Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(11): e0142559, 2015.
Article in English | MEDLINE | ID: mdl-26545116

ABSTRACT

In support of the RNA world hypothesis, previous studies identified trimetaphosphate (Tmp) as a plausible energy source for RNA world organisms. In one of these studies, catalytic RNAs (ribozymes) that catalyze the triphosphorylation of RNA 5'-hydroxyl groups using Tmp were obtained by in vitro selection. One ribozyme (TPR1) was analyzed in more detail. TPR1 catalyzes the triphosphorylation reaction to a rate of 0.013 min-1 under selection conditions (50 mM Tmp, 100 mM MgCl2, 22°C). To identify a triphosphorylation ribozyme that catalyzes faster triphosphorylation, and possibly learn about its secondary structure TPR1 was subjected to a doped selection. The resulting ribozyme, TPR1e, contains seven mutations relative to TPR1, displays a previously unidentified duplex that constrains the ribozyme's structure, and reacts at a 24-fold faster rate than the parent ribozyme. Under optimal conditions (150 mM Tmp, 650 mM MgCl2, 40°C), the triphosphorylation rate of TRP1e reaches 6.8 min-1.


Subject(s)
RNA, Catalytic/metabolism , Base Sequence , Evolution, Molecular , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleic Acid Conformation , Phosphorylation , RNA, Catalytic/chemistry , RNA, Catalytic/genetics
2.
PLoS One ; 9(1): e86473, 2014.
Article in English | MEDLINE | ID: mdl-24466112

ABSTRACT

How does a non-coding RNA evolve in cells? To address this question experimentally we evolved a trans-splicing variant of the group I intron ribozyme from Tetrahymena over 21 cycles of evolution in E.coli cells. Sequence variation was introduced during the evolution by mutagenic and recombinative PCR, and increasingly active ribozymes were selected by their repair of an mRNA mediating antibiotic resistance. The most efficient ribozyme contained four clustered mutations that were necessary and sufficient for maximum activity in cells. Surprisingly, these mutations did not increase the trans-splicing activity of the ribozyme. Instead, they appear to have recruited a cellular protein, the transcription termination factor Rho, and facilitated more efficient translation of the ribozyme's trans-splicing product. In addition, these mutations affected the expression of several other, unrelated genes. These results suggest that during RNA evolution in cells, four mutations can be sufficient to evolve new protein interactions, and four mutations in an RNA molecule can generate a large effect on gene regulation in the cell.


Subject(s)
Evolution, Molecular , Introns/genetics , Protein Biosynthesis/genetics , RNA Splicing/genetics , RNA, Catalytic/genetics , Tetrahymena/enzymology , Trans-Splicing/genetics , Base Sequence , Chloramphenicol/pharmacology , Chloramphenicol O-Acetyltransferase/metabolism , Drug Resistance, Microbial/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Molecular Sequence Data , Mutation/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tetrahymena/genetics , Tetrahymena/growth & development , Transcription, Genetic
3.
RNA ; 20(2): 202-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344321

ABSTRACT

Group I introns are ribozymes (catalytic RNAs) that excise themselves from RNA primary transcripts by catalyzing two successive transesterification reactions. These cis-splicing ribozymes can be converted into trans-splicing ribozymes, which can modify the sequence of a separate substrate RNA, both in vitro and in vivo. Previous work on trans-splicing ribozymes has mostly focused on the 16S rRNA group I intron ribozyme from Tetrahymena thermophila. Here, we test the trans-splicing potential of the tRNA(Ile) group I intron ribozyme from the bacterium Azoarcus. This ribozyme is only half the size of the Tetrahymena ribozyme and folds faster into its active conformation in vitro. Our results showed that in vitro, the Azoarcus and Tetrahymena ribozymes favored the same set of splice sites on a substrate RNA. Both ribozymes showed the same trans-splicing efficiency when containing their individually optimized 5' terminus. In contrast to the previously optimized 5'-terminal design of the Tetrahymena ribozyme, the Azoarcus ribozyme was most efficient with a trans-splicing design that resembled the secondary structure context of the natural cis-splicing Azoarcus ribozyme, which includes base-pairing between the substrate 5' portion and the ribozyme 3' exon. These results suggested preferred trans-splicing interactions for the Azoarcus ribozyme under near-physiological in vitro conditions. Despite the high activity in vitro, however, the splicing efficiency of the Azoarcus ribozyme in Escherichia coli cells was significantly below that of the Tetrahymena ribozyme.


Subject(s)
Azoarcus/genetics , RNA, Bacterial/chemistry , RNA, Catalytic/chemistry , RNA, Messenger/genetics , Trans-Splicing , Azoarcus/enzymology , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , Escherichia coli , Inverted Repeat Sequences , Molecular Sequence Data , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , RNA, Messenger/chemistry , RNA, Protozoan/chemistry , RNA, Protozoan/genetics , RNA, Protozoan/metabolism , Substrate Specificity , Tetrahymena thermophila/enzymology
4.
RNA ; 18(3): 590-602, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22274956

ABSTRACT

Group I introns have been engineered into trans-splicing ribozymes capable of replacing the 3'-terminal portion of an external mRNA with their own 3'-exon. Although this design makes trans-splicing ribozymes potentially useful for therapeutic application, their trans-splicing efficiency is usually too low for medical use. One factor that strongly influences trans-splicing efficiency is the position of the target splice site on the mRNA substrate. Viable splice sites are currently determined using a biochemical trans-tagging assay. Here, we propose a rapid and inexpensive alternative approach to identify efficient splice sites. This approach involves the computation of the binding free energies between ribozyme and mRNA substrate. We found that the computed binding free energies correlate well with the trans-splicing efficiency experimentally determined at 18 different splice sites on the mRNA of chloramphenicol acetyl transferase. In contrast, our results from the trans-tagging assay correlate less well with measured trans-splicing efficiency. The computed free energy components suggest that splice site efficiency depends on the following secondary structure rearrangements: hybridization of the ribozyme's internal guide sequence (IGS) with mRNA substrate (most important), unfolding of substrate proximal to the splice site, and release of the IGS from the 3'-exon (least important). The proposed computational approach can also be extended to fulfill additional design requirements of efficient trans-splicing ribozymes, such as the optimization of 3'-exon and extended guide sequences.


Subject(s)
Computational Biology/methods , RNA Splice Sites , RNA, Catalytic/genetics , Trans-Splicing/genetics , Nucleic Acid Conformation , RNA, Catalytic/metabolism , RNA, Messenger/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...