Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
PLoS Pathog ; 20(6): e1011970, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38885264

RESUMEN

Reactivation from latency plays a significant role in maintaining persistent lifelong Epstein-Barr virus (EBV) infection. Mechanisms governing successful activation and progression of the EBV lytic phase are not fully understood. EBV expresses multiple viral microRNAs (miRNAs) and manipulates several cellular miRNAs to support viral infection. To gain insight into the host miRNAs regulating transitions from EBV latency into the lytic stage, we conducted a CRISPR/Cas9-based screen in EBV+ Burkitt lymphoma (BL) cells using anti-Ig antibodies to crosslink the B cell receptor (BCR) and induce reactivation. Using a gRNA library against >1500 annotated human miRNAs, we identified miR-142 as a key regulator of EBV reactivation. Genetic ablation of miR-142 enhanced levels of immediate early and early lytic gene products in infected BL cells. Ago2-PAR-CLIP experiments with reactivated cells revealed miR-142 targets related to Erk/MAPK signaling, including components directly downstream of the B cell receptor (BCR). Consistent with these findings, disruption of miR-142 enhanced SOS1 levels and Mek phosphorylation in response to surface Ig cross-linking. Effects could be rescued by inhibitors of Mek (cobimetinib) or Raf (dabrafenib). Taken together, these results show that miR-142 functionally regulates SOS1/Ras/Raf/Mek/Erk signaling initiated through the BCR and consequently, restricts EBV entry into the lytic cycle.


Asunto(s)
Sistemas CRISPR-Cas , Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , MicroARNs , Activación Viral , Latencia del Virus , Humanos , Herpesvirus Humano 4/genética , MicroARNs/genética , MicroARNs/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Linfoma de Burkitt/virología , Linfoma de Burkitt/genética , Linfoma de Burkitt/metabolismo , Línea Celular Tumoral
2.
Genes Dev ; 30(18): 2076-2092, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798849

RESUMEN

RNA silencing is a conserved eukaryotic gene expression regulatory mechanism mediated by small RNAs. In Caenorhabditis elegans, the accumulation of a distinct class of siRNAs synthesized by an RNA-dependent RNA polymerase (RdRP) requires the PIR-1 phosphatase. However, the function of PIR-1 in RNAi has remained unclear. Since mammals lack an analogous siRNA biogenesis pathway, an RNA silencing role for the mammalian PIR-1 homolog (dual specificity phosphatase 11 [DUSP11]) was unexpected. Here, we show that the RNA triphosphatase activity of DUSP11 promotes the RNA silencing activity of viral microRNAs (miRNAs) derived from RNA polymerase III (RNAP III) transcribed precursors. Our results demonstrate that DUSP11 converts the 5' triphosphate of miRNA precursors to a 5' monophosphate, promoting loading of derivative 5p miRNAs into Argonaute proteins via a Dicer-coupled 5' monophosphate-dependent strand selection mechanism. This mechanistic insight supports a likely shared function for PIR-1 in C. elegans Furthermore, we show that DUSP11 modulates the 5' end phosphate group and/or steady-state level of several host RNAP III transcripts, including vault RNAs and Alu transcripts. This study shows that steady-state levels of select noncoding RNAs are regulated by DUSP11 and defines a previously unknown portal for small RNA-mediated silencing in mammals, revealing that DUSP11-dependent RNA silencing activities are shared among diverse metazoans.


Asunto(s)
Proteínas Argonautas/metabolismo , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , MicroARNs/metabolismo , ARN no Traducido/metabolismo , Ácido Anhídrido Hidrolasas/metabolismo , Adenoviridae/genética , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Virus de la Leucemia Bovina/genética , Fosforilación , ARN Polimerasa III/metabolismo , ARN Viral/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(32): 8197-8202, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30038017

RESUMEN

Seventy percent of people infected with hepatitis C virus (HCV) will suffer chronic infection, putting them at risk for liver disease, including hepatocellular carcinoma. The full range of mechanisms that render some people more susceptible to chronic infection and liver disease is still being elucidated. XRN exonucleases can restrict HCV replication and may help to resolve HCV infections. However, it is unknown how 5' triphosphorylated HCV transcripts, primary products of the viral polymerase, become susceptible to attack by 5' monophosphate-specific XRNs. Here, we show that the 5' RNA triphosphatase DUSP11 acts on HCV transcripts, rendering them susceptible to XRN-mediated attack. Cells lacking DUSP11 show substantially enhanced HCV replication, and this effect is diminished when XRN expression is reduced. MicroRNA-122 (miR-122), a target of current phase II anti-HCV drugs, is known to protect HCV transcripts against XRNs. We show that HCV replication is less dependent on miR-122 in cells lacking DUSP11. Combined, these results implicate DUSP11 as an important component of XRN-mediated restriction of HCV.


Asunto(s)
Ácido Anhídrido Hidrolasas/metabolismo , Fosfatasas de Especificidad Dual/metabolismo , Exorribonucleasas/metabolismo , Hepacivirus/patogenicidad , Interacciones Huésped-Patógeno/fisiología , MicroARNs/metabolismo , Ácido Anhídrido Hidrolasas/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Fosfatasas de Especificidad Dual/genética , Exorribonucleasas/genética , Técnicas de Inactivación de Genes , Genoma Viral , Hepacivirus/fisiología , Hepatitis C Crónica/genética , Hepatitis C Crónica/virología , Hepatocitos/virología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , ARN Interferente Pequeño/metabolismo , ARN Viral/metabolismo , Replicación Viral/genética
4.
PLoS Pathog ; 14(7): e1007156, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30048533

RESUMEN

MicroRNAs (miRNAs) are small RNAs that regulate diverse biological processes including multiple aspects of the host-pathogen interface. Consequently, miRNAs are commonly encoded by viruses that undergo long-term persistent infection. Papillomaviruses (PVs) are capable of undergoing persistent infection, but as yet, no widely-accepted PV-encoded miRNAs have been described. The incomplete understanding of PV-encoded miRNAs is due in part to lack of tractable laboratory models for most PV types. To overcome this, we have developed miRNA Discovery by forced Genome Expression (miDGE), a new wet bench approach to miRNA identification that screens numerous pathogen genomes in parallel. Using miDGE, we screened over 73 different PV genomes for the ability to code for miRNAs. Our results show that most PVs are unlikely to code for miRNAs and we conclusively demonstrate a lack of PV miRNA expression in cancers associated with infections of several high risk HPVs. However, we identified five different high-confidence or highly probable miRNAs encoded by four different PVs (Human PVs 17, 37, 41 and a Fringilla coelebs PV (FcPV1)). Extensive in vitro assays confirm the validity of these miRNAs in cell culture and two FcPV1 miRNAs are further confirmed to be expressed in vivo in a natural host. We show that miRNAs from two PVs (HPV41 & FcPV1) are able to regulate viral transcripts corresponding to the early region of the PV genome. Combined, these findings identify the first canonical PV miRNAs and support that miRNAs of either host or viral origin are important regulators of the PV life cycle.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Regulación Viral de la Expresión Génica/genética , MicroARNs/genética , Papillomaviridae/genética , ARN Viral/análisis , Células HEK293 , Humanos , Infecciones por Papillomavirus/genética , ARN Viral/genética , Transcriptoma
5.
J Virol ; 92(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29875236

RESUMEN

Polyomaviruses (PyVs) can cause serious disease in immunosuppressed hosts. Several pathogenic PyVs encode microRNAs (miRNAs), small RNAs that regulate gene expression via RNA silencing. Despite recent advances in understanding the activities of PyV miRNAs, the biological functions of PyV miRNAs during in vivo infections are mostly unknown. The studies presented here used murine polyomavirus (MuPyV) as a model to assess the roles of the PyV miRNAs in a natural host. This analysis revealed that a MuPyV mutant that is unable to express miRNAs has enhanced viral DNA loads in select tissues at late times after infection. This is consistent with the PyV miRNAs functioning to reduce viral replication during the persistent phase of infection in a natural host. Additionally, the MuPyV miRNA locus promotes viruria during the acute phase of infection as evidenced by a defect in shedding during infection with the miRNA mutant virus. The viruria defect of the miRNA mutant virus could be rescued by infecting Rag2-/- mice. These findings implicate the miRNA locus as functioning in both the persistent and acute phases of infection and suggest a role for MuPyV miRNA in evading the adaptive immune response.IMPORTANCE MicroRNAs are expressed by diverse viruses, but for only a few is there any understanding of their in vivo function. PyVs can cause serious disease in immunocompromised hosts. Therefore, increased knowledge of how these viruses interact with the immune response is of clinical relevance. Here we show a novel activity for a viral miRNA locus in promoting virus shedding. This work indicates that in addition to any role for the PyV miRNA locus in long-term persistence, it also has biological activity during the acute phase. As this mutant phenotype is alleviated by infection of mice lacking an adaptive immune response, our work also connects the in vivo activity of the PyV miRNA locus to the immune response. Given that PyV-associated disease is associated with alterations in the immune response, our findings help to better understand how the balance between PyVs and the immune response becomes altered in pathogenic states.


Asunto(s)
MicroARNs/metabolismo , Infecciones por Polyomavirus/patología , Infecciones por Polyomavirus/virología , Poliomavirus/patogenicidad , ARN Viral/metabolismo , Orina/virología , Animales , Ratones , MicroARNs/genética , Poliomavirus/genética , ARN Viral/genética , Esparcimiento de Virus
6.
Nucleic Acids Res ; 45(17): e154, 2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-28973449

RESUMEN

Short hairpin RNAs (shRNAs) are effective in generating stable repression of gene expression. RNA polymerase III (RNAP III) type III promoters (U6 or H1) are typically used to drive shRNA expression. While useful for some knockdown applications, the robust expression of U6/H1-driven shRNAs can induce toxicity and generate heterogeneous small RNAs with undesirable off-target effects. Additionally, typical U6/H1 promoters encompass the majority of the ∼270 base pairs (bp) of vector space required for shRNA expression. This can limit the efficacy and/or number of delivery vector options, particularly when delivery of multiple gene/shRNA combinations is required. Here, we develop a compact shRNA (cshRNA) expression system based on retroviral microRNA (miRNA) gene architecture that uses RNAP III type II promoters. We demonstrate that cshRNAs coded from as little as 100 bps of total coding space can precisely generate small interfering RNAs (siRNAs) that are active in the RNA-induced silencing complex (RISC). We provide an algorithm with a user-friendly interface to design cshRNAs for desired target genes. This cshRNA expression system reduces the coding space required for shRNA expression by >2-fold as compared to the typical U6/H1 promoters, which may facilitate therapeutic RNAi applications where delivery vector space is limiting.


Asunto(s)
Marcación de Gen/métodos , Virus de la Leucemia Bovina/genética , MicroARNs/genética , ARN Polimerasa III/genética , ARN Interferente Pequeño/genética , ARN Viral/genética , Algoritmos , Emparejamiento Base , Secuencia de Bases , Regulación de la Expresión Génica , Silenciador del Gen , Genes Reporteros , Vectores Genéticos , Células HEK293 , Humanos , Virus de la Leucemia Bovina/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , MicroARNs/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa III/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Viral/metabolismo , Complejo Silenciador Inducido por ARN/genética , Complejo Silenciador Inducido por ARN/metabolismo , Análisis de Secuencia de ARN , Interfaz Usuario-Computador
7.
RNA ; 20(7): 1068-77, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24854622

RESUMEN

Processing of primary microRNA (pri-miRNA) stem-loops by the Drosha-DGCR8 complex is the initial step in miRNA maturation and crucial for miRNA function. Nonetheless, the underlying mechanism that determines the Drosha cleavage site of pri-miRNAs has remained unclear. Two prevalent but seemingly conflicting models propose that Drosha-DGCR8 anchors to and directs cleavage a fixed distance from either the basal single-stranded (ssRNA) or the terminal loop. However, recent studies suggest that the basal ssRNA and/or the terminal loop may influence the Drosha cleavage site dependent upon the sequence/structure of individual pri-miRNAs. Here, using a panel of closely related pri-miRNA variants, we further examine the role of pri-miRNA structures on Drosha cleavage site selection in cells. Our data reveal that both the basal ssRNA and terminal loop influence the Drosha cleavage site within three pri-miRNAs, the Simian Virus 40 (SV40) pri-miRNA, pri-miR-30a, and pri-miR-16. In addition to the flanking ssRNA regions, we show that an internal loop within the SV40 pri-miRNA stem strongly influences Drosha cleavage position and efficiency. We further demonstrate that the positions of the internal loop, basal ssRNA, and the terminal loop of the SV40 pri-miRNA cooperatively coordinate Drosha cleavage position and efficiency. Based on these observations, we propose that the pri-miRNA stem, defined by internal and flanking structural elements, guides the binding position of Drosha-DGCR8, which consequently determines the cleavage site. This study provides mechanistic insight into pri-miRNA processing in cells that has numerous biological implications and will assist in refining Drosha-dependent shRNA design.


Asunto(s)
MicroARNs/metabolismo , Conformación de Ácido Nucleico , Procesamiento Postranscripcional del ARN , ARN Viral , Ribonucleasa III/metabolismo , Secuencia de Bases , Sitios de Unión/genética , Células HEK293 , Humanos , MicroARNs/química , Datos de Secuencia Molecular , Proteínas/metabolismo , División del ARN/genética , Precursores del ARN/química , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN/genética , ARN Viral/química , ARN Viral/metabolismo , Proteínas de Unión al ARN , Ribonucleasa III/genética , Virus 40 de los Simios/genética
8.
Methods ; 91: 57-68, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26210399

RESUMEN

Many eukaryotes and some viruses encode microRNAs (miRNAs), small RNAs that post-transcriptionally regulate gene expression. While most miRNAs are generated through the activity of RNA Polymerase II (RNAP II) and subsequent processing by Drosha and Dicer, some viral miRNAs utilize alternative pathways of biogenesis. Some members of the herpesvirus and retrovirus families can direct synthesis of miRNAs through RNAP III transcription rather than RNAP II and can utilize atypical enzymes to generate miRNAs. Though the advantages of alternative miRNA biogenesis remain unclear for herpesviruses, the retroviral miRNA biogenesis routes allow the RNAP II transcribed retroviral genome to escape Drosha cleavage while still expressing abundant, biologically-active miRNAs. These RNAP III-derived miRNAs have unique characteristics that allow for their identification and characterization. In this article, we describe procedures to predict, validate, and characterize RNAP III-transcribed miRNAs and other small RNAs, while providing resources that are also useful for canonical miRNAs.


Asunto(s)
Algoritmos , Herpesviridae/metabolismo , MicroARNs/biosíntesis , ARN Viral/biosíntesis , Retroviridae/metabolismo , Eucariontes/metabolismo , Herpesviridae/genética , ARN Polimerasa III/metabolismo , Retroviridae/genética , Transcripción Genética
9.
Nucleic Acids Res ; 42(22): 13949-62, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25428356

RESUMEN

Transcripts possessing a 5'-triphosphate are a hallmark of viral transcription and can trigger the host antiviral response. 5'-triphosphates are also found on common host transcripts transcribed by RNA polymerase III (RNAP III), yet how these transcripts remain non-immunostimulatory is incompletely understood. Most microRNAs (miRNAs) are 5'-monophosphorylated as a result of sequential endonucleolytic processing by Drosha and Dicer from longer RNA polymerase II (RNAP II)-transcribed primary transcripts. In contrast, bovine leukemia virus (BLV) expresses subgenomic RNAP III transcripts that give rise to miRNAs independent of Drosha processing. Here, we demonstrate that each BLV pre-miRNA is directly transcribed by RNAP III from individual, compact RNAP III type II genes. Thus, similar to manmade RNAP III-generated short hairpin RNAs (shRNAs), the BLV pre-miRNAs are initially 5'-triphosphorylated. Nonetheless, the derivative 5p miRNAs and shRNA-generated 5p small RNAs (sRNAs) possess a 5'-monophosphate. Our enzymatic characterization and small RNA sequencing data demonstrate that BLV 5p miRNAs are co-terminal with 5'-triphosphorylated miRNA precursors (pre-miRNAs). Thus, these results identify a 5'-tri-phosphatase activity that is involved in the biogenesis of BLV miRNAs and shRNA-generated sRNAs. This work advances our understanding of retroviral miRNA and shRNA biogenesis and may have implications regarding the immunostimulatory capacity of RNAP III transcripts.


Asunto(s)
Ácido Anhídrido Hidrolasas/metabolismo , Virus de la Leucemia Bovina/genética , ARN Polimerasa III/metabolismo , ARN Interferente Pequeño/biosíntesis , Células HEK293 , Humanos , MicroARNs/biosíntesis , MicroARNs/química , MicroARNs/metabolismo , Fosfatos/análisis , Precursores del ARN/biosíntesis , ARN Interferente Pequeño/metabolismo , ARN Pequeño no Traducido/química , ARN Pequeño no Traducido/metabolismo , Ribonucleasa III/metabolismo
10.
J Virol ; 88(21): 12683-93, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25142594

RESUMEN

UNLABELLED: Several different polyomaviruses (PyVs) encode microRNAs (miRNAs) that regulate viral as well as host gene expression. However, the functions of polyomaviral miRNAs, particularly during in vivo infection, remain poorly understood. Here we identify rare naturally arising PyVs that are severely attenuated or null for miRNA expression. We identify hypomorphic or null strains for miRNA expression from rhesus macaque simian virus 40 (SV40) and human JC virus. These strains were isolated from immunocompromised hosts and derive from insertions or deletions in the viral DNA that preserve the amino acid reading frame of opposing-strand large T antigen gene. Characterization of the SV40 miRNA hypomorph, K661, shows that it is inhibited at the early miRNA biogenesis step of Drosha-mediated processing. Despite having a nonrearranged enhancer, which a previous study has shown renders some PyVs more susceptible to the autoregulatory activities of the miRNA, restoring miRNA expression to K661 has little effect on virus growth in either immortalized or primary monkey kidney cells. Thus, in addition to any effect of accompanying genomic elements, these results suggest that the cellular context also determines susceptibility to PyV miRNA-mediated effects. Combined, these results demonstrate that polyomaviruses lacking miRNAs can arise infrequently and that the functional importance of polyomaviral miRNAs is context dependent, consistent with an activity connected to the immune status of the host. IMPORTANCE: Diverse virus families encode miRNAs, yet much remains unknown about viral miRNA function and contribution to the infectious cycle. Polyomaviruses (PyVs) are small DNA viruses, long known to be important as etiological agents of rare diseases and valuable models of DNA virus infection. Here, in immunosuppressed hosts, we uncover rare naturally arising variants of different PyVs that have lost the ability to express miRNAs. This represents some of the only known natural viruses to have lost miRNA expression. By probing the biogenesis pathways of these variants, we uncover that miRNA expression is lost via small insertions or deletions that render the transcripts resistant to early steps of miRNA biogenesis while preserving the reading frame of the opposing T antigen transcripts. Overall, our study informs how miRNA genes evolve/devolve in viruses and suggests that miRNA function is exquisitely dependent not only on viral genomic context but also on the cellular and host environment.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus JC/genética , MicroARNs/biosíntesis , Infecciones por Polyomavirus/veterinaria , Infecciones por Polyomavirus/virología , Virus 40 de los Simios/genética , Animales , Línea Celular , Humanos , Huésped Inmunocomprometido , Virus JC/aislamiento & purificación , Virus JC/fisiología , Macaca mulatta , MicroARNs/genética , Mutagénesis Insercional , Eliminación de Secuencia , Virus 40 de los Simios/aislamiento & purificación , Virus 40 de los Simios/fisiología , Replicación Viral
11.
PLoS Pathog ; 9(12): e1003818, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24367263

RESUMEN

Torque teno viruses (TTVs) are a group of viruses with small, circular DNA genomes. Members of this family are thought to ubiquitously infect humans, although causal disease associations are currently lacking. At present, there is no understanding of how infection with this diverse group of viruses is so prevalent. Using a combined computational and synthetic approach, we predict and identify miRNA-coding regions in diverse human TTVs and provide evidence for TTV miRNA production in vivo. The TTV miRNAs are transcribed by RNA polymerase II, processed by Drosha and Dicer, and are active in RISC. A TTV mutant defective for miRNA production replicates as well as wild type virus genome; demonstrating that the TTV miRNA is dispensable for genome replication in a cell culture model. We demonstrate that a recombinant TTV genome is capable of expressing an exogenous miRNA, indicating the potential utility of TTV as a small RNA vector. Gene expression profiling of host cells identifies N-myc (and STAT) interactor (NMI) as a target of a TTV miRNA. NMI transcripts are directly regulated through a binding site in the 3'UTR. SiRNA knockdown of NMI contributes to a decreased response to interferon signaling. Consistent with this, we show that a TTV miRNA mediates a decreased response to IFN and increased cellular proliferation in the presence of IFN. Thus, we add Annelloviridae to the growing list of virus families that encode miRNAs, and suggest that miRNA-mediated immune evasion can contribute to the pervasiveness associated with some of these viruses.


Asunto(s)
Interferón Tipo I/genética , MicroARNs/fisiología , Torque teno virus/genética , Anelloviridae/genética , Células Cultivadas , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Perfilación de la Expresión Génica , Variación Genética , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Interferón Tipo I/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/genética
12.
Proc Natl Acad Sci U S A ; 109(8): 3077-82, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22308400

RESUMEN

MicroRNAs (miRNAs) are small RNAs that play a regulatory role in numerous and diverse eukaryotic cellular processes. Virus-encoded miRNAs have garnered much interest, although the functions of most remain to be deciphered. To date, readily detectable, evolutionarily conserved natural miRNAs have only been identified from viruses with DNA genomes. Combined with the fact that most miRNAs are generated from endonucleolytic cleavage of longer transcripts, this finding has led to a common conception that naturally occurring RNA viruses will not encode miRNAs to avoid unproductive cleavage of their genomes or mRNAs. Here we demonstrate that the bovine leukemia virus (BLV), a retrovirus with an RNA genome, encodes a conserved cluster of miRNAs that are transcribed by RNA polymerase III (pol III). Thus, the BLV miRNAs avoid the conundrum of genome/mRNA cleavage because only the subgenomic pol III transcripts are efficiently processed into miRNAs. BLV infection is strongly associated with B-cell tumors in cattle. Because most cells in BLV-associated tumors express little viral mRNAs or proteins, exactly how BLV contributes to tumorigenesis has remained a decades-long unsolved mystery. One BLV miRNA, BLV-miR-B4, shares partial sequence identity and shared common targets with the host miRNA, miR-29. As miR-29 overexpression is associated with B-cell neoplasms that resemble BLV-associated tumors, our findings suggest a possible mechanism contributing to BLV-induced tumorigenesis.


Asunto(s)
Linfocitos B/metabolismo , Virus de la Leucemia Bovina/genética , MicroARNs/genética , Neoplasias/genética , Animales , Secuencia de Bases , Northern Blotting , Bovinos , Biología Computacional , Células HEK293 , Humanos , Datos de Secuencia Molecular , ARN Polimerasa III/metabolismo , Procesamiento Postranscripcional del ARN/genética , Ribonucleasa III/metabolismo , Transcripción Genética
13.
J Virol ; 87(20): 11135-47, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23926342

RESUMEN

Hundreds of virus-encoded microRNAs (miRNAs) have been uncovered, but an in-depth functional understanding is lacking for most. A major challenge for the field is separating those miRNA targets that are biologically relevant from those that are not advantageous to the virus. Here, we show that miRNAs from related variants of the polyomavirus simian vacuolating virus 40 (SV40) have differing host target repertoires (targetomes) while their direct autoregulatory activity on virus-encoded early gene products is completely preserved. These results underscore the importance of miRNA-mediated viral gene autoregulation in some polyomavirus life cycles. More broadly, these findings imply that some host targets of virus-encoded miRNAs are likely to be of little selective advantage to the virus, and our approach provides a strategy for prioritizing relevant targets.


Asunto(s)
Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , MicroARNs/genética , MicroARNs/metabolismo , Virus 40 de los Simios/genética , Animales , Línea Celular , Humanos , ARN Viral/genética , ARN Viral/metabolismo , Virus 40 de los Simios/fisiología
14.
J Virol ; 87(23): 12838-49, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24067953

RESUMEN

3' untranslated regions (UTRs) are known to play an important role in posttranscriptional regulation of gene expression. Here we map the 3' UTRs of Kaposi's sarcoma-associated herpesvirus (KSHV) using next-generation RNA sequencing, 3' rapid amplification of cDNA ends (RACE), and tiled microarray analyses. Chimeric reporters containing the KSHV 3' UTRs show a general trend toward reduced gene expression under conditions of latent infection. Those 3' UTRs with a higher GC content are more likely to be associated with reduced gene expression. KSHV transcripts display an extensive use of shared polyadenylation sites allowing for partially overlapping 3' UTRs and regulatory activities. In addition, a subset of KSHV 3' UTRs is sufficient to convey increased gene expression under conditions of lytic infection. These results suggest a role for viral 3' UTRs in contributing to differential gene expression during latent versus lytic infection.


Asunto(s)
Regiones no Traducidas 3' , Regulación Viral de la Expresión Génica , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/fisiología , Proteínas Virales/genética , Latencia del Virus , Línea Celular , Herpesvirus Humano 8/genética , Humanos , Regiones Promotoras Genéticas , Proteínas Virales/metabolismo , Replicación Viral
15.
PLoS Pathog ; 8(12): e1003018, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23308061

RESUMEN

MicroRNAs (miRNAs) are small RNAs that play important roles in the regulation of gene expression. First described as posttranscriptional gene regulators in eukaryotic hosts, virus-encoded miRNAs were later uncovered. It is now apparent that diverse virus families, most with DNA genomes, but at least some with RNA genomes, encode miRNAs. While deciphering the functions of viral miRNAs has lagged behind their discovery, recent functional studies are bringing into focus these roles. Some of the best characterized viral miRNA functions include subtle roles in prolonging the longevity of infected cells, evading the immune response, and regulating the switch to lytic infection. Notably, all of these functions are particularly important during persistent infections. Furthermore, an emerging view of viral miRNAs suggests two distinct groups exist. In the first group, viral miRNAs mimic host miRNAs and take advantage of conserved networks of host miRNA target sites. In the larger second group, viral miRNAs do not share common target sites conserved for host miRNAs, and it remains unclear what fraction of these targeted transcripts are beneficial to the virus. Recent insights from multiple virus families have revealed new ways of interacting with the host miRNA machinery including noncanonical miRNA biogenesis and new mechanisms of posttranscriptional cis gene regulation. Exciting challenges await the field, including determining the most relevant miRNA targets and parlaying our current understanding of viral miRNAs into new therapeutic strategies. To accomplish these goals and to better grasp miRNA function, new in vivo models that recapitulate persistent infections associated with viral pathogens are required.


Asunto(s)
Regulación Viral de la Expresión Génica , MicroARNs/genética , ARN Viral/genética , Virus/genética , Humanos
16.
RNA ; 16(8): 1540-58, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20566670

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumor virus that encodes 12 precursor microRNAs (pre-miRNAs) that give rise to 17 different known approximately 22-nucleotide (nt) effector miRNAs. Like all herpesviruses, KSHV has two modes of infection: (1) a latent mode whereby only a subset of viral genes are expressed and (2) a lytic mode during which the full remaining viral genes are expressed. To date, KSHV miRNAs have been mostly identified via analysis of cells that are undergoing latent infection. Here, we developed a method to profile small RNAs ( approximately 18-75 nt) from populations of cells undergoing predominantly lytic infection. Using two different next-generation sequencing platforms, we cloned and sequenced both pre-miRNAs and derivative miRNAs. Our analysis shows that the vast majority of viral and host 5p miRNAs are co-terminal with the 5' end of the cloned pre-miRNAs, consistent with both being defined by microprocessor cleavage. We report the complete repertoire (25 total) of 5p and 3p derivative miRNAs from all 12 previously described KSHV pre-miRNAs. Two KSHV pre-miRNAs, pre-miR-K12-8 and pre-miR-K12-12, encode abundant derivative miRNAs from the previously unreported strands of the pre-miRNA. We identify several novel small RNAs of low abundance, including viral miRNA-offset-RNAs (moRNAs), and antisense viral miRNAs (miRNA-AS) that are encoded antisense to previously reported KSHV pre-miRNAs. Finally, we observe widespread antisense transcription relative to known coding sequences during lytic replication. Despite the enormous potential to form double-stranded RNA in KSHV-infected cells, we observe no evidence for the existence of abundant viral-derived small interfering RNAs (siRNAs).


Asunto(s)
Herpesvirus Humano 8/genética , MicroARNs/genética , ARN Viral/genética , Secuencia de Bases , Genes Virales , Genoma , Humanos , Sistemas de Lectura Abierta , ARN/genética , ARN Bicatenario/genética , ARN no Traducido/genética
17.
J Virol ; 85(9): 4487-500, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21345962

RESUMEN

Several different members of the Polyomaviridae, including some human pathogens, encode microRNAs (miRNAs) that lie antisense with respect to the early gene products, the tumor (T) antigens. These miRNAs negatively regulate T antigen expression by directing small interfering RNA (siRNA)-like cleavage of the early transcripts. miRNA mutant viruses of some members of the Polyomaviridae express increased levels of early proteins during lytic infection. However, the importance of miRNA-mediated negative regulation of the T antigens remains uncertain. Bandicoot papillomatosis carcinomatosis virus type 1 (BPCV1) is associated with papillomas and carcinomas in the endangered marsupial the western barred bandicoot (Perameles bougainville). BPCV1 is the founding member of a new group of viruses that remarkably share distinct properties in common with both the polyomavirus and papillomavirus families. Here, we show that BPCV1 encodes, in the same orientation as the papillomavirus-like transcripts, a miRNA located within a long noncoding region (NCR) of the genome. Furthermore, this NCR serves the function of both promoter and template for the primary transcript that gives rise to the miRNA. Unlike the polyomavirus miRNAs, the BPCV1 miRNA is not encoded antisense to the T antigen transcripts but rather lies in a separate, proximal region of the genome. We have mapped the 3' untranslated region (UTR) of the BPCV1 large T antigen early transcript and identified a functional miRNA target site that is imperfectly complementary to the BPCV1 miRNA. Chimeric reporters containing the entire BPCV1 T antigen 3' UTR undergo negative regulation when coexpressed with the BPCV1 miRNA. Notably, the degree of negative regulation observed is equivalent to that of an identical reporter that is engineered to bind to the BPCV1 miRNA with perfect complementarity. We also show that this miRNA and this novel mode of early gene regulation are conserved with the related BPCV2. Finally, papillomatous lesions from a western barred bandicoot express readily detectable levels of this miRNA, stressing its likely importance in vivo. Combined, the alternative mechanisms of negative regulation of T antigen expression between the BPCVs and the polyomaviruses support the importance of miRNA-mediated autoregulation in the life cycles of some divergent polyomaviruses and polyomavirus-like viruses.


Asunto(s)
Regulación Viral de la Expresión Génica , MicroARNs/metabolismo , Polyomaviridae/fisiología , Infecciones por Polyomavirus/veterinaria , ARN Viral/metabolismo , Replicación Viral , Animales , Antígenos Transformadores de Poliomavirus/genética , Sitios de Unión , Marsupiales/virología , Polyomaviridae/aislamiento & purificación , Infecciones por Polyomavirus/virología , ARN Complementario/genética
19.
Virology ; 513: 180-187, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29096160

RESUMEN

Mouse mammary tumor virus (MMTV) induces breast cancer in mice in the absence of known virally-encoded oncogenes. Tumorigenesis by MMTV is thought to occur primarily through insertional mutagenesis, leading to the activation of cellular proto-oncogenes and outgrowth of selected cells. Here we investigated whether MMTV encodes microRNAs (miRNAs) and/or modulates host miRNAs that could contribute to tumorigenesis. High throughput small RNA sequencing analysis of MMTV-infected cells and MMTV-induced mammary tumors demonstrates that MMTV does not encode miRNAs. However, infected tissues have altered levels of several host miRNAs, including increased expression of members of the oncogenic miRNA cluster, miR-17-92. Notably, similar changes in miRNA levels have been previously reported in human breast cancers. Combined, our results demonstrate that virally encoded miRNAs do not contribute to MMTV-mediated tumorigenesis, but that changes in specific host miRNAs in infected cells may contribute to virus replication and tumor biology.


Asunto(s)
Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Neoplasias Mamarias Experimentales/virología , Virus del Tumor Mamario del Ratón/fisiología , MicroARNs/análisis , Animales , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones
20.
mSphere ; 1(2)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27301787

RESUMEN

Despite increasing interest in the biology of noncoding RNAs (ncRNAs), few functions have been uncovered for viral ncRNAs in vivo. In their recent article in mSphere, Feldman and colleagues [E. R. Feldman et al., mSphere 1(2):e00105-15, 2016, doi:10.1128/mSphere.00105-15] demonstrate a highly specific activity of a gammaherpesviral ncRNA in viral dissemination and establishment of latent infection. Their work highlights several interesting features that should be informative to future studies of viral ncRNA.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA