Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Nat Microbiol ; 7(12): 2101-2113, 2022 12.
Article in English | MEDLINE | ID: mdl-36376394

ABSTRACT

After viral entry and reverse transcription, HIV-1 proviruses that fail to integrate are epigenetically silenced, but the underlying mechanism has remained unclear. Using a genome-wide CRISPR/Cas9 knockout screen, we identified the host SMC5/6 complex as essential for this epigenetic silencing. We show that SMC5/6 binds to and then SUMOylates unintegrated chromatinized HIV-1 DNA. Inhibition of SUMOylation, either by point mutagenesis of the SMC5/6 component NSMCE2-a SUMO E3 ligase-or using the SUMOylation inhibitor TAK-981, prevents epigenetic silencing, enables transcription from unintegrated HIV-1 DNA and rescues the replication of integrase-deficient HIV-1. Finally, we show that blocking SMC5/6 complex expression, or inhibiting its SUMOylation activity, suppresses the establishment of latent HIV-1 infections in both CD4+ T cell lines and primary human T cells. Collectively, our data show that the SMC5/6 complex plays a direct role in mediating the establishment of HIV-1 latency by epigenetically silencing integration-competent HIV-1 proviruses before integration.


Subject(s)
HIV Infections , HIV-1 , Humans , HIV-1/physiology , HIV Infections/genetics , Virus Latency/genetics , Proviruses/genetics , Proviruses/metabolism , DNA , Epigenesis, Genetic , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Ligases/genetics
2.
RNA ; 27(11): 1400-1411, 2021 11.
Article in English | MEDLINE | ID: mdl-34376564

ABSTRACT

Pseudouridine (Ψ) is the most common noncanonical ribonucleoside present on mammalian noncoding RNAs (ncRNAs), including rRNAs, tRNAs, and snRNAs, where it contributes ∼7% of the total uridine level. However, Ψ constitutes only ∼0.1% of the uridines present on mRNAs and its effect on mRNA function remains unclear. Ψ residues have been shown to inhibit the detection of exogenous RNA transcripts by host innate immune factors, thus raising the possibility that viruses might have subverted the addition of Ψ residues to mRNAs by host pseudouridine synthase (PUS) enzymes as a way to inhibit antiviral responses in infected cells. Here, we describe and validate a novel antibody-based Ψ mapping technique called photo-crosslinking-assisted Ψ sequencing (PA-Ψ-seq) and use it to map Ψ residues on not only multiple cellular RNAs but also on the mRNAs and genomic RNA encoded by HIV-1. We describe 293T-derived cell lines in which human PUS enzymes previously reported to add Ψ residues to human mRNAs, specifically PUS1, PUS7, and TRUB1/PUS4, were inactivated by gene editing. Surprisingly, while this allowed us to assign several sites of Ψ addition on cellular mRNAs to each of these three PUS enzymes, Ψ sites present on HIV-1 transcripts remained unaffected. Moreover, loss of PUS1, PUS7, or TRUB1 function did not significantly reduce the level of Ψ residues detected on total human mRNA below the ∼0.1% level seen in wild-type cells, thus implying that the PUS enzyme(s) that adds the bulk of Ψ residues to human mRNAs remains to be defined.


Subject(s)
Antibodies, Monoclonal/immunology , Gene Editing , Intramolecular Transferases/metabolism , Pseudouridine/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , RNA, Viral/metabolism , HEK293 Cells , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Humans , Hydro-Lyases/antagonists & inhibitors , Hydro-Lyases/genetics , Hydro-Lyases/immunology , Hydro-Lyases/metabolism , Intramolecular Transferases/antagonists & inhibitors , Intramolecular Transferases/genetics , Intramolecular Transferases/immunology , Pseudouridine/immunology , RNA, Messenger/genetics , RNA, Viral/genetics
3.
Genes Dev ; 35(13-14): 992-1004, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34140354

ABSTRACT

Previous work has demonstrated that the epitranscriptomic addition of m6A to viral transcripts can promote the replication and pathogenicity of a wide range of DNA and RNA viruses, including HIV-1, yet the underlying mechanisms responsible for this effect have remained unclear. It is known that m6A function is largely mediated by cellular m6A binding proteins or readers, yet how these regulate viral gene expression in general, and HIV-1 gene expression in particular, has been controversial. Here, we confirm that m6A addition indeed regulates HIV-1 RNA expression and demonstrate that this effect is largely mediated by the nuclear m6A reader YTHDC1 and the cytoplasmic m6A reader YTHDF2. Both YTHDC1 and YTHDF2 bind to multiple distinct and overlapping sites on the HIV-1 RNA genome, with YTHDC1 recruitment serving to regulate the alternative splicing of HIV-1 RNAs. Unexpectedly, while YTHDF2 binding to m6A residues present on cellular mRNAs resulted in their destabilization as previously reported, YTHDF2 binding to m6A sites on HIV-1 transcripts resulted in a marked increase in the stability of these viral RNAs. Thus, YTHDF2 binding can exert diametrically opposite effects on RNA stability, depending on RNA sequence context.


Subject(s)
HIV-1 , Adenosine/metabolism , Alternative Splicing , HIV-1/genetics , HIV-1/metabolism , RNA Splicing , RNA Stability/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism
4.
RNA ; 27(1): 12-26, 2021 01.
Article in English | MEDLINE | ID: mdl-33028652

ABSTRACT

Identifying small molecules that selectively bind an RNA target while discriminating against all other cellular RNAs is an important challenge in RNA-targeted drug discovery. Much effort has been directed toward identifying drug-like small molecules that minimize electrostatic and stacking interactions that lead to nonspecific binding of aminoglycosides and intercalators to many stem-loop RNAs. Many such compounds have been reported to bind RNAs and inhibit their cellular activities. However, target engagement and cellular selectivity assays are not routinely performed, and it is often unclear whether functional activity directly results from specific binding to the target RNA. Here, we examined the propensities of three drug-like compounds, previously shown to bind and inhibit the cellular activities of distinct stem-loop RNAs, to bind and inhibit the cellular activities of two unrelated HIV-1 stem-loop RNAs: the transactivation response element (TAR) and the rev response element stem IIB (RREIIB). All compounds bound TAR and RREIIB in vitro, and two inhibited TAR-dependent transactivation and RRE-dependent viral export in cell-based assays while also exhibiting off-target interactions consistent with nonspecific activity. A survey of X-ray and NMR structures of RNA-small molecule complexes revealed that aminoglycosides and drug-like molecules form hydrogen bonds with functional groups commonly accessible in canonical stem-loop RNA motifs, in contrast to ligands that specifically bind riboswitches. Our results demonstrate that drug-like molecules can nonspecifically bind stem-loop RNAs most likely through hydrogen bonding and electrostatic interactions and reinforce the importance of assaying for off-target interactions and RNA selectivity in vitro and in cells when assessing novel RNA-binders.


Subject(s)
Aminoglycosides/pharmacology , Genes, env/drug effects , HIV Long Terminal Repeat/drug effects , RNA, Viral/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Base Pairing , Base Sequence , Binding Sites , Biological Assay , Drug Discovery , HIV-1/drug effects , HIV-1/genetics , HIV-1/metabolism , Humans , Hydrogen Bonding , Isoquinolines/chemistry , Isoquinolines/metabolism , Isoquinolines/pharmacology , Nucleic Acid Conformation , Pentamidine/chemistry , Pentamidine/metabolism , Pentamidine/pharmacology , RNA, Viral/genetics , RNA, Viral/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Static Electricity , Transcriptional Activation/drug effects , Yohimbine/chemistry , Yohimbine/metabolism , Yohimbine/pharmacology
5.
mBio ; 11(3)2020 06 02.
Article in English | MEDLINE | ID: mdl-32487757

