Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 159(5): 975-976, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25416937

RESUMEN

HIV-1 virion assembly at the plasma membrane requires the selective recruitment of the viral RNA genome into nascent viral particles while cellular transcripts are excluded. Kutluay et al. now demonstrate that this is a two-step process in which Gag binds sequentially to different sites on the viral genome.


Asunto(s)
VIH-1/fisiología , ARN Viral/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos
2.
Nature ; 617(7962): 835-841, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37198487

RESUMEN

Cellular processes are the product of interactions between biomolecules, which associate to form biologically active complexes1. These interactions are mediated by intermolecular contacts, which if disrupted, lead to alterations in cell physiology. Nevertheless, the formation of intermolecular contacts nearly universally requires changes in the conformations of the interacting biomolecules. As a result, binding affinity and cellular activity crucially depend both on the strength of the contacts and on the inherent propensities to form binding-competent conformational states2,3. Thus, conformational penalties are ubiquitous in biology and must be known in order to quantitatively model binding energetics for protein and nucleic acid interactions4,5. However, conceptual and technological limitations have hindered our ability to dissect and quantitatively measure how conformational propensities affect cellular activity. Here we systematically altered and determined the propensities for forming the protein-bound conformation of HIV-1 TAR RNA. These propensities quantitatively predicted the binding affinities of TAR to the RNA-binding region of the Tat protein and predicted the extent of HIV-1 Tat-dependent transactivation in cells. Our results establish the role of ensemble-based conformational propensities in cellular activity and reveal an example of a cellular process driven by an exceptionally rare and short-lived RNA conformational state.


Asunto(s)
Duplicado del Terminal Largo de VIH , VIH-1 , Conformación de Ácido Nucleico , ARN Viral , Activación Transcripcional , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Duplicado del Terminal Largo de VIH/genética , ARN Viral/química , ARN Viral/genética , ARN Viral/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , VIH-1/genética , VIH-1/metabolismo
3.
Genes Dev ; 35(13-14): 992-1004, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34140354

RESUMEN

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.


Asunto(s)
VIH-1 , Adenosina/metabolismo , Empalme Alternativo , VIH-1/genética , VIH-1/metabolismo , Empalme del ARN , Estabilidad del ARN/genética , ARN Viral/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo
4.
Immunity ; 46(6): 970-972, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28636964

RESUMEN

The importance of RNA interference (RNAi) as a mammalian antiviral defense mechanism has been controversial. Qiu et al. (2017) now present data suggesting that the difficulty of detecting RNAi in virus-infected mammalian cells reflects the expression of highly effective viral suppressors of RNAi.


Asunto(s)
Interferencia de ARN , ARN Interferente Pequeño/genética , Animales , Antivirales , Humanos , Mamíferos/genética , Virus/genética
5.
Nat Immunol ; 14(3): 205-10, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23416678

RESUMEN

Cellular microRNAs serve key roles in the post-transcriptional regulation of almost every cellular gene-regulatory pathway, and it therefore is not surprising that viruses have found ways to subvert this process. Several viruses encode microRNAs that directly downregulate the expression of factors of the innate immune system, including proteins involved in promoting apoptosis and recruiting effector cells of the immune system. Viruses have also evolved the ability to downregulate or upregulate the expression of specific cellular miRNAs to enhance their replication. This Review provides an overview of the present knowledge of the complex interactions of viruses with the microRNA machinery of cells.


Asunto(s)
Evasión Inmune/genética , MicroARNs/genética , Virus , Animales , Apoptosis , Citocinas/biosíntesis , Citocinas/genética , Regulación de la Expresión Génica , Humanos , MicroARNs/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Replicación Viral , Virus/genética , Virus/inmunología , Virus/patogenicidad
6.
Cell ; 136(4): 592-7, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19239880

RESUMEN

Viruses are adept at evolving or co-opting genomic elements that allow them to maximize their replication potential in the infected host. This evolutionary plasticity makes viruses an invaluable system to identify new mechanisms used not only by viruses but also by vertebrate cells to modulate gene expression. Here, I discuss the identification and characterization of viral mRNA structures and noncoding RNAs that have led to important insights into the molecular mechanisms of eukaryotic cells.


Asunto(s)
ARN Viral/metabolismo , Virus/genética , Animales , Células Eucariotas/metabolismo , Células Eucariotas/virología , Regulación de la Expresión Génica , Humanos , ARN Mensajero/química , ARN Mensajero/metabolismo , ARN Viral/química , Virus/metabolismo
8.
RNA ; 27(11): 1400-1411, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34376564

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Edición Génica , Transferasas Intramoleculares/metabolismo , Seudouridina/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Células HEK293 , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Humanos , Hidroliasas/antagonistas & inhibidores , Hidroliasas/genética , Hidroliasas/inmunología , Hidroliasas/metabolismo , Transferasas Intramoleculares/antagonistas & inhibidores , Transferasas Intramoleculares/genética , Transferasas Intramoleculares/inmunología , Seudouridina/inmunología , ARN Mensajero/genética , ARN Viral/genética
9.
RNA ; 27(1): 12-26, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33028652

RESUMEN

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.


Asunto(s)
Aminoglicósidos/farmacología , Genes env/efectos de los fármacos , Duplicado del Terminal Largo de VIH/efectos de los fármacos , ARN Viral/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Aminoglicósidos/química , Aminoglicósidos/metabolismo , Emparejamiento Base , Secuencia de Bases , Sitios de Unión , Bioensayo , Descubrimiento de Drogas , VIH-1/efectos de los fármacos , VIH-1/genética , VIH-1/metabolismo , Humanos , Enlace de Hidrógeno , Isoquinolinas/química , Isoquinolinas/metabolismo , Isoquinolinas/farmacología , Conformación de Ácido Nucleico , Pentamidina/química , Pentamidina/metabolismo , Pentamidina/farmacología , ARN Viral/genética , ARN Viral/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Electricidad Estática , Activación Transcripcional/efectos de los fármacos , Yohimbina/química , Yohimbina/metabolismo , Yohimbina/farmacología
10.
J Virol ; 95(13): e0028521, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33883218

RESUMEN

We previously reported that the normally essential step of integration of the HIV-1 proviral DNA intermediate into the host cell genome becomes dispensable in T cells that express the human T cell leukemia virus 1 (HTLV-1) Tax protein, a known activator of cellular NF-κB. The rescue of integrase (IN)-deficient HIV-1 replication by Tax results from the strong activation of transcription from the long terminal repeat (LTR) promoter on episomal HIV-1 DNA, an effect that is closely correlated with the recruitment of activating epigenetic marks, such as H3Ac, and depletion of repressive epigenetic marks, such as H3K9me3, from chromatinized unintegrated proviruses. In addition, activation of transcription from unintegrated HIV-1 DNA coincides with the recruitment of NF-κB to the two NF-κB binding sites found in the HIV-1 LTR enhancer. Here, we report that the recruitment of NF-κB to unintegrated viral DNA precedes, and is a prerequisite for, Tax-induced changes in epigenetic marks, so that an IN- HIV-1 mutant lacking both LTR NF-κB sites is entirely nonresponsive to Tax and fails to undergo the epigenetic changes listed above. Interestingly, we found that induction of Tax expression at 24 h postinfection, when unintegrated HIV-1 DNA is already fully repressed by inhibitory chromatin modifications, is able to effectively reverse the epigenetic silencing of that DNA and rescue viral gene expression. Finally, we report that heterologous promoters introduced into IN-deficient HIV-1-based vectors are transcriptionally active even in the absence of Tax and do not increase their activity when the HIV-1 promoter and enhancer, located in the LTR U3 region, are deleted, as has been recently proposed. IMPORTANCE Integrase-deficient expression vectors based on HIV-1 are becoming increasingly popular as tools for gene therapy in vivo due to their inability to cause insertional mutagenesis. However, many IN- lentiviral vectors are able to achieve only low levels of gene expression, and methods to increase this low level have not been extensively explored. Here, we analyzed how the HTLV-1 Tax protein is able to rescue the replication of IN- HIV-1 in T cells, and we describe IN- lentiviral vectors, lacking any inserted origin of replication, that are able to express a heterologous gene effectively.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Productos del Gen tax/metabolismo , Integrasa de VIH/genética , VIH-1/genética , Integración Viral/genética , Línea Celular , ADN Viral/genética , Activación Enzimática/genética , Expresión Génica/genética , Células HEK293 , Integrasa de VIH/deficiencia , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Provirus/genética , Replicación Viral/genética
11.
RNA ; 24(9): 1172-1182, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29903832

RESUMEN

While the issue of whether RNA interference (RNAi) ever forms part of the antiviral innate immune response in mammalian somatic cells remains controversial, there is considerable evidence demonstrating that few, if any, viral small interfering RNAs (siRNAs) are produced in infected cells. Moreover, inhibition of RNAi by mutational inactivation of key RNAi factors, such as Dicer or Argonaute 2, fails to enhance virus replication. One potential explanation for this lack of inhibitory effect is that mammalian viruses encode viral suppressors of RNAi (VSRs) that are so effective that viral siRNAs are not produced in infected cells. Indeed, a number of mammalian VSRs have been described, of which the most prominent is the influenza A virus (IAV) NS1 protein, which has not only been reported to inhibit RNAi in plants and insects but also to prevent the production of viral siRNAs in IAV-infected human cells. Here, we confirm that an IAV mutant lacking NS1 indeed differs from wild-type IAV in that it induces the production of readily detectable levels of Dicer-dependent viral siRNAs in infected human cells. However, we also demonstrate that these siRNAs have little if any inhibitory effect on IAV gene expression. This is likely due, at least in part, to their inefficient loading into RNA-induced silencing complexes.


