Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Nat Immunol ; 16(10): 1025-33, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26343537

ABSTRACT

Cytosolic DNA that emerges during infection with a retrovirus or DNA virus triggers antiviral type I interferon responses. So far, only double-stranded DNA (dsDNA) over 40 base pairs (bp) in length has been considered immunostimulatory. Here we found that unpaired DNA nucleotides flanking short base-paired DNA stretches, as in stem-loop structures of single-stranded DNA (ssDNA) derived from human immunodeficiency virus type 1 (HIV-1), activated the type I interferon-inducing DNA sensor cGAS in a sequence-dependent manner. DNA structures containing unpaired guanosines flanking short (12- to 20-bp) dsDNA (Y-form DNA) were highly stimulatory and specifically enhanced the enzymatic activity of cGAS. Furthermore, we found that primary HIV-1 reverse transcripts represented the predominant viral cytosolic DNA species during early infection of macrophages and that these ssDNAs were highly immunostimulatory. Collectively, our study identifies unpaired guanosines in Y-form DNA as a highly active, minimal cGAS recognition motif that enables detection of HIV-1 ssDNA.


Subject(s)
DNA, Complementary/chemistry , DNA, Viral/chemistry , DNA, Viral/immunology , HIV-1/genetics , HIV-1/immunology , Interferon-alpha/immunology , Nucleotidyltransferases/genetics , Animals , Cell Line , Cells, Cultured , DNA, Complementary/genetics , DNA, Complementary/immunology , DNA, Viral/genetics , HEK293 Cells , Humans , Immunization , Mice
2.
EMBO J ; 41(17): e111608, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35833542

ABSTRACT

The SARS-CoV-2 infection cycle is a multistage process that relies on functional interactions between the host and the pathogen. Here, we repurposed antiviral drugs against both viral and host enzymes to pharmaceutically block methylation of the viral RNA 2'-O-ribose cap needed for viral immune escape. We find that the host cap 2'-O-ribose methyltransferase MTr1 can compensate for loss of viral NSP16 methyltransferase in facilitating virus replication. Concomitant inhibition of MTr1 and NSP16 efficiently suppresses SARS-CoV-2 replication. Using in silico target-based drug screening, we identify a bispecific MTr1/NSP16 inhibitor with anti-SARS-CoV-2 activity in vitro and in vivo but with unfavorable side effects. We further show antiviral activity of inhibitors that target independent stages of the host SAM cycle providing the methyltransferase co-substrate. In particular, the adenosylhomocysteinase (AHCY) inhibitor DZNep is antiviral in in vitro, in ex vivo, and in a mouse infection model and synergizes with existing COVID-19 treatments. Moreover, DZNep exhibits a strong immunomodulatory effect curbing infection-induced hyperinflammation and reduces lung fibrosis markers ex vivo. Thus, multispecific and metabolic MTase inhibitors constitute yet unexplored treatment options against COVID-19.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Animals , Antiviral Agents/pharmacology , Inflammation/drug therapy , Methyltransferases/metabolism , Mice , RNA Caps/metabolism , RNA, Viral/genetics , Ribose , Viral Nonstructural Proteins/genetics
3.
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
4.
EMBO Rep ; 23(12): e55648, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36285486

ABSTRACT

Methylation of the mRNA 5' cap by cellular methyltransferases enables efficient translation and avoids recognition by innate immune factors. Coronaviruses encode viral 2'-O-methyltransferases to shield their RNA from host factors. Here, we generate recombinant SARS-CoV-2 harboring a catalytically inactive 2'-O-methyltransferase Nsp16, Nsp16mut, and analyze viral replication in human lung epithelial cells. Although replication is only slightly attenuated, we find SARS-CoV-2 Nsp16mut to be highly immunogenic, resulting in a strongly enhanced release of type I interferon upon infection. The elevated immunogenicity of Nsp16mut is absent in cells lacking the RNA sensor MDA5. In addition, we report that Nsp16mut is highly sensitive to type I IFN treatment and demonstrate that this strong antiviral effect of type I IFN is mediated by the restriction factor IFIT1. Together, we describe a dual role for the 2'-O-methyltransferase Nsp16 during SARS-CoV-2 replication in avoiding efficient recognition by MDA5 and in shielding its RNA from interferon-induced antiviral responses, thereby identifying Nsp16 as a promising target for generating attenuated and highly immunogenic SARS-CoV-2 strains and as a potential candidate for therapeutic intervention.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , RNA , Methyltransferases/genetics , RNA-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics
5.
J Biol Chem ; 298(6): 102004, 2022 06.
Article in English | MEDLINE | ID: mdl-35504352

ABSTRACT

The epithelial sodium channel (ENaC) is a heterotrimer consisting of α-, ß-, and γ-subunits. Channel activation requires proteolytic release of inhibitory tracts from the extracellular domains of α-ENaC and γ-ENaC; however, the proteases involved in the removal of the γ-inhibitory tract remain unclear. In several epithelial tissues, ENaC is coexpressed with the transmembrane serine protease 2 (TMPRSS2). Here, we explored the effect of human TMPRSS2 on human αßγ-ENaC heterologously expressed in Xenopus laevis oocytes. We found that coexpression of TMPRSS2 stimulated ENaC-mediated whole-cell currents by approximately threefold, likely because of an increase in average channel open probability. Furthermore, TMPRSS2-dependent ENaC stimulation was not observed using a catalytically inactive TMPRSS2 mutant and was associated with fully cleaved γ-ENaC in the intracellular and cell surface protein fractions. This stimulatory effect of TMPRSS2 on ENaC was partially preserved when inhibiting its proteolytic activity at the cell surface using aprotinin but was abolished when the γ-inhibitory tract remained attached to its binding site following introduction of two cysteine residues (S155C-Q426C) to form a disulfide bridge. In addition, computer simulations and site-directed mutagenesis experiments indicated that TMPRSS2 can cleave γ-ENaC at sites both proximal and distal to the γ-inhibitory tract. This suggests a dual role of TMPRSS2 in the proteolytic release of the γ-inhibitory tract. Finally, we demonstrated that TMPRSS2 knockdown in cultured human airway epithelial cells (H441) reduced baseline proteolytic activation of endogenously expressed ENaC. Thus, we conclude that TMPRSS2 is likely to contribute to proteolytic ENaC activation in epithelial tissues in vivo.


Subject(s)
Epithelial Sodium Channels , Oocytes , Serine Endopeptidases , Animals , Epithelial Sodium Channels/metabolism , Humans , Ion Transport/physiology , Oocytes/metabolism , Peptide Hydrolases/metabolism , Proteolysis , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Xenopus laevis/genetics
6.
Proc Natl Acad Sci U S A ; 117(30): 17965-17976, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32651277

ABSTRACT

Mobile genetic elements have significantly shaped our genomic landscape. LINE-1 retroelements are the only autonomously active elements left in the human genome. Since new insertions can have detrimental consequences, cells need to efficiently control LINE-1 retrotransposition. Here, we demonstrate that the intrinsic immune factor TRIM5α senses and restricts LINE-1 retroelements. Previously, rhesus TRIM5α has been shown to efficiently block HIV-1 replication, while human TRIM5α was found to be less active. Surprisingly, we found that both human and rhesus TRIM5α efficiently repress human LINE-1 retrotransposition. TRIM5α interacts with LINE-1 ribonucleoprotein complexes in the cytoplasm, which is essential for restriction. In line with its postulated role as pattern recognition receptor, we show that TRIM5α also induces innate immune signaling upon interaction with LINE-1 ribonucleoprotein complexes. The signaling events activate the transcription factors AP-1 and NF-κB, leading to the down-regulation of LINE-1 promoter activity. Together, our findings identify LINE-1 as important target of human TRIM5α, which restricts and senses LINE-1 via two distinct mechanisms. Our results corroborate TRIM5α as pattern recognition receptor and shed light on its previously undescribed activity against mobile genetic elements, such as LINE-1, to protect the integrity of our genome.


Subject(s)
Long Interspersed Nucleotide Elements , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Antiviral Restriction Factors , Gene Expression , Genes, Reporter , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Macaca mulatta , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Signal Transduction , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics
7.
PLoS Pathog ; 16(8): e1008752, 2020 08.
Article in English | MEDLINE | ID: mdl-32760121

ABSTRACT

Members of the family of pyrin and HIN domain containing (PYHIN) proteins play an emerging role in innate immunity. While absent in melanoma 2 (AIM2) acts a cytosolic sensor of non-self DNA and plays a key role in inflammasome assembly, the γ-interferon-inducible protein 16 (IFI16) restricts retroviral gene expression by sequestering the transcription factor Sp1. Here, we show that the remaining two human PYHIN proteins, i.e. myeloid cell nuclear differentiation antigen (MNDA) and pyrin and HIN domain family member 1 (PYHIN1 or IFIX) share this antiretroviral function of IFI16. On average, knock-down of each of these three nuclear PYHIN proteins increased infectious HIV-1 yield from human macrophages by more than an order of magnitude. Similarly, knock-down of IFI16 strongly increased virus transcription and production in primary CD4+ T cells. The N-terminal pyrin domain (PYD) plus linker region containing a nuclear localization signal (NLS) were generally required and sufficient for Sp1 sequestration and anti-HIV-1 activity of IFI16, MNDA and PYHIN1. Replacement of the linker region of AIM2 by the NLS-containing linker of IFI16 resulted in a predominantly nuclear localization and conferred direct antiviral activity to AIM2 while attenuating its ability to form inflammasomes. The reverse change caused nuclear-to-cytoplasmic relocalization of IFI16 and impaired its antiretroviral activity but did not result in inflammasome assembly. We further show that the Zn-finger domain of Sp1 is critical for the interaction with IFI16 supporting that pyrin domains compete with DNA for Sp1 binding. Finally, we found that human PYHIN proteins also inhibit Hepatitis B virus and simian vacuolating virus 40 as well as the LINE-1 retrotransposon. Altogether, our data show that IFI16, PYHIN1 and MNDA restrict HIV-1 and other viral pathogens by interfering with Sp1-dependent gene expression and support an important role of nuclear PYHIN proteins in innate antiviral immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Nucleus/metabolism , HIV Infections/prevention & control , HIV-1/immunology , Macrophages/immunology , Nuclear Proteins/metabolism , Sp1 Transcription Factor/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Nucleus/genetics , DNA, Viral/genetics , HEK293 Cells , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , HIV-1/genetics , HIV-1/isolation & purification , Hep G2 Cells , Humans , Immunity, Innate/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Macrophages/metabolism , Macrophages/virology , Nuclear Proteins/genetics , Sp1 Transcription Factor/genetics , Virus Replication
8.
Retrovirology ; 12: 103, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26667483

ABSTRACT

BACKGROUND: Human SAMHD1 is a triphosphohydrolase that restricts the replication of retroviruses, retroelements and DNA viruses in noncycling cells. While modes of action have been extensively described for human SAMHD1, only little is known about the regulation of SAMHD1 in the mouse. Here, we characterize the antiviral activity of murine SAMHD1 with the help of knockout mice to shed light on the regulation and the mechanism of the SAMHD1 restriction and to validate the SAMHD1 knockout mouse model for the use in future infectivity studies. RESULTS: We found that endogenous mouse SAMHD1 restricts not only HIV-1 but also MLV reporter virus infection at the level of reverse transcription in primary myeloid cells. Similar to the human protein, the antiviral activity of murine SAMHD1 is regulated through phosphorylation at threonine 603 and is limited to nondividing cells. Comparing the susceptibility to infection with intracellular dNTP levels and SAMHD1 phosphorylation in different cell types shows that both functions are important determinants of the antiviral activity of murine SAMHD1. In contrast, we found the proposed RNase activity of SAMHD1 to be less important and could not detect any effect of mouse or human SAMHD1 on the level of incoming viral RNA. CONCLUSION: Our findings show that SAMHD1 in the mouse blocks retroviral infection at the level of reverse transcription and is regulated through cell cycle-dependent phosphorylation. We show that the antiviral restriction mediated by murine SAMHD1 is mechanistically similar to what is known for the human protein, making the SAMHD1 knockout mouse model a valuable tool to characterize the influence of SAMHD1 on the replication of different viruses in vivo.


Subject(s)
HIV-1/physiology , Leukemia Virus, Murine/physiology , Monomeric GTP-Binding Proteins/metabolism , Retroviridae Infections/virology , Reverse Transcription , Animals , Cell Line , Cells, Cultured , Humans , Macrophages/virology , Mice , Mice, Knockout , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/deficiency , Monomeric GTP-Binding Proteins/genetics , Myeloid Cells/virology , Phosphorylation , RNA, Viral/genetics , RNA, Viral/metabolism , SAM Domain and HD Domain-Containing Protein 1 , Threonine/physiology , Virus Replication
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.
Front Immunol ; 14: 1168589, 2023.
Article in English | MEDLINE | ID: mdl-37180175

ABSTRACT

The intracellular restriction factor TRIM5α inhibits endogenous LINE-1 retroelements. It induces innate immune signaling cascades upon sensing of cytoplasmic LINE-1 complexes, thereby underlining its importance for protecting the human genome from harmful retrotransposition events. Here, we show that a frequent SNP within the RING domain of TRIM5α, resulting in the variant H43Y, blocks LINE-1 retrotransposition with higher efficiency compared to TRIM5α WT. Upon sensing of LINE-1 complexes in the cytoplasm, TRIM5α H43Y activates both NF-κB and AP-1 signaling pathways more potently than TRIM5α WT, triggering a strong block of the LINE-1 promoter. Interestingly, the H43Y allele lost its antiviral function suggesting that its enhanced activity against endogenous LINE-1 elements is the driving force behind its maintenance within the population. Thus, our study suggests that the H43Y variant of the restriction factor and sensor TRIM5α persists within the human population since it preserves our genome from uncontrolled LINE-1 retrotransposition with higher efficiency.


Subject(s)
Long Interspersed Nucleotide Elements , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Antiviral Restriction Factors , Immunity, Innate/genetics
11.
J Exp Med ; 220(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36346347

ABSTRACT

Defects in nucleic acid metabolizing enzymes can lead to spontaneous but selective activation of either cGAS/STING or RIG-like receptor (RLR) signaling, causing type I interferon-driven inflammatory diseases. In these pathophysiological conditions, activation of the DNA sensor cGAS and IFN production are linked to spontaneous DNA damage. Physiological, or tonic, IFN signaling on the other hand is essential to functionally prime nucleic acid sensing pathways. Here, we show that low-level chronic DNA damage in mice lacking the Aicardi-Goutières syndrome gene SAMHD1 reduced tumor-free survival when crossed to a p53-deficient, but not to a DNA mismatch repair-deficient background. Increased DNA damage did not result in higher levels of type I interferon. Instead, we found that the chronic interferon response in SAMHD1-deficient mice was driven by the MDA5/MAVS pathway but required functional priming through the cGAS/STING pathway. Our work positions cGAS/STING upstream of tonic IFN signaling in Samhd1-deficient mice and highlights an important role of the pathway in physiological and pathophysiological innate immune priming.


Subject(s)
Interferon Type I , Nucleic Acids , Mice , Animals , SAM Domain and HD Domain-Containing Protein 1/genetics , Immunity, Innate/genetics , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Interferon Type I/metabolism
12.
J Virol ; 84(3): 1387-96, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923175

ABSTRACT

Vpx and Vpr are related lentiviral accessory proteins that enhance virus replication in macrophages and dendritic cells. Both proteins are packaged into virions and mediate their effects in the target cell through an interaction with an E3 ubiquitin ligase that contains DCAF1 and DDB1. When introduced into primary macrophages and dendritic cells in viruslike particles, Vpx can enhance the efficiency of a subsequent infection. Here, we confirm the ability of Vpx to enhance simian immunodeficiency virus (SIV) and human immunodeficiency virus type 1 (HIV-1) infection of macrophages up to 100-fold by using single-cycle reporter viruses and by pretreatment of the cells with Vpx-containing viruslike particles. Vpx was also active in differentiated THP-1 cells but not in other cell lines. Induction of an antiviral state in macrophages with type I interferon significantly magnified the effect of Vpx on HIV-1 infection, suggesting that Vpx helps the virus to overcome an inducible intracellular restriction. Quantitative PCR quantitation of SIV and HIV-1 reverse transcripts in newly infected macrophages showed that the block was at an early step in reverse transcription. In spite of its structural similarity, Vpr was inactive. This difference allowed us to map the functional domains of Vpx with a panel of Vpr/Vpx chimeras. Analysis of the chimeras demonstrated that the amino-terminal domain of Vpx is important for the enhancement of infection. Fine mapping of the region indicated that amino acids at positions 9, 12, and 15 to 17 were required. Although the mutants failed to enhance infection, they retained their ability to interact with DCAF1. These findings suggest that the Vpx amino terminus contains an activation domain that serves as the binding site for a cellular restriction factor.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Virus Activation , Animals , Base Sequence , Cell Line , DNA Primers , HIV-1/metabolism , HIV-1/physiology , Humans , Polymerase Chain Reaction , Simian Immunodeficiency Virus/physiology , Viral Regulatory and Accessory Proteins/chemistry
13.
Viruses ; 13(3)2021 03 02.
Article in English | MEDLINE | ID: mdl-33801276

ABSTRACT

The SAM and HD domain-containing protein 1 (SAMHD1) is a dNTP triphosphohydrolase that plays a crucial role for a variety of different cellular functions. Besides balancing intracellular dNTP concentrations, facilitating DNA damage repair, and dampening excessive immune responses, SAMHD1 has been shown to act as a major restriction factor against various virus species. In addition to its well-described activity against retroviruses such as HIV-1, SAMHD1 has been identified to reduce the infectivity of different DNA viruses such as the herpesviruses CMV and EBV, the poxvirus VACV, or the hepadnavirus HBV. While some viruses are efficiently restricted by SAMHD1, others have developed evasion mechanisms that antagonize the antiviral activity of SAMHD1. Within this review, we summarize the different cellular functions of SAMHD1 and highlight the countermeasures viruses have evolved to neutralize the restriction factor SAMHD1.


Subject(s)
DNA Virus Infections/immunology , Host-Pathogen Interactions/immunology , Retroviridae Infections/immunology , SAM Domain and HD Domain-Containing Protein 1/immunology , DNA Viruses/immunology , Humans , Retroviridae/immunology , Viral Interference
14.
Viruses ; 13(1)2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33445593

ABSTRACT

Although mobile genetic elements, or transposons, have played an important role in genome evolution, excess activity of mobile elements can have detrimental consequences. Already, the enhanced expression of transposons-derived nucleic acids can trigger autoimmune reactions that may result in severe autoinflammatory disorders. Thus, cells contain several layers of protective measures to restrict transposons and to sense the enhanced activity of these "intragenomic pathogens". This review focuses on our current understanding of immunogenic patterns derived from the most active elements in humans, the retrotransposons long interspersed element (LINE)-1 and Alu. We describe the role of known pattern recognition receptors in nucleic acid sensing of LINE-1 and Alu and the possible consequences for autoimmune diseases.


Subject(s)
Retroelements , Alu Elements , Animals , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Host-Pathogen Interactions , Humans , Long Interspersed Nucleotide Elements
15.
Curr HIV/AIDS Rep ; 6(1): 36-42, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19149995

ABSTRACT

Recent advances in understanding the roles of the lentiviral accessory proteins have provided fascinating insight into the molecular biology of the virus and uncovered previously unappreciated innate immune mechanisms by which the host defends itself. HIV-1 and other lentiviruses have developed accessory proteins that counterattack the antiviral defenses in a sort of evolutionary battle. The virus is remarkably adept at co-opting cellular degradative pathways to destroy the protective proteins. This review focuses on recent advances in understanding three of the accessory proteins-virion infectivity factor (Vif), viral protein R (Vpr), and viral protein U (Vpu)-that target different restriction factors to ensure virus replication. These proteins may provide promising targets for the development of novel classes of antiretroviral drugs.


Subject(s)
Gene Products, vpr , HIV-1/physiology , HIV-1/pathogenicity , Virus Replication , vif Gene Products, Human Immunodeficiency Virus , vpr Gene Products, Human Immunodeficiency Virus , Animals , Gene Expression Regulation , Gene Expression Regulation, Viral , Gene Products, vpr/genetics , Gene Products, vpr/metabolism , HIV-1/genetics , HIV-1/metabolism , Humans , Proteins/genetics , Proteins/metabolism , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism , vpr Gene Products, Human Immunodeficiency Virus/genetics , vpr Gene Products, Human Immunodeficiency Virus/metabolism
16.
Nat Microbiol ; 4(12): 2260-2272, 2019 12.
Article in English | MEDLINE | ID: mdl-31548682

ABSTRACT

The host restriction factor sterile alpha motif and histidine-aspartate domain-containing protein 1 (SAMHD1) is an important component of the innate immune system. By regulating the intracellular nucleotide pool, SAMHD1 influences cell division and restricts the replication of viruses that depend on high nucleotide concentrations. Human cytomegalovirus (HCMV) is a pathogenic virus with a tropism for non-dividing myeloid cells, in which SAMHD1 is catalytically active. Here we investigate how HCMV achieves efficient propagation in these cells despite the SAMHD1-mediated dNTP depletion. Our analysis reveals that SAMHD1 has the capability to suppress HCMV replication. However, HCMV has evolved potent countermeasures to circumvent this block. HCMV interferes with SAMHD1 steady-state expression and actively induces SAMHD1 phosphorylation using the viral kinase pUL97 and by hijacking cellular kinases. These actions convert SAMHD1 to its inactive phosphorylated form. This mechanism of SAMHD1 inactivation by phosphorylation might also be used by other viruses to overcome intrinsic immunity.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/metabolism , Macrophages/immunology , SAM Domain and HD Domain-Containing Protein 1/metabolism , Viral Proteins/metabolism , Cytomegalovirus/pathogenicity , HEK293 Cells , Humans , Immunity, Innate , Macrophages/virology , Phosphorylation , SAM Domain and HD Domain-Containing Protein 1/genetics , SAM Domain and HD Domain-Containing Protein 1/pharmacology , THP-1 Cells , Virus Replication/drug effects
17.
Nat Microbiol ; 4(12): 2273-2284, 2019 12.
Article in English | MEDLINE | ID: mdl-31548683

ABSTRACT

The deoxynucleotide triphosphate (dNTP) hydrolase SAMHD1 inhibits retroviruses in non-dividing myeloid cells. Although antiviral activity towards DNA viruses has also been demonstrated, the role of SAMHD1 during cytomegalovirus (CMV) infection remains unclear. To determine the impact of SAMHD1 on the replication of CMV, we used murine CMV (MCMV) to infect a previously established SAMHD1 knockout mouse model and found that SAMHD1 inhibits the replication of MCMV in vivo. By comparing the replication of MCMV in vitro in myeloid cells and fibroblasts from SAMHD1-knockout and control mice, we found that the viral kinase M97 counteracts SAMHD1 after infection by phosphorylating the regulatory residue threonine 603. The phosphorylation of SAMHD1 in infected cells correlated with a reduced level of dNTP hydrolase activity and the loss of viral restriction. Together, we demonstrate that SAMHD1 acts as a restriction factor in vivo and we identify the M97-mediated phosphorylation of SAMHD1 as a previously undescribed viral countermeasure.


Subject(s)
Muromegalovirus/drug effects , Phosphotransferases/metabolism , SAM Domain and HD Domain-Containing Protein 1/antagonists & inhibitors , SAM Domain and HD Domain-Containing Protein 1/metabolism , Animals , Antiviral Agents/pharmacology , Colony-Stimulating Factors/metabolism , Disease Models, Animal , HEK293 Cells , Herpesviridae Infections/drug therapy , Herpesviridae Infections/virology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/enzymology , Muromegalovirus/growth & development , NIH 3T3 Cells , Phosphorylation , Recombinant Proteins , SAM Domain and HD Domain-Containing Protein 1/genetics , Transcriptome , Viral Proteins/metabolism , Virus Replication/drug effects
18.
Cell Host Microbe ; 25(6): 858-872.e13, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31175045

ABSTRACT

The interferon γ-inducible protein 16 (IFI16) is known as immune sensor of retroviral DNA intermediates. We show that IFI16 restricts HIV-1 independently of immune sensing by binding and inhibiting the host transcription factor Sp1 that drives viral gene expression. This antiretroviral activity and ability to bind Sp1 require the N-terminal pyrin domain and nuclear localization of IFI16, but not the HIN domains involved in DNA binding. Highly prevalent clade C HIV-1 strains are more resistant to IFI16 and less dependent on Sp1 than other HIV-1 subtypes. Furthermore, inhibition of Sp1 by IFI16 or pharmacologically by Mithramycin A suppresses reactivation of latent HIV-1 in CD4+ T cells. Finally, IFI16 also inhibits retrotransposition of LINE-1, known to engage Sp1, and murine IFI16 homologs restrict Friend retrovirus replication in mice. Thus, IFI16 restricts retroviruses and retrotransposons by interfering with Sp1-dependent gene expression, and evasion from this restriction may facilitate spread of HIV-1 subtype C.


Subject(s)
HIV-1/immunology , Immunologic Factors/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Transcription, Genetic , Virus Activation , Virus Latency , Animals , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Genotype , HIV-1/classification , HIV-1/genetics , HIV-1/growth & development , Mice
19.
Mob DNA ; 9: 11, 2018.
Article in English | MEDLINE | ID: mdl-29610582

ABSTRACT

BACKGROUND: The restriction factor SAMHD1 regulates intracellular nucleotide level by degrading dNTPs and blocks the replication of retroviruses and DNA viruses in non-cycling cells, like macrophages or dendritic cells. In patients, inactivating mutations in samhd1 are associated with the autoimmune disease Aicardi-Goutières Syndrome (AGS). The accumulation of intracellular nucleic acids derived from endogenous retroelements thriving in the absence of SAMHD1 has been discussed as potential trigger of the autoimmune reaction. In vitro, SAMHD1 has been found to restrict endogenous retroelements, like LINE-1 elements (L1). The mechanism, however, by which SAMHD1 blocks endogenous retroelements, is still unclear. RESULTS: Here, we show that SAMHD1 inhibits the replication of L1 and other endogenous retroelements in cycling cells. By applying GFP- and neomycin-based reporter assays we found that the anti-L1 activity of SAMHD1 is regulated by phosphorylation at threonine 592 (T592). Similar to the block of HIV, the cofactor binding site and the enzymatic active HD domain of SAMHD1 proofed to be essential for restriction of L1 elements. However, phosphorylation at T592 did not correlate with the dNTP hydrolase activity of SAMHD1 in cycling 293T cells suggesting an alternative mechanism of regulation. Interestingly, we found that SAMHD1 binds to ORF2 protein of L1 and that this interaction is regulated by T592 phosphorylation. Together with the finding that the block is also active in cycling cells, our results suggest that the SAMHD1-mediated inhibition of L1 is similar but not identical to HIV restriction. CONCLUSION: Our findings show conclusively that SAMHD1 restricts the replication of endogenous retroelements in vitro. The results suggest that SAMHD1 is important for maintaining genome integrity and support the idea of an enhanced replication of endogenous retroelements in the absence of SAMHD1 in vivo, potentially triggering autoimmune diseases like AGS. Our analysis also contributes to the better understanding of the activities of SAMHD1 in antiviral defense and nucleotide metabolism. The finding that the phosphorylation of SAMHD1 at T592 regulates its activity against retroelements but not necessarily intracellular dNTP level suggests that the dNTP hydrolase activity might not be the only function of SAMHD1 important for its antiviral activity and for controlling autoimmunity.

20.
Curr HIV Res ; 14(3): 225-34, 2016.
Article in English | MEDLINE | ID: mdl-26957197

ABSTRACT

BACKGROUND: The antiviral restriction factor SAM domain and HD domain-containing protein 1 (SAMHD1) is a dNTP triphosphohydrolase and thereby contributes to the regulation of intracellular dNTP levels. SAMHD1 blocks retroviral infection at the level of reverse transcription in myeloid cells and resting CD4+ T cells and is counteracted by the accessory protein Vpx, which is encoded by human immunodeficiency virus 2 (HIV-2) and several simian immunodeficiency virus (SIV) strains. Recently, it has been shown that the antiviral activity of SAMHD1 in myeloid dendritic cells (DC) hampers the induction of an efficient immune response directed against HIV-1. CONCLUSION: Within this review, we will summarize recent advances on the biology of SAMHD1 and its function as an antiviral restriction factor. In addition, we will discuss its role in autoimmunity and the antiviral immune response directed against HIV-1 and will evaluate the possibility of modulating SAMHD1 activity to generate an enhanced antiretroviral immune response.


Subject(s)
HIV Infections/etiology , HIV Infections/metabolism , HIV/physiology , Host-Pathogen Interactions , Immunity, Innate , Monomeric GTP-Binding Proteins/metabolism , Animals , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Autoimmunity , Disease Resistance , Drug Discovery , HIV Infections/drug therapy , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunomodulation , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/genetics , Protein Binding , SAM Domain and HD Domain-Containing Protein 1 , Simian Immunodeficiency Virus/physiology , Viral Regulatory and Accessory Proteins , Virus Replication , vpr Gene Products, Human Immunodeficiency Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL