Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
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.
RNA ; 29(2): 200-216, 2023 02.
Article in English | MEDLINE | ID: mdl-36418172

ABSTRACT

mRNA-based vaccines are relatively new technologies that have been in the field of interest of research centers and pharmaceutical companies in recent years. Such therapeutics are an attractive alternative for DNA-based vaccines since they provide material that can be used with no risk of genomic integration. Additionally, mRNA can be quite easily engineered to introduce modifications for different applications or to modulate its properties, for example, to increase translational efficiency or stability, which is not available for DNA vectors. Here, we describe the use of N2 modified dinucleotide cap analogs as components of mRNA transcripts. The compounds obtained showed very promising biological properties while incorporated into mRNA. The presented N2-guanine modifications within the cap structure ensure proper attachment of the dinucleotide to the transcripts in the IVT reaction, guarantees their incorporation only in the correct orientation, and enables highly efficient translation of mRNA both in the in vitro translation system and in human HEK293 cells.


Subject(s)
Protein Biosynthesis , Vaccines , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA Cap Analogs/chemistry , HEK293 Cells , Dinucleoside Phosphates
4.
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
5.
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
6.
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
7.
Bioorg Med Chem ; 28(13): 115523, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32362385

ABSTRACT

Eukaryotic translation initiation factor 4E (eIF4E) is overexpressed in many cancers deregulating translational control of the cell cycle. mRNA 5' cap analogs targeting eIF4E are small molecules with the potential to counteract elevated levels of eIF4E in cancer cells. However, the practical utility of typical cap analogs is limited because of their reduced cell membrane permeability. Transforming the active analogs into their pronucleotide derivatives is a promising approach to overcome this obstacle. 7-Benzylguanosine monophosphate (bn7GMP) is a cap analog that has been successfully transformed into a cell-penetrating pronucleotide by conjugation of the phosphate moiety with tryptamine. In this work, we explored whether a similar strategy is applicable to other cap analogs, particularly phosphate-modified 7-methylguanine nucleotides. We report the synthesis of six new tryptamine conjugates containing N7-methylguanosine mono- and diphosphate and their analogs modified with thiophosphate moiety. These new potential pronucleotides and the expected products of their activation were characterized by biophysical and biochemical methods to determine their affinity towards eIF4E, their ability to inhibit translation in vitro, their susceptibility to enzymatic degradation and their turnover in cell extract. The results suggest that compounds containing the thiophosphate moiety may act as pronucleotides that release low but sustainable concentrations of 7-methylguanosine 5'-phosphorothioate (m7GMPS), which is a translation inhibitor with in vitro potency higher than bn7GMP.


Subject(s)
Eukaryotic Initiation Factor-4E/genetics , Guanine/analogs & derivatives , Nucleotides/chemistry , Phosphates/chemistry , Tryptamines/chemistry , Endoribonucleases/metabolism , Genetic Variation , Guanine/chemistry , Guanosine/analogs & derivatives , Guanosine/chemistry , Humans , Models, Molecular , Nerve Tissue Proteins/metabolism , Nucleotide Motifs , Nucleotides/genetics , Protein Biosynthesis , RNA Cap Analogs/chemistry , RNA Cap Analogs/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics
8.
Bioorg Chem ; 96: 103583, 2020 03.
Article in English | MEDLINE | ID: mdl-31972465

ABSTRACT

Herein we describe a synthesis of new isoxazole-containing 5' mRNA cap analogues via a cycloaddition reaction. The obtained analogues show a capability to inhibit cap-dependent translation in vitro and are characterized by a new binding mode in which an isoxazolic ring, instead of guanine, is involved in the stacking effect. Our study provides valuable information toward designing new compounds that can be potentially used as anticancer therapeutics.


Subject(s)
Isoxazoles/chemistry , Isoxazoles/pharmacology , Peptide Chain Initiation, Translational/drug effects , RNA Cap Analogs/chemistry , RNA Cap Analogs/pharmacology , Animals , Drug Design , Eukaryotic Initiation Factor-4E/metabolism , Isoxazoles/chemical synthesis , Mice , Molecular Docking Simulation , RNA Cap Analogs/chemical synthesis , Rabbits
9.
Curr Protoc Nucleic Acid Chem ; 79(1): e100, 2019 12.
Article in English | MEDLINE | ID: mdl-31756051

ABSTRACT

This article describes a simple, reliable, efficient, and general method for the synthesis of 7-methylguanosine nucleotides such as 7-methylguanosine 5'-O-monophosphate (m7 GMP), 7-methylguanosine 5'-O-diphosphate (m7 GDP), 7-methyl-2'-deoxyguanosine 5'-O-triphosphate (m7 2'dGTP), and 7-methylguanosine 5'-O-triphosphate (m7 GTP) starting from the corresponding guanosine nucleotide is described. The present protocol involves methylation reaction of guanosine nucleotide using dimethyl sulfate as a methylating agent and water as a solvent at room temperature to provide the corresponding 7-methylguanosine nucleotide in good yields with high purity (>99.5%). It is noteworthy that the present methylation reaction proceeds smoothly under aqueous conditions that is highly regioselective to afford exclusive 7-methylguanosine nucleotide. © 2019 by John Wiley & Sons, Inc. Basic Protocol: Synthesis of 7-methylguanosine nucleotides.


Subject(s)
Guanosine Diphosphate/analogs & derivatives , RNA Cap Analogs/chemical synthesis , Guanosine Diphosphate/chemical synthesis , Guanosine Diphosphate/chemistry , Indicators and Reagents/chemistry , Methylation , RNA Cap Analogs/chemistry , Solvents/chemistry , Sulfuric Acid Esters/chemistry
10.
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
11.
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
12.
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
13.
Bioorg Med Chem ; 26(1): 191-199, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29195795

ABSTRACT

The hydrolysis of nucleoside 5'-monophosphates to the corresponding nucleosides and inorganic phosphate is catalysed by 5'-nucleotidases, thereby contributing to the control of endogenous nucleotide turnover and affecting the fate of exogenously delivered nucleotide- and nucleoside-derived therapeutics in cells. A recently identified nucleotidase cNIIIB shows preference towards 7-methylguanosine monophosphate (m7GMP) as a substrate, which suggests its potential involvement in mRNA degradation. However, the extent of biological functions and the significance of cNIIIB remains to be elucidated. Here, we synthesised a series of m7GMP analogues carrying a 1,2,3-triazole moiety at the 5' position as the potential inhibitors of human cNIIIB. The compounds were synthesised by using the copper-catalysed azide-alkyne cycloaddition (CuAAC) between 5'-azido-5'-deoxy-7-methylguanosine and different phosphate or phosphonate derivatives carrying terminal alkyne. The analogues were evaluated as cNIIIB inhibitors using HPLC and malachite green assays, demonstrating that compound 1a, carrying a 1,2,3-triazoylphosphonate moiety, inhibits cNIIIB activity at micromolar concentrations (IC50 87.8 ±â€¯7.5 µM), while other analogues showed no activity. In addition, compound 1d was identified as an artifical substrate for HscNIIIB. Further characterization of inhibitor 1a revealed that it is poorly recognised by other m7G-binding proteins, eIF4E and DcpS, indicating its selectivity towards cNIIIB. The first inhibitor (1a) and unnatural substrate (1d) of cNIIIB, identified here, can be used as molecular probes for the elucidation of biological roles of cNIIIB, including the verification of its proposed function in mRNA metabolism.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , RNA Cap Analogs/pharmacology , Triazoles/pharmacology , 5'-Nucleotidase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , RNA Cap Analogs/chemical synthesis , RNA Cap Analogs/chemistry , Structure-Activity Relationship , Triazoles/chemistry
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
16.
Methods Mol Biol ; 1428: 31-43, 2016.
Article in English | MEDLINE | ID: mdl-27236790

ABSTRACT

The 7-methylguanosine triphosphate cap present at the 5' ends of eukaryotic mRNAs plays numerous roles in mRNA expression and metabolism. The identification and studies on cap-binding partners can be significantly advanced using tailored chemical tools such as synthetic cap analogues or RNAs carrying modified cap structures. Here we provide protocols for the production of mRNAs specifically labeled within the 5' cap with a nucleoside capable of being photo-activated, either 6-thioguanosine or 7-methyl-6-thioguanosine, which can be used in photo-cross-linking experiments to identify or characterize cap-binding biomolecules. We also describe a protocol for the cross-linking experiments with capped RNAs to map histone H4 cap-binding pocket.


Subject(s)
Guanosine/analogs & derivatives , RNA Cap Analogs/metabolism , RNA, Messenger/chemistry , Thionucleosides/metabolism , Animals , Cross-Linking Reagents , Guanosine/metabolism , Histones/metabolism , Humans , Molecular Structure , Protein Biosynthesis , RNA Cap Analogs/chemistry , RNA, Messenger/metabolism , Ultraviolet Rays
17.
Methods Mol Biol ; 1428: 61-75, 2016.
Article in English | MEDLINE | ID: mdl-27236792

ABSTRACT

Fluorescent mRNA molecules offer a wide range of applications for studying capping/decapping reactions, translation, and other biophysical studies. Furthermore, fluorescent tags prove invaluable for tracking RNA molecules in cells. Here, we describe an efficient synthesis of a fluorescent cap analog, anthranioyl-GTP, its purification, and in vitro cap labeling of transcribed mRNA catalyzed by the recombinant vaccinia capping enzyme to produce anthranioyl-m(7)GpppG-capped RNA.


Subject(s)
RNA Cap Analogs/chemical synthesis , RNA, Messenger/chemistry , Guanosine/analogs & derivatives , Guanosine/chemistry , Molecular Structure , Protein Biosynthesis , RNA Cap Analogs/chemistry , RNA, Messenger/genetics , Spectrometry, Fluorescence , Transcription, Genetic
18.
Acta Crystallogr D Struct Biol ; 72(Pt 2): 245-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26894672

ABSTRACT

The RNA polymerase of influenza virus consists of three subunits: PA, PB1 and PB2. It uses a unique `cap-snatching' mechanism for the transcription of viral mRNAs. The cap-binding domain of the PB2 subunit (PB2cap) in the viral polymerase binds the cap of a host pre-mRNA molecule, while the endonuclease of the PA subunit cleaves the RNA 10-13 nucleotides downstream from the cap. The capped RNA fragment is then used as the primer for viral mRNA transcription. The structure of PB2cap from influenza virus H1N1 A/California/07/2009 and of its complex with the cap analog m(7)GTP were solved at high resolution. Structural changes are observed in the cap-binding site of this new pandemic influenza virus strain, especially the hydrophobic interactions between the ligand and the target protein. m(7)GTP binds deeper in the pocket than some other virus strains, much deeper than the host cap-binding proteins. Analysis of the new H1N1 structures and comparisons with other structures provide new insights into the design of small-molecule inhibitors that will be effective against multiple strains of both type A and type B influenza viruses.


Subject(s)
Viral Proteins/chemistry , Antiviral Agents/chemistry , Catalytic Domain , Crystallography, X-Ray , Guanosine Triphosphate/chemistry , Hydrogen Bonding , Influenza A Virus, H1N1 Subtype/enzymology , Models, Molecular , RNA Cap Analogs/chemistry
19.
Bioorg Med Chem ; 24(6): 1204-8, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26899596

ABSTRACT

The first example of the synthesis of new dinucleotide cap analog containing propargyl group such as m(7,3'-O-propargyl)G[5']ppp[5']G is reported. The effect of propargyl cap analog with standard cap was evaluated with respect to their capping efficiency, in vitro T7 RNA polymerase transcription efficiency, and translation activity using cultured HeLa cells. It is noteworthy that propargyl cap analog outperforms standard cap by 3.1 fold in terms of translational properties. The propargyl cap analog forms a more stable complex with translation initiation factor eIF4E based on the molecular modeling studies.


Subject(s)
Dinucleoside Phosphates/chemistry , Dinucleoside Phosphates/pharmacology , Drug Design , Eukaryotic Initiation Factor-4E/chemistry , Guanosine/analogs & derivatives , RNA Cap Analogs/chemistry , RNA Cap Analogs/pharmacology , DNA-Directed RNA Polymerases/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Guanosine/chemistry , HeLa Cells , Humans , Models, Molecular , RNA Cap Analogs/chemical synthesis , Transcription, Genetic/drug effects , Viral Proteins/metabolism
20.
RNA ; 22(4): 518-29, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26826132

ABSTRACT

Dcp1/2 is the major eukaryotic RNA decapping complex, comprised of the enzyme Dcp2 and activator Dcp1, which removes the 5' m(7)G cap from mRNA, committing the transcript to degradation. Dcp1/2 activity is crucial for RNA quality control and turnover, and deregulation of these processes may lead to disease development. The molecular details of Dcp1/2 catalysis remain elusive, in part because both cap substrate (m(7)GpppN) and m(7)GDP product are bound by Dcp1/2 with weak (mM) affinity. In order to find inhibitors to use in elucidating the catalytic mechanism of Dcp2, we screened a small library of synthetic m(7)G nucleotides (cap analogs) bearing modifications in the oligophosphate chain. One of the most potent cap analogs, m(7)GpSpppSm(7)G, inhibited Dcp1/2 20 times more efficiently than m(7)GpppN or m(7)GDP. NMR experiments revealed that the compound interacts with specific surfaces of both regulatory and catalytic domains of Dcp2 with submillimolar affinities. Kinetics analysis revealed that m(7)GpSpppSm(7)G is a mixed inhibitor that competes for the Dcp2 active site with micromolar affinity. m(7)GpSpppSm(7)G-capped RNA undergoes rapid decapping, suggesting that the compound may act as a tightly bound cap mimic. Our identification of the first small molecule inhibitor of Dcp2 should be instrumental in future studies aimed at understanding the structural basis of RNA decapping and may provide insight toward the development of novel therapeutically relevant decapping inhibitors.


Subject(s)
RNA Cap Analogs/chemistry , Schizosaccharomyces pombe Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical , RNA Cleavage , RNA, Messenger/chemistry , Schizosaccharomyces/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...