Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Nat Rev Mol Cell Biol ; 24(10): 749-769, 2023 10.
Article in English | MEDLINE | ID: mdl-37474727

ABSTRACT

RNA helicases are highly conserved proteins that use nucleoside triphosphates to bind or remodel RNA, RNA-protein complexes or both. RNA helicases are classified into the DEAD-box, DEAH/RHA, Ski2-like, Upf1-like and RIG-I families, and are the largest class of enzymes active in eukaryotic RNA metabolism - virtually all aspects of gene expression and its regulation involve RNA helicases. Mutation and dysregulation of these enzymes have been linked to a multitude of diseases, including cancer and neurological disorders. In this Review, we discuss the regulation and functional mechanisms of RNA helicases and their roles in eukaryotic RNA metabolism, including in transcription regulation, pre-mRNA splicing, ribosome assembly, translation and RNA decay. We highlight intriguing models that link helicase structure, mechanisms of function (such as local strand unwinding, translocation, winching, RNA clamping and displacing RNA-binding proteins) and biological roles, including emerging connections between RNA helicases and cellular condensates formed through liquid-liquid phase separation. We also discuss associations of RNA helicases with human diseases and recent efforts towards the design of small-molecule inhibitors of these pivotal regulators of eukaryotic gene expression.


Subject(s)
RNA Helicases , RNA , Humans , RNA Helicases/genetics , RNA Helicases/metabolism , RNA/metabolism , Eukaryotic Cells/metabolism , DNA Helicases , Saccharomyces cerevisiae/genetics
2.
Mol Cell ; 81(20): 4191-4208.e8, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34686314

ABSTRACT

To survive, mammalian cells must adapt to environmental challenges. While the cellular response to mild stress has been widely studied, how cells respond to severe stress remains unclear. We show here that under severe hyperosmotic stress, cells enter a transient hibernation-like state in anticipation of recovery. We demonstrate this adaptive pausing response (APR) is a coordinated cellular response that limits ATP supply and consumption through mitochondrial fragmentation and widespread pausing of mRNA translation. This pausing is accomplished by ribosome stalling at translation initiation codons, which keeps mRNAs poised to resume translation upon recovery. We further show that recovery from severe stress involves ISR (integrated stress response) signaling that permits cell cycle progression, resumption of growth, and reversal of mitochondria fragmentation. Our findings indicate that cells can respond to severe stress via a hibernation-like mechanism that preserves vital elements of cellular function under harsh environmental conditions.


Subject(s)
Cell Proliferation , Fibroblasts/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Osmotic Pressure , Protein Biosynthesis , Ribosomes/metabolism , Adaptation, Physiological , Adenosine Triphosphate/metabolism , Animals , Codon, Initiator , Fibroblasts/pathology , HEK293 Cells , Humans , Kinetics , Mice , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Ribosomes/genetics , Signal Transduction
3.
Nat Rev Mol Cell Biol ; 16(9): 533-44, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26285679

ABSTRACT

To fully understand the regulation of gene expression, it is critical to quantitatively define whether and how RNA-binding proteins (RBPs) discriminate between alternative binding sites in RNAs. Here, we describe new methods that measure protein binding to large numbers of RNA variants, and ways to analyse and interpret data obtained by these approaches, including affinity distributions and free energy landscapes. We discuss how the new methodologies and the associated concepts enable the development of inclusive, quantitative models for RNA-protein interactions that transcend the traditional binary classification of RBPs as either specific or nonspecific.


Subject(s)
RNA-Binding Proteins/physiology , RNA/physiology , Animals , Base Sequence , Humans , Kinetics , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Conformation , RNA/chemistry , RNA-Binding Proteins/chemistry , Thermodynamics
4.
Nature ; 591(7848): 152-156, 2021 03.
Article in English | MEDLINE | ID: mdl-33568810

ABSTRACT

Gene expression in higher eukaryotic cells orchestrates interactions between thousands of RNA-binding proteins (RBPs) and tens of thousands of RNAs1. The kinetics by which RBPs bind to and dissociate from their RNA sites are critical for the coordination of cellular RNA-protein interactions2. However, these kinetic parameters have not been experimentally measured in cells. Here we show that time-resolved RNA-protein cross-linking with a pulsed femtosecond ultraviolet laser, followed by immunoprecipitation and high-throughput sequencing, allows the determination of binding and dissociation kinetics of the RBP DAZL for thousands of individual RNA-binding sites in cells. This kinetic cross-linking and immunoprecipitation (KIN-CLIP) approach reveals that DAZL resides at individual binding sites for time periods of only seconds or shorter, whereas the binding sites remain DAZL-free for markedly longer. The data also indicate that DAZL binds to many RNAs in clusters of multiple proximal sites. The effect of DAZL on mRNA levels and ribosome association correlates with the cumulative probability of DAZL binding in these clusters. Integrating kinetic data with mRNA features quantitatively connects DAZL-RNA binding to DAZL function. Our results show how kinetic parameters for RNA-protein interactions can be measured in cells, and how these data link RBP-RNA binding to the cellular function of RBPs.


Subject(s)
RNA-Binding Proteins/metabolism , RNA/metabolism , 3' Untranslated Regions/genetics , Animals , Binding Sites/genetics , Cell Line , Kinetics , Lasers , Mice , Protein Binding , RNA/genetics , RNA-Binding Proteins/genetics , Ribosomes/metabolism
5.
Development ; 150(20)2023 10 15.
Article in English | MEDLINE | ID: mdl-37306388

ABSTRACT

The eIF4E family of translation initiation factors bind 5' methylated caps and act as the limiting step for mRNA translation. The canonical eIF4E1A is required for cell viability, yet other related eIF4E families exist and are utilized in specific contexts or tissues. Here, we describe a family called Eif4e1c, for which we find roles during heart development and regeneration in zebrafish. The Eif4e1c family is present in all aquatic vertebrates but is lost in all terrestrial species. A core group of amino acids shared over 500 million years of evolution forms an interface along the protein surface, suggesting that Eif4e1c functions in a novel pathway. Deletion of eif4e1c in zebrafish caused growth deficits and impaired survival in juveniles. Mutants surviving to adulthood had fewer cardiomyocytes and reduced proliferative responses to cardiac injury. Ribosome profiling of mutant hearts demonstrated changes in translation efficiency of mRNA for genes known to regulate cardiomyocyte proliferation. Although eif4e1c is broadly expressed, its disruption had most notable impact on the heart and at juvenile stages. Our findings reveal context-dependent requirements for translation initiation regulators during heart regeneration.


Subject(s)
Heart Injuries , Myocytes, Cardiac , Animals , Zebrafish/genetics , Eukaryotic Initiation Factor-4E/genetics , Cell Proliferation/genetics
6.
Cell ; 145(6): 890-901, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663793

ABSTRACT

Many steps in nuclear RNA processing, surveillance, and degradation require TRAMP, a complex containing the poly(A) polymerase Trf4p, the Zn-knuckle protein Air2p, and the RNA helicase Mtr4p. TRAMP polyadenylates RNAs designated for decay or trimming by the nuclear exosome. It has been unclear how polyadenylation by TRAMP differs from polyadenylation by conventional poly(A) polymerase, which produces poly(A) tails that stabilize RNAs. Using reconstituted S. cerevisiae TRAMP, we show that TRAMP inherently suppresses poly(A) addition after only 3-4 adenosines. This poly(A) tail length restriction is controlled by Mtr4p. The helicase detects the number of 3'-terminal adenosines and, over several adenylation steps, elicits precisely tuned adjustments of ATP affinities and rate constants for adenylation and TRAMP dissociation. Our data establish Mtr4p as a critical regulator of polyadenylation by TRAMP and reveal that an RNA helicase can control the activity of another enzyme in a highly complex fashion and in response to features in RNA.


Subject(s)
DEAD-box RNA Helicases/metabolism , Polyadenylation , RNA, Fungal/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adenosine/metabolism , DNA-Directed DNA Polymerase/metabolism , Multiprotein Complexes/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics
7.
Nature ; 559(7712): 130-134, 2018 07.
Article in English | MEDLINE | ID: mdl-29950728

ABSTRACT

The conserved and essential DEAD-box RNA helicase Ded1p from yeast and its mammalian orthologue DDX3 are critical for the initiation of translation1. Mutations in DDX3 are linked to tumorigenesis2-4 and intellectual disability5, and the enzyme is targeted by a range of viruses6. How Ded1p and its orthologues engage RNAs during the initiation of translation is unknown. Here we show, by integrating transcriptome-wide analyses of translation, RNA structure and Ded1p-RNA binding, that the effects of Ded1p on the initiation of translation are connected to near-cognate initiation codons in 5' untranslated regions. Ded1p associates with the translation pre-initiation complex at the mRNA entry channel and repressing the activity of Ded1p leads to the accumulation of RNA structure in 5' untranslated regions, the initiation of translation from near-cognate start codons immediately upstream of these structures and decreased protein synthesis from the corresponding main open reading frames. The data reveal a program for the regulation of translation that links Ded1p, the activation of near-cognate start codons and mRNA structure. This program has a role in meiosis, in which a marked decrease in the levels of Ded1p is accompanied by the activation of the alternative translation initiation sites that are seen when the activity of Ded1p is repressed. Our observations indicate that Ded1p affects translation initiation by controlling the use of near-cognate initiation codons that are proximal to mRNA structure in 5' untranslated regions.


Subject(s)
5' Untranslated Regions/genetics , Codon, Initiator/genetics , DEAD-box RNA Helicases/metabolism , Peptide Chain Initiation, Translational/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Cross-Linking Reagents/chemistry , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/metabolism
8.
RNA ; 27(4): 465-476, 2021 04.
Article in English | MEDLINE | ID: mdl-33408095

ABSTRACT

The 3' to 5' exonuclease Pop2p (Caf1p) is part of the CCR4-NOT deadenylation complex that removes poly(A) tails from mRNAs in cells. Pop2p is structurally conserved in eukaryotes, but Saccharomyces cerevisiae Pop2p harbors noncanonical amino acids in its catalytic center. The enzymatic properties of S. cerevisiae Pop2p are not well defined. Here we characterize the RNA exonuclease activity of recombinant S. cerevisiae Pop2p. We find that S. cerevisiae Pop2p degrades RNAs via two alternative reactions pathways, one generating nucleotides with 5'-phosphates and RNA intermediates with 3'-hydroxyls, and the other generating nucleotides with 3'-phosphates and RNA intermediates with 3'-phosphates. The enzyme is not able to initiate the reaction on RNAs with a 3'-phosphate, which leads to accumulation of RNAs with 3'-phosphates that can exceed 10 nt and are resistant to further degradation by S. cerevisiae Pop2p. We further demonstrate that S. cerevisiae Pop2p degrades RNAs in three reaction phases: an initial distributive phase, a second processive phase and a third phase during which processivity gradually declines. We also show that mutations of subsets of amino acids in the catalytic center, including those previously thought to inactivate the enzyme, moderately reduce, but not eliminate activity. Only mutation of all five amino acids in the catalytic center diminishes activity of Pop2p to background levels. Collectively, our results reveal robust exonuclease activity of S. cerevisiae Pop2p with unusual enzymatic properties, characterized by alternative degradation pathways, multiple reaction phases and functional redundancy of amino acids in the catalytic core.


Subject(s)
Amino Acids/chemistry , RNA Stability , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Ribonucleases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Adenosine Monophosphate/metabolism , Amino Acid Substitution , Amino Acids/metabolism , Binding Sites , Catalytic Domain , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Models, Molecular , Mutation , Phosphates/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA, Messenger/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Ribonucleases/chemistry , Ribonucleases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity
9.
Nat Rev Mol Cell Biol ; 12(8): 505-16, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21779027

ABSTRACT

RNA helicases of the DEAD box family are present in all eukaryotic cells and in many bacteria and Archaea. These highly conserved enzymes are required for RNA metabolism from transcription to degradation and are therefore important players in gene expression. DEAD box proteins use ATP to unwind short duplex RNA in an unusual fashion and remodel RNA-protein complexes, but they can also function as ATP-dependent RNA clamps to provide nucleation centres that establish larger RNA-protein complexes. Structural, mechanistic and molecular biological studies have started to reveal how these conserved proteins can perform such diverse functions and how accessory proteins have a central role in their regulation.


Subject(s)
DEAD-box RNA Helicases/metabolism , Adenosine Triphosphate/metabolism , Animals , Binding Sites , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4A/metabolism , Humans , Models, Biological , Models, Molecular , Nucleocytoplasmic Transport Proteins/metabolism , Protein Biosynthesis , Protein Structure, Tertiary , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Saccharomyces cerevisiae/metabolism
10.
Methods ; 204: 376-385, 2022 08.
Article in English | MEDLINE | ID: mdl-35429628

ABSTRACT

RNA helicases are the largest class of enzymes in eukaryotic RNA metabolism. In cells, protein cofactors regulate RNA helicase functions and impact biochemical helicase activities. Understanding how cofactors affect enzymatic activities of RNA helicases is thus critical for delineating physical roles and regulation of RNA helicases in cells. Here, we discuss approaches and conceptual considerations for the design of experiments to interrogate cofactor effects on RNA helicase activities in vitro. We outline the mechanistic frame for helicase reactions, discuss optimization of experimental setup and reaction parameters for measuring cofactor effects on RNA helicase activities, and provide basic guides to data analysis and interpretation. The described approaches are also instructive for determining the impact of small molecule inhibitors of RNA helicases.


Subject(s)
DEAD-box RNA Helicases , RNA , DEAD-box RNA Helicases/chemistry , RNA/chemistry
11.
Cell ; 135(7): 1224-36, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19109894

ABSTRACT

Alternative splicing makes a major contribution to proteomic diversity in higher eukaryotes with approximately 70% of genes encoding two or more isoforms. In most cases, the molecular mechanisms responsible for splice site choice remain poorly understood. Here, we used a randomization-selection approach in vitro to identify sequence elements that could silence a proximal strong 5' splice site located downstream of a weakened 5' splice site. We recovered two exonic and four intronic motifs that effectively silenced the proximal 5' splice site both in vitro and in vivo. Surprisingly, silencing was only observed in the presence of the competing upstream 5' splice site. Biochemical evidence strongly suggests that the silencing motifs function by altering the U1 snRNP/5' splice site complex in a manner that impairs commitment to specific splice site pairing. The data indicate that perturbations of non-rate-limiting step(s) in splicing can lead to dramatic shifts in splice site choice.


Subject(s)
Alternative Splicing , Gene Expression Regulation , RNA Splice Sites , Exons , Genetic Techniques , HeLa Cells , Humans , Models, Biological
12.
Mol Cell ; 59(4): 541-52, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26212457

ABSTRACT

Most aspects of RNA metabolism involve DEAD-box RNA helicases, enzymes that bind and remodel RNA and RNA-protein complexes in an ATP-dependent manner. Here we show that the DEAD-box helicase Ded1p oligomerizes in the cell and in vitro, and unwinds RNA as a trimer. Two protomers bind the single-stranded region of RNA substrates and load a third protomer to the duplex, which then separates the strands. ATP utilization differs between the strand-separating protomer and those bound to the single-stranded region. Binding of the eukaryotic initiation factor 4G to Ded1p interferes with oligomerization and thereby modulates unwinding activity and RNA affinity of the helicase. Our data reveal a strict division of labor between the Ded1p protomers in the oligomer. This mode of oligomerization fundamentally differs from other helicases. Oligomerization represents a previously unappreciated level of regulation for DEAD-box helicase activities.


Subject(s)
DEAD-box RNA Helicases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Adenosine Triphosphate/chemistry , Biocatalysis , DEAD-box RNA Helicases/physiology , Hydrolysis , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/chemistry , RNA, Double-Stranded/chemistry , Saccharomyces cerevisiae Proteins/physiology
13.
Proc Natl Acad Sci U S A ; 117(2): 982-992, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31879344

ABSTRACT

The exoribonuclease Rrp6p is critical for RNA decay in the nucleus. While Rrp6p acts on a large range of diverse substrates, it does not indiscriminately degrade all RNAs. How Rrp6p accomplishes this task is not understood. Here, we measure Rrp6p-RNA binding and degradation kinetics in vitro at single-nucleotide resolution and find an intrinsic substrate selectivity that enables Rrp6p to discriminate against specific RNAs. RNA length and the four 3'-terminal nucleotides contribute most to substrate selectivity and collectively enable Rrp6p to discriminate between different RNAs by several orders of magnitude. The most pronounced discrimination is seen against RNAs ending with CCA-3'. These RNAs correspond to 3' termini of uncharged tRNAs, which are not targeted by Rrp6p in cells. The data show that in contrast to many other proteins that use substrate selectivity to preferentially interact with specific RNAs, Rrp6p utilizes its selectivity to discriminate against specific RNAs. This ability allows Rrp6p to target diverse substrates while avoiding a subset of RNAs.


Subject(s)
Exoribonucleases/metabolism , RNA Stability , RNA-Binding Proteins/metabolism , RNA/metabolism , Escherichia coli , Exoribonucleases/chemistry , Exosome Multienzyme Ribonuclease Complex/metabolism , Kinetics , RNA/chemistry , RNA, Transfer/metabolism , Substrate Specificity
14.
RNA ; 26(5): 541-549, 2020 05.
Article in English | MEDLINE | ID: mdl-32014999

ABSTRACT

The PI3K/Akt/mTOR kinase pathway is extensively deregulated in human cancers. One critical node under regulation of this signaling axis is eukaryotic initiation factor (eIF) 4F, a complex involved in the control of translation initiation rates. eIF4F-dependent addictions arise during tumor initiation and maintenance due to increased eIF4F activity-generally in response to elevated PI3K/Akt/mTOR signaling flux. There is thus much interest in exploring eIF4F as a small molecule target for the development of new anticancer drugs. The DEAD-box RNA helicase, eIF4A, is an essential subunit of eIF4F, and several potent small molecules (rocaglates, hippuristanol, pateamine A) affecting its activity have been identified and shown to demonstrate anticancer activity in vitro and in vivo in preclinical models. Recently, a number of new small molecules have been reported as having the capacity to target and inhibit eIF4A. Here, we undertook a comparative analysis of their biological activity and specificity relative to the eIF4A inhibitor, hippuristanol.


Subject(s)
Antineoplastic Agents/chemistry , Eukaryotic Initiation Factor-4A/chemistry , Neoplasms/drug therapy , Small Molecule Libraries/chemistry , Sterols/chemistry , Antineoplastic Agents/pharmacology , Benzofurans/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Epoxy Compounds/chemistry , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Eukaryotic Initiation Factor-4F/chemistry , Humans , Macrolides/chemistry , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/genetics , Small Molecule Libraries/pharmacology , Sterols/pharmacology , TOR Serine-Threonine Kinases/genetics , Thiazoles/chemistry
15.
Nucleic Acids Res ; 48(14): 8063-8073, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32609821

ABSTRACT

The mechanism for how internal ribosome entry sites (IRESs) recruit ribosomes to initiate translation of an mRNA is not completely understood. We investigated how a 40S subunit was recruited by the cricket paralysis virus intergenic region (CrPV IGR) IRES to form a stable 40S-IRES complex. Kinetic binding studies revealed that formation of the complex between the CrPV IGR and the 40S subunit consisted of two-steps: an initial fast binding step of the IRES to the 40S ribosomal subunit, followed by a slow unimolecular reaction consistent with a conformational change that stabilized the complex. We further showed that the ribosomal protein S25 (eS25), which is required by functionally and structurally diverse IRESs, impacts both steps of the complex formation. Mutations in eS25 that reduced CrPV IGR IRES activity either decreased 40S-IRES complex formation, or increased the rate of the conformational change that was required to form a stable 40S-IRES complex. Our data are consistent with a model in which eS25 facilitates initial binding of the CrPV IGR IRES to the 40S while ensuring that the conformational change stabilizing the 40S-IRES complex does not occur prematurely.


Subject(s)
Internal Ribosome Entry Sites , Ribosomal Proteins/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Binding Sites , DNA, Intergenic/genetics , DNA, Intergenic/metabolism , Dicistroviridae/genetics , Mutation , Protein Binding , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/genetics , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
16.
Methods ; 178: 3-10, 2020 06 01.
Article in English | MEDLINE | ID: mdl-31494245

ABSTRACT

To understand the regulation of gene expression it is critical to determine how proteins interact with and discriminate between different RNAs. In this review, we discuss experimental techniques that utilize high throughput approaches to characterize the interactions of proteins with large numbers of RNAs in vitro. We describe the underlying principles for the main methods, briefly discuss their scope and limitations, and outline how insight from the techniques contributes to our understanding of specificity for RNA-protein interactions.


Subject(s)
High-Throughput Nucleotide Sequencing/trends , High-Throughput Screening Assays/trends , RNA-Binding Proteins/isolation & purification , RNA/genetics , RNA/isolation & purification , RNA-Binding Proteins/genetics
17.
18.
RNA ; 24(12): 1693-1705, 2018 12.
Article in English | MEDLINE | ID: mdl-30262458

ABSTRACT

The DEAD-box RNA helicase Dbp2p is highly conserved in eukaryotes and has been implicated in transcription, ribosome biogenesis, mRNP assembly, nuclear export, and long noncoding RNA (lncRNA) function. It is not understood how Dbp2p performs these seemingly unrelated biological roles. An important step toward addressing this question is the determination of cellular RNA binding sites of Dbp2p. Here, we identify transcriptome-wide RNA binding sites of Dbp2p from Saccharomyces cerevisiae using UV-crosslinking, denaturing tandem affinity purification, and next generation sequencing. We find that Dbp2p crosslinks to mRNAs and ribosomal RNAs, and markedly to noncoding RNAs, including snoRNA, snRNAs, and tRNAs. In snoRNAs, Dbp2p preferentially crosslinks at sites near the 3' ends. These sites coincide with regions where RNA-DNA hybrids (R-loops) form and with binding sites of Sen1p, another RNA helicase that functions in transcription termination and 3' processing of noncoding RNAs. We show that Dbp2p interacts in an RNA-independent manner with Sen1p in vivo. Dbp2p crosslinks to tRNAs and other RNAs also at sites where R-loops form. Collectively, our data link Dbp2p to noncoding RNAs, Sen1p, and R-loops. The transcriptome-wide connection to R-loops provides a unifying theme for diverse cellular roles of Dbp2p.


Subject(s)
DEAD-box RNA Helicases/chemistry , Proteasome Endopeptidase Complex/chemistry , RNA, Untranslated/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Binding Sites , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Fungal , Proteasome Endopeptidase Complex/genetics , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/genetics , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Untranslated/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
19.
Curr Genet ; 65(2): 453-456, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30483885

ABSTRACT

Upstream open reading frames (uORFs) in 5' UTRs of eukaryotic mRNAs are increasingly recognized as important elements that regulate cellular protein synthesis. Since uORFs can start from non-AUG codons, an enormous number of potential uORF initiation sites exists in 5'UTRs. However, only a subset of these sites is used and it has been unclear how actual start sites are selected. Studies of the DEAD-box helicase Ded1p from S. cerevisiae show that translation of uORFs with non-AUG initiation codons occurs upstream of mRNA structures that emerge with defective Ded1p. The data designate mRNA structure as important determinant for non-AUG initiation sites of uORFs. Ded1p can control this RNA structure and thereby regulate uORF translation.


Subject(s)
5' Untranslated Regions , DNA Helicases/metabolism , Open Reading Frames , RNA, Messenger/chemistry , RNA, Messenger/genetics , Codon , Gene Expression Regulation, Fungal , Meiosis/genetics , Protein Biosynthesis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
20.
RNA ; 23(10): 1502-1511, 2017 10.
Article in English | MEDLINE | ID: mdl-28694328

ABSTRACT

Recognition of RNA by RNA processing enzymes and RNA binding proteins often involves cooperation between multiple subunits. However, the interdependent contributions of RNA and protein subunits to molecular recognition by ribonucleoproteins are relatively unexplored. RNase P is an endonuclease that removes 5' leaders from precursor tRNAs and functions in bacteria as a dimer formed by a catalytic RNA subunit (P RNA) and a protein subunit (C5 in E. coli). The P RNA subunit contacts the tRNA body and proximal 5' leader sequences [N(-1) and N(-2)] while C5 binds distal 5' leader sequences [N(-3) to N(-6)]. To determine whether the contacts formed by P RNA and C5 contribute independently to specificity or exhibit cooperativity or anti-cooperativity, we compared the relative kcat/Km values for all possible combinations of the six proximal 5' leader nucleotides (n = 4096) for processing by the E. coli P RNA subunit alone and by the RNase P holoenzyme. We observed that while the P RNA subunit shows specificity for 5' leader nucleotides N(-2) and N(-1), the presence of the C5 protein reduces the contribution of P RNA to specificity, but changes specificity at N(-2) and N(-3). The results reveal that the contribution of C5 protein to RNase P processing is controlled by the identity of N(-2) in the pre-tRNA 5' leader. The data also clearly show that pairing of the 5' leader with the 3' ACCA of tRNA acts as an anti-determinant for RNase P cleavage. Comparative analysis of genomically encoded E. coli tRNAs reveals that both anti-determinants are subject to negative selection in vivo.


Subject(s)
Escherichia coli Proteins/metabolism , RNA Precursors/metabolism , RNA, Transfer/metabolism , Ribonuclease P/metabolism , Escherichia coli Proteins/chemistry , Nucleotides/chemistry , Nucleotides/metabolism , RNA Precursors/chemistry , RNA, Transfer/chemistry , RNA, Transfer, Met/chemistry , RNA, Transfer, Met/metabolism , Ribonuclease P/chemistry , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL