Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
Add more filters

Publication year range
1.
Nature ; 622(7982): 402-409, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37758951

ABSTRACT

Transposable elements are genomic parasites that expand within and spread between genomes1. PIWI proteins control transposon activity, notably in the germline2,3. These proteins recognize their targets through small RNA co-factors named PIWI-interacting RNAs (piRNAs), making piRNA biogenesis a key specificity-determining step in this crucial genome immunity system. Although the processing of piRNA precursors is an essential step in this process, many of the molecular details remain unclear. Here, we identify an endoribonuclease, precursor of 21U RNA 5'-end cleavage holoenzyme (PUCH), that initiates piRNA processing in the nematode Caenorhabditis elegans. Genetic and biochemical studies show that PUCH, a trimer of Schlafen-like-domain proteins (SLFL proteins), executes 5'-end piRNA precursor cleavage. PUCH-mediated processing strictly requires a 7-methyl-G cap (m7G-cap) and a uracil at position three. We also demonstrate how PUCH interacts with PETISCO, a complex that binds to piRNA precursors4, and that this interaction enhances piRNA production in vivo. The identification of PUCH concludes the search for the 5'-end piRNA biogenesis factor in C. elegans and uncovers a type of RNA endonuclease formed by three SLFL proteins. Mammalian Schlafen (SLFN) genes have been associated with immunity5, exposing a molecular link between immune responses in mammals and deeply conserved RNA-based mechanisms that control transposable elements.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Endoribonucleases , Piwi-Interacting RNA , Animals , Argonaute Proteins/metabolism , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , DNA Transposable Elements/genetics , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Holoenzymes/chemistry , Holoenzymes/metabolism , Piwi-Interacting RNA/chemistry , Piwi-Interacting RNA/genetics , Piwi-Interacting RNA/metabolism , RNA Cap Analogs/chemistry , RNA Cap Analogs/metabolism
2.
Nature ; 614(7947): 358-366, 2023 02.
Article in English | MEDLINE | ID: mdl-36725932

ABSTRACT

The mRNA cap structure is a major site of dynamic mRNA methylation. mRNA caps exist in either the Cap1 or Cap2 form, depending on the presence of 2'-O-methylation on the first transcribed nucleotide or both the first and second transcribed nucleotides, respectively1,2. However, the identity of Cap2-containing mRNAs and the function of Cap2 are unclear. Here we describe CLAM-Cap-seq, a method for transcriptome-wide mapping and quantification of Cap2. We find that unlike other epitranscriptomic modifications, Cap2 can occur on all mRNAs. Cap2 is formed through a slow continuous conversion of mRNAs from Cap1 to Cap2 as mRNAs age in the cytosol. As a result, Cap2 is enriched on long-lived mRNAs. Large increases in the abundance of Cap1 leads to activation of RIG-I, especially in conditions in which expression of RIG-I is increased. The methylation of Cap1 to Cap2 markedly reduces the ability of RNAs to bind to and activate RIG-I. The slow methylation rate of Cap2 allows Cap2 to accumulate on host mRNAs, yet ensures that low levels of Cap2 occur on newly expressed viral RNAs. Overall, these results reveal an immunostimulatory role for Cap1, and that Cap2 functions to reduce activation of the innate immune response.


Subject(s)
Cellular Senescence , Epigenome , Mammals , Methylation , RNA Caps , RNA, Messenger , Animals , Cytosol/metabolism , DEAD Box Protein 58 , Gene Expression Profiling , Immunity, Innate , Mammals/genetics , Mammals/metabolism , Nucleotides/chemistry , Nucleotides/genetics , Nucleotides/metabolism , Receptors, Immunologic , RNA Cap Analogs/chemistry , RNA Cap Analogs/genetics , RNA Cap Analogs/metabolism , RNA Caps/chemistry , RNA Caps/genetics , RNA Caps/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome , Time Factors
3.
Org Biomol Chem ; 22(33): 6763-6790, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39105613

ABSTRACT

The trimethylguanosine (TMG) cap is a motif present inter alia at the 5' end of small nuclear RNAs, which are involved in RNA splicing. The TMG cap plays a crucial role in RNA processing and stability as it protects the RNA molecule from degradation by exonucleases and facilitates its export from the nucleus. Additionally, the TMG cap plays a role in the recognition of snRNA by snurportin, a protein that facilitates nuclear import. TMG cap analogs are used in biochemical experiments as molecular tools to substitute the natural TMG cap. To expand the range of available TMG-based tools, here we conjugated the TMG cap to Fluorescent Molecular Rotors (FMRs) to open the possibility of detecting protein-ligand interactions in vitro and, potentially, in vivo, particularly visualizing interactions with snurportin. Consequently, we report the synthesis of 34 differently modified TMG cap-FMR conjugates and their evaluation as molecular probes for snurportin. As FMRs we selected three GFP-like chromophores (derived from green fluorescent protein) and one julolidine derivative. The evaluation of binding affinities for snurportin showed unexpectedly a strong stabilizing effect for TMGpppG-derived dinucleotides containing the FMR at the 2'-O-position of guanosine. These newly discovered compounds are potent snurportin ligands with nanomolar KD (dissociation constant) values, which are two orders of magnitude lower than that of natural TMGpppG. The effect is diminished by ∼50-fold for the corresponding 3'-regioisomers. To deepen the understanding of the structure-activity relationship, we synthesized and tested FMR conjugates lacking the TMG cap moiety. These studies, supported by molecular docking, suggested that the enhanced affinity arises from additional hydrophobic contacts provided by the FMR moiety. The strongest snurportin ligand, which also gave the greatest fluorescence enhancement (Fm/F0) when saturated with the protein, were tested in living cells to detect interactions and visualize complexes by fluorescence lifetime monitoring. This approach has potential applications in the study of RNA processing and RNA-protein interactions.


Subject(s)
Fluorescent Dyes , Guanosine , Ligands , Humans , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Guanosine/analogs & derivatives , Guanosine/chemistry , Guanosine/metabolism , RNA Cap Analogs/chemistry , RNA Cap Analogs/chemical synthesis , RNA Cap Analogs/metabolism , HeLa Cells , Molecular Structure
4.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-33972410

ABSTRACT

The genome of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) coronavirus has a capping modification at the 5'-untranslated region (UTR) to prevent its degradation by host nucleases. These modifications are performed by the Nsp10/14 and Nsp10/16 heterodimers using S-adenosylmethionine as the methyl donor. Nsp10/16 heterodimer is responsible for the methylation at the ribose 2'-O position of the first nucleotide. To investigate the conformational changes of the complex during 2'-O methyltransferase activity, we used a fixed-target serial synchrotron crystallography method at room temperature. We determined crystal structures of Nsp10/16 with substrates and products that revealed the states before and after methylation, occurring within the crystals during the experiments. Here we report the crystal structure of Nsp10/16 in complex with Cap-1 analog (m7GpppAm2'-O). Inhibition of Nsp16 activity may reduce viral proliferation, making this protein an attractive drug target.


Subject(s)
RNA Caps/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , SARS-CoV-2/chemistry , Crystallography , Methylation , Methyltransferases/chemistry , Methyltransferases/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , RNA Cap Analogs/chemistry , RNA Cap Analogs/metabolism , RNA Caps/chemistry , RNA, Messenger/chemistry , RNA, Viral/chemistry , S-Adenosylhomocysteine/chemistry , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Synchrotrons , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/metabolism
5.
Chem Rec ; 22(8): e202200005, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35420257

ABSTRACT

The recent FDA approval of the mRNA vaccine for severe acute respiratory syndrome coronavirus (SARS-CoV-2) emphasizes the importance of mRNA as a powerful tool for therapeutic applications. The chemically modified mRNA cap analogs contain a unique cap structure, m7 G[5']ppp[5']N (where N=G, A, C or U), present at the 5'-end of many eukaryotic cellular and viral RNAs and several non-coding RNAs. The chemical modifications on cap analog influence orientation's nature, translational efficiency, nuclear stability, and binding affinity. The recent invention of a trinucleotide cap analog provides groundbreaking research in the area of mRNA analogs. Notably, trinucleotide cap analogs outweigh dinucleotide cap analogs in terms of capping efficiency and translational properties. This review focuses on the recent development in the synthesis of various dinucleotide cap analogs such as dinucleotide containing a triazole moiety, phosphorothiolate cap, biotinylated cap, cap analog containing N1 modification, cap analog containing N2 modification, dinucleotide containing fluorescence probe and TAT, bacterial caps, and trinucleotide cap analogs. In addition, the biological applications of these novel cap analogs are delineated.


Subject(s)
COVID-19 , Vaccines , COVID-19/prevention & control , Humans , RNA Cap Analogs/chemistry , RNA Cap Analogs/metabolism , RNA, Messenger/chemistry , SARS-CoV-2 , Vaccines, Synthetic , mRNA Vaccines
6.
PLoS Pathog ; 15(5): e1007829, 2019 05.
Article in English | MEDLINE | ID: mdl-31136637

ABSTRACT

Rift Valley fever virus (RVFV) belongs to the family of Phenuiviridae within the order of Bunyavirales. The virus may cause fatal disease both in livestock and humans, and therefore, is of great economical and public health relevance. In analogy to the influenza virus polymerase complex, the bunyavirus L protein is assumed to bind to and cleave off cap structures of cellular mRNAs to prime viral transcription. However, even though the presence of an endonuclease in the N-terminal domain of the L protein has been demonstrated for several bunyaviruses, there is no evidence for a cap-binding site within the L protein. We solved the structure of a C-terminal 117 amino acid-long domain of the RVFV L protein by X-ray crystallography. The overall fold of the domain shows high similarity to influenza virus PB2 cap-binding domain and the putative non-functional cap-binding domain of reptarenaviruses. Upon co-crystallization with m7GTP, we detected the cap-analogue bound between two aromatic side chains as it has been described for other cap-binding proteins. We observed weak but specific interaction with m7GTP rather than GTP in vitro using isothermal titration calorimetry. The importance of m7GTP-binding residues for viral transcription was validated using a RVFV minigenome system. In summary, we provide structural and functional evidence for a cap-binding site located within the L protein of a virus from the Bunyavirales order.


Subject(s)
RNA Cap Analogs/metabolism , RNA Caps/chemistry , RNA Caps/metabolism , Rift Valley fever virus/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , Protein Domains , RNA Caps/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , Sequence Homology , Viral Proteins/genetics
7.
Nucleic Acids Res ; 47(11): 5973-5987, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31066441

ABSTRACT

Association of the initiation factor eIF4E with the mRNA cap structure is a key step for translation. Trypanosomatids present six eIF4E homologues, showing a low conservation and also differing significantly from the IF4Es of multicellular eukaryotes. On the mRNA side, while in most eukaryotes the mRNA contains cap-0 (7-methyl-GTP), the trypanosomatid mRNA features a cap-4, which is formed by a cap-0, followed by the AACU sequence containing 2'-O-ribose methylations and base methylations on nucleotides 1 and 4. The studies on eIF4E-cap-4 interaction have been hindered by the difficulty to synthesize this rather elaborated cap-4 sequence. To overcome this problem, we applied a liquid-phase oligonucleotide synthesis strategy and describe for the first time the crystal structure of a trypanosomatid eIF4E (T. cruzi EIF4E5) in complex with cap-4. The TcEIF4E5-cap-4 structure allowed a detailed description of the binding mechanism, revealing the interaction mode for the AACU sequence, with the bases packed in a parallel stacking conformation and involved, together with the methyl groups, in hydrophobic contacts with the protein. This binding mechanism evidences a distinct cap interaction mode in comparison with previously described eIF4E structures and may account for the difference of TcEIF4E5-cap-4 dissociation constant in comparison with other eIF4E homologues.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , RNA Caps/chemistry , Trypanosoma cruzi/chemistry , Animals , Carrier Proteins/metabolism , Crystallography, X-Ray , DNA Methylation , Humans , Ligands , Models, Molecular , Nucleotides/chemistry , Oligonucleotides , Protein Binding , RNA Cap Analogs/metabolism , RNA, Messenger/metabolism , Schistosoma mansoni/metabolism , Temperature , Trypanosoma/metabolism
8.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884522

ABSTRACT

Leishmania parasites are digenetic protists that shuffle between sand fly vectors and mammalian hosts, transforming from flagellated extracellular promastigotes that reside within the intestinal tract of female sand flies to the obligatory intracellular and non-motile amastigotes within mammalian macrophages. Stage differentiation is regulated mainly by post-transcriptional mechanisms, including translation regulation. Leishmania parasites encode six different cap-binding proteins, LeishIF4E1-6, that show poor conservation with their counterparts from higher eukaryotes and among themselves. In view of the changing host milieu encountered throughout their life cycle, we propose that each LeishIF4E has a unique role, although these functions may be difficult to determine. Here we characterize LeishIF4E-6, a unique eIF4E ortholog that does not readily associate with m7GTP cap in either of the tested life forms of the parasite. We discuss the potential effect of substituting two essential tryptophan residues in the cap-binding pocket, expected to be involved in the cap-binding activity, as judged from structural studies in the mammalian eIF4E. LeishIF4E-6 binds to LeishIF4G-5, one of the five eIF4G candidates in Leishmania. However, despite this binding, LeishIF4E-6 does not appear to function as a translation factor. Its episomal overexpression causes a general reduction in the global activity of protein synthesis, which was not observed in the hemizygous deletion mutant generated by CRISPR-Cas9. This genetic profile suggests that LeishIF4E-6 has a repressive role. The interactome of LeishIF4E-6 highlights proteins involved in RNA metabolism such as the P-body marker DHH1, PUF1 and an mRNA-decapping enzyme that is homologous to the TbALPH1.


Subject(s)
Eukaryotic Initiation Factor-4F/metabolism , Leishmania/metabolism , Protozoan Proteins/metabolism , RNA Cap Analogs/genetics , RNA Cap-Binding Proteins/metabolism , Amino Acid Sequence , Eukaryotic Initiation Factor-4F/chemistry , Eukaryotic Initiation Factor-4F/genetics , Leishmania/genetics , Leishmania/growth & development , Protein Biosynthesis , Protein Conformation , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , RNA Cap Analogs/metabolism , RNA Cap-Binding Proteins/genetics , Sequence Homology
9.
Biochem Biophys Res Commun ; 533(3): 391-396, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32962861

ABSTRACT

The interferon-induced proteins with tetratricopeptide repeats (IFITs) are a family of RNA-binding proteins that are very highly expressed during antiviral response of immune system. IFIT proteins recognize and tightly bind foreign RNA particles. These are primarily viral RNAs ended with triphosphate at the 5' or lacking methylation of the first cap-proximal nucleotide but also in vitro transcribed RNA synthesized in the laboratory. Recognition of RNA by IFIT proteins leads to the formation of stable RNA/IFIT complexes and translational shut off of non-self transcripts. Here, we present a fluorescent-based assay to study the interaction between RNA molecules and IFIT family proteins. We have particularly focused on two representatives of this family: IFIT1 and IFIT5. We found a probe that competitively with RNA binds the positively charged tunnel in these IFIT proteins. The use of this probe for IFIT titration allowed us to evaluate the differences in binding affinities of mRNAs with different variants of 5' ends.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Anilino Naphthalenesulfonates/chemistry , Biological Assay , Fluorescent Dyes/chemistry , Neoplasm Proteins/chemistry , RNA Cap-Binding Proteins/chemistry , RNA Caps/chemistry , RNA-Binding Proteins/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Binding, Competitive , Humans , Hydrogen Bonding , Kinetics , Molecular Docking Simulation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Binding , Protein Conformation , RNA Cap Analogs/chemistry , RNA Cap Analogs/metabolism , RNA Cap-Binding Proteins/genetics , RNA Cap-Binding Proteins/metabolism , RNA Caps/genetics , RNA Caps/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Spectrometry, Fluorescence , Static Electricity , Thermodynamics
10.
RNA ; 24(5): 633-642, 2018 05.
Article in English | MEDLINE | ID: mdl-29483298

ABSTRACT

Human Nudt16 (hNudt16) is a member of the Nudix family of hydrolases, comprising enzymes catabolizing various substrates including canonical (d)NTPs, oxidized (d)NTPs, nonnucleoside polyphosphates, and capped mRNAs. Decapping activity of the Xenopus laevis (X29) Nudt16 homolog was observed in the nucleolus, with a high specificity toward U8 snoRNA. Subsequent studies have reported cytoplasmic localization of mammalian Nudt16 with cap hydrolysis activity initiating RNA turnover, similar to Dcp2. The present study focuses on hNudt16 and its hydrolytic activity toward dinucleotide cap analogs and short capped oligonucleotides. We performed a screening assay for potential dinucleotide and oligonucleotide substrates for hNudt16. Our data indicate that dinucleotide cap analogs and capped oligonucleotides containing guanine base in the first transcribed nucleotide are more susceptible to enzymatic digestion by hNudt16 than their counterparts containing adenine. Furthermore, unmethylated dinucleotides (GpppG and ApppG) and respective oligonucleotides (GpppG-16nt and GpppA-16nt) were hydrolyzed by hNudt16 with greater efficiency than were m7GpppG and m7GpppG-16nt. In conclusion, we found that hNudt16 hydrolysis of dinucleotide cap analogs and short capped oligonucleotides displayed a broader spectrum specificity than is currently known.


Subject(s)
Endoribonucleases/metabolism , Pyrophosphatases/metabolism , RNA Cap Analogs/metabolism , Humans , Hydrolysis , Oligonucleotides/chemistry , Oligonucleotides/metabolism , RNA Cap Analogs/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Substrate Specificity
11.
Nucleic Acids Res ; 46(13): 6893-6908, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30053226

ABSTRACT

The interaction of the eukaryotic initiation factor 4G (eIF4G) with the cap-binding protein eIF4E initiates cap-dependent translation and is regulated by the 4E-binding proteins (4E-BPs), which compete with eIF4G to repress translation. Metazoan eIF4G and 4E-BPs interact with eIF4E via canonical and non-canonical motifs that bind to the dorsal and lateral surface of eIF4E in a bipartite recognition mode. However, previous studies pointed to mechanistic differences in how fungi and metazoans regulate protein synthesis. We present crystal structures of the yeast eIF4E bound to two yeast 4E-BPs, p20 and Eap1p, as well as crystal structures of a fungal eIF4E-eIF4G complex. We demonstrate that the core principles of molecular recognition of eIF4E are in fact highly conserved among translational activators and repressors in eukaryotes. Finally, we reveal that highly specialized structural motifs do exist and serve to modulate the affinity of protein-protein interactions that regulate cap-dependent translation initiation in fungi.


Subject(s)
Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4G/chemistry , Gene Expression Regulation, Fungal , Peptide Chain Initiation, Translational , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/genetics , Transcription Factors/chemistry , Amino Acid Motifs , Binding, Competitive , Chaetomium/genetics , Conserved Sequence , Crystallography, X-Ray , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Evolution, Molecular , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Models, Molecular , Protein Binding , Protein Conformation , RNA Cap Analogs/metabolism , Recombinant Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Scattering, Small Angle , Sequence Alignment , Species Specificity , Structure-Activity Relationship , Transcription Factors/metabolism
12.
Nucleic Acids Res ; 46(2): 956-971, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29202182

ABSTRACT

Influenza polymerase uses short capped primers snatched from nascent Pol II transcripts to initiate transcription of viral mRNAs. Here we describe crystal structures of influenza A and B polymerase bound to a capped primer in a configuration consistent with transcription initiation ('priming state') and show by functional assays that conserved residues from both the PB2 midlink and cap-binding domains are important for positioning the capped RNA. In particular, mutation of PB2 Arg264, which interacts with the triphosphate linkage in the cap, significantly and specifically decreases cap-dependent transcription. We also compare the configuration of the midlink and cap-binding domains in the priming state with their very different relative arrangement (called the 'apo' state) in structures where the potent cap-binding inhibitor VX-787, or a close analogue, is bound. In the 'apo' state the inhibitor makes additional interactions to the midlink domain that increases its affinity beyond that to the cap-binding domain alone. The comparison suggests that the mechanism of resistance of certain mutations that allow virus to escape from VX-787, notably PB2 N510T, can only be rationalized if VX-787 has a dual mode of action, direct inhibition of capped RNA binding as well as stabilization of the transcriptionally inactive 'apo' state.


Subject(s)
RNA Cap Analogs/metabolism , RNA Caps/metabolism , RNA Polymerase II/metabolism , RNA/metabolism , Viral Proteins/metabolism , Binding Sites/genetics , Crystallography, X-Ray , HEK293 Cells , Humans , Indoles/metabolism , Indoles/pharmacology , Influenza A virus/enzymology , Protein Binding , Pyridines , Pyrimidines , Pyrroles , RNA/chemistry , RNA/genetics , RNA Cap Analogs/pharmacology , RNA Caps/chemistry , RNA Caps/genetics , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Viral Proteins/chemistry , Viral Proteins/genetics
13.
RNA ; 23(5): 683-695, 2017 05.
Article in English | MEDLINE | ID: mdl-28159804

ABSTRACT

The 5' cap structure of eukaryotic mRNA is critical for its processing, transport, translation, and stability. The many functions of the cap and the fact that most, if not all, mRNA carries the same type of cap makes it difficult to analyze cap function in vivo at individual steps of gene expression. We have used the lariat capping ribozyme (LCrz) from the myxomycete Didymium to replace the mRNA m7G cap of a single reporter mRNA species with a tiny lariat in which the first and the third nucleotide are joined by a 2', 5' phosphodiester bond. We show that the ribozyme functions in vivo in the budding yeast Saccharomyces cerevisiae presumably without cofactors and that lariat capping occurs cotranscriptionally. The lariat-capped reporter mRNA is efficiently exported to the cytoplasm where it is found to be oligoadenylated and evenly distributed. Both the oligoadenylated form and a lariat-capped mRNA with a templated poly(A) tail translates poorly, underlining the critical importance of the m7G cap in translation. Finally, the lariat-capped RNA exhibits a threefold longer half-life compared to its m7G-capped counterpart, consistent with a key role for the m7G cap in mRNA turnover. Our study emphasizes important activities of the m7G cap and suggests new utilities of lariat capping as a molecular tool in vivo.


Subject(s)
RNA Cap Analogs/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Cell Nucleus/metabolism , Green Fluorescent Proteins/genetics , Half-Life , Protein Biosynthesis , RNA Cap Analogs/physiology , RNA Transport , RNA, Catalytic/metabolism , RNA, Messenger/chemistry , Saccharomyces cerevisiae/metabolism
14.
Nucleic Acids Res ; 45(15): 8661-8675, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28666355

ABSTRACT

Analogues of the mRNA 5'-cap are useful tools for studying mRNA translation and degradation, with emerging potential applications in novel therapeutic interventions including gene therapy. We report the synthesis of novel mono- and dinucleotide cap analogues containing dihalogenmethylenebisphosphonate moiety (i.e. one of the bridging O atom substituted with CCl2 or CF2) and their properties in the context of cellular translational and decapping machineries, compared to phosphate-unmodified and previously reported CH2-substituted caps. The analogues were bound tightly to eukaryotic translation initiation factor 4E (eIF4E), with CCl2-substituted analogues having the highest affinity. When incorporated into mRNA, the CCl2-substituted dinucleotide most efficiently promoted cap-dependent translation. Moreover, the CCl2-analogues were potent inhibitors of translation in rabbit reticulocyte lysate. The crystal structure of eIF4E in complex with the CCl2-analogue revealed a significantly different ligand conformation compared to that of the unmodified cap analogue, which likely contributes to the improved binding. Both CCl2- and CF2- analogues showed lower susceptibility to hydrolysis by the decapping scavenger enzyme (DcpS) and, when incorporated into RNA, conferred stability against major cellular decapping enzyme (Dcp2) to transcripts. Furthermore, the use of difluoromethylene cap analogues was exemplified by the development of 19F NMR assays for DcpS activity and eIF4E binding.


Subject(s)
Endoribonucleases/metabolism , Protein Biosynthesis/drug effects , RNA Cap Analogs/pharmacology , RNA Processing, Post-Transcriptional/drug effects , RNA, Messenger/drug effects , Animals , Binding Sites/drug effects , Crystallography, X-Ray , Dinucleoside Phosphates/chemistry , Dinucleoside Phosphates/metabolism , Eukaryotic Initiation Factor-4E/metabolism , HeLa Cells , Humans , Mice , Models, Molecular , RNA Cap Analogs/chemistry , RNA Cap Analogs/metabolism , RNA Caps/chemistry , RNA Caps/drug effects , RNA Caps/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism
15.
Molecules ; 24(10)2019 May 17.
Article in English | MEDLINE | ID: mdl-31108861

ABSTRACT

The mRNA 5' cap consists of N7-methylguanosine bound by a 5',5'-triphosphate bridge to the first nucleotide of the transcript. The cap interacts with various specific proteins and participates in all key mRNA-related processes, which may be of therapeutic relevance. There is a growing demand for new biophysical and biochemical methods to study cap-protein interactions and identify the factors which inhibit them. The development of such methods can be aided by the use of properly designed fluorescent molecular probes. Herein, we synthesized a new class of m7Gp3G cap derivatives modified with an alkyne handle at the N1-position of guanosine and, using alkyne-azide cycloaddition, we functionalized them with fluorescent tags to obtain potential probes. The cap derivatives and probes were evaluated in the context of two cap-binding proteins, eukaryotic translation initiation factor (eIF4E) and decapping scavenger (DcpS). Biochemical and biophysical studies revealed that N1-propargyl moiety did not significantly disturb cap-protein interaction. The fluorescent properties of the probes turned out to be in line with microscale thermophoresis (MST)-based binding assays.


Subject(s)
RNA Cap Analogs/chemical synthesis , RNA Cap-Binding Proteins/metabolism , Click Chemistry , Cycloaddition Reaction , Guanosine/chemistry , Humans , RNA Cap Analogs/chemistry , RNA Cap Analogs/metabolism , RNA Cap-Binding Proteins/chemistry
16.
J Virol ; 91(6)2017 03 15.
Article in English | MEDLINE | ID: mdl-28053102

ABSTRACT

The GDP polyribonucleotidyltransferase (PRNTase) domain of the multifunctional L protein of rhabdoviruses, such as vesicular stomatitis virus (VSV) and rabies virus, catalyzes the transfer of 5'-phospho-RNA (pRNA) from 5'-triphospho-RNA (pppRNA) to GDP via a covalent enzyme-pRNA intermediate to generate a 5'-cap structure (GpppA). Here, using an improved oligo-RNA capping assay with the VSV L protein, we showed that the Michaelis constants for GDP and pppAACAG (VSV mRNA-start sequence) are 0.03 and 0.4 µM, respectively. A competition assay between GDP and GDP analogues in the GpppA formation and pRNA transfer assay using GDP analogues as pRNA acceptors indicated that the PRNTase domain recognizes the C-2-amino group, but not the C-6-oxo group, N-1-hydrogen, or N-7-nitrogen, of GDP for the cap formation. 2,6-Diaminopurine-riboside (DAP), 7-deazaguanosine (7-deaza-G), and 7-methylguanosine (m7G) diphosphates efficiently accepted pRNA, resulting in the formation of DAPpppA, 7-deaza-GpppA, and m7GpppA (cap 0), respectively. Furthermore, either the 2'- or 3'-hydroxyl group of GDP was found to be required for efficient pRNA transfer. A 5'-diphosphate form of antiviral ribavirin weakly inhibited the GpppA formation but did not act as a pRNA acceptor. These results indicate that the PRNTase domain has a unique guanosine-binding mode different from that of eukaryotic mRNA capping enzyme, guanylyltransferase. IMPORTANCE mRNAs of nonsegmented negative-strand (NNS) RNA viruses, such as VSV, possess a fully methylated cap structure, which is required for mRNA stability, efficient translation, and evasion of antiviral innate immunity in host cells. GDP polyribonucleotidyltransferase (PRNTase) is an unconventional mRNA capping enzyme of NNS RNA viruses that is distinct from the eukaryotic mRNA capping enzyme, guanylyltransferase. In this study, we studied the pRNA acceptor specificity of VSV PRNTase using various GDP analogues and identified chemical groups of GDP as essential for the substrate activity. The findings presented here are useful not only for understanding the mechanism of the substrate recognition with PRNTase but also for designing antiviral agents targeting this enzyme.


Subject(s)
Guanosine Diphosphate/metabolism , RNA Cap Analogs/metabolism , RNA, Messenger/metabolism , RNA-Dependent RNA Polymerase/metabolism , Vesiculovirus/enzymology , Viral Proteins/metabolism , Kinetics , Substrate Specificity
17.
Org Biomol Chem ; 16(36): 6741-6748, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30187040

ABSTRACT

Augmenting the mRNA translation efficiency and stability by replacing the standard 7-methylguanosine 5'-cap with properly designed analogues is a viable strategy for increasing the in vivo expression of proteins from exogenously delivered mRNA. However, the development of novel cap analogues with superior biological properties is hampered by the challenges associated with the synthesis of such highly modified nucleotides. To provide a simpler alternative to traditional methods for cap analogue preparation, we have recently proposed a click-chemistry-based strategy for the synthesis of dinucleotide cap analogues and identified several triazole-containing compounds with promising biochemical properties. Here, we further explored the concept of CuAAC-mediated cap synthesis by designing and studying 'second generation' triazole-modified caps, which were derived from the most promising 'first generation' compounds by modifying the oligophosphate chain length, altering the position of the triazole moiety, or replacing chemically labile P-N bonds with P-O bonds. The biochemical properties of the new analogues were evaluated by determining their affinity for eIF4E, susceptibility to hDcp2-catalysed decapping, and translation efficiencies in vitro and in cultured cells. The results led to identification of cap analogues that have superior translational properties compared to standard caps and the parent triazole-modified compounds as well as provided directions for future improvements.


Subject(s)
Protein Biosynthesis/drug effects , RNA Cap Analogs/chemistry , RNA Cap Analogs/pharmacology , Triazoles/chemistry , Drug Design , Drug Stability , Endoribonucleases/metabolism , Eukaryotic Initiation Factor-4E/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , RNA Cap Analogs/metabolism , RNA Stability , RNA, Messenger/genetics
18.
J Biol Chem ; 291(1): 363-70, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26559973

ABSTRACT

Influenza virus polymerase catalyzes the transcription of viral mRNAs by a process known as "cap-snatching," where the 5'-cap of cellular pre-mRNA is recognized by the PB2 subunit and cleaved 10-13 nucleotides downstream of the cap by the endonuclease PA subunit. Although this mechanism is common to both influenza A (FluA) and influenza B (FluB) viruses, FluB PB2 recognizes a wider range of cap structures including m(7)GpppGm-, m(7)GpppG-, and GpppG-RNA, whereas FluA PB2 utilizes methylated G-capped RNA specifically. Biophysical studies with isolated PB2 cap-binding domain (PB2(cap)) confirm that FluB PB2 has expanded mRNA cap recognition capability, although the affinities toward m(7)GTP are significantly reduced when compared with FluA PB2. The x-ray co-structures of the FluB PB2(cap) with bound cap analogs m(7)GTP and GTP reveal an inverted GTP binding mode that is distinct from the cognate m(7)GTP binding mode shared between FluA and FluB PB2. These results delineate the commonalities and differences in the cap-binding site between FluA and FluB PB2 and will aid structure-guided drug design efforts to identify dual inhibitors of both FluA and FluB PB2.


Subject(s)
Influenza B virus/enzymology , Protein Subunits/metabolism , RNA Caps/metabolism , Viral Proteins/metabolism , Calorimetry , Crystallography, X-Ray , Fluorometry , Influenza A virus/enzymology , Models, Molecular , Pliability , Protein Subunits/chemistry , RNA Cap Analogs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solutions , Viral Proteins/chemistry
19.
Biochim Biophys Acta ; 1864(10): 1292-303, 2016 10.
Article in English | MEDLINE | ID: mdl-27374989

ABSTRACT

The majority of eukaryotic mRNAs are translated in a cap-dependent manner, which requires recognition of the mRNA 5' cap by eIF4E protein. Multiple eIF4E family members have been identified in most eukaryotic organisms. Drosophila melanogaster (Dm) has eight eIF4E related proteins; seven of them belong to Class I and one to Class II. Their biological roles with the exception of Dm eIF4E-1, Dm eIF4E-3 and Dm 4EHP, remain unknown. Here, we compare the molecular basis of Dm eIF4E's interactions with cap and eIF4G peptide by using homology modelling and fluorescence binding assays with various cap analogues. We found that despite the presence of conserved key residues responsible for cap recognition, the differences in binding different cap analogues among Class I Dm eIF4E isoforms are up to 14-fold. The highest affinity for cap analogues was observed for Dm eIF4E-3. We suggest that Dm eIF4E-3 and Dm eIF4E-5 bind the second nucleoside of the cap in an unusual manner via stacking interactions with a histidine or a phenylalanine residue, respectively. Moreover, the analysis of ternary complexes of eIF4G peptide-eIF4E-cap analogue showed cooperativity between eIF4G and cap binding only for Dm eIF4E-4, which exhibits the lowest affinity for cap analogues among all Dm eIF4Es.


Subject(s)
Drosophila melanogaster/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Protein Isoforms/metabolism , RNA Caps/metabolism , Amino Acid Sequence , Animals , Binding Sites/physiology , Eukaryotic Initiation Factor-4G/metabolism , Histidine/metabolism , Models, Molecular , Peptides/metabolism , Phenylalanine/metabolism , Protein Binding/physiology , Protein Structure, Tertiary , RNA Cap Analogs/metabolism , Sequence Alignment
20.
Nucleic Acids Res ; 43(1): 482-92, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25432955

ABSTRACT

Eukaryotic 5' mRNA cap structures participate to the post-transcriptional control of gene expression before being released by the two main mRNA decay pathways. In the 3'-5' pathway, the exosome generates free cap dinucleotides (m7GpppN) or capped oligoribonucleotides that are hydrolyzed by the Scavenger Decapping Enzyme (DcpS) forming m7GMP. In the 5'-3' pathway, the decapping enzyme Dcp2 generates m7GDP. We investigated the fate of m7GDP and m7GpppN produced by RNA decay in extracts and cells. This defined a pathway involving DcpS, NTPs and the nucleoside diphosphate kinase for m7GDP elimination. Interestingly, we identified and characterized in vitro and in vivo a new scavenger decapping enzyme involved in m7GpppN degradation. We show that activities mediating cap elimination identified in yeast are essentially conserved in human. Their alteration may contribute to pathologies, possibly through the interference of cap (di)nucleotide with cellular function.


Subject(s)
Acid Anhydride Hydrolases/metabolism , Endoribonucleases/metabolism , Neoplasm Proteins/metabolism , RNA Caps/metabolism , RNA Stability , RNA, Messenger/metabolism , Adenosine Triphosphate/metabolism , Dinucleoside Phosphates/metabolism , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/metabolism , HEK293 Cells , Humans , N-Glycosyl Hydrolases/metabolism , Nucleoside-Diphosphate Kinase/metabolism , RNA Cap Analogs/metabolism , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL