Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Science ; 371(6529)2021 02 05.
Article in English | MEDLINE | ID: mdl-33414189

ABSTRACT

Polyadenylate [poly(A)] tail addition to the 3' end of a wide range of RNAs is a highly conserved modification that plays a central role in cellular RNA function. Elements for nuclear expression (ENEs) are cis-acting RNA elements that stabilize poly(A) tails by sequestering them in RNA triplex structures. A crystal structure of a double ENE from a rice hAT transposon messenger RNA complexed with poly(A)28 at a resolution of 2.89 angstroms reveals multiple modes of interaction with poly(A), including major-groove triple helices, extended minor-groove interactions with RNA double helices, a quintuple-base motif that transitions poly(A) from minor-groove associations to major-groove triple helices, and a poly(A) 3'-end binding pocket. Our findings both expand the repertoire of motifs involved in long-range RNA interactions and provide insights into how polyadenylation can protect an RNA's extreme 3' end.


Subject(s)
Poly A/chemistry , Polyadenylation , RNA Stability , RNA, Messenger/chemistry , Crystallization , Nucleic Acid Conformation , Oryza
2.
Mol Cell ; 65(1): 191-201, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-28017590

ABSTRACT

Nonsense-mediated decay (NMD) degrades mRNAs containing a premature termination codon (PTC). PTCs are a frequent cause of human genetic diseases, and the NMD pathway is known to modulate disease severity. Since partial NMD attenuation can potentially enhance nonsense suppression therapies, better definition of human-specific NMD is required. However, the majority of NMD factors were first discovered in model organisms and then subsequently identified by homology in human. Sensitivity and throughput limitations of existing approaches have hindered systematic forward genetic screening for NMD factors in human cells. We developed a method of in vivo amplification of NMD reporter fluorescence (Fireworks) that enables CRISPR-based forward genetic screening for NMD pathway defects in human cells. The Fireworks genetic screen identifies multiple known NMD factors and numerous human candidate genes, providing a platform for discovery of additional key factors in human mRNA degradation.


Subject(s)
Cell Separation/methods , Flow Cytometry , Green Fluorescent Proteins/biosynthesis , Nonsense Mediated mRNA Decay , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , CRISPR-Cas Systems , Codon, Nonsense , Genotype , Green Fluorescent Proteins/genetics , HeLa Cells , High-Throughput Screening Assays , Humans , Mutation , Phenotype , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Time Factors , Transfection
3.
RNA ; 22(11): 1785-1792, 2016 11.
Article in English | MEDLINE | ID: mdl-27659050

ABSTRACT

The proximity ligation assay (PLA) is an immune staining method that detects protein-protein interactions in fixed cells. We describe here RNA-PLA, a simple adaptation of this technology that allows the detection of specific RNA-protein interactions in fixed cells by using a DNA oligonucleotide that hybridizes to a target RNA in combination with an antibody that recognizes the protein bound to the target RNA. Stable and transient RNA-protein interactions can be readily detected by generation of a fluorescent signal in discrete compartments in intact fixed cells with high specificity. We demonstrate that this approach requires the colocalization of the binding protein and its RNA target in the same cellular compartment, use of an oligonucleotide complementary to the target RNA, and the presence of a binding site for the protein in the target RNA.


Subject(s)
RNA, Small Nuclear/metabolism , RNA-Binding Proteins/metabolism , Cell Compartmentation , HEK293 Cells , HeLa Cells , Humans
4.
Cell Rep ; 15(6): 1266-76, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27134163

ABSTRACT

The ENE (element for nuclear expression) is a cis-acting RNA structure that protects viral or cellular noncoding RNAs (ncRNAs) from nuclear decay through triple-helix formation with the poly(A) tail or 3'-terminal A-rich tract. We expanded the roster of nine known ENEs by bioinformatic identification of ∼200 distinct ENEs that reside in transposable elements (TEs) of numerous non-metazoan and one fish species and in four Dicistrovirus genomes. Despite variation within the ENE core, none of the predicted triple-helical stacks exceeds five base triples. Increased accumulation of reporter transcripts in human cells demonstrated functionality for representative ENEs. Location close to the poly(A) tail argues that ENEs are active in TE transcripts. Their presence in intronless, but not intron-containing, hAT transposase genes supports the idea that TEs acquired ENEs to counteract the RNA-destabilizing effects of intron loss, a potential evolutionary consequence of TE horizontal transfer in organisms that couple RNA silencing to splicing deficits.


Subject(s)
DNA Transposable Elements/genetics , Fungi/genetics , Nucleic Acid Conformation , Plants/genetics , RNA, Fungal/chemistry , RNA, Plant/chemistry , Base Sequence , Cell Nucleus/genetics , Conserved Sequence/genetics , HEK293 Cells , Humans , Introns/genetics , RNA Stability/genetics , RNA, Fungal/genetics , RNA, Messenger/genetics , RNA, Plant/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Transposases/genetics , Transposases/metabolism
5.
Genes Dev ; 29(14): 1552-64, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26220997

ABSTRACT

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.


Subject(s)
Herpesvirus 2, Saimiriine/genetics , MicroRNAs/metabolism , RNA 3' End Processing/physiology , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Herpesvirus 2, Saimiriine/metabolism , Host-Pathogen Interactions/genetics , Humans , Immunoprecipitation , MicroRNAs/chemistry , MicroRNAs/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , RNA 3' End Processing/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Small Nuclear/metabolism , Transcription, Genetic
6.
Cell ; 155(7): 1568-80, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24360278

ABSTRACT

MicroRNAs (miRNAs) are short RNA gene regulators typically produced from primary transcripts that are cleaved by the nuclear microprocessor complex, with the resulting precursor miRNA hairpins exported by exportin 5 and processed by cytoplasmic Dicer to yield two (5p and 3p) miRNAs. Here, we document microprocessor-independent 7-methylguanosine (m(7)G)-capped pre-miRNAs, whose 5' ends coincide with transcription start sites and 3' ends are most likely generated by transcription termination. By establishing a small RNA Cap-seq method that employs the cap-binding protein eIF4E, we identified a group of murine m(7)G-capped pre-miRNAs genome wide. The m(7)G-capped pre-miRNAs are exported via the PHAX-exportin 1 pathway. After Dicer cleavage, only the 3p-miRNA is efficiently loaded onto Argonaute to form a functional microRNP. This unusual miRNA biogenesis pathway, which differs in pre-miRNA synthesis, nuclear-cytoplasmic transport, and guide strand selection, enables the development of shRNA expression constructs that produce a single 3p-siRNA.


Subject(s)
MicroRNAs/genetics , RNA Caps , Animals , Argonaute Proteins/metabolism , Base Sequence , Biosynthetic Pathways , DEAD-box RNA Helicases/metabolism , Genome-Wide Association Study , Guanosine/analogs & derivatives , Guanosine/metabolism , Humans , Karyopherins/metabolism , Mice , MicroRNAs/chemistry , MicroRNAs/metabolism , Molecular Sequence Data , RNA Polymerase II/metabolism , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Ribonuclease III/metabolism , Transcription Termination, Genetic , Exportin 1 Protein
7.
Proc Natl Acad Sci U S A ; 109(52): 21313-8, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236153

ABSTRACT

The multiprotein exon junction complex (EJC) that is deposited upstream of spliced junctions orchestrates downstream events in the life of a metazoan mRNA, including its surveillance via the nonsense-mediated decay (NMD) pathway. However, the mechanism by which the spliceosome mediates EJC formation is not well understood. We show that human eIF4G-like spliceosomal protein (h)CWC22 directly interacts with the core EJC component eIF4AIII in vitro and in vivo; mutations at the predicted hCWC22/eIF4AIII interface disrupt association. In vivo depletion of hCWC22, as for yeast Cwc22p, causes a splicing defect, resulting in decreased levels of mature cellular mRNAs. Nonetheless, hCWC22 depletion yields increased levels of spliced RNA from the unusual nonsense codon-containing U22 host gene, which is a natural substrate of NMD. To test whether hCWC22 acts in NMD through coupling splicing to EJC deposition, we searched for mutations in hCWC22 that affect eIF4AIII deposition without affecting splicing. Addition of hCWC22(G168Y) with a mutation at the putative hCWC22/eIF4AIII interface exacerbates the defect in splicing-dependent deposition of eIF4AIII(T334V) with a mutation reported to be in direct contact with mRNA, linking hCWC22 to the process of EJC deposition in vitro. Importantly, the addition of hCWC22(G168Y) affects deposition of eIF4AIII(T334V) without inhibiting splicing or the efficiency of deposition of the endogenous eF4AIII(WT) in the same reaction, demonstrating hCWC22's specific role in eIF4AIII deposition in addition to its role in splicing. The essential splicing factor CWC22 has, therefore, acquired functions in EJC assembly and NMD during evolution from single-celled to complex eukaryotes.


Subject(s)
Carrier Proteins/metabolism , Exons/genetics , Nonsense Mediated mRNA Decay/genetics , RNA Splicing/genetics , Spliceosomes/metabolism , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/metabolism , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Mutation/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Peptidylprolyl Isomerase , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism
8.
Cell Rep ; 2(1): 26-32, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22840393

ABSTRACT

Abundant expression of the long noncoding (lnc) PAN (polyadenylated nuclear) RNA by the human oncogenic gammaherpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV) depends on a cis-element called the expression and nuclear retention element (ENE). The ENE upregulates PAN RNA by inhibiting its rapid nuclear decay through triple-helix formation with the poly(A) tail. Using structure-based bioinformatics, we identified six ENE-like elements in evolutionarily diverse viral genomes. Five are in double-stranded DNA viruses, including mammalian herpesviruses, insect polydnaviruses, and a protist mimivirus. One is in an insect picorna-like positive-strand RNA virus, suggesting that the ENE can counteract cytoplasmic as well as nuclear RNA decay pathways. Functionality of four of the ENEs was demonstrated by increased accumulation of an intronless polyadenylated reporter transcript in human cells. Identification of these ENEs enabled the discovery of PAN RNA homologs in two additional gammaherpesviruses, RRV and EHV2. Our findings demonstrate that searching for structural elements can lead to rapid identification of lncRNAs.


Subject(s)
Conserved Sequence , Nucleic Acid Conformation , RNA Stability/genetics , RNA Viruses/genetics , RNA, Untranslated , Regulatory Elements, Transcriptional/genetics , Base Sequence , Cell Nucleus/genetics , Cell Nucleus/metabolism , Conserved Sequence/physiology , Genome, Viral/genetics , HEK293 Cells , Humans , Models, Biological , Molecular Sequence Data , RNA Viruses/chemistry , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Untranslated/chemistry , RNA, Untranslated/genetics
9.
Mol Cell ; 34(1): 47-57, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19285445

ABSTRACT

Small Cajal body (CB)-specific RNPs (scaRNPs) function in posttranscriptional modification of small nuclear (sn)RNAs. An RNA element, the CAB box, facilitates CB localization of H/ACA scaRNPs. Using a related element in Drosophila C/D scaRNAs, we purified a fly WD40 repeat protein that UV crosslinks to RNA in a C/D CAB box-dependent manner and associates with C/D and mixed domain C/D-H/ACA scaRNAs. Its human homolog, WDR79, associates with C/D, H/ACA, and mixed domain scaRNAs, as well as with telomerase RNA. WDR79's binding to human H/ACA and mixed domain scaRNAs is CAB box dependent, and its association with mixed domain RNAs also requires the ACA motif, arguing for additional interactions of WDR79 with H/ACA core proteins. We demonstrate a requirement for WDR79 binding in the CB localization of a scaRNA. This and other recent reports establish WDR79 as a central player in the localization and processing of nuclear RNPs.


Subject(s)
Coiled Bodies/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Amino Acid Motifs , Animals , Base Sequence , Cell Line , Chromatography, Affinity , Coiled Bodies/ultrastructure , Drosophila Proteins/analysis , Drosophila Proteins/chemistry , Drosophila melanogaster/genetics , HeLa Cells , Humans , Molecular Sequence Data , Nucleic Acid Conformation , RNA-Binding Proteins/analysis , RNA-Binding Proteins/chemistry , Recombinant Fusion Proteins/analysis , Regulatory Sequences, Ribonucleic Acid , Ribonucleoproteins/analysis , Sequence Alignment
10.
RNA ; 14(8): 1459-62, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18567814

ABSTRACT

A small fraction of premessenger RNA introns in certain eukaryotes is excised by the minor spliceosome, which contains low-abundance small nuclear ribonucleoproteins (snRNPs). Recently, it was suggested that minor-class snRNPs are localized to and function in the cytoplasm of vertebrate cells. To test whether U12-type splicing occurs in the cytoplasm of Xenopus oocytes, we performed microinjections of the well-characterized P120 minor-class splicing substrate into the nucleus or into the cytoplasm. Our results demonstrate that accurate splicing of this U12-dependent intron occurs exclusively in the nuclear compartment of the oocyte, where U12 and U6atac snRNPs are primarily localized. We further demonstrate that splicing of both a major-class and a minor-class intron is inhibited after nuclear envelope breakdown during meiosis.


Subject(s)
Cell Nucleus/metabolism , Oocytes/metabolism , RNA Splicing , Xenopus laevis/metabolism , Animals , Oocytes/cytology , RNA, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes
11.
Proc Natl Acad Sci U S A ; 104(25): 10412-7, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17563387

ABSTRACT

We previously demonstrated that the Kaposi's sarcoma-associated herpesvirus polyadenylated nuclear RNA contains a 79-nt cis-acting element, the ENE, which allows intronless polyadenylated transcripts to accumulate to high nuclear levels by protecting them from rapid degradation. We proposed a model based on the predicted structure of the ENE in which a U-rich internal loop hybridizes with the 3'-polyadenylate (polyA) tail to sequester it from exonucleolytic attack. We have tested this model by mutational analysis of the ENE. Point mutations in the predicted U-rich internal loop and in the flanking stems abolish the ENE's ability to (i) interact with the polyA tail, (ii) inhibit deadenylation in vitro, and (iii) stabilize transcripts in vivo. In all but one case, compensatory mutations in the flanking stems restore ENE activities, demonstrating the importance of these stems and uncovering a unique role for the loop-proximal G-C base pair in the lower stem. Increasing the U content of the U-rich internal loop surprisingly decreases stability in vivo but does not affect deadenylation in vitro, comparable to the effects of deleting certain "unstructured" regions of the ENE. Taken together, our data support the formation of the proposed ENE secondary structure in vivo and argue that the specific ENE structure inhibits rapid RNA decay in cis by engaging in a limited set of base-pairing interactions with the polyA tail.


Subject(s)
Poly A/metabolism , RNA Stability , RNA, Viral/genetics , RNA, Viral/metabolism , Regulatory Sequences, Ribonucleic Acid/genetics , Base Pairing , Base Sequence , HeLa Cells , Humans , Nucleic Acid Conformation , Point Mutation , Polyadenylation
12.
Mol Cell ; 24(6): 943-53, 2006 Dec 28.
Article in English | MEDLINE | ID: mdl-17189195

ABSTRACT

Cellular RNAs are subject to quality-control pathways that insure the fidelity of gene expression. We previously identified a 79 nt element, the ENE, that is essential for the nuclear accumulation of a viral polyadenylated nuclear (PAN) RNA. Here, we show that intron-less polyadenylated transcripts such as PAN RNA and beta-globin cRNA exhibit two-component exponential decay kinetics in which some transcripts are rapidly degraded (t(1/2) = approximately 15 min) while others decay more slowly (t(1/2) = approximately 3 hr). Inclusion of the ENE protects such transcripts from rapid decay in a poly(A)-dependent fashion. The ENE inhibits deadenylation and decay in nuclear extract and prevents deadenylation of naked RNA by a purified deadenylase, likely through snoRNA-like intramolecular hybridization with the poly(A) tail. The ENE causes increased accumulation of splicing-defective beta-globin pre-mRNAs in vivo. We propose that the ENE-controlled rapid-decay mechanism for polyadenylated transcripts comprises a nuclear pre-mRNA surveillance system in mammalian cells.


Subject(s)
Elements , RNA Stability , RNA, Messenger/genetics , RNA, Viral/metabolism , Base Sequence , Globins/genetics , Humans , Molecular Sequence Data , Mutation , Polyadenylation , RNA Splice Sites , Signal Transduction , Transcription, Genetic
13.
Mol Cell ; 23(5): 673-84, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16949364

ABSTRACT

Pre-mRNA splicing in vertebrates is molecularly linked to other processes. We previously reported that splicing is required for efficient assembly of intron-encoded box C/D small nucleolar ribonucleoprotein (snoRNP). In the spliceosomal C1 complex, snoRNP proteins efficiently assemble onto snoRNA sequences if they are located about 50 nt upstream of the intron branchpoint. Here, we identify the splicing factor responsible for coupling snoRNP assembly to intron excision. Intron binding protein (IBP) 160, a helicase-like protein previously detected in the spliceosomal C1 complex, binds the pre-mRNA in a sequence-independent manner, contacting nucleotides 33-40 upstream of the intron branch site, regardless of whether a snoRNA is present. Depletion of IBP160 abrogates snoRNP assembly in vitro. IBP160 binding directly to a snoRNA located too close to the intron branch site interferes with snoRNP assembly. Thus, IBP160 is the key factor linking snoRNP biogenesis and perhaps other postsplicing events to pre-mRNA splicing.


Subject(s)
DNA-Binding Proteins/metabolism , Introns/genetics , RNA Precursors/metabolism , RNA Splicing/genetics , Ribonucleoproteins, Small Nucleolar/metabolism , Spliceosomes/chemistry , Spliceosomes/metabolism , Animals , Base Sequence , HeLa Cells , Humans , Mice , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , RNA Helicases/metabolism , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/biosynthesis
14.
Proc Natl Acad Sci U S A ; 101(52): 17976-81, 2004 Dec 28.
Article in English | MEDLINE | ID: mdl-15608055

ABSTRACT

Metazoan cells have two pathways for intron removal involving the U2- and U12-type spliceosomes, which contain mostly nonoverlapping sets of small nuclear ribonucleoproteins. We show that in vitro splicing of a U12-type intron assembles an exon junction complex (EJC) that is comparably positioned and contains many of the same components as that deposited by the U2-type spliceosome. The presence of a U12-type intron downstream of a premature termination codon within an open reading frame (ORF) induces nonsense-mediated decay of the mRNA in vivo. These findings suggest a common pathway for EJC assembly by the two spliceosomes and highlight the evolutionary age of the EJC and its downstream functions in gene expression.


Subject(s)
Introns , RNA Splicing , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics , Cell Line , Cell Nucleus/metabolism , Codon, Nonsense , DNA, Complementary/metabolism , Evolution, Molecular , Exons , Gene Expression Regulation , HeLa Cells , Humans , Immunoprecipitation , Mutagenesis, Site-Directed , Open Reading Frames , Plasmids/metabolism , RNA/chemistry , RNA/metabolism , RNA Precursors/metabolism , RNA, Small Nuclear/metabolism , Ribonuclease H/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/metabolism , Time Factors , Transfection
15.
Mol Cell ; 12(1): 113-23, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12887897

ABSTRACT

In mammalian cells, all small nucleolar RNAs (snoRNAs) that guide rRNA modification are encoded within the introns of host genes. An optimal position about 70 nts upstream of the 3' splice site of the host intron is critical for efficient expression of box C/D snoRNAs in vivo, suggesting synergy with splicing. Here, we have used a coupled in vitro splicing-snoRNA processing system to demonstrate that assembly of box C/D snoRNP proteins is the step affected by snoRNA location, and that active splicing is essential for snoRNP assembly. Splicing blockage experiments further reveal that snoRNP proteins bind specifically at the spliceosomal C1 complex stage. In contrast, splicing-independent snoRNP assembly can occur in vitro on snoRNAs that possess stable external stems. In vivo analyses confirm that a stable stem can compensate for the unusual position of those few box C/D snoRNAs located far from the 3' splice site of their host intron.


Subject(s)
Eukaryotic Cells/metabolism , Introns/genetics , Mammals/genetics , RNA Splicing/genetics , RNA, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/biosynthesis , Animals , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation/genetics , Humans , Molecular Structure , Protein Binding/genetics , RNA Splice Sites/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Ribonucleoproteins, Small Nuclear/genetics , Spliceosomes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...