ABSTRACT

Integration of the proviral DNA intermediate into the host cell genome normally represents an essential step in the retroviral life cycle. While the reason(s) for this requirement remains unclear, it is known that unintegrated proviral DNA is epigenetically silenced. Here, we demonstrate that human immunodeficiency virus 1 (HIV-1) mutants lacking a functional integrase (IN) can mount a robust, spreading infection in cells expressing the Tax transcription factor encoded by human T-cell leukemia virus 1 (HTLV-1). In these cells, HIV-1 forms episomal DNA circles, analogous to hepatitis B virus (HBV) covalently closed circular DNAs (cccDNAs), that are transcriptionally active and fully capable of supporting viral replication. In the presence of Tax, induced NF-κB proteins are recruited to the long terminal repeat (LTR) promoters present on unintegrated HIV-1 DNA, and this recruitment in turn correlates with the loss of inhibitory epigenetic marks and the acquisition of activating marks on histones bound to viral DNA. Therefore, HIV-1 is capable of replication in the absence of integrase function if the epigenetic silencing of unintegrated viral DNA can be prevented or reversed.IMPORTANCE While retroviral DNA is synthesized normally after infection by integrase-deficient viruses, the resultant episomal DNA is then epigenetically silenced. Here, we show that expression of the Tax transcription factor encoded by a second human retrovirus, HTLV-1, prevents or reverses the epigenetic silencing of unintegrated HIV-1 DNA and instead induces the addition of activating epigenetic marks and the recruitment of NF-κB/Rel proteins to the HIV-1 LTR promoter. Moreover, in the presence of Tax, the HIV-1 DNA circles that form in the absence of integrase function are not only efficiently transcribed but also support a spreading, pathogenic integrase-deficient (IN-) HIV-1 infection. Thus, retroviruses have the potential to replicate without integration, as is indeed seen with HBV. Moreover, these data suggest that integrase inhibitors may be less effective in the treatment of HIV-1 infections in individuals who are also coinfected with HTLV-1.


Subject(s)
Epigenesis, Genetic , HIV Integrase/genetics , HIV-1/genetics , Virus Integration/genetics , Virus Replication/genetics , A549 Cells , Genes, pX/genetics , HEK293 Cells , HIV-1/physiology , HeLa Cells , Human T-lymphotropic virus 1/genetics , Humans , Promoter Regions, Genetic , THP-1 Cells , Transcription, Genetic
6.
Cell Rep ; 30(8): 2472-2480.e4, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32101729

ABSTRACT

Low-abundance short-lived non-native conformations referred to as excited states (ESs) are increasingly observed in vitro and implicated in the folding and biological activities of regulatory RNAs. We developed an approach for assessing the relative abundance of RNA ESs within the functional cellular context. Nuclear magnetic resonance (NMR) spectroscopy was used to estimate the degree to which substitution mutations bias conformational equilibria toward the inactive ES in vitro. The cellular activity of the ES-stabilizing mutants was used as an indirect measure of the conformational equilibria within the functional cellular context. Compensatory mutations that restore the ground-state conformation were used to control for changes in sequence. Using this approach, we show that the ESs of two regulatory RNAs from HIV-1, the transactivation response element (TAR) and the Rev response element (RRE), likely form in cells with abundances comparable to those measured in vitro, and their targeted stabilization may provide an avenue for developing anti-HIV therapeutics.


Subject(s)
Cells/metabolism , Nucleic Acid Conformation , Cellular Microenvironment , Genes, env , HEK293 Cells , HeLa Cells , Humans , RNA Stability
7.
Cell Host Microbe ; 26(2): 217-227.e6, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31415754

ABSTRACT

How the covalent modification of mRNA ribonucleotides, termed epitranscriptomic modifications, alters mRNA function remains unclear. One issue has been the difficulty of quantifying these modifications. Using purified HIV-1 genomic RNA, we show that this RNA bears more epitranscriptomic modifications than the average cellular mRNA, with 5-methylcytosine (m5C) and 2'O-methyl modifications being particularly prevalent. The methyltransferase NSUN2 serves as the primary writer for m5C on HIV-1 RNAs. NSUN2 inactivation inhibits not only m5C addition to HIV-1 transcripts but also viral replication. This inhibition results from reduced HIV-1 protein, but not mRNA, expression, which in turn correlates with reduced ribosome binding to viral mRNAs. In addition, loss of m5C dysregulates the alternative splicing of viral RNAs. These data identify m5C as a post-transcriptional regulator of both splicing and function of HIV-1 mRNA, thereby affecting directly viral gene expression.


Subject(s)
5-Methylcytosine/pharmacology , Gene Expression Regulation, Viral , HIV-1/genetics , RNA, Viral/metabolism , Transcriptome , 5-Methylcytosine/metabolism , CD4-Positive T-Lymphocytes , Gene Expression Regulation, Viral/drug effects , HEK293 Cells , Humans , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Methyltransferases/pharmacology , RNA Splicing , RNA, Messenger/metabolism , RNA, Viral/drug effects , Virion , Virus Replication/drug effects
8.
mBio ; 10(3)2019 06 11.
Article in English | MEDLINE | ID: mdl-31186331

ABSTRACT

While it has been known for several years that viral RNAs are subject to the addition of several distinct covalent modifications to individual nucleotides, collectively referred to as epitranscriptomic modifications, the effect of these editing events on viral gene expression has been controversial. Here, we report the purification of murine leukemia virus (MLV) genomic RNA to homogeneity and show that this viral RNA contains levels of N6-methyladenosine (m6A), 5-methylcytosine (m5C), and 2'O-methylated (Nm) ribonucleotides that are an order of magnitude higher than detected on bulk cellular mRNAs. Mapping of m6A and m5C residues on MLV transcripts identified multiple discrete editing sites and allowed the construction of MLV variants bearing silent mutations that removed a subset of these sites. Analysis of the replication potential of these mutants revealed a modest but significant attenuation in viral replication in 3T3 cells in culture. Consistent with a positive role for m6A and m5C in viral replication, we also demonstrate that overexpression of the key m6A reader protein YTHDF2 enhances MLV replication, while downregulation of the m5C writer NSUN2 inhibits MLV replication.IMPORTANCE The data presented in the present study demonstrate that MLV RNAs bear an exceptionally high level of the epitranscriptomic modifications m6A, m5C, and Nm, suggesting that these each facilitate some aspect of the viral replication cycle. Consistent with this hypothesis, we demonstrate that mutational removal of a subset of these m6A or m5C modifications from MLV transcripts inhibits MLV replication in cis, and a similar result was also observed upon manipulation of the level of expression of key cellular epitranscriptomic cofactors in trans Together, these results argue that the addition of several different epitranscriptomic modifications to viral transcripts stimulates viral gene expression and suggest that MLV has therefore evolved to maximize the level of these modifications that are added to viral RNAs.


Subject(s)
Adenosine/chemistry , Cytosine/chemistry , DNA Methylation , Leukemia Virus, Murine/genetics , Virus Replication , Gene Expression , Genome, Viral , Leukemia Virus, Murine/physiology , Methyltransferases/metabolism , RNA, Messenger , RNA, Viral/genetics
9.
Virology ; 520: 116-126, 2018 07.
Article in English | MEDLINE | ID: mdl-29857168

ABSTRACT

DNA editing using CRISPR/Cas has emerged as a potential treatment for diseases caused by pathogenic human DNA viruses. One potential target is HIV-1, which replicates via a chromosomally integrated DNA provirus. While CRISPR/Cas can protect T cells from de novo HIV-1 infection, HIV-1 frequently becomes resistant due to mutations in the chosen single guide RNA (sgRNA) target site. To address this problem, we asked whether an sgRNA targeted to a conserved, functionally critical HIV-1 sequence might prevent the selection of escape mutants. We report that two sgRNAs specific for the HIV-1 transactivation response (TAR) element produce opposite results: the TAR2 sgRNA rapidly selects for mutants that retain TAR function, but are no longer inhibited by Cas9, while the TAR1 sgRNA fails to select any replication competent TAR mutants, most probably because it is targeted to a region of TAR that is disrupted by even minor mutations.


Subject(s)
CRISPR-Cas Systems , HIV Long Terminal Repeat , HIV-1/genetics , Proviruses/genetics , Virus Inactivation , Clustered Regularly Interspaced Short Palindromic Repeats , HIV-1/physiology , Humans , Mutation , Proviruses/physiology , RNA, Guide, Kinetoplastida/genetics , Transcriptional Activation , Virus Integration/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
11.
Cell Host Microbe ; 22(3): 377-386.e5, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28910636

ABSTRACT

Many viral RNAs are modified by methylation of the N6 position of adenosine (m6A). m6A is thought to regulate RNA splicing, stability, translation, and secondary structure. Influenza A virus (IAV) expresses m6A-modified RNAs, but the effects of m6A on this segmented RNA virus remain unclear. We demonstrate that global inhibition of m6A addition inhibits IAV gene expression and replication. In contrast, overexpression of the cellular m6A "reader" protein YTHDF2 increases IAV gene expression and replication. To address whether m6A residues modulate IAV RNA function in cis, we mapped m6A residues on the IAV plus (mRNA) and minus (vRNA) strands and used synonymous mutations to ablate m6A on both strands of the hemagglutinin (HA) segment. These mutations inhibited HA mRNA and protein expression while leaving other IAV mRNAs and proteins unaffected, and they also resulted in reduced IAV pathogenicity in mice. Thus, m6A residues in IAV transcripts enhance viral gene expression.


Subject(s)
Influenza A virus/physiology , Influenza, Human/virology , RNA, Viral/genetics , Virus Replication , Animals , DNA Methylation , Epigenesis, Genetic , Female , Host-Pathogen Interactions , Humans , Influenza A virus/genetics , Influenza, Human/genetics , Influenza, Human/metabolism , Methylation , Mice, Inbred C57BL , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcriptome
12.
mBio ; 8(1)2017 01 17.
Article in English | MEDLINE | ID: mdl-28096489

ABSTRACT

Analysis of the incorporation of cellular microRNAs (miRNAs) into highly purified HIV-1 virions revealed that this largely, but not entirely, mirrored the level of miRNA expression in the producer CD4+ T cells. Specifically, of the 58 cellular miRNAs detected at significant levels in the producer cells, only 5 were found in virions at a level 2- to 4-fold higher than that predicted on the basis of random cytoplasmic sampling. Of note, these included two miRNAs, miR-155 and miR-92a, that were reported previously to at least weakly bind HIV-1 transcripts. To test whether miRNA binding to the HIV-1 genome can induce virion incorporation, artificial miRNA target sites were introduced into the viral genome and a 10- to 40-fold increase in the packaging of the cognate miRNAs into virions was then observed, leading to the recruitment of up to 1.6 miRNA copies per virion. Importantly, this high level of incorporation significantly inhibited HIV-1 virion infectivity. These results suggest that target sites for cellular miRNAs can inhibit RNA virus replication at two distinct steps, i.e., during infection and during viral gene expression, thus explaining why a range of different RNA viruses appear to have evolved to avoid cellular miRNA binding to their genome. IMPORTANCE: The genomes of RNA viruses have the potential to interact with cellular miRNAs, which could lead to their incorporation into virions, with unknown effects on virion function. Here, it is demonstrated that wild-type HIV-1 virions essentially randomly incorporate low levels of the miRNAs expressed by infected cells. However, the specific incorporation of high levels of individual cellular miRNAs can be induced by insertion of cognate target sites into the viral genome. Of note, this results in a modest but significant inhibition of virion infectivity. These data imply that cellular miRNAs have the potential to inhibit viral replication by interfering with not only viral mRNA function but also virion infectivity.


Subject(s)
Antiviral Agents/analysis , HIV-1/growth & development , HIV-1/physiology , MicroRNAs/analysis , Virion/chemistry , Virus Assembly , Cell Line , Humans
13.
RNA ; 23(2): 153-160, 2017 02.
Article in English | MEDLINE | ID: mdl-27837013

ABSTRACT

While mammalian somatic cells are incapable of mounting an effective RNA interference (RNAi) response to viral infections, plants and invertebrates are able to generate high levels of viral short interfering RNAs (siRNAs) that can control many infections. In Drosophila, the RNAi response is mediated by the Dicer 2 enzyme (dDcr2) acting in concert with two cofactors called Loqs-PD and R2D2. To examine whether a functional RNAi response could be mounted in human somatic cells, we expressed dDcr2, in the presence or absence of Loqs-PD and/or R2D2, in a previously described human cell line, NoDice/ΔPKR, that lacks functional forms of human Dicer (hDcr) and PKR. We observed significant production of ∼21-nt long siRNAs, derived from a cotransfected double stranded RNA (dsRNA) expression vector, that were loaded into the human RNA-induced silencing complex (RISC) and were able to significantly reduce the expression of a cognate indicator gene. Surprisingly, dDcr2 was able to produce siRNAs even in the absence of Loqs-PD, which is thought to be required for dsRNA cleavage by dDcr2. This result may be explained by our finding that dDcr2 is able to bind the human Loqs-PD homolog TRBP when expressed in human cells in the absence of Loqs-PD. We conclude that it is possible to at least partially rescue the ability of mammalian somatic cells to express functional siRNAs using gene products of invertebrate origin.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , RNA Helicases/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Ribonuclease III/genetics , Animals , Cell Engineering , Cell Line , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Genetic Complementation Test , Humans , Nuclear Receptor Coactivators/genetics , Nuclear Receptor Coactivators/metabolism , Protein Binding , RNA Helicases/metabolism , RNA, Small Interfering/biosynthesis , RNA-Binding Proteins/metabolism , RNA-Induced Silencing Complex/biosynthesis , RNA-Induced Silencing Complex/genetics , Ribonuclease III/deficiency , Ribonuclease III/metabolism , Transgenes , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
14.
Cell Host Microbe ; 19(5): 675-85, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27117054

ABSTRACT

Covalent addition of a methyl group to adenosine N(6) (m(6)A) is an evolutionarily conserved and common RNA modification that is thought to modulate several aspects of RNA metabolism. While the presence of multiple m(6)A editing sites on diverse viral RNAs was reported starting almost 40 years ago, how m(6)A editing affects virus replication has remained unclear. Here, we used photo-crosslinking-assisted m(6)A sequencing techniques to precisely map several m(6)A editing sites on the HIV-1 genome and report that they cluster in the HIV-1 3' untranslated region (3' UTR). Viral 3' UTR m(6)A sites or analogous cellular m(6)A sites strongly enhanced mRNA expression in cis by recruiting the cellular YTHDF m(6)A "reader" proteins. Reducing YTHDF expression inhibited, while YTHDF overexpression enhanced, HIV-1 protein and RNA expression, and virus replication in CD4+ T cells. These data identify m(6)A editing and the resultant recruitment of YTHDF proteins as major positive regulators of HIV-1 mRNA expression.


Subject(s)
HIV-1/genetics , HIV-1/metabolism , RNA Editing , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , 3' Untranslated Regions , CD4-Positive T-Lymphocytes/virology , Cell Line , Cloning, Molecular , Gene Expression Regulation, Viral , Genome, Viral , HEK293 Cells , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Virus Replication/drug effects
15.
Proc Natl Acad Sci U S A ; 112(52): E7249-56, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26668372

ABSTRACT

Whereas several mammalian proteins can restrict the replication of HIV-1 and other viruses, these are often not expressed in relevant target cells. A potential method to inhibit viral replication might therefore be to use synthetic transcription factors to induce restriction factor expression. In particular, mutants of the RNA-guided DNA binding protein Cas9 that have lost their DNA cleavage activity could be used to recruit transcription activation domains to specific promoters. However, initial experiments revealed only weak activation unless multiple promoter-specific single guide RNAs (sgRNAs) were used. Recently, the recruitment of multiple transcription activation domains by a single sgRNA, modified to contain MS2-derived stem loops that recruit fusion proteins consisting of the MS2 coat protein linked to transcription activation domains, was reported to induce otherwise silent cellular genes. Here, we demonstrate that such "synergistic activation mediators" can induce the expression of two restriction factors, APOBEC3G (A3G) and APOBEC3B (A3B), in human cells that normally lack these proteins. We observed modest activation of endogenous A3G or A3B expression using single sgRNAs but high expression when two sgRNAs were used. Whereas the induced A3G and A3B proteins both blocked infection by an HIV-1 variant lacking a functional vif gene by inducing extensive dC-to-dU editing, only the induced A3B protein inhibited wild-type HIV-1. These data demonstrate that Cas9-derived transcriptional activators have the potential to be used for screens for endogenous genes that affect virus replication and raise the possibility that synthetic transcription factors might prove clinically useful if efficient delivery mechanisms could be developed.


Subject(s)
CRISPR-Cas Systems/genetics , Cytidine Deaminase/genetics , Transcriptional Activation , APOBEC-3G Deaminase , Base Sequence , Blotting, Western , Cell Line , Cytidine Deaminase/metabolism , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Minor Histocompatibility Antigens , Reverse Transcriptase Polymerase Chain Reaction , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism
16.
Proc Natl Acad Sci U S A ; 112(50): E6945-54, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26621737

ABSTRACT

Although RNA interference (RNAi) functions as a potent antiviral innate-immune response in plants and invertebrates, mammalian somatic cells appear incapable of mounting an RNAi response and few, if any, small interfering RNAs (siRNAs) can be detected. To examine why siRNA production is inefficient, we have generated double-knockout human cells lacking both Dicer and protein kinase RNA-activated. Using these cells, which tolerate double-stranded RNA expression, we show that a mutant form of human Dicer lacking the amino-terminal helicase domain can process double-stranded RNAs to produce high levels of siRNAs that are readily detectable by Northern blot, are loaded into RNA-induced silencing complexes, and can effectively and specifically inhibit the expression of cognate mRNAs. Remarkably, overexpression of this mutant Dicer, but not wild-type Dicer, also resulted in a partial inhibition of Influenza A virus-but not poliovirus-replication in human cells.


Subject(s)
RNA, Small Interfering/physiology , Ribonuclease III/genetics , Sequence Deletion , HEK293 Cells , Humans , RNA, Viral/genetics , Ribonuclease III/chemistry
17.
Methods ; 91: 82-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26291065

ABSTRACT

RNA-guided endonucleases or CRISPR/Cas systems have been widely employed for gene engineering/DNA editing applications, and have recently been used against a variety of dsDNA viruses as a potential therapeutic. However, in vivo delivery to specific tissue reservoirs using adeno-associated virus (AAV) vectors is problematic due to the large coding requirement for the principal effector commonly used in these applications, Streptococcus pyogenes (Spy) Cas9. Here we describe design of a minimal CRISPR/Cas system that is capable of multiplexing and can be packaged into a single AAV vector. This system consists of the small Type II Cas9 protein from Staphylococcus aureus (Sau) driven by a truncated CMV promoter/enhancer, and flanked 3' by a poly(A) addition signal, as well as two sgRNA expression cassettes driven by either U6 or ∼70-bp tRNA-derived Pol III promoters. Specific protocols for construction of these AAV vector scaffolds, shuttle cloning of their contents into AAV and lentiviral backbones, and a quantitative luciferase assay capable of screening for optimal sgRNAs, are detailed. These protocols can facilitate construction of AAV vectors that have optimal multiplexed sgRNA expression and function. These will have potential utility in multiplex applications, including in antiviral therapy in tissues chronically infected with a pathogenic DNA virus.


Subject(s)
Antiviral Agents/therapeutic use , CRISPR-Cas Systems , Genetic Therapy/methods , Genetic Vectors , Virus Diseases/therapy , Animals , Dependovirus/genetics , Humans , Promoter Regions, Genetic , Staphylococcus aureus/genetics , Virus Diseases/genetics
18.
RNA ; 21(9): 1683-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26187160

ABSTRACT

The in vivo application of CRISPR/Cas-based DNA editing technology will require the development of efficient delivery methods that likely will be dependent on adeno-associated virus (AAV)-based viral vectors. However, AAV vectors have only a modest, ∼4.7-kb packaging capacity, which will necessitate the identification and characterization of highly active Cas9 proteins that are substantially smaller than the prototypic Streptococcus pyogenes Cas9 protein, which covers ∼4.2 kb of coding sequence, as well as the development of single guide RNA (sgRNA) expression cassettes substantially smaller than the current ∼360 bp size. Here, we report that small, ∼70-bp tRNA promoters can be used to express high levels of tRNA:sgRNA fusion transcripts that are efficiently and precisely cleaved by endogenous tRNase Z to release fully functional sgRNAs. Importantly, cells stably expressing functional tRNA:sgRNA precursors did not show a detectable change in the level of endogenous tRNA expression. This novel sgRNA expression strategy should greatly facilitate the construction of effective AAV-based Cas9/sgRNA vectors for future in vivo use.


Subject(s)
CRISPR-Associated Proteins/metabolism , Genetic Engineering/methods , Promoter Regions, Genetic , RNA, Guide, Kinetoplastida/genetics , RNA, Transfer/genetics , Animals , HEK293 Cells , Humans , RNA, Guide, Kinetoplastida/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Sequence Analysis, RNA
19.
Virology ; 476: 196-205, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25553515

ABSTRACT

Hepatitis B virus (HBV) remains a major human pathogen, with over 240 million individuals suffering from chronic HBV infections. These can persist for decades due to the lack of therapies that can effectively target the stable viral covalently closed circular (ccc) DNA molecules present in infected hepatocytes. Using lentiviral transduction of a bacterial Cas9 gene and single guide RNAs (sgRNAs) specific for HBV, we observed effective inhibition of HBV DNA production in in vitro models of both chronic and de novo HBV infection. Cas9/sgRNA combinations specific for HBV reduced total viral DNA levels by up to ~1000-fold and HBV cccDNA levels by up to ~10-fold and also mutationally inactivated the majority of the residual viral DNA. Together, these data provide proof of principle for the hypothesis that CRISPR/Cas systems have the potential to serve as effective tools for the depletion of the cccDNA pool in chronically HBV infected individuals.


Subject(s)
CRISPR-Cas Systems , DNA, Viral/metabolism , Gene Targeting/methods , Hepatitis B virus/genetics , Hepatitis B/virology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , DNA, Viral/genetics , Deoxyribonuclease I/metabolism , Down-Regulation , Hepatitis B/therapy , Hepatitis B virus/physiology , Humans , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/genetics , Virus Replication
20.
J Virol ; 88(20): 11965-72, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25100830

ABSTRACT

High-risk human papillomaviruses (HPVs), including HPV-16 and HPV-18, are the causative agents of cervical carcinomas and are linked to several other tumors of the anogenital and oropharyngeal regions. The majority of HPV-induced tumors contain integrated copies of the normally episomal HPV genome that invariably retain intact forms of the two HPV oncogenes E6 and E7. E6 induces degradation of the cellular tumor suppressor p53, while E7 destabilizes the retinoblastoma (Rb) protein. Previous work has shown that loss of E6 function in cervical cancer cells induces p53 expression as well as downstream effectors that induce apoptosis and cell cycle arrest. Similarly, loss of E7 allows increased Rb expression, leading to cell cycle arrest and senescence. Here, we demonstrate that expression of a bacterial Cas9 RNA-guided endonuclease, together with single guide RNAs (sgRNAs) specific for E6 or E7, is able to induce cleavage of the HPV genome, resulting in the introduction of inactivating deletion and insertion mutations into the E6 or E7 gene. This results in the induction of p53 or Rb, leading to cell cycle arrest and eventual cell death. Both HPV-16- and HPV-18-transformed cells were found to be responsive to targeted HPV genome-specific DNA cleavage. These data provide a proof of principle for the idea that vector-delivered Cas9/sgRNA combinations could represent effective treatment modalities for HPV-induced cancers. Importance: Human papillomaviruses (HPVs) are the causative agents of almost all cervical carcinomas and many other tumors, including many head and neck cancers. In these cancer cells, the HPV DNA genome is integrated into the cellular genome, where it expresses high levels of two viral oncogenes, called E6 and E7, that are required for cancer cell growth and viability. Here, we demonstrate that the recently described bacterial CRISPR/Cas RNA-guided endonuclease can be reprogrammed to target and destroy the E6 or E7 gene in cervical carcinoma cells transformed by HPV, resulting in cell cycle arrest, leading to cancer cell death. We propose that viral vectors designed to deliver E6- and/or E7-specific CRISPR/Cas to tumor cells could represent a novel and highly effective tool to treat and eliminate HPV-induced cancers.


Subject(s)
Alphapapillomavirus/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DNA-Binding Proteins/genetics , Endonucleases/metabolism , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Repressor Proteins/genetics , Uterine Cervical Neoplasms/genetics , Alphapapillomavirus/isolation & purification , Base Sequence , Cell Line, Tumor , DNA, Viral/genetics , Female , Humans , Molecular Sequence Data , Uterine Cervical Neoplasms/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...