Asunto(s)
ARN Helicasas DEAD-box/genética , Virus de la Influenza A/fisiología , Interferencia de ARN , Ribonucleasa III/genética , Proteínas no Estructurales Virales/genética , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/genética , Mutación , ARN Viral/genética , Análisis de Secuencia de ARN , Replicación Viral
12.
13.
PLoS Pathog ; 14(2): e1006919, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29447282

RESUMEN

Polyomaviruses are a family of small DNA tumor viruses that includes several pathogenic human members, including Merkel cell polyomavirus, BK virus and JC virus. As is characteristic of DNA tumor viruses, gene expression in polyomaviruses is temporally regulated into an early phase, consisting of the viral regulatory proteins, and a late phase, consisting of the viral structural proteins. Previously, the late transcripts expressed by the prototypic polyomavirus simian virus 40 (SV40) were reported to contain several adenosines bearing methyl groups at the N6 position (m6A), although the precise location of these m6A residues, and their phenotypic effects, have not been investigated. Here, we first demonstrate that overexpression of the key m6A reader protein YTHDF2 induces more rapid viral replication, and larger viral plaques, in SV40 infected BSC40 cells, while mutational inactivation of the endogenous YTHDF2 gene, or the m6A methyltransferase METTL3, has the opposite effect, thus suggesting a positive role for m6A in the regulation of SV40 gene expression. To directly test this hypothesis, we mapped sites of m6A addition on SV40 transcripts and identified two m6A sites on the viral early transcripts and eleven m6A sites on the late mRNAs. Using synonymous mutations, we inactivated the majority of the m6A sites on the SV40 late mRNAs and observed that the resultant viral mutant replicated more slowly than wild type SV40. Alternative splicing of SV40 late mRNAs was unaffected by the reduction in m6A residues and our data instead suggest that m6A enhances the translation of viral late transcripts. Together, these data argue that the addition of m6A residues to the late transcripts encoded by SV40 plays an important role in enhancing viral gene expression and, hence, replication.


Asunto(s)
Adenosina/análogos & derivados , Procesamiento Postranscripcional del ARN/fisiología , ARN Mensajero/metabolismo , Virus 40 de los Simios/genética , Proteínas Estructurales Virales/genética , Replicación Viral/genética , Células A549 , Adenosina/metabolismo , Animales , Células Cultivadas , Chlorocebus aethiops , Regulación Viral de la Expresión Génica , Genes Virales , Células HEK293 , Humanos , Metilación , ARN Viral/metabolismo , Virus 40 de los Simios/metabolismo , Células Vero
14.
Annu Rev Med ; 68: 401-411, 2017 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-27576009

RESUMEN

The emergence of the CRISPR/Cas system of antiviral adaptive immunity in bacteria as a facile system for gene editing in mammalian cells may well lead to gene editing becoming a novel treatment for a range of human diseases, especially those caused by deleterious germline mutations. Another potential target for gene editing are DNA viruses that cause chronic pathogenic diseases that cannot be cured by using currently available drugs. We review the current state of this field and discuss the potential advantages and problems with using a gene editing approach as a treatment for diseases caused by DNA viruses.


Asunto(s)
Sistemas CRISPR-Cas , Infecciones por Virus ADN/terapia , Virus ADN/genética , Edición Génica , Infecciones por VIH/terapia , VIH-1/genética , Terapia Genética/métodos , Vectores Genéticos , Virus de la Hepatitis B/genética , Hepatitis B Crónica/terapia , Herpes Genital/terapia , Herpes Simple/terapia , Herpesvirus Humano 1/genética , Herpesvirus Humano 2/genética , Humanos , Papillomaviridae/genética , Infecciones por Papillomavirus/terapia
15.
RNA ; 23(2): 153-160, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27837013

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , ARN Helicasas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética , Animales , Ingeniería Celular , Línea Celular , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Prueba de Complementación Genética , Humanos , Coactivadores de Receptor Nuclear/genética , Coactivadores de Receptor Nuclear/metabolismo , Unión Proteica , ARN Helicasas/metabolismo , ARN Interferente Pequeño/biosíntesis , Proteínas de Unión al ARN/metabolismo , Complejo Silenciador Inducido por ARN/biosíntesis , Complejo Silenciador Inducido por ARN/genética , Ribonucleasa III/deficiencia , Ribonucleasa III/metabolismo , Transgenes , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética
16.
Genes Dev ; 25(18): 1881-94, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21896651

RESUMEN

Analyses of small RNA expression profiles have revealed that several DNA viruses-including particularly, herpesviruses-express high levels of multiple viral microRNAs (miRNAs) in infected cells. Here, I review our current understanding of how viral miRNAs influence viral replication and pathogenesis and discuss how viruses reshape the pattern of cellular miRNA expression. Indeed, viruses are now known to both activate and repress the expression of specific cellular miRNAs, and disrupting this process can perturb the ability of viruses to replicate normally. In addition, it is now clear that virally encoded miRNAs play a key role in inhibiting antiviral innate immune responses and can also promote cell transformation in culture. While our understanding of how viruses interact with miRNAs remains somewhat rudimentary, it is nevertheless already clear that these interactions can play a critical role in mediating viral pathogenesis and therefore may represent novel and highly specific targets for therapeutic intervention.


Asunto(s)
Virus ADN/fisiología , MicroARNs/genética , MicroARNs/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Complejo Silenciador Inducido por ARN/metabolismo , Animales , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/virología , Virus ADN/genética , Regulación Viral de la Expresión Génica , Humanos , Fenotipo , ARN Mensajero/metabolismo , Replicación Viral/genética
17.
J Virol ; 91(9)2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28250115

RESUMEN

Although it has been known for over 40 years that eukaryotic mRNAs bear internal base modifications, it is only in the last 5 years that the importance of these modifications has begun to come into focus. The most common mRNA modification, the addition of a methyl group to the N6 position of adenosine (m6A), has been shown to affect splicing, translation, and stability, and m6A is also essential for embryonic development in organisms ranging from plants to mice. While all viral transcripts examined so far have been found to be extensively m6A modified, the role, if any, of m6A in regulating viral gene expression and replication was previously unknown. However, recent data generated using HIV-1 as a model system strongly suggest that sites of m6A addition not only are evolutionarily conserved but also enhance virus replication. It is therefore likely that the field of viral epitranscriptomics, which can be defined as the study of functionally relevant posttranscriptional modifications of viral RNA transcripts that do not change the nucleotide sequence of that RNA, is poised for a major expansion in scientific interest and may well fundamentally change our understanding of how viral replication is regulated.


Asunto(s)
Adenosina/análogos & derivados , Virus ADN/genética , Procesamiento Postranscripcional del ARN/genética , Virus ARN/genética , ARN Mensajero/genética , ARN Viral/genética , Adenosina/genética , VIH-1/genética , Humanos , Metilación , Metiltransferasas/metabolismo
18.
Nature ; 552(7684): 184-185, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29239386
19.
Mol Cell ; 37(1): 135-42, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20129062

RESUMEN

Canonical primary microRNA (pri-miRNA) precursors are transcribed by RNA polymerase II and then processed by the Drosha endonuclease to generate approximately 60 nt pre-miRNA hairpins. Pre-miRNAs in turn are cleaved by Dicer to generate mature miRNAs. Previously, some short introns, called miRtrons, were reported to fold into pre-miRNA hairpins after splicing and debranching, and miRNAs can also be excised by Dicer cleavage of rare endogenous short hairpin RNAs. Here we report that the miRNAs encoded by murine gamma-herpesvirus 68 (MHV68) are also generated via atypical mechanisms. Specifically, MHV68 miRNAs are transcribed from RNA polymerase III promoters located within adjacent viral tRNA-like sequences. The resultant pri-miRNAs, which bear a 5' tRNA moiety, are not processed by Drosha but instead by cellular tRNase Z, which cleaves 3' to the tRNA to liberate pre-miRNA hairpins that are then processed by Dicer to yield the mature viral miRNAs.


Asunto(s)
MicroARNs/biosíntesis , ARN Viral/biosíntesis , Rhadinovirus/genética , Línea Celular , Endorribonucleasas/metabolismo , Humanos , MicroARNs/química , Conformación de Ácido Nucleico , Interferencia de ARN , ARN Polimerasa III/fisiología , ARN de Transferencia/metabolismo , ARN Viral/química , Ribonucleasa III/metabolismo
20.
Proc Natl Acad Sci U S A ; 112(50): E6945-54, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621737

RESUMEN

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.


Asunto(s)
ARN Interferente Pequeño/fisiología , Ribonucleasa III/genética , Eliminación de Secuencia , Células HEK293 , Humanos , ARN Viral/genética , Ribonucleasa III/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA