activation of IFN-alpha, IFN-beta, IL-28, and IL-29 genes. TLR7/8 agonist weakly activated IFN-alpha, IFN-beta, IL-28, and IL-29 genes, whereas TLR2 agonist was not able to activate these genes. IFN-alpha enhanced TLR responsiveness in macrophages by up-regulating the expression of TLR3, TLR4, and TLR7. IFN-alpha also enhanced the expression of TLR signaling molecules MyD88, TIR domain-containing adaptor inducing IFN-beta, IkappaB kinase-epsilon, receptor interacting protein 1, and IFN regulatory factor 7. Furthermore, the activation of transcription factor IFN regulatory factor 3 by TLR3 and TLR4 agonists was dependent on IFN-alpha pretreatment. In conclusion, our results suggest that IFN-alpha sensitizes cells to microbial recognition by up-regulating the expression of several TLRs as well as adapter molecules and kinases involved in TLR signaling.


Subject(s)
Gene Expression Regulation/immunology , Interferon-alpha/physiology , Interferon-beta/genetics , Interleukins/genetics , Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Antiviral Agents/physiology , Cell Line , Cell-Free System/immunology , Cells, Cultured , Cytokines , DNA-Binding Proteins/metabolism , Humans , Influenza A virus/immunology , Interferon Regulatory Factor-1 , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Interferon-beta/biosynthesis , Interferon-gamma/biosynthesis , Interferons , Interleukins/biosynthesis , Ligands , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Membrane Glycoproteins/agonists , Membrane Glycoproteins/biosynthesis , NF-kappa B/metabolism , Phosphoproteins/metabolism , Receptors, Cell Surface/agonists , Receptors, Cell Surface/biosynthesis , Sendai virus/immunology , Signal Transduction/immunology , Toll-Like Receptor 2 , Toll-Like Receptor 3 , Toll-Like Receptor 4 , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Toll-Like Receptor 9 , Toll-Like Receptors
9.
J Immunol ; 174(3): 1574-9, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15661918

ABSTRACT

CD8(+) cells from HIV-infected individuals showing the CD8(+) cell noncytotoxic antiviral response unexpectedly revealed mRNA for VCAM-1, a cell surface molecule found on endothelial cells. Uninfected subjects had undetectable levels of VCAM-1 mRNA in their CD8(+) cells. Flow cytometry analysis showed that up to 12% of the CD8(+) cells from HIV-positive individuals expressed VCAM-1 compared with 0.8% of the CD8(+) cells of HIV-negative individuals. Enrichment of the CD8(+)VCAM-1(+) cell population and subsequent coculture with CD4(+) cells acutely infected with HIV-1 showed that the VCAM-1(+)CD8(+) cells were able to suppress viral replication with 50% less input cells than the unseparated CD8(+) cell population. This study demonstrates, for the first time, the expression of VCAM-1 on CD8(+) cells. Moreover, the CD8(+)VCAM-1(+) cells show enhanced CD8(+) cell noncytotoxic antiviral response activity that could have clinical importance in HIV infection.


Subject(s)
Adjuvants, Immunologic/biosynthesis , Anti-HIV Agents/metabolism , Antiviral Agents/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Up-Regulation/immunology , Vascular Cell Adhesion Molecule-1/biosynthesis , Acute Disease , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/physiology , Antiviral Agents/genetics , Antiviral Agents/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/virology , Cells, Cultured , Coculture Techniques , HIV Seronegativity/immunology , HIV Seropositivity/genetics , HIV Seropositivity/immunology , HIV Seropositivity/metabolism , HIV-1/growth & development , HIV-1/immunology , Humans , Interleukin-15/pharmacology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , RNA, Messenger/biosynthesis , T-Lymphocytes/immunology , T-Lymphocytes/virology , Up-Regulation/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/physiology
10.
J Immunol ; 174(3): 1587-93, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15661920

ABSTRACT

Human CMV is often associated with transplant rejection and opportunistic infections such as pneumonia in immunosuppressed patients. Current anti-CMV therapies, although effective, show relatively high toxicity, which seriously limits their long-term use. In this study, we provide evidence that leukotriene B(4) (LTB(4)) plays an important role in the fight against murine CMV (MCMV) infection in vivo. Intravenous administration of 50 and 500 ng/kg/day of LTB(4) to mice infected with a lethal dose of MCMV significantly increases their survival (50 and 70%, respectively), compared with the placebo-treated group (10% of survival). In mice infected with a sublethal dose of MCMV and treated daily with 50 ng/kg/day of LTB(4), the salivary gland viral loads were found to be reduced by 66% compared with the control group. Furthermore, using an allogeneic bone marrow transplantation mouse model, the frequency of MCMV reactivation from latently infected mice was much lower (38%) in LTB(4) (500 ng/kg)-treated mice than in the placebo-treated group (78%). Finally, in experiments using 5-lipoxygenase-deficient mice, MCMV viral loads in salivary glands were found to be higher in animals unable to produce leukotrienes than in the control groups, supporting a role of endogenous 5-lipoxygenase products, possibly LTB(4), in host defense against CMV infection.


Subject(s)
Antiviral Agents/therapeutic use , Bone Marrow Transplantation/adverse effects , Cytomegalovirus Infections/prevention & control , Leukotriene B4/therapeutic use , Muromegalovirus/physiology , Virus Activation , Virus Latency , Animals , Antiviral Agents/deficiency , Antiviral Agents/genetics , Antiviral Agents/physiology , Arachidonate 5-Lipoxygenase/deficiency , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/physiology , Bone Marrow Transplantation/immunology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/mortality , Cytomegalovirus Infections/virology , Female , Graft Rejection/genetics , Graft Rejection/prevention & control , Graft vs Host Disease/genetics , Graft vs Host Disease/prevention & control , Injections, Intravenous , Leukotriene B4/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Knockout , Salivary Glands/drug effects , Salivary Glands/virology , Spleen/cytology , Spleen/transplantation , Spleen/virology , Viral Load , Virus Activation/genetics , Virus Activation/immunology , Virus Latency/genetics , Virus Latency/immunology
11.
Curr Biol ; 15(2): 166-70, 2005 Jan 26.
Article in English | MEDLINE | ID: mdl-15668174

ABSTRACT

The antiretroviral activity of the cellular enzyme APOBEC3G has been attributed to the excessive deamination of cytidine (C) to uridine (U) in minus strand reverse transcripts, a process resulting in guanosine (G) to adenosine (A) hypermutation of plus strand DNAs. The HIV-1 Vif protein counteracts APOBEC3G by inducing proteasomal degradation and exclusion from virions through recruitment of a cullin5 ECS E3 ubiquitin ligase complex. APOBEC3G belongs to the APOBEC protein family, members of which possess consensus (H/C)-(A/V)-E-(X)24-30-P-C-(X)2-C cytidine deaminase motifs. Earlier analyses of APOBEC-1 have defined specific residues that are important for zinc coordination, proton transfer, and, therefore, catalysis within this motif. Because APOBEC3G contains two such motifs, we used site-directed mutagenesis of conserved residues to assess each region's contribution to anti-HIV-1 activity. Surprisingly, whereas either the N- or C-terminal domain could confer antiviral function in tissue culture-based infectivity assays, only an intact C-terminal motif was essential for DNA mutator activity. These findings reveal the nonequivalency of APOBEC3G's N- and C-terminal domains and imply that APOBEC3G-mediated DNA editing may not always be necessary for antiviral activity. Accordingly, we propose that APOBEC3G can achieve an anti-HIV-1 effect through an undescribed mechanism that is distinct from cytidine deamination.


Subject(s)
Antiviral Agents/physiology , Cytidine Deaminase/metabolism , Gene Products, vif/metabolism , HIV-1 , Mutation/physiology , Proteins/physiology , APOBEC-3G Deaminase , Amino Acid Motifs , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Mutagenesis, Site-Directed , Mutation/genetics , Nucleoside Deaminases , Protein Structure, Tertiary , Repressor Proteins , Virion/metabolism , vif Gene Products, Human Immunodeficiency Virus
12.
J Immunol ; 173(10): 6274-83, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15528366

ABSTRACT

The aim of this study was to investigate the mechanism of HIV-1 neutralization using monocyte-derived macrophages (MDM) in comparison to PBMC as target cells. For this purpose, we analyzed neutralizing activities of different human polyclonal IgG samples purified from sera of HIV-1-infected individuals using a single cycle infection assay. We found an increase of the neutralizing titer when macrophages vs PBMC were used as target cells. Moreover, polyclonal IgG from HIV-1-infected patients that are not able to neutralize virus when PBMC are used as target cells strongly inhibit MDM infection. Similar results were obtained with neutralizing mAbs. To explore the participation of FcgammaRs in HIV-1 inhibition, F(ab')(2) and Fab of these Igs were produced. Results indicated that both F(ab')(2) and Fab are less effective to inhibit virus replication in MDM. Moreover, competition experiments with Fc fragments of IgG from healthy donors or with purified monoclonal anti-human FcgammaRs Ab strengthen the participation of the FcgammaRs, and in particular of FcgammaRI (CD64) in HIV-1 inhibition on MDM. Mechanisms by which HIV-specific IgG inhibit virus replication in cultured macrophages are proposed and the benefit of inducing such Abs by vaccination is discussed.


Subject(s)
Antiviral Agents/physiology , HIV Infections/immunology , HIV-1/immunology , Immunoglobulin G/isolation & purification , Immunoglobulin G/physiology , Macrophages/immunology , Macrophages/metabolism , Receptors, IgG/physiology , Anti-HIV Agents/pharmacology , Binding, Competitive/immunology , Cells, Cultured , Chemokines/biosynthesis , Humans , Immunoglobulin A/isolation & purification , Immunoglobulin A/physiology , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fab Fragments/physiology , Immunoglobulin G/biosynthesis , Macrophages/virology , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Neutralization Tests , Phagocytosis/immunology , Protein Binding/immunology , Receptors, IgG/immunology , Receptors, IgG/metabolism , Virus Replication/immunology
13.
J Immunol ; 173(10): 6303-11, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15528369

ABSTRACT

A large proportion of the CD8(+) T cell pool in persons chronically infected with HIV consists of cells that show features of replicative senescence, an end stage characterized by irreversible cell cycle arrest, multiple genetic and functional changes, and shortened telomeres. The objective of our research was to determine whether constitutive expression of the gene for the human telomerase (hTERT) can prevent senescence-induced impairments in human virus-specific CD8(+) T cells, particularly in the context of HIV-1 disease. Our results indicate that hTERT-expressing HIV-specific CD8(+) lymphocytes show both an enhanced and sustained capacity to inhibit HIV-1 replication in in vitro coculture experiments, as well as prolonged ability to produce IFN-gamma and TNF-alpha in response to stimulation with HIV-1-derived peptides, as compared with vector-transduced controls. Loss of CD28 expression, the signature change of replicative senescence in cell culture, was retarded in those CD8(+) T cell cultures that had high levels of CD28 at the time of hTERT transduction. These findings suggest that telomere shortening may be the primary driving force behind several aspects of CD8(+) T cell dysfunction associated with replicative senescence. We also demonstrate reduced accumulation of the p16(INK4a) and p21(WAF1) cell cycle inhibitors in hTERT-transduced lymphocytes, providing a possible mechanism by which stable hTERT expression is able to circumvent the senescence barrier in CD8(+) T cells. Given the key role of CD8(+) T cell function in controlling a variety of acute and latent viral infections, approaches to retard the functional decrements associated with replicative senescence may lead to novel types of immunotherapy.


Subject(s)
Adjuvants, Immunologic/physiology , Antiviral Agents/physiology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HIV-1/immunology , Telomerase/physiology , Telomere/metabolism , Adjuvants, Immunologic/biosynthesis , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/metabolism , Antiviral Agents/biosynthesis , Antiviral Agents/genetics , Antiviral Agents/metabolism , CD28 Antigens/biosynthesis , CD28 Antigens/physiology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/virology , Cell Cycle/genetics , Cell Cycle/immunology , Cell Proliferation , Cells, Cultured , Cellular Senescence/genetics , Cellular Senescence/immunology , Cytotoxicity, Immunologic/genetics , DNA-Binding Proteins , Growth Inhibitors/antagonists & inhibitors , Growth Inhibitors/biosynthesis , HIV-1/growth & development , HIV-1/physiology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/physiology , Telomerase/biosynthesis , Telomerase/genetics , Telomerase/metabolism , Telomere/enzymology , Telomere/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/physiology , Up-Regulation/immunology , Virus Replication/genetics , Virus Replication/immunology
14.
J Immunol ; 173(6): 4108-19, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15356161

ABSTRACT

Adaptive cellular immunity is required to clear HSV-1 infection in the periphery. Myeloid dendritic cells (DCs) are the first professional Ag-presenting cell to encounter the virus after primary and secondary infection and thus the consequences of their infection are important in understanding the pathogenesis of the disease and the response to the virus. Following HSV-1 infection, both uninfected and infected human DCs acquire a more mature phenotype. In this study, we demonstrate that type I IFN secreted from myeloid DC mediates bystander activation of the uninfected DCs. Furthermore, we confirm that this IFN primes DCs for elevated IL-12 p40 and p70 secretion. However, secretion of IFN is not responsible for the acquisition of a mature phenotype by HSV-1-infected DC. Rather, virus binding to a receptor on the cell surface induces DC maturation directly, through activation of the NF-kappaB and p38 MAPK pathways. The binding of HSV glycoprotein D is critical to the acquisition of a mature phenotype and type I IFN secretion. The data therefore demonstrate that DCs can respond to HSV exposure directly through recognition of viral envelope structures. In the context of natural HSV infection, the coupling of viral entry to the activation of DC signaling pathways is likely to be counterbalanced by viral disruption of DC maturation. However, the parallel release of type I IFN may result in paracrine activation so that the DCs are nonetheless able to mount an adaptive immune response.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Herpesvirus 1, Human/immunology , Interferon Type I/metabolism , Myeloid Cells/immunology , Myeloid Cells/virology , Paracrine Communication/immunology , Antigens, CD/biosynthesis , Antiviral Agents/physiology , B7-2 Antigen , Cell Differentiation/immunology , Cell Membrane/enzymology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/virology , Cells, Cultured , Dendritic Cells/metabolism , Enzyme Activation/immunology , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/radiation effects , Humans , Interferon Type I/antagonists & inhibitors , Interferon Type I/physiology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Myeloid Cells/metabolism , NF-kappa B/metabolism , Neutralization Tests , Ultraviolet Rays , Up-Regulation/immunology , Viral Envelope Proteins/physiology , Virus Inactivation/radiation effects , p38 Mitogen-Activated Protein Kinases
15.
J Immunol ; 173(7): 4618-26, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383596

ABSTRACT

During HIV-1 infection or vaccination, HIV-1 envelope spikes elicit Ab responses. Neutralizing Abs block viral entry by recognizing epitopes on spikes critical for their interaction with receptor, coreceptors or fusion. In contrast, nonneutralizing Abs fail to do so because they recognize epitopes either buried or exposed but not critical for viral entry. Previously, we produced a high-affinity human mAb against the cluster II determinant of gp41. This Ab or its recombinant Fab and single-chain Fv have been repeatedly shown to bind to HIV-1 gp160 or gp41, but fail to block viral entry. We report that, surprisingly, expression of this nonneutralizing anti-HIV-1 gp41 single-chain Fv on the surface of human CD4 T cells markedly inhibits HIV-1 replication and cell-cell fusion. The inhibition targets the HIV-1 envelope at the level of viral entry, regardless of HIV-1 tropism. Although this bona fide nonneutralizing Ab does not neutralize HIV-1 entry when produced as a soluble protein, it acts as a neutralizing Ab when expressed on the cell surface. Expressing Abs on the surface of HIV-1-susceptible cells can be a new way to fight HIV-1.


Subject(s)
Antiviral Agents/physiology , HIV Antibodies/physiology , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , HIV-1/pathogenicity , Receptors, Antigen, B-Cell/physiology , Antiviral Agents/biosynthesis , Cell Line, Tumor , Cell Membrane/immunology , Cell Membrane/virology , Disease Susceptibility/immunology , Gene Products, env/antagonists & inhibitors , Genetic Vectors , Giant Cells/immunology , Giant Cells/virology , Green Fluorescent Proteins , HIV Antibodies/biosynthesis , HIV Fusion Inhibitors/immunology , HIV-1/physiology , Humans , Immunoglobulin Fragments/biosynthesis , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/physiology , Luminescent Proteins/genetics , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Neutralization Tests , Receptors, Antigen, B-Cell/biosynthesis , Transduction, Genetic , Transgenes , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/genetics , Virus Replication/immunology
16.
Curr Biol ; 14(15): 1385-91, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15296757

ABSTRACT

APOBEC3G (CEM15 ) deaminates cytosine to uracil in nascent retroviral cDNA. The potency of this cellular defense is evidenced by a dramatic reduction in viral infectivity and the occurrence of high frequencies of retroviral genomic-strand G --> A transition mutations. The overwhelming dinucleotide hypermutation preference of APOBEC3G acting upon a variety of model retroviral substrates is 5'-GG --> -AG. However, a distinct 5'-GA --> -AA bias, which is difficult to attribute to APOBEC3G alone, prevails in HIV-1 sequences derived from infected individuals (e.g., ). Here, we show that APOBEC3F is also a potent retroviral restrictor but that its activity, unlike that of APOBEC3G, is partially resistant to HIV-1 Vif and results in a clear 5'-GA --> -AA retroviral hypermutation preference. This bias is also apparent in a bacterial mutation assay, suggesting that it is an intrinsic APOBEC3F property. Moreover, APOBEC3F and APOBEC3G appear to be coordinately expressed in a wide range of human tissues and are independently able to inhibit retroviral infection. Thus, APOBEC3F and APOBEC3G are likely to function alongside one another in the provision of an innate immune defense, with APOBEC3F functioning as the major contributor to HIV-1 hypermutation in vivo.


Subject(s)
Antiviral Agents/physiology , Cytosine Deaminase/metabolism , HIV-1/genetics , Mutagenesis/genetics , Proteins/metabolism , APOBEC-3G Deaminase , Amino Acid Sequence , Base Sequence , Blotting, Northern , Cytidine Deaminase , DNA Primers , Genes, vif/genetics , HIV-1/metabolism , Humans , Molecular Sequence Data , Nucleoside Deaminases , Point Mutation/genetics , Proteins/genetics , Repressor Proteins , Sequence Alignment , Sequence Analysis, DNA , Virus Replication/genetics , Virus Replication/physiology
17.
J Gen Virol ; 85(Pt 8): 2315-2326, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15269373

ABSTRACT

The interferon-induced murine Mx1 GTPase is a nuclear protein. It specifically inhibits influenza A viruses at the step of primary transcription, a process known to occur in the nucleus of infected cells. However, the exact mechanism of inhibition is still poorly understood. The Mx1 GTPase has previously been shown to accumulate in distinct nuclear dots that are spatially associated with promyelocytic leukaemia protein (PML) nuclear bodies (NBs), but the significance of this association is not known. Here it is reported that, in cells lacking PML and, as a consequence, PML NBs, Mx1 still formed nuclear dots. These dots were indistinguishable from the dots observed in wild-type cells, indicating that intact PML NBs are not required for Mx1 dot formation. Furthermore, Mx1 retained its antiviral activity against influenza A virus in these PML-deficient cells, which were fully permissive for influenza A virus. Nuclear Mx proteins from other species showed a similar subnuclear distribution. This was also the case for the human MxA GTPase when this otherwise cytoplasmic protein was translocated into the nucleus by virtue of a foreign nuclear localization signal. Human MxA and mouse Mx1 do not interact or form heterooligomers. Yet, they co-localized to a large degree when co-expressed in the nucleus. Taken together, these findings suggest that Mx1 dots represent distinct nuclear domains ('Mx nuclear domains') that are frequently associated with, but functionally independent of, PML NBs.


Subject(s)
Antiviral Agents/physiology , GTP-Binding Proteins/physiology , Influenza A virus/growth & development , Neoplasm Proteins/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , Cell Nucleus/chemistry , GTP-Binding Proteins/analysis , Mice , Myxovirus Resistance Proteins , Promyelocytic Leukemia Protein , Tumor Suppressor Proteins
18.
J Virol ; 78(15): 8238-44, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15254195

ABSTRACT

Human APOBEC3G (huAPOBEC3G), also known as CEM15, is a broad antiretroviral host factor that deaminates dC to dU in the minus strand DNA of human immunodeficiency virus type 1 (HIV-1), other lentiviruses, and murine leukemia virus (MLV), thereby creating G-to-A hypermutation in the plus strand DNA to inhibit the infectivity of these viruses. In this study, we examined the antiretroviral function of a murine homologue of APOBEC3G (muAPOBEC3G) on several retrovirus systems with different producer cells. MuAPOBEC3G did not suppress the infectivity of murine retroviral vectors produced from human or murine cells, whereas it showed antiviral activity on both wild-type and Deltavif virions of HIV-1 in human cells. In contrast, huAPOBEC3G showed broad antiviral activity on HIV-1 and murine retroviral vectors produced from human cells as well as murine cells. These data suggested that muAPOBEC3G does not possess antiretroviral activity on murine retroviruses and has a different target specificity from that of huAPOBEC3G and that huAPOBEC3G works as a broad antiviral factor not only in human cells but also in murine cells. A functional interaction study between human and murine APOBEC3G supported the former hypothesis. Furthermore, studies on the expression of APOBEC3G in producer cells and its incorporation into virions revealed that muAPOBEC3G is incorporated into HIV-1 virions but not into MLV virions. Thus, muAPOBEC3G cannot suppress the infectivity of murine retrovirus because it is not incorporated into virions. We suggest that murine retroviruses can replicate in murine target cells expressing muAPOBEC3G because they are not targets for this enzyme.


Subject(s)
Antiviral Agents/physiology , Proteins/physiology , APOBEC-3G Deaminase , Animals , Cell Line , Cytidine Deaminase , HIV-1/pathogenicity , Humans , Leukemia Virus, Murine/pathogenicity , Mice , Nucleoside Deaminases , Repressor Proteins , Species Specificity , Virion/pathogenicity
19.
AIDS Res Hum Retroviruses ; 20(6): 600-7, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15242536

ABSTRACT

Human saliva contains multiple components that inhibit HIV-1 infection in vitro, which may contribute to low oral HIV-1 transmission. Salivary agglutinin (SAG) is a high-molecular-weight glycoprotein encoded by DMBT-1 and identical to gp340, a member of the lung scavange receptor, cysteine-rich receptor family. gp340 binds to surfactants A and D, which is believed to function in the clearance of microorganisms from the lung, as part of the innate immune response. Previously we reported that SAG (gp340) specifically inhibits HIV-1 infection with broad activity against diverse HIV-1 isolates. This gp340 inhibitory activity is mediated by binding to viral gp120 and involves a region different from the CD4-binding site on gp120. Here, we report that the gp340-binding region is localized to a linear, highly conserved sequence near the stem of the V3 loop that is critical for chemokine receptor interaction during viral binding and infection. The interaction of gp340 with gp120 is enhanced by prebinding of sCD4 to gp120, suggesting that gp340 inhibitory activity is mediated by blocking access of the gp120 to the chemokine receptor.


Subject(s)
Agglutinins/metabolism , Antiviral Agents/physiology , HIV Envelope Protein gp120/metabolism , Peptide Fragments/metabolism , Receptors, CCR5/metabolism , Receptors, Cell Surface/metabolism , Antiviral Agents/metabolism , Binding Sites , CD4 Antigens/metabolism , Calcium-Binding Proteins , DNA-Binding Proteins , Humans , Models, Molecular , Protein Binding , Tumor Suppressor Proteins
20.
J Virol ; 78(11): 6073-6, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15141007

ABSTRACT

Recently, APOBEC3G has been identified as a host factor that blocks retroviral replication. It introduces G to A hypermutations in newly synthesized minus strand viral cDNA at the step of reverse transcription in target cells. Here, we identified the human APOBEC3F protein as another host factor that blocks human immunodeficiency virus type 1 (HIV-1) replication. Similar to APOBEC3G, APOBEC3F also induced G to A hypermutations in HIV genomic DNA, and the viral Vif protein counteracted its activity. Thus, APOBEC family members might have evolved as a general defense mechanism of the body against retroviruses, retrotransposons, and other mobile genetic elements.


Subject(s)
Antiviral Agents/physiology , Apolipoproteins B/genetics , Cytidine Deaminase/physiology , HIV-1/physiology , Virus Replication , APOBEC-1 Deaminase , APOBEC-3G Deaminase , Amino Acid Sequence , Base Sequence , Cytidine Deaminase/chemistry , Gene Products, vif/physiology , HIV-1/genetics , Humans , Molecular Sequence Data , Mutation , Nucleoside Deaminases , Proteins/chemistry , Repressor Proteins , Transcription, Genetic , vif Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...