Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Cell ; 185(20): 3671-3688.e23, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36113466

ABSTRACT

Bacteria encode reverse transcriptases (RTs) of unknown function that are closely related to group II intron-encoded RTs. We found that a Pseudomonas aeruginosa group II intron-like RT (G2L4 RT) with YIDD instead of YADD at its active site functions in DNA repair in its native host and when expressed in Escherichia coli. G2L4 RT has biochemical activities strikingly similar to those of human DNA repair polymerase Î¸ and uses them for translesion DNA synthesis and double-strand break repair (DSBR) via microhomology-mediated end-joining (MMEJ). We also found that a group II intron RT can function similarly in DNA repair, with reciprocal active-site substitutions showing isoleucine favors MMEJ and alanine favors primer extension in both enzymes. These DNA repair functions utilize conserved structural features of non-LTR-retroelement RTs, including human LINE-1 and other eukaryotic non-LTR-retrotransposon RTs, suggesting such enzymes may have inherent ability to function in DSBR in a wide range of organisms.


Subject(s)
RNA-Directed DNA Polymerase , Retroelements , Alanine/genetics , DNA End-Joining Repair , DNA Repair , DNA-Directed RNA Polymerases/genetics , Humans , Introns , Isoleucine/genetics , RNA-Directed DNA Polymerase/chemistry
2.
Mol Cell ; 72(4): 700-714.e8, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30344094

ABSTRACT

Prokaryotic CRISPR-Cas systems provide adaptive immunity by integrating portions of foreign nucleic acids (spacers) into genomic CRISPR arrays. Cas6 proteins then process CRISPR array transcripts into spacer-derived RNAs (CRISPR RNAs; crRNAs) that target Cas nucleases to matching invaders. We find that a Marinomonas mediterranea fusion protein combines three enzymatic domains (Cas6, reverse transcriptase [RT], and Cas1), which function in both crRNA biogenesis and spacer acquisition from RNA and DNA. We report a crystal structure of this divergent Cas6, identify amino acids required for Cas6 activity, show that the Cas6 domain is required for RT activity and RNA spacer acquisition, and demonstrate that CRISPR-repeat binding to Cas6 regulates RT activity. Co-evolution of putative interacting surfaces suggests a specific structural interaction between the Cas6 and RT domains, and phylogenetic analysis reveals repeated, stable association of free-standing Cas6s with CRISPR RTs in multiple microbial lineages, indicating that a functional interaction between these proteins preceded evolution of the fusion.


Subject(s)
CRISPR-Associated Proteins/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , RNA-Directed DNA Polymerase/physiology , Base Sequence/genetics , CRISPR-Cas Systems/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA , Endonucleases , Marinomonas/genetics , Marinomonas/metabolism , Phylogeny , RNA/biosynthesis , Substrate Specificity
3.
Mol Cell ; 68(5): 926-939.e4, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29153391

ABSTRACT

Bacterial group II intron reverse transcriptases (RTs) function in both intron mobility and RNA splicing and are evolutionary predecessors of retrotransposon, telomerase, and retroviral RTs as well as the spliceosomal protein Prp8 in eukaryotes. Here we determined a crystal structure of a full-length thermostable group II intron RT in complex with an RNA template-DNA primer duplex and incoming deoxynucleotide triphosphate (dNTP) at 3.0-Å resolution. We find that the binding of template-primer and key aspects of the RT active site are surprisingly different from retroviral RTs but remarkably similar to viral RNA-dependent RNA polymerases. The structure reveals a host of features not seen previously in RTs that may contribute to distinctive biochemical properties of group II intron RTs, and it provides a prototype for many related bacterial and eukaryotic non-LTR retroelement RTs. It also reveals how protein structural features used for reverse transcription evolved to promote the splicing of both group II and spliceosomal introns.


Subject(s)
Bacterial Proteins/chemistry , Evolution, Molecular , RNA Splicing , RNA-Directed DNA Polymerase/chemistry , Temperature , Transcription, Genetic , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Enzyme Stability , Introns , Models, Molecular , Nucleic Acid Conformation , Nucleic Acid Heteroduplexes/chemistry , Nucleic Acid Heteroduplexes/genetics , Nucleic Acid Heteroduplexes/metabolism , Protein Binding , Protein Denaturation , Protein Interaction Domains and Motifs , RNA/chemistry , RNA/genetics , RNA/metabolism , RNA-Directed DNA Polymerase/genetics , RNA-Directed DNA Polymerase/metabolism , Retroelements , Spliceosomes/chemistry , Spliceosomes/enzymology , Spliceosomes/genetics , Structure-Activity Relationship
4.
Genes Dev ; 30(18): 2076-2092, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27798849

ABSTRACT

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.


Subject(s)
Argonaute Proteins/metabolism , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , MicroRNAs/metabolism , RNA, Untranslated/metabolism , Acid Anhydride Hydrolases/metabolism , Adenoviridae/genetics , Gene Knockout Techniques , HEK293 Cells , Humans , Leukemia Virus, Bovine/genetics , Phosphorylation , RNA Polymerase III/metabolism , RNA, Viral/metabolism
5.
J Biol Chem ; 297(2): 100971, 2021 08.
Article in English | MEDLINE | ID: mdl-34280434

ABSTRACT

Reverse transcriptases (RTs) can switch template strands during complementary DNA synthesis, enabling them to join discontinuous nucleic acid sequences. Template switching (TS) plays crucial roles in retroviral replication and recombination, is used for adapter addition in RNA-Seq, and may contribute to retroelement fitness by increasing evolutionary diversity and enabling continuous complementary DNA synthesis on damaged templates. Here, we determined an X-ray crystal structure of a TS complex of a group II intron RT bound simultaneously to an acceptor RNA and donor RNA template-DNA primer heteroduplex with a 1-nt 3'-DNA overhang. The structure showed that the 3' end of the acceptor RNA binds in a pocket formed by an N-terminal extension present in non-long terminal repeat-retroelement RTs and the RT fingertips loop, with the 3' nucleotide of the acceptor base paired to the 1-nt 3'-DNA overhang and its penultimate nucleotide base paired to the incoming dNTP at the RT active site. Analysis of structure-guided mutations identified amino acids that contribute to acceptor RNA binding and a phenylalanine residue near the RT active site that mediates nontemplated nucleotide addition. Mutation of the latter residue decreased multiple sequential template switches in RNA-Seq. Our results provide new insights into the mechanisms of TS and nontemplated nucleotide addition by RTs, suggest how these reactions could be improved for RNA-Seq, and reveal common structural features for TS by non-long terminal repeat-retroelement RTs and viral RNA-dependent RNA polymerases.


Subject(s)
Crystallography, X-Ray/methods , DNA, Complementary/genetics , Geobacillus stearothermophilus/enzymology , Introns , RNA, Bacterial/genetics , RNA-Directed DNA Polymerase/chemistry , Retroelements/genetics , Geobacillus stearothermophilus/chemistry , Models, Molecular , RNA-Directed DNA Polymerase/metabolism , Templates, Genetic
6.
PLoS Genet ; 15(7): e1008273, 2019 07.
Article in English | MEDLINE | ID: mdl-31329584

ABSTRACT

5' ends are important for determining the fate of RNA molecules. BCDIN3D is an RNA phospho-methyltransferase that methylates the 5' monophosphate of specific RNAs. In order to gain new insights into the molecular function of BCDIN3D, we performed an unbiased analysis of its interacting RNAs by Thermostable Group II Intron Reverse Transcriptase coupled to next generation sequencing (TGIRT-seq). Our analyses showed that BCDIN3D interacts with full-length phospho-methylated tRNAHis and miR-4454. Interestingly, we found that miR-4454 is not synthesized from its annotated genomic locus, which is a primer-binding site for an endogenous retrovirus, but rather by Dicer cleavage of mature tRNAHis. Sequence analysis revealed that miR-4454 is identical to the 3' end of tRNAHis. Moreover, we were able to generate this 'miRNA' in vitro through incubation of mature tRNAHis with Dicer. As found previously for several pre-miRNAs, a 5'P-tRNAHis appears to be a better substrate for Dicer cleavage than a phospho-methylated tRNAHis. Moreover, tRNAHis 3'-fragment/'miR-4454' levels increase in cells depleted for BCDIN3D. Altogether, our results show that in addition to microRNAs, BCDIN3D regulates tRNAHis 3'-fragment processing without negatively affecting tRNAHis's canonical function of aminoacylation.


Subject(s)
DEAD-box RNA Helicases/genetics , High-Throughput Nucleotide Sequencing/methods , Methyltransferases/genetics , RNA, Transfer, His/metabolism , Ribonuclease III/genetics , Cell Line , Humans , MicroRNAs/genetics , Sequence Analysis, RNA , Transfer RNA Aminoacylation
7.
J Biol Chem ; 294(51): 19764-19784, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31712313

ABSTRACT

The reverse transcriptases (RTs) encoded by mobile group II introns and other non-LTR retroelements differ from retroviral RTs in being able to template-switch efficiently from the 5' end of one template to the 3' end of another with little or no complementarity between the donor and acceptor templates. Here, to establish a complete kinetic framework for the reaction and to identify conditions that more efficiently capture acceptor RNAs or DNAs, we used a thermostable group II intron RT (TGIRT; GsI-IIC RT) that can template switch directly from synthetic RNA template/DNA primer duplexes having either a blunt end or a 3'-DNA overhang end. We found that the rate and amplitude of template switching are optimal from starter duplexes with a single nucleotide 3'-DNA overhang complementary to the 3' nucleotide of the acceptor RNA, suggesting a role for nontemplated nucleotide addition of a complementary nucleotide to the 3' end of cDNAs synthesized from natural templates. Longer 3'-DNA overhangs progressively decreased the template-switching rate, even when complementary to the 3' end of the acceptor template. The reliance on only a single bp with the 3' nucleotide of the acceptor together with discrimination against mismatches and the high processivity of group II intron RTs enable synthesis of full-length DNA copies of nucleic acids beginning directly at their 3' end. We discuss the possible biological functions of the template-switching activity of group II intron- and other non-LTR retroelement-encoded RTs, as well as the optimization of this activity for adapter addition in RNA- and DNA-Seq protocols.


Subject(s)
Introns , Nucleotides/genetics , RNA-Directed DNA Polymerase/metabolism , RNA-Seq/methods , Retroelements/genetics , Templates, Genetic , Animals , DNA Primers , DNA Transposable Elements , Genetic Complementation Test , Insecta , Kinetics , RNA/genetics , Retroviridae/genetics , Temperature , Exome Sequencing
8.
Nat Methods ; 14(1): 75-82, 2017 01.
Article in English | MEDLINE | ID: mdl-27819661

ABSTRACT

Coupling of structure-specific in vivo chemical modification to next-generation sequencing is transforming RNA secondary structure studies in living cells. The dominant strategy for detecting in vivo chemical modifications uses reverse transcriptase truncation products, which introduce biases and necessitate population-average assessments of RNA structure. Here we present dimethyl sulfate (DMS) mutational profiling with sequencing (DMS-MaPseq), which encodes DMS modifications as mismatches using a thermostable group II intron reverse transcriptase. DMS-MaPseq yields a high signal-to-noise ratio, can report multiple structural features per molecule, and allows both genome-wide studies and focused in vivo investigations of even low-abundance RNAs. We apply DMS-MaPseq for the first analysis of RNA structure within an animal tissue and to identify a functional structure involved in noncanonical translation initiation. Additionally, we use DMS-MaPseq to compare the in vivo structure of pre-mRNAs with their mature isoforms. These applications illustrate DMS-MaPseq's capacity to dramatically expand in vivo analysis of RNA structure.


Subject(s)
Genome, Human , High-Throughput Nucleotide Sequencing/methods , RNA-Binding Proteins/genetics , RNA/chemistry , RNA/genetics , Sulfuric Acid Esters/chemistry , Computational Biology , HEK293 Cells , Humans , Mutation/genetics , Nucleic Acid Conformation , Protein Biosynthesis , Sequence Analysis, RNA
9.
RNA ; 24(7): 950-965, 2018 07.
Article in English | MEDLINE | ID: mdl-29703781

ABSTRACT

Comparing the abundance of one RNA molecule to another is crucial for understanding cellular functions but most sequencing techniques can target only specific subsets of RNA. In this study, we used a new fragmented ribodepleted TGIRT sequencing method that uses a thermostable group II intron reverse transcriptase (TGIRT) to generate a portrait of the human transcriptome depicting the quantitative relationship of all classes of nonribosomal RNA longer than 60 nt. Comparison between different sequencing methods indicated that FRT is more accurate in ranking both mRNA and noncoding RNA than viral reverse transcriptase-based sequencing methods, even those that specifically target these species. Measurements of RNA abundance in different cell lines using this method correlate with biochemical estimates, confirming tRNA as the most abundant nonribosomal RNA biotype. However, the single most abundant transcript is 7SL RNA, a component of the signal recognition particle. Structured noncoding RNAs (sncRNAs) associated with the same biological process are expressed at similar levels, with the exception of RNAs with multiple functions like U1 snRNA. In general, sncRNAs forming RNPs are hundreds to thousands of times more abundant than their mRNA counterparts. Surprisingly, only 50 sncRNA genes produce half of the non-rRNA transcripts detected in two different cell lines. Together the results indicate that the human transcriptome is dominated by a small number of highly expressed sncRNAs specializing in functions related to translation and splicing.


Subject(s)
RNA, Untranslated/metabolism , Transcriptome , Cell Line, Tumor , High-Throughput Nucleotide Sequencing , Humans , Proteins/genetics , RNA, Messenger/metabolism , RNA, Small Nucleolar/metabolism , RNA, Transfer/metabolism , RNA-Directed DNA Polymerase , Ribonucleoproteins/metabolism , Sequence Analysis, RNA
10.
Nucleic Acids Res ; 46(1): e1, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29036654

ABSTRACT

Cellular accumulation of repetitive RNA occurs in several dominantly-inherited genetic disorders. Expanded CUG, CCUG or GGGGCC repeats are expressed in myotonic dystrophy type 1 (DM1), myotonic dystrophy type 2 (DM2), or familial amyotrophic lateral sclerosis, respectively. Expanded repeat RNAs (ER-RNAs) exert a toxic gain-of-function and are prime therapeutic targets in these diseases. However, efforts to quantify ER-RNA levels or monitor knockdown are confounded by stable structure and heterogeneity of the ER-RNA tract and background signal from non-expanded repeats. Here, we used a thermostable group II intron reverse transcriptase (TGIRT-III) to convert ER-RNA to cDNA, followed by quantification on slot blots. We found that TGIRT-III was capable of reverse transcription (RTn) on enzymatically synthesized ER-RNAs. By using conditions that limit cDNA synthesis from off-target sequences, we observed hybridization signals on cDNA slot blots from DM1 and DM2 muscle samples but not from healthy controls. In transgenic mouse models of DM1 the cDNA slot blots accurately reflected the differences of ER-RNA expression across different transgenic lines, and showed therapeutic reductions in skeletal and cardiac muscle, accompanied by improvements of the DM1-associated splicing defects. TGIRT-III was also active on CCCCGG- and GGGGCC-repeats, suggesting that ER-RNA analysis is feasible for several repeat expansion disorders.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Introns/genetics , Myotonic Dystrophy/genetics , RNA-Directed DNA Polymerase/genetics , RNA/genetics , Repetitive Sequences, Nucleic Acid/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Base Sequence , Electrophoresis, Capillary , Electrophoresis, Polyacrylamide Gel , Enzyme Stability , Humans , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myotonic Dystrophy/metabolism , RNA Splicing , RNA-Directed DNA Polymerase/metabolism , Temperature
11.
Proc Natl Acad Sci U S A ; 114(43): E8987-E8995, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073095

ABSTRACT

RNA is secreted from cells enclosed within extracellular vesicles (EVs). Defining the RNA composition of EVs is challenging due to their coisolation with contaminants, lack of knowledge of the mechanisms of RNA sorting into EVs, and limitations of conventional RNA-sequencing methods. Here we present our observations using thermostable group II intron reverse transcriptase sequencing (TGIRT-seq) to characterize the RNA extracted from HEK293T cell EVs isolated by flotation gradient ultracentrifugation and from exosomes containing the tetraspanin CD63 further purified from the gradient fractions by immunoisolation. We found that EV-associated transcripts are dominated by full-length, mature transfer RNAs (tRNAs) and other small noncoding RNAs (ncRNAs) encapsulated within vesicles. A substantial proportion of the reads mapping to protein-coding genes, long ncRNAs, and antisense RNAs were due to DNA contamination on the surface of vesicles. Nevertheless, sequences mapping to spliced mRNAs were identified within HEK293T cell EVs and exosomes, among the most abundant being transcripts containing a 5' terminal oligopyrimidine (5' TOP) motif. Our results indicate that the RNA-binding protein YBX1, which is required for the sorting of selected miRNAs into exosomes, plays a role in the sorting of highly abundant small ncRNA species, including tRNAs, Y RNAs, and Vault RNAs. Finally, we obtained evidence for an EV-specific tRNA modification, perhaps indicating a role for posttranscriptional modification in the sorting of some RNA species into EVs. Our results suggest that EVs and exosomes could play a role in the purging and intercellular transfer of excess free RNAs, including full-length tRNAs and other small ncRNAs.


Subject(s)
Exosomes/physiology , RNA, Small Untranslated/metabolism , Y-Box-Binding Protein 1/metabolism , Animals , DNA/chemistry , DNA/metabolism , Exosomes/chemistry , Extracellular Vesicles , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Y-Box-Binding Protein 1/genetics
12.
BMC Genomics ; 19(1): 510, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29969991

ABSTRACT

BACKGROUND: Alignment-free RNA quantification tools have significantly increased the speed of RNA-seq analysis. However, it is unclear whether these state-of-the-art RNA-seq analysis pipelines can quantify small RNAs as accurately as they do with long RNAs in the context of total RNA quantification. RESULT: We comprehensively tested and compared four RNA-seq pipelines for accuracy of gene quantification and fold-change estimation. We used a novel total RNA benchmarking dataset in which small non-coding RNAs are highly represented along with other long RNAs. The four RNA-seq pipelines consisted of two commonly-used alignment-free pipelines and two variants of alignment-based pipelines. We found that all pipelines showed high accuracy for quantifying the expression of long and highly-abundant genes. However, alignment-free pipelines showed systematically poorer performance in quantifying lowly-abundant and small RNAs. CONCLUSION: We have shown that alignment-free and traditional alignment-based quantification methods perform similarly for common gene targets, such as protein-coding genes. However, we have identified a potential pitfall in analyzing and quantifying lowly-expressed genes and small RNAs with alignment-free pipelines, especially when these small RNAs contain biological variations.


Subject(s)
RNA/chemistry , Sequence Analysis, RNA/methods , Algorithms , Area Under Curve , High-Throughput Nucleotide Sequencing , RNA/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , ROC Curve
13.
Nat Methods ; 12(9): 835-837, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26214130

ABSTRACT

Despite its biological importance, tRNA has not been adequately sequenced by standard methods because of its abundant post-transcriptional modifications and stable structure, which interfere with cDNA synthesis. We achieved efficient and quantitative tRNA sequencing in HEK293T cells by using engineered demethylases to remove base methylations and a highly processive thermostable group II intron reverse transcriptase to overcome these obstacles. Our method, DM-tRNA-seq, should be applicable to investigations of tRNA in all organisms.


Subject(s)
Algorithms , Gene Library , High-Throughput Nucleotide Sequencing/methods , RNA, Transfer/genetics , Base Sequence , HEK293 Cells , Humans , Molecular Sequence Data
14.
RNA ; 22(4): 597-613, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26826130

ABSTRACT

Next-generation RNA sequencing (RNA-seq) has revolutionized our ability to analyze transcriptomes. Current RNA-seq methods are highly reproducible, but each has biases resulting from different modes of RNA sample preparation, reverse transcription, and adapter addition, leading to variability between methods. Moreover, the transcriptome cannot be profiled comprehensively because highly structured RNAs, such as tRNAs and snoRNAs, are refractory to conventional RNA-seq methods. Recently, we developed a new method for strand-specific RNA-seq using thermostable group II intron reverse transcriptases (TGIRTs). TGIRT enzymes have higher processivity and fidelity than conventional retroviral reverse transcriptases plus a novel template-switching activity that enables RNA-seq adapter addition during cDNA synthesis without using RNA ligase. Here, we obtained TGIRT-seq data sets for well-characterized human RNA reference samples and compared them to previous data sets obtained for these RNAs by the Illumina TruSeq v2 and v3 methods. We find that TGIRT-seq recapitulates the relative abundance of human transcripts and RNA spike-ins in ribo-depleted, fragmented RNA samples comparably to non-strand-specific TruSeq v2 and better than strand-specific TruSeq v3. Moreover, TGIRT-seq is more strand specific than TruSeq v3 and eliminates sampling biases from random hexamer priming, which are inherent to TruSeq. The TGIRT-seq data sets also show more uniform 5' to 3' gene coverage and identify more splice junctions, particularly near the 5' ends of mRNAs, than do the TruSeq data sets. Finally, TGIRT-seq enables the simultaneous profiling of mRNAs and lncRNAs in the same RNA-seq experiment as structured small ncRNAs, including tRNAs, which are essentially absent with TruSeq.


Subject(s)
RNA-Directed DNA Polymerase/chemistry , Base Sequence , Gene Expression Profiling/standards , Humans , RNA Splice Sites , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Reference Standards , Sequence Analysis, RNA/standards
15.
RNA ; 22(1): 111-28, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26554030

ABSTRACT

Next-generation RNA-sequencing (RNA-seq) has revolutionized transcriptome profiling, gene expression analysis, and RNA-based diagnostics. Here, we developed a new RNA-seq method that exploits thermostable group II intron reverse transcriptases (TGIRTs) and used it to profile human plasma RNAs. TGIRTs have higher thermostability, processivity, and fidelity than conventional reverse transcriptases, plus a novel template-switching activity that can efficiently attach RNA-seq adapters to target RNA sequences without RNA ligation. The new TGIRT-seq method enabled construction of RNA-seq libraries from <1 ng of plasma RNA in <5 h. TGIRT-seq of RNA in 1-mL plasma samples from a healthy individual revealed RNA fragments mapping to a diverse population of protein-coding gene and long ncRNAs, which are enriched in intron and antisense sequences, as well as nearly all known classes of small ncRNAs, some of which have never before been seen in plasma. Surprisingly, many of the small ncRNA species were present as full-length transcripts, suggesting that they are protected from plasma RNases in ribonucleoprotein (RNP) complexes and/or exosomes. This TGIRT-seq method is readily adaptable for profiling of whole-cell, exosomal, and miRNAs, and for related procedures, such as HITS-CLIP and ribosome profiling.


Subject(s)
High-Throughput Nucleotide Sequencing , Introns , RNA-Directed DNA Polymerase/metabolism , RNA/blood , Enzyme Stability , Hot Temperature , Humans
16.
Nature ; 490(7418): 121-5, 2012 Oct 04.
Article in English | MEDLINE | ID: mdl-22940866

ABSTRACT

DEAD-box proteins are the largest family of nucleic acid helicases, and are crucial to RNA metabolism throughout all domains of life. They contain a conserved 'helicase core' of two RecA-like domains (domains (D)1 and D2), which uses ATP to catalyse the unwinding of short RNA duplexes by non-processive, local strand separation. This mode of action differs from that of translocating helicases and allows DEAD-box proteins to remodel large RNAs and RNA-protein complexes without globally disrupting RNA structure. However, the structural basis for this distinctive mode of RNA unwinding remains unclear. Here, structural, biochemical and genetic analyses of the yeast DEAD-box protein Mss116p indicate that the helicase core domains have modular functions that enable a novel mechanism for RNA-duplex recognition and unwinding. By investigating D1 and D2 individually and together, we find that D1 acts as an ATP-binding domain and D2 functions as an RNA-duplex recognition domain. D2 contains a nucleic-acid-binding pocket that is formed by conserved DEAD-box protein sequence motifs and accommodates A-form but not B-form duplexes, providing a basis for RNA substrate specificity. Upon a conformational change in which the two core domains join to form a 'closed state' with an ATPase active site, conserved motifs in D1 promote the unwinding of duplex substrates bound to D2 by excluding one RNA strand and bending the other. Our results provide a comprehensive structural model for how DEAD-box proteins recognize and unwind RNA duplexes. This model explains key features of DEAD-box protein function and affords a new perspective on how the evolutionarily related cores of other RNA and DNA helicases diverged to use different mechanisms.


Subject(s)
DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , Nucleic Acid Conformation , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Base Sequence , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Evolution, Molecular , GC Rich Sequence/genetics , Models, Molecular , Protein Structure, Tertiary , RNA, Double-Stranded/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity
17.
PLoS Genet ; 11(8): e1005422, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26241656

ABSTRACT

Mobile bacterial group II introns are evolutionary ancestors of spliceosomal introns and retroelements in eukaryotes. They consist of an autocatalytic intron RNA (a "ribozyme") and an intron-encoded reverse transcriptase, which function together to promote intron integration into new DNA sites by a mechanism termed "retrohoming". Although mobile group II introns splice and retrohome efficiently in bacteria, all examined thus far function inefficiently in eukaryotes, where their ribozyme activity is limited by low Mg2+ concentrations, and intron-containing transcripts are subject to nonsense-mediated decay (NMD) and translational repression. Here, by using RNA polymerase II to express a humanized group II intron reverse transcriptase and T7 RNA polymerase to express intron transcripts resistant to NMD, we find that simply supplementing culture medium with Mg2+ induces the Lactococcus lactis Ll.LtrB intron to retrohome into plasmid and chromosomal sites, the latter at frequencies up to ~0.1%, in viable HEK-293 cells. Surprisingly, under these conditions, the Ll.LtrB intron reverse transcriptase is required for retrohoming but not for RNA splicing as in bacteria. By using a genetic assay for in vivo selections combined with deep sequencing, we identified intron RNA mutations that enhance retrohoming in human cells, but <4-fold and not without added Mg2+. Further, the selected mutations lie outside the ribozyme catalytic core, which appears not readily modified to function efficiently at low Mg2+ concentrations. Our results reveal differences between group II intron retrohoming in human cells and bacteria and suggest constraints on critical nucleotide residues of the ribozyme core that limit how much group II intron retrohoming in eukaryotes can be enhanced. These findings have implications for group II intron use for gene targeting in eukaryotes and suggest how differences in intracellular Mg2+ concentrations between bacteria and eukarya may have impacted the evolution of introns and gene expression mechanisms.


Subject(s)
Retroelements , Active Transport, Cell Nucleus , Bacterial Proteins/genetics , Base Sequence , Cell Survival , Directed Molecular Evolution , HEK293 Cells , Humans , Introns , Inverted Repeat Sequences , Molecular Sequence Data , Nonsense Mediated mRNA Decay , Plasmids/genetics , RNA-Directed DNA Polymerase/genetics
18.
J Biol Chem ; 291(22): 11911-27, 2016 May 27.
Article in English | MEDLINE | ID: mdl-27036943

ABSTRACT

The mitochondrial tyrosyl-tRNA synthetases (mtTyrRSs) of Pezizomycotina fungi, a subphylum that includes many pathogenic species, are bifunctional proteins that both charge mitochondrial tRNA(Tyr) and act as splicing cofactors for autocatalytic group I introns. Previous studies showed that one of these proteins, Neurospora crassa CYT-18, binds group I introns by using both its N-terminal catalytic and C-terminal anticodon binding domains and that the catalytic domain uses a newly evolved group I intron binding surface that includes an N-terminal extension and two small insertions (insertions 1 and 2) with distinctive features not found in non-splicing mtTyrRSs. To explore how this RNA binding surface diverged to accommodate different group I introns in other Pezizomycotina fungi, we determined x-ray crystal structures of C-terminally truncated Aspergillus nidulans and Coccidioides posadasii mtTyrRSs. Comparisons with previous N. crassa CYT-18 structures and a structural model of the Aspergillus fumigatus mtTyrRS showed that the overall topology of the group I intron binding surface is conserved but with variations in key intron binding regions, particularly the Pezizomycotina-specific insertions. These insertions, which arose by expansion of flexible termini or internal loops, show greater variation in structure and amino acids potentially involved in group I intron binding than do neighboring protein core regions, which also function in intron binding but may be more constrained to preserve mtTyrRS activity. Our results suggest a structural basis for the intron specificity of different Pezizomycotina mtTyrRSs, highlight flexible terminal and loop regions as major sites for enzyme diversification, and identify targets for therapeutic intervention by disrupting an essential RNA-protein interaction in pathogenic fungi.


Subject(s)
Aspergillus nidulans/enzymology , Coccidioides/enzymology , Introns/genetics , Mitochondria/enzymology , RNA Splicing/genetics , Tyrosine-tRNA Ligase/chemistry , Tyrosine-tRNA Ligase/metabolism , Amino Acid Sequence , Aspergillus nidulans/genetics , Aspergillus nidulans/growth & development , Catalytic Domain , Coccidioides/genetics , Coccidioides/growth & development , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Neurospora crassa/enzymology , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Tyrosine-tRNA Ligase/genetics
19.
PLoS Biol ; 12(12): e1002028, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25536042

ABSTRACT

The Neurospora crassa mitochondrial tyrosyl-tRNA synthetase (mtTyrRS; CYT-18 protein) evolved a new function as a group I intron splicing factor by acquiring the ability to bind group I intron RNAs and stabilize their catalytically active RNA structure. Previous studies showed: (i) CYT-18 binds group I introns by using both its N-terminal catalytic domain and flexibly attached C-terminal anticodon-binding domain (CTD); and (ii) the catalytic domain binds group I introns specifically via multiple structural adaptations that occurred during or after the divergence of Peziomycotina and Saccharomycotina. However, the function of the CTD and how it contributed to the evolution of splicing activity have been unclear. Here, small angle X-ray scattering analysis of CYT-18 shows that both CTDs of the homodimeric protein extend outward from the catalytic domain, but move inward to bind opposite ends of a group I intron RNA. Biochemical assays show that the isolated CTD of CYT-18 binds RNAs non-specifically, possibly contributing to its interaction with the structurally different ends of the intron RNA. Finally, we find that the yeast mtTyrRS, which diverged from Pezizomycotina fungal mtTyrRSs prior to the evolution of splicing activity, binds group I intron and other RNAs non-specifically via its CTD, but lacks further adaptations needed for group I intron splicing. Our results suggest a scenario of constructive neutral (i.e., pre-adaptive) evolution in which an initial non-specific interaction between the CTD of an ancestral fungal mtTyrRS and a self-splicing group I intron was "fixed" by an intron RNA mutation that resulted in protein-dependent splicing. Once fixed, this interaction could be elaborated by further adaptive mutations in both the catalytic domain and CTD that enabled specific binding of group I introns. Our results highlight a role for non-specific RNA binding in the evolution of RNA-binding proteins.


Subject(s)
Evolution, Molecular , Fungal Proteins/metabolism , Neurospora crassa/enzymology , RNA Splicing/genetics , RNA, Fungal/metabolism , Tyrosine-tRNA Ligase/metabolism , Amino Acid Sequence , Fungal Proteins/chemistry , Introns/genetics , Mitochondria/enzymology , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA, Catalytic/metabolism , RNA, Fungal/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Scattering, Small Angle , Sequence Alignment , Sequence Deletion , X-Ray Diffraction
20.
Mol Cell ; 35(5): 598-609, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19748356

ABSTRACT

The yeast DEAD box protein Mss116p is a general RNA chaperone that functions in mitochondrial group I and II intron splicing, translational activation, and RNA end processing. Here we determined high-resolution X-ray crystal structures of Mss116p complexed with an RNA oligonucleotide and ATP analogs AMP-PNP, ADP-BeF(3)(-), or ADP-AlF(4)(-). The structures show the entire helicase core acting together with a functionally important C-terminal extension. In all structures, the helicase core is in a closed conformation with a wedge alpha helix bending RNA 3' of the central bound nucleotides, as in previous DEAD box protein structures. Notably, Mss116p's C-terminal extension also bends RNA 5' of the central nucleotides, resulting in RNA crimping. Despite reported functional differences, we observe few structural changes in ternary complexes with different ATP analogs. The structures constrain models of DEAD box protein function and reveal a strand separation mechanism in which a protein uses two wedges to act as a molecular crimper.


Subject(s)
Adenosine Triphosphate/chemistry , DEAD-box RNA Helicases/chemistry , Poly U/chemistry , RNA/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/chemistry , Binding Sites , Crystallography, X-Ray , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Models, Molecular , Nucleic Acid Conformation , Organometallic Compounds/chemistry , Poly U/metabolism , Protein Conformation , Protein Structure, Tertiary , RNA/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL