Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Gen Virol ; 100(4): 656-661, 2019 04.
Article in English | MEDLINE | ID: mdl-30767852

ABSTRACT

The release of porcine endogenous retrovirus (PERV) particles from pig cells is a potential risk factor during xenotransplantation by way of productively infecting the human transplant recipient. Potential countermeasures against PERV replication are restriction factors that block retroviral replication. SAMHD1 is a triphosphohydrolase that depletes the cellular pool of dNTPs in non-cycling cells starving retroviral reverse transcription. We investigated the antiviral activity of human SAMHD1 against PERV and found that SAMHD1 potently restricts its reverse transcription in human monocytes, monocyte-derived dendritic cells (MDDC), or macrophages (MDM) and in monocytic THP-1 cells. Degradation of SAMHD1 by SIVmac Vpx or CRISPR/Cas9 knock-out of SAMHD1 allowed for PERV reverse transcription. Addition of deoxynucleosides alleviated the SAMHD1-mediated restriction suggesting that SAMHD1-mediated degradation of dNTPs restricts PERV replication in these human immune cells. In conclusion, our findings highlight SAMHD1 as a potential barrier to PERV transmission from pig transplants to human recipients during xenotransplantation.


Subject(s)
Endogenous Retroviruses/physiology , Heterografts/metabolism , Heterografts/virology , SAM Domain and HD Domain-Containing Protein 1/metabolism , Animals , CRISPR-Cas Systems/physiology , Cell Line , HEK293 Cells , Humans , Macrophages/metabolism , Macrophages/virology , Monocytes/metabolism , Monocytes/virology , Reverse Transcription/physiology , Swine , THP-1 Cells , Transplantation, Heterologous/methods , Virus Replication/physiology
2.
Front Microbiol ; 9: 178, 2018.
Article in English | MEDLINE | ID: mdl-29487579

ABSTRACT

Initial indications that retroviruses are connected to neoplastic transformation were seen more than a century ago. This concept has also been tested for endogenized retroviruses (ERVs) that are abundantly expressed in many transformed cells. In healthy cells, ERV expression is commonly prevented by DNA methylation and other epigenetic control mechanisms. ERVs are remnants of former exogenous forms that invaded the germ line of the host and have since been vertically transmitted. Several examples of ERV-induced genomic recombination events and dysregulation of cellular genes that contribute to tumor formation have been well documented. Moreover, evidence is accumulating that certain ERV proteins have oncogenic properties. In contrast to these implications for supporting cancer induction, a recent string of papers has described favorable outcomes of increasing human ERV (HERV) RNA and DNA abundance by treatment of cancer cells with methyltransferase inhibitors. Analogous to an infecting agent, the ERV-derived nucleic acids are sensed in the cytoplasm and activate innate immune responses that drive the tumor cell into apoptosis. This "viral mimicry" induced by epigenetic drugs might offer novel therapeutic approaches to help target cancer cells that are normally difficult to treat using standard chemotherapy. In this review, we discuss both the detrimental and the new beneficial role of HERV reactivation in terms of its implications for cancer.

3.
J Mol Biol ; 429(8): 1171-1191, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28315663

ABSTRACT

The retroviral restriction factors of the APOBEC3 (A3) cytidine deaminase family catalyze the deamination of cytidines in single-stranded viral DNA. APOBEC3C (A3C) is a strong antiviral factor against viral infectivity factor (vif)-deficient simian immunodeficiency virus Δvif, which is, however, a weak inhibitor against human immunodeficiency virus (HIV)-1 for reasons unknown. The precise link between the antiretroviral effect of A3C and its catalytic activity is incompletely understood. Here, we show that the S61P mutation in human A3C (A3C.S61P) boosted hypermutation in the viral genomes of simian immunodeficiency virus Δvif and murine leukemia virus but not in human immunodeficiency virus HIV-1Δvif. The enhanced antiviral activity of A3C.S61P correlated with enhanced in vitro cytidine deamination. Furthermore, the S61P mutation did not change the substrate specificity of A3C, ribonucleoprotein complex formation, self-association, Zinc coordination, or viral incorporation features. We propose that local structural changes induced by the serine-to-proline substitution are responsible for the gain of catalytic activity of A3C.S61P. Our results are a first step toward an understanding of A3C's DNA binding capacity, deamination-dependent editing, and antiviral functions at the molecular level. We conclude that the enhanced enzymatic activity of A3C is insufficient to restrict HIV-1, indicating an unknown escape mechanism of HIV-1.


Subject(s)
Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , HIV-1/pathogenicity , Amino Acid Substitution , Animals , Cytidine Deaminase/genetics , Cytosine/metabolism , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA, Viral/metabolism , HEK293 Cells/virology , HIV-1/genetics , Host-Pathogen Interactions , Humans , Leukemia Virus, Murine/metabolism , Leukemia Virus, Murine/pathogenicity , Pan troglodytes , Protein Conformation , Simian Immunodeficiency Virus/metabolism , Simian Immunodeficiency Virus/pathogenicity , Zinc/metabolism , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism
4.
J Biol Chem ; 292(3): 1068-1080, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27920203

ABSTRACT

The triphosphohydrolase SAMHD1 (sterile α motif and histidine-aspartate domain-containing protein 1) restricts HIV-1 replication in nondividing myeloid cells by depleting the dNTP pool, preventing reverse transcription. SAMHD1 is also reported to have ribonuclease activity that degrades the virus genomic RNA. Human SAMHD1 is regulated by phosphorylation of its carboxyl terminus at Thr-592, which abrogates its antiviral function yet has only a small effect on its phosphohydrolase activity. In the mouse, SAMHD1 is expressed as two isoforms (ISF1 and ISF2) that differ at the carboxyl terminus due to alternative splicing of the last coding exon. In this study we characterized the biochemical and antiviral properties of the two mouse isoforms of SAMHD1. Both are antiviral in nondividing cells. Mass spectrometry analysis showed that SAMHD1 is phosphorylated at several amino acid residues, one of which (Thr-634) is homologous to Thr-592. Phosphomimetic mutation at Thr-634 of ISF1 ablates its antiviral activity yet has little effect on phosphohydrolase activity in vitro dGTP caused ISF1 to tetramerize, activating its catalytic activity. In contrast, ISF2, which lacks the phosphorylation site, was significantly more active, tetramerized, and was active without added dGTP. Neither isoform nor human SAMHD1 had detectable RNase activity in vitro or affected HIV-1 genomic RNA stability in newly infected cells. These data support a model in which SAMHD1 catalytic activity is regulated through tetramer stabilization by the carboxyl-terminal tail, phosphorylation destabilizing the complexes and inactivating the enzyme. ISF2 may serve to reduce the dNTP pool to very low levels as a means of restricting virus replication.


Subject(s)
HIV Infections/enzymology , HIV-1/physiology , Monomeric GTP-Binding Proteins/metabolism , Protein Multimerization , RNA, Viral/metabolism , Virus Replication/physiology , Amino Acid Substitution , Animals , HIV Infections/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Models, Molecular , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/genetics , Mutation, Missense , Phosphorylation , RNA, Viral/genetics , SAM Domain and HD Domain-Containing Protein 1 , U937 Cells
5.
Retrovirology ; 13(1): 83, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27905985

ABSTRACT

BACKGROUND: Monocytes, the primary myeloid cell-type in peripheral blood, are resistant to HIV-1 infection as a result of the lentiviral restriction factor SAMHD1. Toll-like receptors recognize microbial pathogen components, inducing the expression of antiviral host proteins and proinflammatory cytokines. TLR agonists that mimic microbial ligands have been found to have activity against HIV-1 in macrophages. The induction of restriction factors in monocytes by TLR agonist activation has not been well studied. To analyze restriction factor induction by TLR activation in monocytes, we used the imidazoquinoline TLR7/8 agonist R848 and infected with HIV-1 reporter virus that contained packaged viral accessory protein Vpx, which allows the virus to escape SAMHD1-mediated restriction.  RESULTS: R848 prevented the replication of Vpx-containing HIV-1 and HIV-2 in peripheral blood mononuclear cells and monocytes. The block was post-entry but prior to reverse transcription of the viral genomic RNA. The restriction was associated with destabilization of the genomic RNA molecules of the in-coming virus particle. R848 treatment of activated T cells did not protect them from infection but treated monocytes produced high levels of proinflammatory cytokines, including type-I IFN that protected bystander activated T cells from infection. CONCLUSION: The activation of TLR7/8 induces two independent restrictions to HIV-1 replication in monocytes: a cell-intrinsic block that acts post-entry to prevent reverse transcription; and a cell-extrinsic block, in which monocytes produce high levels of proinflammatory cytokines (primarily type-I IFN) that protects bystander monocytes and T lymphocytes. The cell-intrinsic block may result from the induction of a novel restriction factor, which can be termed Lv5 and acts by destabilizing the in-coming viral genomic RNA, either by the induction of a host ribonuclease or by disrupting the viral capsid. TLR agonists are being developed for therapeutic use to diminish the size of the latent provirus reservoir in HIV-1 infected individuals. Such drugs may both induce latent provirus expression and restrict virus replication during treatment.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Imidazoles/pharmacology , Monocytes/drug effects , Monocytes/virology , Cell Line , HEK293 Cells , HIV-1/physiology , Humans , Interferon Type I/biosynthesis , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Lymphocyte Activation/drug effects , Monocytes/immunology , Monomeric GTP-Binding Proteins/genetics , RNA, Viral , Reverse Transcription , SAM Domain and HD Domain-Containing Protein 1 , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Virus Replication/drug effects
6.
PLoS One ; 11(6): e0155422, 2016.
Article in English | MEDLINE | ID: mdl-27249646

ABSTRACT

APOBEC4 (A4) is a member of the AID/APOBEC family of cytidine deaminases. In this study we found a high mRNA expression of A4 in human testis. In contrast, there were only low levels of A4 mRNA detectable in 293T, HeLa, Jurkat or A3.01 cells. Ectopic expression of A4 in HeLa cells resulted in mostly cytoplasmic localization of the protein. To test whether A4 has antiviral activity similar to that of proteins of the APOBEC3 (A3) subfamily, A4 was co-expressed in 293T cells with wild type HIV-1 and HIV-1 luciferase reporter viruses. We found that A4 did not inhibit the replication of HIV-1 but instead enhanced the production of HIV-1 in a dose-dependent manner and seemed to act on the viral LTR. A4 did not show detectable cytidine deamination activity in vitro and weakly interacted with single-stranded DNA. The presence of A4 in virus producer cells enhanced HIV-1 replication by transiently transfected A4 or stably expressed A4 in HIV-susceptible cells. APOBEC4 was capable of similarly enhancing transcription from a broad spectrum of promoters, regardless of whether they were viral or mammalian. We hypothesize that A4 may have a natural role in modulating host promoters or endogenous LTR promoters.


Subject(s)
Cytidine Deaminase/physiology , HIV-1/physiology , Virus Replication/physiology , Cell Line , Cytidine/metabolism , Cytidine Deaminase/metabolism , Deamination , HIV Long Terminal Repeat , Humans , Male , Promoter Regions, Genetic , Testis/metabolism
7.
Nucleic Acids Res ; 42(1): 396-416, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24101588

ABSTRACT

LINE-1 (L1) retrotransposons are mobile genetic elements whose extensive proliferation resulted in the generation of ≈ 34% of the human genome. They have been shown to be a cause of single-gene diseases. Moreover, L1-encoded endonuclease can elicit double-strand breaks that may lead to genomic instability. Mammalian cells adopted strategies restricting mobility and deleterious consequences of uncontrolled retrotransposition. The human APOBEC3 protein family of polynucleotide cytidine deaminases contributes to intracellular defense against retroelements. APOBEC3 members inhibit L1 retrotransposition by 35-99%. However, genomic L1 retrotransposition events that occurred in the presence of L1-restricting APOBEC3 proteins are devoid of detectable G-to-A hypermutations, suggesting one or multiple deaminase-independent L1 restricting mechanisms. We set out to uncover the mechanism of APOBEC3C (A3C)-mediated L1 inhibition and found that it is deaminase independent, requires an intact dimerization site and the RNA-binding pocket mutation R122A abolishes L1 restriction by A3C. Density gradient centrifugation of L1 ribonucleoprotein particles, subcellular co-localization of L1-ORF1p and A3C and co-immunoprecipitation experiments indicate that an RNA-dependent physical interaction between L1 ORF1p and A3C dimers is essential for L1 restriction. Furthermore, we demonstrate that the amount of L1 complementary DNA synthesized by L1 reverse transcriptase is reduced by ≈ 50% if overexpressed A3C is present.


Subject(s)
Cytidine Deaminase/metabolism , Long Interspersed Nucleotide Elements , Proteins/metabolism , RNA-Directed DNA Polymerase/metabolism , Carrier Proteins/analysis , Cytidine Deaminase/chemistry , Cytidine Deaminase/genetics , Cytoplasmic Granules/chemistry , Cytoplasmic Granules/enzymology , DNA Helicases , HeLa Cells , Humans , Mutation , Poly-ADP-Ribose Binding Proteins , Protein Multimerization , Proteins/analysis , Proteins/chemistry , RNA Helicases , RNA Recognition Motif Proteins
8.
J Virol ; 87(21): 11741-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23986575

ABSTRACT

The deoxynucleoside triphosphohydrolase SAMHD1 restricts retroviral replication in myeloid cells. Human immunodeficiency virus type 2 (HIV-2) and a simian immunodeficiency virus from rhesus macaques (SIVmac) encode Vpx, a virion-packaged accessory protein that counteracts SAMHD1 by inducing its degradation. SAMHD1 is thought to work by depleting the pool of intracellular deoxynucleoside triphosphates but has also been reported to have exonuclease activity that could allow it to degrade the viral genomic RNA or viral reverse-transcribed DNA. To induce the degradation of SAMHD1, Vpx co-opts the cullin4a-based E3 ubiquitin ligase, CRL4. E3 ubiquitin ligases are regulated by the covalent attachment of the ubiquitin-like protein Nedd8 to the cullin subunit. Neddylation can be prevented by MLN4924, a drug that inhibits the nedd8-activating enzyme. We report that MLN4924 inhibits the neddylation of CRL4, blocking Vpx-induced degradation of SAMHD1 and maintaining the restriction. Removal of the drug several hours postinfection released the block. Similarly, Vpx-containing virus-like particles and deoxynucleosides added to the cells more than 24 h postinfection released the SAMHD1-mediated block. Taken together, these findings support deoxynucleoside triphosphate pool depletion as the primary mechanism of SAMHD1 restriction and argue against a nucleolytic mechanism, which would not be reversible.


Subject(s)
Cullin Proteins/metabolism , HIV-1/immunology , Monomeric GTP-Binding Proteins/metabolism , Ubiquitins/metabolism , Virus Replication , Cell Line , Cullin Proteins/immunology , Cyclopentanes/metabolism , Enzyme Inhibitors/metabolism , HIV-1/physiology , Humans , Monomeric GTP-Binding Proteins/immunology , NEDD8 Protein , Protein Processing, Post-Translational/drug effects , Pyrimidines/metabolism , SAM Domain and HD Domain-Containing Protein 1 , Ubiquitins/antagonists & inhibitors
9.
Retrovirology ; 10: 26, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23497255

ABSTRACT

BACKGROUND: SAMHD1 is a triphosphohydrolase that restricts the replication of HIV-1 and SIV in myeloid cells. In macrophages and dendritic cells, SAMHD1 restricts virus replication by diminishing the deoxynucleotide triphosphate pool to a level below that which supports lentiviral reverse transcription. HIV-2 and related SIVs encode the accessory protein Vpx to induce the proteasomal degradation of SAMHD1 following virus entry. While SAMHD1 has been shown to restrict HIV-1 and SIV, the breadth of its restriction is not known and whether other viruses have a means to counteract the restriction has not been determined. RESULTS: We show that SAMHD1 restricts a wide array of divergent retroviruses, including the alpha, beta and gamma classes. Murine leukemia virus was restricted by SAMHD1 in macrophages yet removal of SAMHD1 did not alleviate the block to infection because of an additional block to viral nuclear import. Prototype foamy virus (PFV) and Human T cell leukemia virus type I (HTLV-1) were the only retroviruses tested that were not restricted by SAMHD1. PFV reverse transcribes predominantly prior to entry and thus is unaffected by the dNTP level in the target cell. It is possible that HTLV-1 has a mechanism to render the virus resistant to SAMHD1-mediated restriction. CONCLUSION: The results suggest that SAMHD1 has broad anti-retroviral activity against which most viruses have not found an escape.


Subject(s)
Macrophages/virology , Monomeric GTP-Binding Proteins/pharmacology , Myeloid Cells/virology , Retroviridae/drug effects , Retroviridae/pathogenicity , Virus Replication/drug effects , Cell Line , Dendritic Cells/metabolism , Dendritic Cells/virology , HIV-1/drug effects , HIV-1/physiology , Humans , Jurkat Cells , Macrophages/immunology , Monomeric GTP-Binding Proteins/metabolism , Myeloid Cells/metabolism , Retroviridae/classification , Retroviridae/physiology , SAM Domain and HD Domain-Containing Protein 1
10.
J Virol ; 86(23): 12552-60, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22973040

ABSTRACT

Sterile alpha motif domain- and HD domain-containing protein 1 (SAMHD1) is a deoxynucleoside triphosphohydrolase that restricts the replication of lentiviruses in myeloid cells by hydrolyzing the cellular deoxynucleotide triphosphates to a level below that which is required for reverse transcription. Human immunodeficiency virus type 2 (HIV-2) and some simian immunodeficiency viruses (SIVs) encode the accessory protein viral protein X (Vpx) that counteracts SAMHD1. Vpx recruits SAMHD1 to a cullin4A-RING E3 ubiquitin ligase (CRL4), which targets the enzyme for proteasomal degradation. Vpx and SAMHD1 both localize to the nucleus of the cell. We identified the nuclear localization sequence (NLS) of SAMHD1 as a conserved KRPR sequence at amino acid residues 11 to 14. SAMHD1 lacking a functional NLS localized to the cytoplasm but retained its triphosphohydrolase and antiviral activities. However, cytoplasmic SAMHD1 was resistant to Vpx-induced degradation, and its antiviral activity was not counteracted by Vpx. Cytoplasmic SAMHD1 interacted with Vpx and retained it in the cytoplasm. The inhibition of nuclear export with leptomycin B did not impair the ability of Vpx to degrade SAMHD1. These findings suggest that SAMHD1 is targeted by Vpx for ubiquitination and degradation in the nucleus.


Subject(s)
Cell Nucleus/metabolism , Monomeric GTP-Binding Proteins/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Amino Acid Sequence/genetics , Cullin Proteins/metabolism , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Microscopy, Fluorescence , Nuclear Localization Signals/genetics , Plasmids/genetics , SAM Domain and HD Domain-Containing Protein 1 , Ubiquitination
11.
Nat Immunol ; 13(3): 223-228, 2012 Feb 12.
Article in English | MEDLINE | ID: mdl-22327569

ABSTRACT

SAMHD1 restricts the infection of dendritic and other myeloid cells by human immunodeficiency virus type 1 (HIV-1), but in lentiviruses of the simian immunodeficiency virus of sooty mangabey (SIVsm)-HIV-2 lineage, SAMHD1 is counteracted by the virion-packaged accessory protein Vpx. Here we found that SAMHD1 restricted infection by hydrolyzing intracellular deoxynucleoside triphosphates (dNTPs), lowering their concentrations to below those required for the synthesis of the viral DNA by reverse transcriptase (RT). SAMHD1-mediated restriction was alleviated by the addition of exogenous deoxynucleosides. An HIV-1 with a mutant RT with low affinity for dNTPs was particularly sensitive to SAMHD1-mediated restriction. Vpx prevented the SAMHD1-mediated decrease in dNTP concentration and induced the degradation of human and rhesus macaque SAMHD1 but had no effect on mouse SAMHD1. Nucleotide-pool depletion could be a general mechanism for protecting cells from infectious agents that replicate through a DNA intermediate.


Subject(s)
HIV-1/physiology , Monomeric GTP-Binding Proteins/metabolism , Nucleotides/metabolism , Virus Replication , Animals , Cell Line , Humans , Intracellular Space/metabolism , Macaca mulatta , Macrophages/immunology , Mice , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/immunology , SAM Domain and HD Domain-Containing Protein 1
12.
J Virol ; 84(14): 7312-24, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20444897

ABSTRACT

To get more insight into the role of APOBEC3 (A3) cytidine deaminases in the species-specific restriction of feline immunodeficiency virus (FIV) of the domestic cat, we tested the A3 proteins present in big cats (puma, lion, tiger, and lynx). These A3 proteins were analyzed for expression and sensitivity to the Vif protein of FIV. While A3Z3s and A3Z2-Z3s inhibited Deltavif FIV, felid A3Z2s did not show any antiviral activity against Deltavif FIV or wild-type (wt) FIV. All felid A3Z3s and A3Z2-Z3s were sensitive to Vif of the domestic cat FIV. Vif also induced depletion of felid A3Z2s. Tiger A3s showed a moderate degree of resistance against the Vif-mediated counter defense. These findings may imply that the A3 restriction system does not play a major role to prevent domestic cat FIV transmission to other Felidae. In contrast to the sensitive felid A3s, many nonfelid A3s actively restricted wt FIV replication. To test whether Vif(FIV) can protect also the distantly related human immunodeficiency virus type 1 (HIV-1), a chimeric HIV-1.Vif(FIV) was constructed. This HIV-1.Vif(FIV) was replication competent in nonpermissive feline cells expressing human CD4/CCR5 that did not support the replication of wt HIV-1. We conclude that the replication of HIV-1 in some feline cells is inhibited only by feline A3 restriction factors and the absence of the appropriate receptor or coreceptor.


Subject(s)
Cytosine Deaminase/metabolism , Felidae/metabolism , Felidae/virology , Feline Acquired Immunodeficiency Syndrome/virology , Gene Products, vif/metabolism , Immunodeficiency Virus, Feline/metabolism , Isoenzymes/metabolism , Animals , Cats , Cell Line , Cytosine Deaminase/genetics , Felidae/genetics , Gene Products, vif/genetics , Humans , Immunodeficiency Virus, Feline/genetics , Isoenzymes/genetics , RNA Splicing , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 106(29): 12079-84, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19581596

ABSTRACT

Human APOBEC3 (A3) proteins form part of the intrinsic immunity to retroviruses. Carrying 1 or 2 copies of a cytidine deaminase motif, A3s act by deamination of retroviral genomes during reverse transcription. HIV-1 overcomes this inhibition by the Vif protein, which prevents incorporation of A3 into virions. In this study we modeled and probed the structure of APOBEC3C (A3C), a single-domain A3 with strong antilentiviral activity. The 3-dimensional protein model was used to predict the effect of mutations on antiviral activity, which was tested in a Deltavif simian immunodeficiency virus (SIV) reporter virus assay. We found that A3C activity requires protein dimerization for antiviral activity against SIV. Furthermore, by using a structure-based algorithm for automated pocket extraction, we detected a putative substrate binding pocket of A3C distal from the zinc-coordinating deaminase motif. Mutations in this region diminished antiviral activity by excluding A3C from virions. We found evidence that the small 5.8S RNA specifically binds to this locus and mediates incorporation of A3C into virus particles.


Subject(s)
Capsid/metabolism , Cytosine Deaminase/chemistry , Cytosine Deaminase/metabolism , Models, Molecular , RNA/metabolism , APOBEC Deaminases , Binding Sites , Cell Line , Cytidine Deaminase , Humans , Immunoblotting , Mutant Proteins/metabolism , Protein Multimerization , Protein Processing, Post-Translational , Protein Structure, Secondary , vif Gene Products, Human Immunodeficiency Virus/metabolism
14.
Retrovirology ; 6: 38, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19371434

ABSTRACT

Human APOBEC3G is an antiretroviral protein that was described to act via deamination of retroviral cDNA. However, it was suggested that APOBEC proteins might act with antiviral activity by yet other mechanisms and may also possess RNA deamination activity. As a consequence there is an ongoing debate whether APOBEC proteins might also act with antiviral activity on other RNA viruses. Influenza A viruses are single-stranded RNA viruses, capable of inducing a variety of antiviral gene products. In searching for novel antiviral genes against these pathogens, we detected a strong induction of APOBEC3G but not APOBEC3F gene transcription in infected cells. This upregulation appeared to be induced by the accumulation of viral RNA species within the infected cell and occurred in an NF-kappaB dependent, but MAP kinase independent manner. It further turned out that APOBEC expression is part of a general IFNbeta response to infection. However, although strongly induced, APOBEC3G does not negatively affect influenza A virus propagation.


Subject(s)
Antiviral Agents , Cytidine Deaminase/metabolism , Influenza A Virus, H1N1 Subtype , Influenza A virus/pathogenicity , Up-Regulation , Virus Replication/drug effects , APOBEC-3G Deaminase , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Cell Line , Cytidine Deaminase/genetics , Cytidine Deaminase/pharmacology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Influenza A virus/drug effects , Influenza A virus/physiology , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Viral/metabolism , RNA, Viral/pharmacology
15.
J Biol Chem ; 284(9): 5819-26, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19074429

ABSTRACT

The APOBEC3 cytidine deaminases are part of the intrinsic defense of cells against retroviruses. Lentiviruses and spumaviruses have evolved essential accessory proteins, Vif and Bet, respectively, which counteract the APOBEC3 proteins. We show here that Bet of the Prototype foamy virus inhibits the antiviral APOBEC3C activity by a mechanism distinct to Vif: Bet forms a complex with APOBEC3C without inducing its degradation. Bet abolished APOBEC3C dimerization as shown by coimmunoprecipitation and cross-linking experiments. These findings implicate a physical interaction between Bet and the APOBEC3C. Subsequently, we identified the Bet interaction domain in human APOBEC3C in the predicted APOBEC3C dimerization site. Taken together, these data support the hypothesis that Bet inhibits incorporation of APOBEC3Cs into retroviral particles. Bet likely achieves this by trapping APOBEC3C protein in complexes rendering them unavailable for newly generated viruses due to direct immobilization.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Cytidine Deaminase/metabolism , Proviruses/genetics , Retroviridae Proteins/physiology , APOBEC-3G Deaminase , Animals , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Cross-Linking Reagents , Dimerization , Gene Products, vif/physiology , Humans , Immunoblotting , Immunoprecipitation , Kidney/cytology , Kidney/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Macaca mulatta , Macaca nemestrina , Pan troglodytes , Simian foamy virus , Transfection , Virus Assembly , Virus Replication/drug effects , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...