Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
ChemMedChem ; 19(7): e202300600, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38235959

ABSTRACT

All widely used mRNA vaccines against COVID-19 contain in their sequence 1-methylpseudouridine (m1Ψ) instead of uridine. In this publication, we report two high resolution crystal structures (at up to 1.01 and 1.32 Å, respectively) of one such double-stranded 12-mer RNA sequence crystallized in two crystal forms. The structures are compared with similar structures which do not contain this modification. Additionally, the X-ray structure of 1-methyl-pseudouridine itself was determined.


Subject(s)
Pseudouridine , Pseudouridine/analogs & derivatives , RNA , Humans , Pseudouridine/chemistry , mRNA Vaccines , COVID-19 Vaccines
2.
Nature ; 625(7993): 189-194, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38057663

ABSTRACT

In vitro-transcribed (IVT) mRNAs are modalities that can combat human disease, exemplified by their use as vaccines for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). IVT mRNAs are transfected into target cells, where they are translated into recombinant protein, and the biological activity or immunogenicity of the encoded protein exerts an intended therapeutic effect1,2. Modified ribonucleotides are commonly incorporated into therapeutic IVT mRNAs to decrease their innate immunogenicity3-5, but their effects on mRNA translation fidelity have not been fully explored. Here we demonstrate that incorporation of N1-methylpseudouridine into mRNA results in +1 ribosomal frameshifting in vitro and that cellular immunity in mice and humans to +1 frameshifted products from BNT162b2 vaccine mRNA translation occurs after vaccination. The +1 ribosome frameshifting observed is probably a consequence of N1-methylpseudouridine-induced ribosome stalling during IVT mRNA translation, with frameshifting occurring at ribosome slippery sequences. However, we demonstrate that synonymous targeting of such slippery sequences provides an effective strategy to reduce the production of frameshifted products. Overall, these data increase our understanding of how modified ribonucleotides affect the fidelity of mRNA translation, and although there are no adverse outcomes reported from mistranslation of mRNA-based SARS-CoV-2 vaccines in humans, these data highlight potential off-target effects for future mRNA-based therapeutics and demonstrate the requirement for sequence optimization.


Subject(s)
Frameshifting, Ribosomal , Pseudouridine , RNA, Messenger , Animals , Humans , Mice , BNT162 Vaccine/adverse effects , BNT162 Vaccine/genetics , BNT162 Vaccine/immunology , Frameshifting, Ribosomal/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Pseudouridine/analogs & derivatives , Pseudouridine/metabolism , Ribosomes/metabolism , Protein Biosynthesis
3.
Nucleic Acids Res ; 50(13): 7202-7215, 2022 07 22.
Article in English | MEDLINE | ID: mdl-34933339

ABSTRACT

Expression of therapeutically important proteins has benefited dramatically from the advent of chemically modified mRNAs that feature decreased lability and immunogenicity. This had a momentous effect on the rapid development of COVID-19 mRNA vaccines. Incorporation of the naturally occurring pseudouridine (Ψ) or N1-methyl-pseudouridine (N1mΨ) into in vitro transcribed mRNAs prevents the activation of unwanted immune responses by blocking eIF2α phosphorylation, which inhibits translation. Here, we report that Ψs in luciferase (Luc) mRNA exacerbate translation pausing in nuclease-untreated rabbit reticulocyte lysate (uRRL) and promote the formation of high-order-ribosome structures. The major deceleration of elongation occurs at the Ψ-rich nucleotides 1294-1326 of Ψ-Luc mRNA and results in premature termination of translation. The impairment of translation is mainly due to the shortage of membranous components. Supplementing uRRL with canine microsomal membranes (CMMs) relaxes the impediments to ribosome movement, resolves collided ribosomes, and greatly enhances full-size luciferase production. CMMs also strongly stimulated an extremely inefficient translation of N1mΨ-Luc mRNA in uRRL. Evidence is presented that translational pausing can promote membrane recruitment of polysomes with nascent polypeptides that lack a signal sequence. Our results highlight an underappreciated role of membrane binding to polysomes in the prevention of ribosome collision and premature release of nascent polypeptides.


Subject(s)
COVID-19 , Intracellular Membranes/metabolism , Peptide Chain Elongation, Translational , Pseudouridine , RNA, Messenger , Animals , Dogs , In Vitro Techniques , Peptides/metabolism , Pseudouridine/analogs & derivatives , Pseudouridine/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Rabbits
4.
J Mol Biol ; 433(15): 167097, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34107280

ABSTRACT

DNA glycosylases remove damaged or modified nucleobases by cleaving the N-glycosyl bond and the correct nucleotide is restored through subsequent base excision repair. In addition to excising threatening lesions, DNA glycosylases contribute to epigenetic regulation by mediating DNA demethylation and perform other important functions. However, the catalytic mechanism remains poorly defined for many glycosylases, including MBD4 (methyl-CpG binding domain IV), a member of the helix-hairpin-helix (HhH) superfamily. MBD4 excises thymine from G·T mispairs, suppressing mutations caused by deamination of 5-methylcytosine, and it removes uracil and modified uracils (e.g., 5-hydroxymethyluracil) mispaired with guanine. To investigate the mechanism of MBD4 we solved high-resolution structures of enzyme-DNA complexes at three stages of catalysis. Using a non-cleavable substrate analog, 2'-deoxy-pseudouridine, we determined the first structure of an enzyme-substrate complex for wild-type MBD4, which confirms interactions that mediate lesion recognition and suggests that a catalytic Asp, highly conserved in HhH enzymes, binds the putative nucleophilic water molecule and stabilizes the transition state. Observation that mutating the Asp (to Gly) reduces activity by 2700-fold indicates an important role in catalysis, but probably not one as the nucleophile in a double-displacement reaction, as previously suggested. Consistent with direct-displacement hydrolysis, a structure of the enzyme-product complex indicates a reaction leading to inversion of configuration. A structure with DNA containing 1-azadeoxyribose models a potential oxacarbenium-ion intermediate and suggests the Asp could facilitate migration of the electrophile towards the nucleophilic water. Finally, the structures provide detailed snapshots of the HhH motif, informing how these ubiquitous metal-binding elements mediate DNA binding.


Subject(s)
DNA/metabolism , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/metabolism , Pseudouridine/analogs & derivatives , Catalytic Domain , Crystallography, X-Ray , DNA/chemistry , Endodeoxyribonucleases/genetics , Epigenesis, Genetic , Humans , Models, Molecular , Mutation , Protein Conformation
5.
Mol Ther ; 29(9): 2769-2781, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33992803

ABSTRACT

It is well established that memory CD8 T cells protect susceptible strains of mice from mousepox, a lethal viral disease caused by ectromelia virus (ECTV), the murine counterpart to human variola virus. While mRNA vaccines induce protective antibody (Ab) responses, it is unknown whether they also induce protective memory CD8 T cells. We now show that immunization with different doses of unmodified or N(1)-methylpseudouridine-modified mRNA (modified mRNA) in lipid nanoparticles (LNP) encoding the ECTV gene EVM158 induced similarly strong CD8 T cell responses to the epitope TSYKFESV, albeit unmodified mRNA-LNP had adverse effects at the inoculation site. A single immunization with 10 µg modified mRNA-LNP protected most susceptible mice from mousepox, and booster vaccination increased the memory CD8 T cell pool, providing full protection. Moreover, modified mRNA-LNP encoding TSYKFESV appended to green fluorescent protein (GFP) protected against wild-type ECTV infection while lymphocytic choriomeningitis virus glycoprotein (GP) modified mRNA-LNP protected against ECTV expressing GP epitopes. Thus, modified mRNA-LNP can be used to create protective CD8 T cell-based vaccines against viral infections.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Viral Proteins/genetics , mRNA Vaccines/administration & dosage , Animals , Drug Compounding , Ectromelia, Infectious/immunology , Immunization, Secondary , Immunologic Memory , Liposomes , Male , Mice , Nanoparticles , Peptides/chemistry , Peptides/genetics , Peptides/immunology , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/pharmacology , mRNA Vaccines/chemistry , mRNA Vaccines/pharmacology
6.
Science ; 371(6525): 145-153, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33414215

ABSTRACT

The ability to control autoreactive T cells without inducing systemic immune suppression is the major goal for treatment of autoimmune diseases. The key challenge is the safe and efficient delivery of pharmaceutically well-defined antigens in a noninflammatory context. Here, we show that systemic delivery of nanoparticle-formulated 1 methylpseudouridine-modified messenger RNA (m1Ψ mRNA) coding for disease-related autoantigens results in antigen presentation on splenic CD11c+ antigen-presenting cells in the absence of costimulatory signals. In several mouse models of multiple sclerosis, the disease is suppressed by treatment with such m1Ψ mRNA. The treatment effect is associated with a reduction of effector T cells and the development of regulatory T cell (Treg cell) populations. Notably, these Treg cells execute strong bystander immunosuppression and thus improve disease induced by cognate and noncognate autoantigens.


Subject(s)
Bystander Effect/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Immunosuppression Therapy/methods , Multiple Sclerosis/therapy , Vaccines, Synthetic/therapeutic use , Animals , Antigen-Presenting Cells , Autoantigens/genetics , Inflammation/immunology , Mice , Mice, Inbred C57BL , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , RNA, Messenger/adverse effects , RNA, Messenger/chemistry , RNA, Messenger/genetics , T-Lymphocytes, Regulatory/immunology , Vaccines, Synthetic/adverse effects , mRNA Vaccines
7.
Nucleic Acids Res ; 48(6): e35, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32090264

ABSTRACT

Synthetic messenger RNA (mRNA) tools often use pseudouridine and 5-methyl cytidine as substitutions for uridine and cytidine to avoid the immune response and cytotoxicity induced by introducing mRNA into cells. However, the influence of base modifications on the functionality of the RNA tools is poorly understood. Here we show that synthetic mRNA switches containing N1-methylpseudouridine (m1Ψ) as a substitution of uridine substantially out-performed all other modified bases studied, exhibiting enhanced microRNA and protein sensitivity, better cell-type separation ability, and comparably low immune stimulation. We found that the observed phenomena stem from the high protein expression from m1Ψ containing mRNA and efficient translational repression in the presence of target microRNAs or proteins. In addition, synthetic gene circuits with m1Ψ significantly improve performance in cells. These findings indicate that synthetic mRNAs with m1Ψ modification have enormous potentials in the research and application of biofunctional RNA tools.


Subject(s)
Cells/metabolism , Pseudouridine/analogs & derivatives , RNA, Messenger/metabolism , Base Sequence , Cell Line , Humans , Immunity , MicroRNAs/genetics , MicroRNAs/metabolism , Pseudouridine/metabolism , RNA-Binding Proteins/metabolism
8.
Nat Biotechnol ; 37(7): 803-809, 2019 07.
Article in English | MEDLINE | ID: mdl-31267113

ABSTRACT

The ability to predict the impact of cis-regulatory sequences on gene expression would facilitate discovery in fundamental and applied biology. Here we combine polysome profiling of a library of 280,000 randomized 5' untranslated regions (UTRs) with deep learning to build a predictive model that relates human 5' UTR sequence to translation. Together with a genetic algorithm, we use the model to engineer new 5' UTRs that accurately direct specified levels of ribosome loading, providing the ability to tune sequences for optimal protein expression. We show that the same approach can be extended to chemically modified RNA, an important feature for applications in mRNA therapeutics and synthetic biology. We test 35,212 truncated human 5' UTRs and 3,577 naturally occurring variants and show that the model predicts ribosome loading of these sequences. Finally, we provide evidence of 45 single-nucleotide variants (SNVs) associated with human diseases that substantially change ribosome loading and thus may represent a molecular basis for disease.


Subject(s)
5' Untranslated Regions , Protein Biosynthesis , RNA, Messenger/genetics , Base Sequence , Gene Expression Regulation , Humans , Models, Genetic , Pseudouridine/analogs & derivatives , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Reproducibility of Results , Ribosomes
9.
Cell Chem Biol ; 25(5): 540-549.e4, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29551347

ABSTRACT

Pseudouridimycin (PUM) is a selective nucleoside-analog inhibitor of bacterial RNA polymerase with activity against Gram-positive and Gram-negative bacteria. PUM, produced by Streptomyces sp. ID38640, consists of a formamidinylated, N-hydroxylated Gly-Gln dipeptide conjugated to 5'-aminopseudouridine. We report the characterization of the PUM gene cluster. Bioinformatic analysis and mutational knockouts of pum genes with analysis of accumulated intermediates, define the PUM biosynthetic pathway. The work provides the first biosynthetic pathway of a C-nucleoside antibiotic and reveals three unexpected features: production of free pseudouridine by the dedicated pseudouridine synthase, PumJ; nucleoside activation by specialized oxidoreductases and aminotransferases; and peptide-bond formation by amide ligases. A central role in the PUM biosynthetic pathway is played by the PumJ, which represents a divergent branch within the TruD family of pseudouridine synthases. PumJ-like sequences are associated with diverse gene clusters likely to govern the biosynthesis of different classes of C-nucleoside antibiotics.


Subject(s)
Anti-Bacterial Agents/metabolism , Biosynthetic Pathways , Nucleosides/analogs & derivatives , Streptomyces/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Intramolecular Transferases/genetics , Intramolecular Transferases/metabolism , Multigene Family , Nucleosides/metabolism , Pseudouridine/analogs & derivatives , Pseudouridine/genetics , Pseudouridine/metabolism , Streptomyces/enzymology , Streptomyces/genetics
10.
Nucleic Acids Res ; 45(10): 6023-6036, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28334758

ABSTRACT

Certain chemical modifications confer increased stability and low immunogenicity to in vitro transcribed mRNAs, thereby facilitating expression of therapeutically important proteins. Here, we demonstrate that N1-methyl-pseudouridine (N1mΨ) outperforms several other nucleoside modifications and their combinations in terms of translation capacity. Through extensive analysis of various modified transcripts in cell-free translation systems, we deconvolute the different components of the effect on protein expression independent of mRNA stability mechanisms. We show that in addition to turning off the immune/eIF2α phosphorylation-dependent inhibition of translation, the incorporated N1mΨ nucleotides dramatically alter the dynamics of the translation process by increasing ribosome pausing and density on the mRNA. Our results indicate that the increased ribosome loading of modified mRNAs renders them more permissive for initiation by favoring either ribosome recycling on the same mRNA or de novo ribosome recruitment.


Subject(s)
Eukaryotic Initiation Factor-2/physiology , Polyribosomes/metabolism , Protein Biosynthesis , Pseudouridine/analogs & derivatives , RNA, Messenger/genetics , Animals , Cell Line , Cell-Free System , Enzyme Activation , Fibroblasts , HEK293 Cells , HeLa Cells , Humans , Mice , Phosphorylation , Protein Processing, Post-Translational , Pseudouridine/metabolism , RNA/metabolism , RNA Stability , RNA, Messenger/chemistry , Transfection , eIF-2 Kinase/metabolism
11.
Methods Mol Biol ; 1499: 109-121, 2017.
Article in English | MEDLINE | ID: mdl-27987145

ABSTRACT

In recent years, numerous studies have demonstrated the outstanding abilities of mRNA to elicit potent immune responses against pathogens, making it a viable new platform for vaccine development (reviewed in Weissman, Expert Rev Vaccines 14:265-281, 2015; Sahin et al., Nat Rev Drug Discov 13:759-780, 2014). The incorporation of modified nucleosides in mRNA has many advantages and is currently undergoing a renaissance in the field of therapeutic protein delivery. Its use in a vaccine against infectious diseases has only begun to be described, but offers advantages for the generation of potent and long-lived antibody responses. FPLC purification and substitution of modified nucleosides in the mRNA make it non-inflammatory and highly translatable (Kariko et al., Immunity 23:165-175, 2005; Kariko et al., Mol Ther 16:1833-1840, 2008; Kariko et al., Nucleic Acids Research 39:e142, 2011) that are crucial features for therapeutic relevance. Formulation of the mRNA in lipid nanoparticles (LNPs) protects it from degradation enabling high levels of protein production for extended periods of time (Pardi et al., J Control Release, 2015). Here, we describe a simple vaccination method using LNP-encapsulated 1-methylpseudouridine-containing FPLC purified mRNA in mice. Furthermore, we describe the evaluation of antigen-specific T and B cell responses elicited by this vaccine format.


Subject(s)
Communicable Diseases/immunology , Nucleosides/immunology , RNA, Messenger/immunology , Vaccines/immunology , Animals , Antibody Formation/immunology , B-Lymphocytes/immunology , Chromatography, High Pressure Liquid/methods , Lipids/chemistry , Mice , Nanoparticles/chemistry , Nucleosides/chemistry , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , RNA, Messenger/chemistry , T-Lymphocytes/immunology , Vaccination/methods , Vaccines/chemistry
12.
J Control Release ; 217: 337-44, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26342664

ABSTRACT

Messenger RNA as a therapeutic modality is becoming increasingly popular in the field of gene therapy. The realization that nucleobase modifications can greatly enhance the properties of mRNA by reducing the immunogenicity and increasing the stability of the RNA molecule (the Kariko paradigm) has been pivotal for this revolution. Here we find that mRNAs containing the N(1)-methylpseudouridine (m1Ψ) modification alone and/or in combination with 5-methylcytidine (m5C) outperformed the current state-of-the-art pseudouridine (Ψ) and/or m5C/Ψ-modified mRNA platform by providing up to ~44-fold (when comparing double modified mRNAs) or ~13-fold (when comparing single modified mRNAs) higher reporter gene expression upon transfection into cell lines or mice, respectively. We show that (m5C/)m1Ψ-modified mRNA resulted in reduced intracellular innate immunogenicity and improved cellular viability compared to (m5C/)Ψ-modified mRNA upon in vitro transfection. The enhanced capability of (m5C/)m1Ψ-modified mRNA to express proteins may at least partially be due to the increased ability of the mRNA to evade activation of endosomal Toll-like receptor 3 (TLR3) and downstream innate immune signaling. We believe that the (m5C/)m1Ψ-mRNA platform presented here may serve as a new standard in the field of modified mRNA-based therapeutics.


Subject(s)
Cytidine/analogs & derivatives , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , RNA, Messenger/chemistry , Animals , Cell Line , Cell Line, Tumor , Cell Survival , Cytidine/chemistry , Humans , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Mice, Inbred BALB C , RNA, Messenger/pharmacology , Transfection
13.
J Control Release ; 217: 345-51, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26264835

ABSTRACT

In recent years, in vitro transcribed messenger RNA (mRNA) has emerged as a potential therapeutic platform. To fulfill its promise, effective delivery of mRNA to specific cell types and tissues needs to be achieved. Lipid nanoparticles (LNPs) are efficient carriers for short-interfering RNAs and have entered clinical trials. However, little is known about the potential of LNPs to deliver mRNA. Here, we generated mRNA-LNPs by incorporating HPLC purified, 1-methylpseudouridine-containing mRNA comprising codon-optimized firefly luciferase into stable LNPs. Mice were injected with 0.005-0.250mg/kg doses of mRNA-LNPs by 6 different routes and high levels of protein translation could be measured using in vivo imaging. Subcutaneous, intramuscular and intradermal injection of the LNP-encapsulated mRNA translated locally at the site of injection for up to 10days. For several days, high levels of protein production could be achieved in the lung from the intratracheal administration of mRNA. Intravenous and intraperitoneal and to a lesser extent intramuscular and intratracheal deliveries led to trafficking of mRNA-LNPs systemically resulting in active translation of the mRNA in the liver for 1-4 days. Our results demonstrate that LNPs are appropriate carriers for mRNA in vivo and have the potential to become valuable tools for delivering mRNA encoding therapeutic proteins.


Subject(s)
Luciferases, Firefly/metabolism , Nanoparticles/administration & dosage , Pseudouridine/analogs & derivatives , RNA, Messenger/administration & dosage , Animals , Cells, Cultured , Dendritic Cells/metabolism , Female , HEK293 Cells , Humans , Kinetics , Luciferases, Firefly/genetics , Lung/metabolism , Mice, Inbred BALB C , Nanoparticles/chemistry , Phosphatidylethanolamines/chemistry , Pseudouridine/chemistry , RNA, Messenger/chemistry , RNA, Messenger/pharmacokinetics , Transfection/methods
14.
Nat Neurosci ; 17(5): 738-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24686785

ABSTRACT

Visual input often arrives in a noisy and discontinuous stream, owing to head and eye movements, occlusion, lighting changes, and many other factors. Yet the physical world is generally stable; objects and physical characteristics rarely change spontaneously. How then does the human visual system capitalize on continuity in the physical environment over time? We found that visual perception in humans is serially dependent, using both prior and present input to inform perception at the present moment. Using an orientation judgment task, we found that, even when visual input changed randomly over time, perceived orientation was strongly and systematically biased toward recently seen stimuli. Furthermore, the strength of this bias was modulated by attention and tuned to the spatial and temporal proximity of successive stimuli. These results reveal a serial dependence in perception characterized by a spatiotemporally tuned, orientation-selective operator-which we call a continuity field-that may promote visual stability over time.


Subject(s)
Attention/physiology , Judgment/physiology , Orientation/physiology , Pattern Recognition, Visual/physiology , Space Perception/physiology , Adult , Cues , Female , Functional Laterality , Humans , Male , Mental Recall/physiology , Photic Stimulation , Pseudouridine/analogs & derivatives , Psychophysics , Time Factors , Vision, Ocular , Visual Pathways/physiology , Young Adult
15.
RNA ; 18(3): 421-33, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22274953

ABSTRACT

The methylation of pseudouridine (Ψ) at position 54 of tRNA, producing m(1)Ψ, is a hallmark of many archaeal species, but the specific methylase involved in the formation of this modification had yet to be characterized. A comparative genomics analysis had previously identified COG1901 (DUF358), part of the SPOUT superfamily, as a candidate for this missing methylase family. To test this prediction, the COG1901 encoding gene, HVO_1989, was deleted from the Haloferax volcanii genome. Analyses of modified base contents indicated that while m(1)Ψ was present in tRNA extracted from the wild-type strain, it was absent from tRNA extracted from the mutant strain. Expression of the gene encoding COG1901 from Halobacterium sp. NRC-1, VNG1980C, complemented the m(1)Ψ minus phenotype of the ΔHVO_1989 strain. This in vivo validation was extended with in vitro tests. Using the COG1901 recombinant enzyme from Methanocaldococcus jannaschii (Mj1640), purified enzyme Pus10 from M. jannaschii and full-size tRNA transcripts or TΨ-arm (17-mer) fragments as substrates, the sequential pathway of m(1)Ψ54 formation in Archaea was reconstituted. The methylation reaction is AdoMet dependent. The efficiency of the methylase reaction depended on the identity of the residue at position 55 of the TΨ-loop. The presence of Ψ55 allowed the efficient conversion of Ψ54 to m(1)Ψ54, whereas in the presence of C55, the reaction was rather inefficient and no methylation reaction occurred if a purine was present at this position. These results led to renaming the Archaeal COG1901 members as TrmY proteins.


Subject(s)
Archaea/enzymology , Archaea/genetics , Intramolecular Transferases/metabolism , RNA, Archaeal/metabolism , RNA, Transfer/metabolism , tRNA Methyltransferases/metabolism , Base Pairing , Base Sequence , Gene Deletion , Genes, Archaeal , Haloferax volcanii/genetics , Haloferax volcanii/metabolism , Inverted Repeat Sequences/genetics , Methanococcales/genetics , Methanococcales/metabolism , Methylation , Phylogeny , Protein Conformation , Pseudouridine/analogs & derivatives , Pseudouridine/metabolism , RNA Processing, Post-Transcriptional , RNA, Archaeal/chemistry , RNA, Transfer/chemistry
16.
J Bacteriol ; 193(1): 154-62, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21037010

ABSTRACT

The Escherichia coli rluD gene encodes a pseudouridine synthase responsible for the pseudouridine (Ψ) modifications at positions 1911, 1915, and 1917 in helix 69 of 23S rRNA. It has been reported that deletion of rluD in K-12 strains of E. coli is associated with extremely slow growth, increased readthrough of stop codons, and defects in 50S ribosomal subunit assembly and 30S-50S subunit association. Suppressor mutations in the prfB and prfC genes encoding release factor 2 (RF2) and RF3 that restore the wild type-growth rate and also correct the ribosomal defects have now been isolated. These suppressors link helix 69 Ψ residues with the termination phase of protein synthesis. However, further genetic analysis reported here also reveals that the slow growth and other defects associated with inactivation of rluD in E. coli K-12 strains are due to a defective RF2 protein, with a threonine at position 246, which is present in all K-12 strains. This is in contrast to the more typical alanine found at this position in most bacterial RF2s, including those of other E. coli strains. Inactivation of rluD in E. coli strains containing the prfB allele from E. coli B or in Salmonella enterica, both carrying an RF2 with Ala246, has negligible effects on growth, termination, or ribosome function. The results indicate that, in contrast to those in wild bacteria, termination functions in E. coli K-12 strains carrying a partially defective RF2 protein are especially susceptible to perturbation of ribosome-RF interactions, such as that caused by loss of h69 Ψ modifications.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Hydro-Lyases/metabolism , Ribosomes/metabolism , Salmonella enterica/metabolism , Alleles , Base Sequence , Escherichia coli/classification , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , Models, Molecular , Mutation , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Phenotype , Protein Conformation , Pseudouridine/analogs & derivatives , Pseudouridine/genetics , Pseudouridine/metabolism , Ribosomes/genetics , Salmonella enterica/classification , Salmonella enterica/genetics
17.
Nucleic Acids Res ; 38(7): 2387-98, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20047967

ABSTRACT

Nep1 (Emg1) is a highly conserved nucleolar protein with an essential function in ribosome biogenesis. A mutation in the human Nep1 homolog causes Bowen-Conradi syndrome-a severe developmental disorder. Structures of Nep1 revealed a dimer with a fold similar to the SPOUT-class of RNA-methyltransferases suggesting that Nep1 acts as a methyltransferase in ribosome biogenesis. The target for this putative methyltransferase activity has not been identified yet. We characterized the RNA-binding specificity of Methanocaldococcus jannaschii Nep1 by fluorescence- and NMR-spectroscopy as well as by yeast three-hybrid screening. Nep1 binds with high affinity to short RNA oligonucleotides corresponding to nt 910-921 of M. jannaschii 16S rRNA through a highly conserved basic surface cleft along the dimer interface. Nep1 only methylates RNAs containing a pseudouridine at a position corresponding to a previously identified hypermodified N1-methyl-N3-(3-amino-3-carboxypropyl) pseudouridine (m1acp3-Psi) in eukaryotic 18S rRNAs. Analysis of the methylated nucleoside by MALDI-mass spectrometry, HPLC and NMR shows that the methyl group is transferred to the N1 of the pseudouridine. Thus, Nep1 is the first identified example of an N1-specific pseudouridine methyltransferase. This enzymatic activity is also conserved in human Nep1 suggesting that Nep1 is the methyltransferase in the biosynthesis of m1acp3-Psi in eukaryotic 18S rRNAs.


Subject(s)
Archaeal Proteins/chemistry , Methanococcales/enzymology , Methyltransferases/chemistry , Nuclear Proteins/chemistry , Pseudouridine/metabolism , RNA, Ribosomal/metabolism , Archaeal Proteins/metabolism , Base Sequence , Binding Sites , Consensus Sequence , Humans , Methanococcales/genetics , Methylation , Methyltransferases/metabolism , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/metabolism , Pseudouridine/analogs & derivatives , Pseudouridine/analysis , RNA, Fungal/chemistry , RNA, Fungal/metabolism , RNA, Ribosomal/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Spectrometry, Fluorescence , Two-Hybrid System Techniques
18.
RNA ; 11(9): 1420-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16120833

ABSTRACT

The helix 69 (H69) region of the large subunit (28S) rRNA of Homo sapiens contains five pseudouridine (Psi) residues out of 19 total nucleotides (26%), three of which are universally or highly conserved. In this study, the effects of this abundant modified nucleotide on the structure and stability of H69 were compared with those of uridine. The role of a loop nucleotide substitution from A in bacteria (position 1918 in Escherichia coli 23S rRNA) to G in eukaryotes (position in 3734 in H. sapiens) was also examined. The thermodynamic parameters were obtained through UV melting studies, and differences in the modified and unmodified RNA structures were examined by 1H NMR and circular dichroism spectroscopy. In addition, a [1,3-15N]Psi phosphoramidite was used to generate H69 analogs with site-specific 15N labels. By using this approach, different Psi residues can be clearly distinguished from one another in 1H NMR experiments. The effects of pseudouridine on H. sapiens H69 are consistent with previous studies on tRNA, rRNA, and snRNA models in which the nucleotide offers stabilization of duplex regions through PsiN1H-mediated hydrogen bonds. The overall secondary structure and base-pairing patterns of human H69 are similar to the bacterial RNA, consistent with the idea that ribosome structure and function are highly conserved. Nonetheless, pseudouridine-containing RNAs have subtle differences in their structures and stabilities compared to the corresponding uridine-containing analogs, suggesting possible roles for Psi such as maintaining translation fidelity.


Subject(s)
Nucleic Acid Conformation , Point Mutation , RNA, Ribosomal, 28S/genetics , Circular Dichroism , Humans , Magnetic Resonance Spectroscopy , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , Pseudouridine/genetics , Pseudouridine/metabolism , RNA, Ribosomal, 28S/chemistry , RNA, Ribosomal, 28S/metabolism , Temperature , Thermodynamics
19.
Bioorg Med Chem ; 13(24): 6777-81, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16125393

ABSTRACT

The conformations of 3-methyluridine and 3-methylpseudouridine are determined using a combination of sugar proton coupling constants from 1D NMR spectra and 1D NOE difference spectroscopy. Both C2'-endo and C3'-endo conformations are observed for 3-methyluridine (59:41, 37 degrees C, D2O) and 3-methylpseudouridine (51:49, 37 degrees C, D2O). 3-Methyluridine preferentially adopts an anti conformation in solution, whereas 3-methylpseudouridine is primarily in a syn conformation. anti/syn-Relationships are deduced by 1D NOE difference spectroscopy.


Subject(s)
Pseudouridine/analogs & derivatives , RNA/chemistry , Uridine/analogs & derivatives , Circular Dichroism , Magnetic Resonance Spectroscopy , Molecular Conformation , Pseudouridine/chemistry , Solutions/chemistry , Temperature , Uridine/chemistry
20.
RNA ; 11(2): 210-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15659360

ABSTRACT

The number and position of the pseudouridines of Haloarcula marismortui and Deinococcus radiodurans large subunit RNA have been determined by a combination of total nucleoside analysis by HPLC-mass spectrometry and pseudouridine sequencing by the reverse transcriptase method and by LC/MS/MS. Three pseudouridines were found in H. marismortui, located at positions 1956, 1958, and 2621 corresponding to Escherichia coli positions 1915, 1917, and 2586, respectively. The three pseudouridines are all in locations found in other organisms. Previous reports of a larger number of pseudouridines in this organism were incorrect. Three pseudouridines and one 3-methyl pseudouridine (m3Psi) were found in D. radiodurans 23S RNA at positions 1894, 1898 (m3Psi), 1900, and 2584, the m3Psi site being determined by a novel application of mass spectrometry. These positions correspond to E. coli positions 1911, 1915, 1917, and 2605, which are also pseudouridines in E. coli (1915 is m3Psi). The pseudouridines in the helix 69 loop, residues 1911, 1915, and 1917, are in positions highly conserved among all phyla. Pseudouridine 2584 in D. radiodurans is conserved in eubacteria and a chloroplast but is not found in archaea or eukaryotes, whereas pseudouridine 2621 in H. marismortui is more conserved in eukaryotes and is not found in eubacteria. All the pseudoridines are near, but not exactly at, nucleotides directly involved in various aspects of ribosome function. In addition, two D. radiodurans Psi synthases responsible for the four Psi were identified.


Subject(s)
Deinococcus/chemistry , Haloarcula marismortui/chemistry , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , RNA, Archaeal/chemistry , RNA, Bacterial/chemistry , RNA, Ribosomal, 23S/chemistry , Base Sequence , Deinococcus/genetics , Escherichia coli/chemistry , Escherichia coli/genetics , Haloarcula marismortui/genetics , Hydro-Lyases/genetics , Nucleic Acid Conformation , RNA, Archaeal/genetics , RNA, Bacterial/genetics , RNA, Ribosomal, 23S/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...