Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros











Intervalo de año de publicación
1.
RNA ; 27(10): 1140-1147, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34253686

RESUMEN

Human metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a nuclear long noncoding RNA (lncRNA) that is highly overexpressed in many cancer tissues and plays important roles in tumor progression and metastasis. The MALAT1 primary transcript contains evolutionarily conserved structural elements in its 3'-terminal region: a triple helix forming element called element for nuclear expression (ENE) and a downstream tRNA-like structure called mascRNA. Instead of being polyadenylated, mature MALAT1 is generated by recognition and processing of the mascRNA by RNase P. A genomically encoded A-rich tract at the new 3' end of MALAT1, which is generated upon RNase P cleavage, forms a triple helical structure with the upstream ENE. Triplex formation is vital for stabilization of the mature transcript and for subsequent accumulation and oncogenic activity of MALAT1. Here, we demonstrate that efficient 3'-end maturation of MALAT1 is dependent on an interaction between the A-rich tract and the mascRNA 3' trailer. Using mutational analyses of cell-based reporter accumulation, we show that an extended mascRNA acceptor stem and formation of a single bulged A 5' to the RNase P cleavage site are required for efficient maturation of the nascent MALAT1 3' end. Our results should benefit the development of therapeutic approaches to cancer through targeting MALAT1.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Neoplasias Pulmonares/genética , Procesamiento de Término de ARN 3' , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN de Transferencia/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Emparejamiento Base , Secuencia de Bases , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Conformación de Ácido Nucleico , Estabilidad del ARN , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , ARN de Transferencia/metabolismo , Ribonucleasa P/genética , Ribonucleasa P/metabolismo
2.
J Vis Exp ; (150)2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31524859

RESUMEN

Mechanistic insight arrives from careful study and quantification of specific RNAs and proteins. The relative locations of these biomolecules throughout the cell at specific times can be captured with fluorescence in situ hybridization (FISH) and immunofluorescence (IF). During lytic herpesvirus infection, the virus hijacks the host cell to preferentially express viral genes, causing changes in cell morphology and behavior of biomolecules. Lytic activities are centered in nuclear factories, termed viral replication compartments, which are discernable only with FISH and IF. Here we describe an adaptable protocol of RNA FISH and IF techniques for Kaposi's sarcoma-associated herpesvirus (KSHV)-infected cells, both adherent and in suspension. The method includes steps for the development of specific anti-sense oligonucleotides, double RNA FISH, RNA FISH with IF, and quantitative calculations of fluorescence intensities. This protocol has been successfully applied to multiple cell types, uninfected cells, latent cells, lytic cells, time-courses, and cells treated with inhibitors to analyze the spatiotemporal activities of specific RNAs and proteins from both the human host and KSHV.


Asunto(s)
Infecciones por Herpesviridae/genética , Herpesvirus Humano 8/fisiología , Hibridación Fluorescente in Situ/métodos , Línea Celular , Núcleo Celular/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , Humanos , Proteínas Nucleares/metabolismo , Replicación Viral/genética
3.
PLoS Pathog ; 14(11): e1007389, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30383841

RESUMEN

During lytic replication of Kaposi's sarcoma-associated herpesvirus (KSHV), a nuclear viral long noncoding RNA known as PAN RNA becomes the most abundant polyadenylated transcript in the cell. Knockout or knockdown of KSHV PAN RNA results in loss of late lytic viral gene expression and, consequently, reduction of progeny virion release from the cell. Here, we demonstrate that knockdown of PAN RNA from the related Rhesus macaque rhadinovirus (RRV) phenocopies that of KSHV PAN RNA. These two PAN RNA homologs, although lacking significant nucleotide sequence conservation, can functionally substitute for each other to rescue phenotypes associated with the absence of PAN RNA expression. Because PAN RNA is exclusively nuclear, previous studies suggested that it directly interacts with host and viral chromatin to modulate gene expression. We studied KSHV and RRV PAN RNA homologs using capture hybridization analysis of RNA targets (CHART) and observed their association with host chromatin, but the loci differ between PAN RNA homologs. Accordingly, we find that KSHV PAN RNA is undetectable in chromatin following cell fractionation. Thus, modulation of gene expression at specific chromatin loci appears not to be the primary, nor the pertinent function of this viral long noncoding RNA. PAN RNA represents a cautionary tale for the investigation of RNA association with chromatin whereby cross-linking of DNA spatially adjacent to an abundant nuclear RNA gives the appearance of specific interactions. Similarly, PAN RNA expression does not affect viral transcription factor complex expression or activity, which is required for generation of the late lytic viral mRNAs. Rather, we provide evidence for an alternative model of PAN RNA function whereby knockdown of KSHV or RRV PAN RNA results in compromised nuclear mRNA export thereby reducing the cytoplasmic levels of viral mRNAs available for production of late lytic viral proteins.


Asunto(s)
ARN Largo no Codificante/genética , Rhadinovirus/genética , Animales , Línea Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Regulación Viral de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen/métodos , Células HEK293 , Herpesviridae/genética , Infecciones por Herpesviridae/genética , Herpesvirus Humano 8/genética , Interacciones Huésped-Patógeno , Humanos , Macaca mulatta/virología , ARN Mensajero/genética , ARN Nuclear/genética , ARN Viral/genética , Infecciones Tumorales por Virus , Proteínas Virales/metabolismo , Replicación Viral
4.
Hum Genomics ; 12(1): 32, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29945683

RESUMEN

Naturally occurring stress-induced transcriptional readthrough is a recently discovered phenomenon, in which stress conditions lead to dramatic induction of long transcripts as a result of transcription termination failure. In 2015, we reported the induction of such downstream of gene (DoG) containing transcripts upon osmotic stress in human cells, while others observed similar transcripts in virus-infected and cancer cells. Using the rigorous methodology Cap-Seq, we demonstrated that DoGs result from transcriptional readthrough, not de novo initiation. More recently, we presented a genome-wide comparison of NIH3T3 mouse cells subjected to osmotic, heat, and oxidative stress and concluded that massive induction of transcriptional readthrough is a hallmark of the mammalian stress response. In their recent letter, Huang and Liu in contrast claim that DoG transcripts result from novel transcription initiation near the ends of genes. Their conclusions rest on analyses of a publicly available transcription start site (TSS-Seq) dataset from unstressed NIH3T3 cells. Here, we present evidence that this dataset identifies not only true transcription start sites, TSSs, but also 5'-ends of numerous snoRNAs, which are generally processed from introns in mammalian cells. We show that failure to recognize these erroneous assignments in the TSS-Seq dataset, as well as ignoring published Cap-Seq data on TSS mapping during osmotic stress, have led to misinterpretation by Huang and Liu. We conclude that, contrary to the claims made by Huang and Liu, TSS-Seq reads near gene ends cannot explain the existence of DoGs, nor their stress-mediated induction. Rather it is, as we originally demonstrated, transcriptional readthrough that leads to the formation of DoGs.


Asunto(s)
ARN Nucleolar Pequeño/genética , Estrés Fisiológico/genética , Sitio de Iniciación de la Transcripción , Transcriptoma/genética , Animales , Regulación de la Expresión Génica/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas
5.
J Virol ; 92(13)2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29643239

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV), like other herpesviruses, replicates within the nuclei of its human cell host and hijacks host machinery for expression of its genes. The activities that culminate in viral DNA synthesis and assembly of viral proteins into capsids physically concentrate in nuclear areas termed viral replication compartments. We sought to better understand the spatiotemporal regulation of viral RNAs during the KSHV lytic phase by examining and quantifying the subcellular localization of select viral transcripts. We found that viral mRNAs, as expected, localized to the cytoplasm throughout the lytic phase. However, dependent on active viral DNA replication, viral transcripts also accumulated in the nucleus, often in foci in and around replication compartments, independent of the host shutoff effect. Our data point to involvement of the viral long noncoding polyadenylated nuclear (PAN) RNA in the localization of an early, intronless viral mRNA encoding ORF59-58 to nuclear foci that are associated with replication compartments.IMPORTANCE Late in the lytic phase, mRNAs from Kaposi's sarcoma-associated herpesvirus accumulate in the host cell nucleus near viral replication compartments, centers of viral DNA synthesis and virion production. This work contributes spatiotemporal data on herpesviral mRNAs within the lytic host cell and suggests a mechanism for viral RNA accumulation. Our findings indicate that the mechanism is independent of the host shutoff effect and splicing but dependent on active viral DNA synthesis and in part on the viral noncoding RNA, PAN RNA. PAN RNA is essential for the viral life cycle, and its contribution to the nuclear accumulation of viral messages may facilitate propagation of the virus.


Asunto(s)
Núcleo Celular/metabolismo , Replicación del ADN , ADN Viral/metabolismo , Poli A/metabolismo , ARN Mensajero/metabolismo , ARN Nuclear/metabolismo , ARN no Traducido/metabolismo , Núcleo Celular/genética , Células Cultivadas , ADN Viral/genética , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/fisiología , Humanos , Poli A/genética , ARN Mensajero/genética , ARN Nuclear/genética , ARN no Traducido/genética , ARN Viral/genética , ARN Viral/metabolismo , Sarcoma de Kaposi/virología , Replicación Viral
6.
Bio Protoc ; 8(3)2018 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-29527542

RESUMEN

Cellular quiescence (also known as G0 arrest) is characterized by reduced DNA replication, increased autophagy, and increased expression of cyclin-dependent kinase p27Kip1. Quiescence is essential for wound healing, organ regeneration, and preventing neoplasia. Previous findings indicate that microRNAs (miRNAs) play an important role in regulating cellular quiescence. Our recent publication demonstrated the existence of an alternative miRNA biogenesis pathway in primary human foreskin fibroblast (HFF) cells during quiescence. Indeed, we have identified a group of pri-miRNAs (whose mature miRNAs were found induced during quiescence) modified with a 2,2,7-trimethylguanosine (TMG)-cap by the trimethylguanosine synthase 1 (TGS1) protein and transported to the cytoplasm by the Exportin-1 (XPO1) protein. We used an antibody against (TMG)-caps (which does not cross-react with the (m7G)-caps that most pri-miRNAs or mRNAs contain [Luhrmann et al., 1982]) to perform RNA immunoprecipitations from total RNA extracts of proliferating or quiescent HFFs. The novelty of this assay is the specific isolation of pri-miRNAs as well as other non-coding RNAs containing a TMG-cap modification.

7.
Genes Dev ; 31(10): 957-958, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28637691

RESUMEN

Post-transcriptional modification of RNA nucleosides has been implicated as a pivotal regulator of mRNA biology. In this issue of Genes & Development, Ke and colleagues (pp. 990-1006) provide insights into the temporal and spatial distribution of N6-methyladenosine (m6A) in RNA transcripts by analyzing different subcellular fractions. Using a recently developed biochemical approach for detecting m6A, the researchers show that m6A methylations are enriched in exons and are added to transcripts prior to splicing. Although m6A addition is widely thought to be readily reversible, they demonstrate in HeLa cells that once RNA is released from chromatin, the modifications are surprisingly static. This study integrates data from previous publications to clarify conflicting conclusions regarding the role of m6A in mRNA biogenesis and function. Ke and colleagues found that m6A methylation levels negatively correlate with transcript half-life but are not required for most pre-mRNA splicing events.


Asunto(s)
Adenosina/metabolismo , Empalme del ARN/fisiología , ARN Mensajero/metabolismo , Adenosina/análisis , Animales , Exones/genética , Células HeLa , Humanos , Metilación , Metiltransferasas/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(25): E4961-E4970, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28584122

RESUMEN

The reversible state of proliferative arrest known as "cellular quiescence" plays an important role in tissue homeostasis and stem cell biology. By analyzing the expression of miRNAs and miRNA-processing factors during quiescence in primary human fibroblasts, we identified a group of miRNAs that are induced during quiescence despite markedly reduced expression of Exportin-5, a protein required for canonical miRNA biogenesis. The biogenesis of these quiescence-induced miRNAs is independent of Exportin-5 and depends instead on Exportin-1. Moreover, these quiescence-induced primary miRNAs (pri-miRNAs) are modified with a 2,2,7-trimethylguanosine (TMG)-cap, which is known to bind Exportin-1, and knockdown of Exportin-1 or trimethylguanosine synthase 1, responsible for (TMG)-capping, inhibits their biogenesis. Surprisingly, in quiescent cells Exportin-1-dependent pri-miR-34a is present in the cytoplasm together with a small isoform of Drosha, implying the existence of a different miRNA processing pathway in these cells. Our findings suggest that during quiescence the canonical miRNA biogenesis pathway is down-regulated and specific miRNAs are generated by an alternative pathway to regulate genes involved in cellular growth arrest.


Asunto(s)
Vías Biosintéticas/genética , Proliferación Celular/genética , Carioferinas/genética , MicroARNs/genética , Receptores Citoplasmáticos y Nucleares/genética , Vías Biosintéticas/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citoplasma/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Guanosina/análogos & derivados , Guanosina/farmacología , Células HEK293 , Células HeLa , Humanos , Proteína Exportina 1
9.
RNA Biol ; 14(5): 632-636, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-26861889

RESUMEN

In recent years, the realization that most of the genome is transcribed has transformed the study of mammalian gene expression. Much effort has gone into investigating how this pervasive transcription is regulated and what the functions of the resulting transcripts are, if any. We recently discovered that stress-induced transcriptional readthrough generates very long downstream of gene containing transcripts (DoGs), which may explain up to 20% of intergenic transcription. DoGs are induced by osmotic stress at the level of transcription by a mechanism that depends on calcium release from the endoplasmic reticulum mediated by IP3 receptors. Here, we discuss DoG induction and function in the context of the literature, with special focus on 2 outstanding questions. First, we discuss possible molecular mechanisms underlying DoG induction through reduced transcription termination. Second, we explore how DoGs may function in maintaining euchromatin after nuclear scaffold stress. In short, we review important aspects of DoG biogenesis and function, and provide an outlook for continued DoG study.


Asunto(s)
ARN Largo no Codificante , Transcripción Genética , Animales , Calcio/metabolismo , Línea Celular Tumoral , Eucromatina/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Ratones , Matriz Nuclear , Presión Osmótica , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Análisis de Secuencia de ARN , Terminación de la Transcripción Genética
10.
Proc Natl Acad Sci U S A ; 113(49): 14013-14018, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27872311

RESUMEN

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a cancer-promoting long noncoding RNA, accumulates in cells by using a 3'-triple-helical RNA stability element for nuclear expression (ENE). The ENE, a stem-loop structure containing a U-rich internal loop, interacts with a downstream A-rich tract (ENE+A) to form a blunt-ended triple helix composed of nine U•A-U triples interrupted by a C•G-C triple and C-G doublet. This unique structure prompted us to explore the possibility of protein binding. Native gel-shift assays revealed a shift in radiolabeled MALAT1 ENE+A RNA upon addition of HEK293T cell lysate. Competitive gel-shift assays suggested that protein binding depends not only on the triple-helical structure but also its nucleotide composition. Selection from the lysate using a biotinylated-RNA probe followed by mass spectrometry identified methyltransferase-like protein 16 (METTL16), a putative RNA methyltransferase, as an interacting protein of the MALAT1 ENE+A. Gel-shift assays confirmed the METTL16-MALAT1 ENE+A interaction in vitro: Binding was observed with recombinant METTL16, but diminished in lysate depleted of METTL16, and a supershift was detected after adding anti-METTL16 antibody. Importantly, RNA immunoprecipitation after in vivo UV cross-linking and an in situ proximity ligation assay for RNA-protein interactions confirmed an association between METTL16 and MALAT1 in cells. METTL16 is an abundant (∼5 × 105 molecules per cell) nuclear protein in HeLa cells. Its identification as a triple-stranded RNA binding protein supports the formation of RNA triple helices inside cells and suggests the existence of a class of triple-stranded RNA binding proteins, which may enable the discovery of additional cellular RNA triple helices.


Asunto(s)
Metiltransferasas/metabolismo , ARN Largo no Codificante/metabolismo , Células HEK293 , Células HeLa , Humanos , Metiltransferasas/genética , Conformación de Ácido Nucleico , Pliegue del ARN/fisiología , Estabilidad del ARN , ARN Largo no Codificante/genética
11.
RNA ; 22(8): 1181-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27335146

RESUMEN

Herpesvirus saimiri, an oncogenic herpesvirus, during latency produces seven small nuclear RNAs, called the Herpesvirus saimiri U RNAs (HSUR1-7). HSUR1 mediates degradation of the host microRNA, miR-27, via a process that requires imperfect base-pairing. The decreased levels of miR-27 lead to prolonged T-cell activation and likely contribute to oncogenesis. To gain insight into HSUR1-mediated degradation of miR-27, we probed the in vivo secondary structure of HSUR1 and coupled this with bioinformatic structural analyses. The results suggest that HSUR1 adopts a conformation different than previously believed and that the region complementary to miR-27 lacks stable structure. To determine whether HSUR1 structural flexibility is important for its ability to mediate miR-27 degradation, we performed structurally informative mutagenic analyses of HSUR1. HSUR1 mutants in which the miR-27 binding site sequence is preserved, but sequestered in predicted helices, lose their ability to decrease miR-27 levels. These results indicate that the HSUR1 miR27-binding region must be available in a conformationally flexible segment for noncoding RNA function.


Asunto(s)
Genes Virales , Herpesvirus Saimiriino 2/metabolismo , MicroARNs/metabolismo , ARN Nuclear Pequeño/metabolismo , Animales , Callithrix , Herpesvirus Saimiriino 2/genética
12.
RNA ; 22(5): 743-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26952103

RESUMEN

Triple-stranded RNA was first deduced to form in vitro more than 50 years ago and has since been implicated in RNA catalysis, stability, and small molecule binding. Despite the emerging biological significance of RNA triple helices, it remains unclear how their nucleotide composition contributes to their thermodynamic stability and cellular function. To investigate these properties, we used in vitro RNA electrophoretic mobility shift assays (EMSAs) and in vivo intronless ß-globin reporter assays to measure the relative contribution of 20 RNA base triples (N•A-U, N•G-C, N•C-G, N•U-A, and N•G-U) to triple-helical stability. These triples replaced a single internal U•A-U within the known structure of the triple-helical RNA stability element of human metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), which contains 10 major-groove base triples. In addition to the canonical C•G-C triple, the noncanonical base triples U•G-C, U•G-U, C•C-G, and U•C-G exhibited at least 30% stability relative to the wild-type U•A-U base triple in both assays. Of these triples, only U•A-U, C•G-C, and U•G-C, when tested as four successive triples, formed stabilizing structures that allowed accumulation of the intronless ß-globin reporter. Overall, we find that Hoogsteen-position pyrimidines support triple helix stability and function and that thermodynamic stability, based on EMSA results, is necessary but not sufficient for stabilization activity of the MALAT1 triple helix in cells. These results suggest that additional RNA triple helices containing noncanonical triples likely exist in nature.


Asunto(s)
Pirimidinas/química , ARN Largo no Codificante/genética , Termodinámica
13.
Proc Natl Acad Sci U S A ; 113(12): 3221-6, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26951683

RESUMEN

Epstein-Barr virus (EBV) produces a highly abundant noncoding RNA called EBV-encoded RNA 2 (EBER2) that interacts indirectly with the host transcription factor paired box protein 5 (PAX5) to regulate viral latent membrane protein 1/2 (LMP1/2) gene expression as well as EBV lytic replication. To identify intermediary proteins, we isolated EBER2-PAX5-containing complexes and analyzed the protein components by mass spectrometry. The top candidates include three host proteins splicing factor proline and glutamine rich (SFPQ), non-POU domain-containing octamer-binding protein (NONO), and RNA binding motif protein 14 (RBM14), all reported to be components of nuclear bodies called paraspeckles. In vivo RNA-protein crosslinking indicates that SFPQ and RBM14 contact EBER2 directly. Binding studies using recombinant proteins demonstrate that SFPQ and NONO associate with PAX5, potentially bridging its interaction with EBER2. Similar to EBER2 or PAX5 depletion, knockdown of any of the three host RNA-binding proteins results in the up-regulation of viral LMP2A mRNA levels, supporting a physiologically relevant interaction of these newly identified factors with EBER2 and PAX5. Identification of these EBER2-interacting proteins enables the search for cellular noncoding RNAs that regulate host gene expression in a manner similar to EBER2.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , ARN Viral/genética , Proteínas de Unión al ARN/metabolismo , Genes Virales , Células HEK293 , Humanos , Unión Proteica , ARN Viral/metabolismo
14.
J Virol ; 89(21): 10901-11, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26292323

RESUMEN

UNLABELLED: In latently infected marmoset T cells, Herpesvirus saimiri (HVS) expresses six microRNAs (known as miR-HSURs [H. saimiri U-rich RNAs]). The viral miR-HSURs are processed from chimeric primary transcripts, each containing a noncoding U-rich RNA (HSUR) and a pre-miRNA hairpin. To uncover the functions of miR-HSURs, we identified mRNA targets in infected cells using high-throughput sequencing of RNA isolated by cross-linking immunoprecipitation (HITS-CLIP). HITS-CLIP revealed hundreds of robust Argonaute (Ago) binding sites mediated by miR-HSURs that map to the host genome but few in the HVS genome. Gene ontology analysis showed that several pathways regulating the cell cycle are enriched among cellular targets of miR-HSURs. Interestingly, miR-HSUR4-3p represses expression of the p300 transcriptional coactivator by binding the open reading frame of its mRNA. miR-HSUR5-3p directly regulates BiP, an endoplasmic reticulum (ER)-localized chaperone facilitating maturation of major histocompatibility complex class I (MHC-I) and the antiviral response. miR-HSUR5-3p also robustly downregulates WEE1, a key negative regulator of cell cycle progression, leading to reduced phosphorylation of its substrate, cyclin-dependent kinase (Cdk1). Consistently, inhibition of miR-HSUR5-3p in HVS-infected cells decreases their proliferation. Together, our results shed light on the roles of viral miRNAs in cellular transformation and viral latency. IMPORTANCE: Viruses express miRNAs during various stages of infection, suggesting that viral miRNAs play critical roles in the viral life cycle. Compared to protein-coding genes, the functions of viral miRNAs are not well understood. This is because it has been challenging to identify their mRNA targets. Here, we focused on the functions of the recently discovered HVS miRNAs, called miR-HSURs. HVS is an oncogenic gammaherpesvirus that causes acute T-cell lymphomas and leukemias in New World primates and transforms human T cells. A better understanding of HVS biology will help advance our knowledge of virus-induced oncogenesis. Because numerous cellular miRNAs play crucial roles in cancer, viral miRNAs from the highly oncogenic HVS might also be important for transformation. Here, we found that the miR-HSURs preferentially modulate expression of host cell cycle regulators, as well as antiviral response factors. Our work provides further insight into the functions of herpesviral miRNAs in virus-induced oncogenesis and latency.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Herpesvirus Saimiriino 2/genética , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Linfocitos T/metabolismo , Animales , Sitios de Unión/genética , Western Blotting , Proteína Quinasa CDC2/metabolismo , Callithrix , Chaperón BiP del Retículo Endoplásmico , Células HEK293 , Proteínas de Choque Térmico/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunoprecipitación , Luciferasas , MicroARNs/genética , Fosforilación , ARN Mensajero/genética
15.
Bioessays ; 37(9): 936-41, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26200477

RESUMEN

High-fidelity binding of transcription factors (TFs) to DNA target sites is fundamental for proper regulation of cellular processes, as well as for the maintenance of cell identity. Recognition of cognate binding motifs in the genome is attributed by and large to the DNA binding domains of TFs. As an additional mode of conferring binding specificity, noncoding RNAs (ncRNAs) have been proposed to assist associated TFs in finding their binding sites by interacting with either DNA or RNA in the vicinity of their target loci. However, a well-documented example of such a mechanism was lacking until we recently reported that a ncRNA made by Epstein-Barr virus uses an RNA-RNA interaction with nascent transcripts generated from the viral genome to facilitate the recruitment of an interacting TF, PAX5, to viral DNA. This proof-of-principle finding suggests that cellular ncRNAs may likewise function in guiding interacting TFs to chromatin target sites.


Asunto(s)
ARN no Traducido/metabolismo , Factores de Transcripción/metabolismo , Silenciador del Gen , Herpesvirus Humano 4/genética , Humanos
16.
Genes Dev ; 29(14): 1552-64, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26220997

RESUMEN

Herpesvirus saimiri (HVS) is an oncogenic γ-herpesvirus that produces microRNAs (miRNAs) by cotranscription of precursor miRNA (pre-miRNA) hairpins immediately downstream from viral small nuclear RNAs (snRNA). The host cell Integrator complex, which recognizes the snRNA 3' end processing signal (3' box), generates the 5' ends of HVS pre-miRNA hairpins. Here, we identify a novel 3' box-like sequence (miRNA 3' box) downstream from HVS pre-miRNAs that is essential for miRNA biogenesis. In vivo knockdown and rescue experiments confirmed that the 3' end processing of HVS pre-miRNAs also depends on Integrator activity. Interaction between Integrator and HVS primary miRNA (pri-miRNA) substrates that contain only the miRNA 3' box was confirmed by coimmunoprecipitation and an in situ proximity ligation assay (PLA) that we developed to localize specific transient RNA-protein interactions inside cells. Surprisingly, in contrast to snRNA 3' end processing, HVS pre-miRNA 3' end processing by Integrator can be uncoupled from transcription, enabling new approaches to study Integrator enzymology.


Asunto(s)
Herpesvirus Saimiriino 2/genética , MicroARNs/metabolismo , Procesamiento de Término de ARN 3'/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Herpesvirus Saimiriino 2/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Inmunoprecipitación , MicroARNs/química , MicroARNs/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Procesamiento de Término de ARN 3'/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Nuclear Pequeño/metabolismo , Transcripción Genética
17.
Methods ; 91: 48-56, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26116541

RESUMEN

This review covers several computational methods for discovering structured non-coding RNAs in viruses and modeling their putative secondary structures. Here we will use examples from two target viruses to highlight these approaches: influenza A virus-a relatively small, segmented RNA virus; and Epstein-Barr virus-a relatively large DNA virus with a complex transcriptome. Each system has unique challenges to overcome and unique characteristics to exploit. From these particular cases, generically useful approaches can be derived for the study of additional viral targets.


Asunto(s)
Simulación por Computador , ARN no Traducido/química , ARN Viral/química , Herpesvirus Humano 4/metabolismo , Virus de la Influenza A/metabolismo , Conformación de Ácido Nucleico , ARN no Traducido/metabolismo
18.
PLoS One ; 10(6): e0124638, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121143

RESUMEN

In Epstein-Barr virus (EBV) latent infection, the EBV-encoded RNAs EBER1 and EBER2 accumulate in the host cell nucleus to ~10(6) copies. While the expression of EBERs in cell lines is associated with transformation, a mechanistic explanation of their roles in EBV latency remains elusive. To identify EBER-specific gene expression features, we compared the proteome and mRNA transcriptome from BJAB cells (an EBV-negative B lymphoma cell line) stably transfected with an empty plasmid or with one carrying both EBER genes. We identified ~1800 proteins with at least 2 SILAC pair measurements, of which only 8 and 12 were up- and downregulated ≥ 2-fold, respectively. One upregulated protein was PIK3AP1, a B-cell specific protein adapter known to activate the PI3K-AKT signaling pathway, which regulates alternative splicing and translation in addition to its pro-survival effects. In the mRNA-seq data, the mRNA levels for some of the proteins changing in the SILAC data did not change. We instead observed isoform switch events. We validated the most relevant findings with biochemical assays. These corroborated the upregulation of PIK3AP1 and AKT activation in BJAB cells expressing high levels of both EBERs and EBNA1 (a surrogate of Burkitt's lymphoma EBV latency I) relative to those expressing only EBNA1. The mRNA-seq data in these cells showed multiple upregulated oncogenes whose mRNAs are enriched for 3´-UTR AU-rich elements (AREs), such as ccl3, ccr7, il10, vegfa and zeb1. The CCL3, CCR7, IL10 and VEGFA proteins promote cell proliferation and are associated with EBV-mediated lymphomas. In EBV latency, ZEB1 represses the transcription of ZEBRA, an EBV lytic phase activation factor. We previously found that EBER1 interacts with AUF1 in vivo and proposed stabilization of ARE-containing mRNAs. Thus, the ~10(6) copies of EBER1 may promote not only cell proliferation due to an increase in the levels of ARE-containing genes like ccl3, ccr7, il10, and vegfa, but also the maintenance of latency, through higher levels of zeb1.


Asunto(s)
Herpesvirus Humano 4/genética , ARN Viral/genética , Línea Celular Tumoral , Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr/genética , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Genes Virales , Herpesvirus Humano 4/fisiología , Humanos , Linfoma de Células B/virología , Oncogenes , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , ARN Viral/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Latencia del Virus/genética
19.
Cell ; 160(4): 607-618, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25662012

RESUMEN

EBER2 is an abundant nuclear noncoding RNA expressed by the Epstein-Barr virus (EBV). Probing its possible chromatin localization by CHART revealed EBER2's presence at the terminal repeats (TRs) of the latent EBV genome, overlapping previously identified binding sites for the B cell transcription factor PAX5. EBER2 interacts with PAX5 and is required for the localization of PAX5 to the TRs. EBER2 knockdown phenocopies PAX5 depletion in upregulating the expression of LMP2A/B and LMP1, genes nearest the TRs. Knockdown of EBER2 also decreases EBV lytic replication, underscoring the essential role of the TRs in viral replication. Recruitment of the EBER2-PAX5 complex is mediated by base-pairing between EBER2 and nascent transcripts from the TR locus. The interaction is evolutionarily conserved in the related primate herpesvirus CeHV15 despite great sequence divergence. Using base-pairing with nascent RNA to guide an interacting transcription factor to its DNA target site is a previously undescribed function for a trans-acting noncoding RNA.


Asunto(s)
Herpesvirus Humano 4/metabolismo , Factor de Transcripción PAX5/metabolismo , ARN Viral/metabolismo , Secuencia de Bases , Ensayo de Cambio de Movilidad Electroforética , Técnicas de Silenciamiento del Gen , Herpesvirus Humano 4/genética , Humanos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Viral/química , ARN Viral/genética , Secuencias Repetidas en Tándem , Proteínas de la Matriz Viral/genética , Replicación Viral
20.
Cancer Biol Ther ; 16(1): 21-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25455629

RESUMEN

The transcriptome is extensively and dynamically regulated by a network of RNA modifying factors. RNA editing enzymes APOBEC (apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like) and ADAR (adenosine deaminase, RNA-specific) irreversibly recode primary RNA sequences, whereas newly described methylases (writers) and de-methylases (erasers) dynamically alter RNA molecules in response to environmental conditions. RNA modifications can affect RNA splicing, nuclear-cytoplasmic transport, translation, and regulation of gene expression by RNA interference. In addition, tRNA base modifications, processing, and regulated cleavage have been shown to alter global patterns of mRNA translation in response to cellular stress pathways. Recent studies, some of which were discussed at this workshop, have rekindled interest in the emerging roles of RNA modifications in health and disease. On September 10th, 2014, the Division of Cancer Biology, NCI sponsored a workshop to explore the role of epitranscriptomic RNA modifications and tRNA processing in cancer progression. The workshop attendees spanned a scientific range including chemists, virologists, and RNA and cancer biologists. The goal of the workshop was to explore the interrelationships between RNA editing, epitranscriptomics, and RNA processing and the enzymatic pathways that regulate these activities in cancer initiation and progression. At the conclusion of the workshop, a general discussion focused on defining the major challenges and opportunities in this field, as well as identifying the tools, technologies, resources and community efforts required to accelerate research in this emerging area.


Asunto(s)
Epigénesis Genética , Neoplasias/genética , Neoplasias/patología , Edición de ARN , Transcriptoma , Desaminasas APOBEC-1 , Animales , Citidina Desaminasa/metabolismo , Progresión de la Enfermedad , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA