Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.230
Filter
1.
Sci Signal ; 17(837): eadi9844, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38771918

ABSTRACT

Oligoadenylate synthetase 3 (OAS3) and ribonuclease L (RNase L) are components of a pathway that combats viral infection in mammals. Upon detection of viral double-stranded RNA (dsRNA), OAS3 synthesizes 2'-5'-oligo(A), which activates the RNase domain of RNase L by promoting the homodimerization and oligomerization of RNase L monomers. Activated RNase L rapidly degrades all cellular mRNAs, shutting off several cellular processes. We sought to understand the molecular mechanisms underlying the rapid activation of RNase L in response to viral infection. Through superresolution microscopy and live-cell imaging, we showed that OAS3 and RNase L concentrated into higher-order cytoplasmic complexes known as dsRNA-induced foci (dRIF) in response to dsRNA or infection with dengue virus, Zika virus, or West Nile virus. The concentration of OAS3 and RNase L at dRIF corresponded with the activation of RNase L-mediated RNA decay. We showed that dimerized/oligomerized RNase L concentrated in a liquid-like shell surrounding a core OAS3-dRIF structure and dynamically exchanged with the cytosol. These data establish that the condensation of dsRNA, OAS3, and RNase L into dRIF is a molecular switch that promotes the rapid activation of RNase L upon detection of dsRNA in mammalian cells.


Subject(s)
2',5'-Oligoadenylate Synthetase , Endoribonucleases , RNA, Double-Stranded , Zika Virus , Endoribonucleases/metabolism , Endoribonucleases/genetics , Endoribonucleases/chemistry , Humans , 2',5'-Oligoadenylate Synthetase/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/chemistry , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , Zika Virus/metabolism , Animals , Dengue Virus/metabolism , RNA, Viral/metabolism , RNA, Viral/genetics , RNA Stability , West Nile virus/metabolism , West Nile virus/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/virology , Enzyme Activation , HeLa Cells , HEK293 Cells
2.
Nucleic Acids Res ; 52(10): 5438-5450, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38716860

ABSTRACT

In recent years, several noncanonical RNA caps derived from cofactors and metabolites have been identified. Purine-containing RNA caps have been extensively studied, with multiple decapping enzymes identified and efficient capture and sequencing protocols developed for nicotinamide adenine dinucleotide (NAD)-RNA, which allowed for a stepwise elucidation of capping functions. Despite being identified as an abundant noncanonical RNA-cap, UDP-sugar-capped RNA remains poorly understood, which is partly due to its complex in vitro preparation. Here, we describe a scalable synthesis of sugar-capped uridine-guanosine dinucleotides from readily available protected building blocks and their enzymatic conversion into several cell wall precursor-capped dinucleotides. We employed these capped dinucleotides in T7 RNA polymerase-catalyzed in vitro transcription reactions to efficiently generate RNAs capped with uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), its N-azidoacetyl derivative UDP-GlcNAz, and various cell wall precursors. We furthermore identified four enzymes capable of processing UDP-GlcNAc-capped RNA in vitro: MurA, MurB and MurC from Escherichia coli can sequentially modify the sugar-cap structure and were used to introduce a bioorthogonal, clickable moiety, and the human Nudix hydrolase Nudt5 was shown to efficiently decap UDP-GlcNAc-RNA. Our findings underscore the importance of efficient synthetic methods for capped model RNAs. Additionally, we provide useful enzymatic tools that could be utilized in the development and application of UDP-GlcNAc capture and sequencing protocols. Such protocols are essential for deepening our understanding of the widespread yet enigmatic GlcNAc modification of RNA and its physiological significance.


Subject(s)
RNA Caps , Uridine Diphosphate N-Acetylglucosamine , Uridine Diphosphate N-Acetylglucosamine/metabolism , RNA Caps/metabolism , Endoribonucleases/metabolism , Endoribonucleases/chemistry , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/chemistry , Humans , Escherichia coli/genetics , Escherichia coli/enzymology , Escherichia coli/metabolism , Viral Proteins
3.
J Biol Chem ; 300(3): 105729, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38336296

ABSTRACT

RNase P and RNase mitochondrial RNA processing (MRP) are ribonucleoproteins (RNPs) that consist of a catalytic RNA and a varying number of protein cofactors. RNase P is responsible for precursor tRNA maturation in all three domains of life, while RNase MRP, exclusive to eukaryotes, primarily functions in rRNA biogenesis. While eukaryotic RNase P is associated with more protein cofactors and has an RNA subunit with fewer auxiliary structural elements compared to its bacterial cousin, the double-anchor precursor tRNA recognition mechanism has remarkably been preserved during evolution. RNase MRP shares evolutionary and structural similarities with RNase P, preserving the catalytic core within the RNA moiety inherited from their common ancestor. By incorporating new protein cofactors and RNA elements, RNase MRP has established itself as a distinct RNP capable of processing ssRNA substrates. The structural information on RNase P and MRP helps build an evolutionary trajectory, depicting how emerging protein cofactors harmonize with the evolution of RNA to shape different functions for RNase P and MRP. Here, we outline the structural and functional relationship between RNase P and MRP to illustrate the coevolution of RNA and protein cofactors, a key driver for the extant, diverse RNP world.


Subject(s)
Endoribonucleases , Evolution, Molecular , Protein Subunits , RNA, Catalytic , Ribonuclease P , Coenzymes , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Ribonuclease P/chemistry , Ribonuclease P/metabolism , RNA Processing, Post-Transcriptional , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Substrate Specificity , Eukaryota/enzymology
4.
Arch Biochem Biophys ; 751: 109836, 2024 01.
Article in English | MEDLINE | ID: mdl-38000493

ABSTRACT

Fungal ribotoxins are extracellular RNases that inactivate ribosomes by cleaving a single phosphodiester bond at the universally conserved sarcin-ricin loop of the large rRNA. However, to reach the ribosomes, they need to cross the plasma membrane. It is there where these toxins show their cellular specificity, being especially active against tumoral or virus-infected cells. Previous studies have shown that fungal ribotoxins interact with negatively charged membranes, typically containing phosphatidylserine or phosphatidylglycerol. This ability is rooted on their long, non-structured, positively charged loops, and its N-terminal ß-hairpin. However, its effect on complex lipid mixtures, including sphingophospholipids or cholesterol, remains poorly studied. Here, wild-type α-sarcin was used to evaluate its interaction with a variety of membranes not assayed before, which resemble much more closely mammalian cell membranes. The results confirm that α-sarcin is particularly sensitive to charge density on the vesicle surface. Its ability to induce vesicle aggregation is strongly influenced by both the lipid headgroup and the degree of saturation of the fatty acid chains. Acyl chain length is indeed particularly important for lipid mixing. Finally, cholesterol plays an important role in diluting the concentration of available negative charges and modulates the ability of α-sarcin to cross the membrane.


Subject(s)
Endoribonucleases , Fungal Proteins , Cholesterol , Endoribonucleases/chemistry , Fungal Proteins/chemistry , Lipids
5.
Nucleic Acids Res ; 52(3): 1298-1312, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38117986

ABSTRACT

Bacteria harbor diverse mechanisms to defend themselves against their viral predators, bacteriophages. In response, phages can evolve counter-defense systems, most of which are poorly understood. In T4-like phages, the gene tifA prevents bacterial defense by the type III toxin-antitoxin (TA) system toxIN, but the mechanism by which TifA inhibits ToxIN remains unclear. Here, we show that TifA directly binds both the endoribonuclease ToxN and RNA, leading to the formation of a high molecular weight ribonucleoprotein complex in which ToxN is inhibited. The RNA binding activity of TifA is necessary for its interaction with and inhibition of ToxN. Thus, we propose that TifA inhibits ToxN during phage infection by trapping ToxN on cellular RNA, particularly the abundant 16S rRNA, thereby preventing cleavage of phage transcripts. Taken together, our results reveal a novel mechanism underlying inhibition of a phage-defensive RNase toxin by a small, phage-encoded protein.


Subject(s)
Bacteriophages , Toxin-Antitoxin Systems , Antitoxins/genetics , Bacteriophages/metabolism , Endoribonucleases/genetics , Endoribonucleases/chemistry , RNA, Ribosomal, 16S
6.
Science ; 382(6674): 1036-1041, 2023 12.
Article in English | MEDLINE | ID: mdl-38033086

ABSTRACT

Prokaryotic type III CRISPR-Cas antiviral systems employ cyclic oligoadenylate (cAn) signaling to activate a diverse range of auxiliary proteins that reinforce the CRISPR-Cas defense. Here we characterize a class of cAn-dependent effector proteins named CRISPR-Cas-associated messenger RNA (mRNA) interferase 1 (Cami1) consisting of a CRISPR-associated Rossmann fold sensor domain fused to winged helix-turn-helix and a RelE-family mRNA interferase domain. Upon activation by cyclic tetra-adenylate (cA4), Cami1 cleaves mRNA exposed at the ribosomal A-site thereby depleting mRNA and leading to cell growth arrest. The structures of apo-Cami1 and the ribosome-bound Cami1-cA4 complex delineate the conformational changes that lead to Cami1 activation and the mechanism of Cami1 binding to a bacterial ribosome, revealing unexpected parallels with eukaryotic ribosome-inactivating proteins.


Subject(s)
Bacteria , Bacterial Proteins , CRISPR-Associated Proteins , CRISPR-Cas Systems , Endoribonucleases , Bacteria/enzymology , Bacterial Proteins/chemistry , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/classification , RNA, Messenger/chemistry , Signal Transduction , Endoribonucleases/chemistry , Protein Domains
7.
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
8.
J Biol Chem ; 299(9): 105138, 2023 09.
Article in English | MEDLINE | ID: mdl-37544645

ABSTRACT

Through its role in intron cleavage, tRNA splicing endonuclease (TSEN) plays a critical function in the maturation of intron-containing pre-tRNAs. The catalytic mechanism and core requirement for this process is conserved between archaea and eukaryotes, but for decades, it has been known that eukaryotic TSENs have evolved additional modes of RNA recognition, which have remained poorly understood. Recent research identified new roles for eukaryotic TSEN, including processing or degradation of additional RNA substrates, and determined the first structures of pre-tRNA-bound human TSEN complexes. These recent discoveries have changed our understanding of how the eukaryotic TSEN targets and recognizes substrates. Here, we review these recent discoveries, their implications, and the new questions raised by these findings.


Subject(s)
Endoribonucleases , Eukaryota , RNA Precursors , RNA Splicing , RNA, Transfer , Humans , Introns/genetics , Nucleic Acid Conformation , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Substrate Specificity , Eukaryota/enzymology , Endoribonucleases/chemistry , Endoribonucleases/metabolism
9.
Int J Biol Macromol ; 249: 126110, 2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37536419

ABSTRACT

Human ribonuclease (RNase) 1 and bovine RNase A are the proto-types of the secretory "pancreatic-type" (pt)-RNase super-family. RNase A can oligomerize through the 3D domain swapping (DS) mechanism upon acetic acid (HAc) lyophilisation, producing enzymatically active oligomeric conformers by swapping both N- and C-termini. Also some RNase 1 mutants were found to self-associate through 3D-DS, however forming only N-swapped dimers. Notably, enzymatically active dimers and larger oligomers of wt-RNase 1 were collected here, in higher amount than RNase A, from HAc lyophilisation. In particular, RNase 1 self-associates through the 3D-DS of its N-terminus and, at a higher extent, of the C-terminus. Since RNase 1 is four-residues longer than RNase A, we further analyzed its oligomerization tendency in a mutant lacking the last four residues. The C-terminus role has been investigated also in amphibian onconase (ONC®), a pt-RNase that can form only a N-swapped dimer, since its C-terminus, that is three-residues longer than RNase A, is locked by a disulfide bond. While ONC mutants designed to unlock or cut this constraint were almost unable to dimerize, the RNase 1 mutant self-associated at a higher extent than the wt, suggesting a specific role of the C-terminus in the oligomerization of different RNases. Overall, RNase 1 reaches here the highest ability, among pt-RNases, to extensively self-associate through 3D-DS, paving the way for new investigations on the structural and biological properties of its oligomers.


Subject(s)
Ribonuclease, Pancreatic , Ribonucleases , Humans , Animals , Cattle , Ribonuclease, Pancreatic/chemistry , Ribonucleases/chemistry , Endoribonucleases/genetics , Endoribonucleases/chemistry , Protein Domains , Dimerization
10.
Nature ; 618(7963): 169-179, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225982

ABSTRACT

Target occupancy is often insufficient to elicit biological activity, particularly for RNA, compounded by the longstanding challenges surrounding the molecular recognition of RNA structures by small molecules. Here we studied molecular recognition patterns between a natural-product-inspired small-molecule collection and three-dimensionally folded RNA structures. Mapping these interaction landscapes across the human transcriptome defined structure-activity relationships. Although RNA-binding compounds that bind to functional sites were expected to elicit a biological response, most identified interactions were predicted to be biologically inert as they bind elsewhere. We reasoned that, for such cases, an alternative strategy to modulate RNA biology is to cleave the target through a ribonuclease-targeting chimera, where an RNA-binding molecule is appended to a heterocycle that binds to and locally activates RNase L1. Overlay of the substrate specificity for RNase L with the binding landscape of small molecules revealed many favourable candidate binders that might be bioactive when converted into degraders. We provide a proof of concept, designing selective degraders for the precursor to the disease-associated microRNA-155 (pre-miR-155), JUN mRNA and MYC mRNA. Thus, small-molecule RNA-targeted degradation can be leveraged to convert strong, yet inactive, binding interactions into potent and specific modulators of RNA function.


Subject(s)
Endoribonucleases , MicroRNAs , RNA, Messenger , Humans , Genes, jun/genetics , Genes, myc/genetics , MicroRNAs/antagonists & inhibitors , MicroRNAs/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Nucleic Acid Conformation , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Structure-Activity Relationship , Substrate Specificity , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Transcriptome
11.
Vet Microbiol ; 280: 109720, 2023 May.
Article in English | MEDLINE | ID: mdl-36921497

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is an enveloped positive-stranded RNA virus which causes serious economic losses to pig industry worldwide. Type I IFN induces expression of interferon-stimulated genes 15 (ISG15) to inhibit virus replication. To survive in the host, PRRSV has evolved to antagonize the antiviral response of ISGylation. Previous studies have reported that nonstructural protein 2 of PRRSV inhibits the ISGylation and antiviral function of ISG15 depending on its ovarian tumor (OTU) domain/papain-like protease domain (PLP2). However, whether there are other PRRSV proteins inhibiting ISGylation of cellular proteins is less well understood. In this study, we first found that PRRSV Nsp11 decreased ISGylation of cellular proteins. Meanwhile, the expression level of ISG15 was significantly inhibited by Nsp11. Further mechanistic studies demonstrated that the transcription of ISG15 was reduced by endoribonuclease activity of Nsp11. Finally, we found that the Nsp11-induced degradation of ISG15 was partially relied on autophagy-lysosome system. Taken together, PRRSV Nsp11 antagonizes the antiviral response of ISG15 by its endoribonuclease activity to promote PRRSV replication. Our results reveal a novel mechanism that PRRSV inhibits ISGylation of cellular proteins and impairs host innate immune response.


Subject(s)
Interferon Type I , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Swine Diseases , Animals , Swine , Porcine respiratory and reproductive syndrome virus/metabolism , Antiviral Agents/pharmacology , Cell Line , Endoribonucleases/genetics , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Immunity, Innate , Viral Nonstructural Proteins/metabolism , Virus Replication
12.
Acta Pharmacol Sin ; 44(1): 234-243, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35840659

ABSTRACT

Inositol-requiring enzyme 1α (IRE1α) is the most conserved endoplasmic reticulum (ER) stress sensor with two catalytic domains, kinase and RNase, in its cytosolic portion. IRE1α inhibitors have been used to improve existing clinical treatments against various cancers. In this study we identified toxoflavin (TXF) as a new-type potent small molecule IRE1α inhibitor. We used luciferase reporter systems to screen compounds that inhibited the IRE1α-XBP1s signaling pathway. As a result, TXF was found to be the most potent IRE1α RNase inhibitor with an IC50 value of 0.226 µM. Its inhibitory potencies on IRE1α kinase and RNase were confirmed in a series of cellular and in vitro biochemical assays. Kinetic analysis showed that TXF caused time- and reducing reagent-dependent irreversible inhibition on IRE1α, implying that ROS might participate in the inhibition process. ROS scavengers decreased the inhibition of IRE1α by TXF, confirming that ROS mediated the inhibition process. Mass spectrometry analysis revealed that the thiol groups of four conserved cysteine residues (CYS-605, CYS-630, CYS-715 and CYS-951) in IRE1α were oxidized to sulfonic groups by ROS. In molecular docking experiments we affirmed the binding of TXF with IRE1α, and predicted its binding site, suggesting that the structure of TXF itself participates in the inhibition of IRE1α. Interestingly, CYS-951 was just near the docked site. In addition, the RNase IC50 and ROS production in vitro induced by TXF and its derivatives were negative correlated (r = -0.872). In conclusion, this study discovers a new type of IRE1α inhibitor that targets a predicted new alternative site located in the junction between RNase domain and kinase domain, and oxidizes conserved cysteine residues of IRE1α active sites to inhibit IRE1α. TXF could be used as a small molecule tool to study IRE1α's role in ER stress.


Subject(s)
Endoribonucleases , Protein Serine-Threonine Kinases , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Inositol , Reactive Oxygen Species , Cysteine , Kinetics , Molecular Docking Simulation , Ribonucleases/metabolism , Endoplasmic Reticulum Stress/physiology , Enzyme Inhibitors/pharmacology , Oxidative Stress
13.
Cell Biochem Funct ; 40(8): 926-934, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36203381

ABSTRACT

The quick widespread of the coronavirus and speedy upsurge in the tally of cases demand the fast development of effective drugs. The uridine-directed endoribonuclease activity of nonstructural protein 15 (Nsp15) of the coronavirus is responsible for the invasion of the host immune system. Therefore, developing potential inhibitors against Nsp15 is a promising strategy. In this concern, the in silico approach can play a significant role, as it is fast and cost-effective in comparison to the trial and error approaches of experimental investigations. In this study, six turmeric derivatives (curcuminoids) were chosen for in silico analysis. The molecular interactions, pharmacokinetics, and drug-likeness of all the curcuminoids were measured. Further, the stability of Nsp15-curcuminoids complexes was appraised by employing molecular dynamics (MD) simulations and MM-PBSA approaches. All the molecules were affirmed to have strong interactions and pharmacokinetic profile. The MD simulations data stated that the Nsp15-curcuminoids complexes were stable during simulations. All the curcuminoids showed stable and high binding affinity, and these curcuminoids could be admitted as potential modulators for Nsp15 inhibition.


Subject(s)
COVID-19 , Viral Nonstructural Proteins , Humans , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , SARS-CoV-2/metabolism , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Antiviral Agents/pharmacology
14.
Nucleic Acids Res ; 50(18): 10665-10679, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36130271

ABSTRACT

The RNA-binding protein tristetraprolin (TTP) is a potent activator of mRNA decay, specifically for transcripts bearing AU-rich elements (AREs) in their 3'-untranslated regions. TTP functions as a mediator for mRNA decay by interacting with the decay machinery and recruiting it to the target ARE-mRNA. In this study, we report a weak, but direct interaction between TTP and the human decapping enzyme DCP2, which impacts the stability of ARE transcripts. The TTP-DCP2 interaction is unusual as it involves intrinsically disordered regions (IDRs) of both binding partners. We show that the IDR of DCP2 has a propensity for oligomerization and liquid-liquid phase separation in vitro. Binding of TTP to DCP2 leads to its partitioning into phase-separated droplets formed by DCP2, suggesting that molecular crowding might facilitate the weak interaction between the two proteins and enable assembly of a decapping-competent mRNA-protein complex on TTP-bound transcripts in cells. Our studies underline the role of weak interactions in the cellular interaction network and their contribution towards cellular functionality.


Subject(s)
Endoribonucleases/chemistry , RNA Stability , Tristetraprolin/chemistry , 3' Untranslated Regions , Endoribonucleases/metabolism , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Tristetraprolin/genetics , Tristetraprolin/metabolism
15.
J Mol Biol ; 434(20): 167796, 2022 10 30.
Article in English | MEDLINE | ID: mdl-35995266

ABSTRACT

Global sequencing efforts from the ongoing COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, continue to provide insight into the evolution of the viral genome. Coronaviruses encode 16 nonstructural proteins, within the first two-thirds of their genome, that facilitate viral replication and transcription as well as evasion of the host immune response. However, many of these viral proteins remain understudied. Nsp15 is a uridine-specific endoribonuclease conserved across all coronaviruses. The nuclease activity of Nsp15 helps the virus evade triggering an innate immune response. Understanding how Nsp15 has changed over the course of the pandemic, and how mutations affect its RNA processing function, will provide insight into the evolution of an oligomerization-dependent endoribonuclease and inform drug design. In combination with previous structural data, bioinformatics analyses of 1.9 + million SARS-CoV-2 sequences revealed mutations across Nsp15's three structured domains (N-terminal, Middle, EndoU). Selected Nsp15 variants were characterized biochemically and compared to wild type Nsp15. We found that mutations to important catalytic residues decreased cleavage activity but increased the hexamer/monomer ratio of the recombinant protein. Many of the highly prevalent variants we analyzed led to decreased nuclease activity as well as an increase in the inactive, monomeric form. Overall, our work establishes how Nsp15 variants seen in patient samples affect nuclease activity and oligomerization, providing insight into the effect of these variants in vivo.


Subject(s)
COVID-19 , Endoribonucleases , SARS-CoV-2 , Uridylate-Specific Endoribonucleases , Viral Nonstructural Proteins , COVID-19/virology , Endoribonucleases/chemistry , Endoribonucleases/genetics , Humans , Recombinant Proteins/chemistry , SARS-CoV-2/enzymology , Uridylate-Specific Endoribonucleases/chemistry , Uridylate-Specific Endoribonucleases/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
16.
Nucleic Acids Res ; 50(18): e106, 2022 10 14.
Article in English | MEDLINE | ID: mdl-35871301

ABSTRACT

With the rapid growth of synthetic messenger RNA (mRNA)-based therapeutics and vaccines, the development of analytical tools for characterization of long, complex RNAs has become essential. Tandem liquid chromatography-mass spectrometry (LC-MS/MS) permits direct assessment of the mRNA primary sequence and modifications thereof without conversion to cDNA or amplification. It relies upon digestion of mRNA with site-specific endoribonucleases to generate pools of short oligonucleotides that are then amenable to MS-based sequence analysis. Here, we showed that the uridine-specific human endoribonuclease hRNase 4 improves mRNA sequence coverage, in comparison with the benchmark enzyme RNase T1, by producing a larger population of uniquely mappable cleavage products. We deployed hRNase 4 to characterize mRNAs fully substituted with 1-methylpseudouridine (m1Ψ) or 5-methoxyuridine (mo5U), as well as mRNAs selectively depleted of uridine-two key strategies to reduce synthetic mRNA immunogenicity. Lastly, we demonstrated that hRNase 4 enables direct assessment of the 5' cap incorporation into in vitro transcribed mRNA. Collectively, this study highlights the power of hRNase 4 to interrogate mRNA sequence, identity, and modifications by LC-MS/MS.


Subject(s)
Endoribonucleases/chemistry , RNA, Messenger/chemistry , Sequence Analysis, RNA/methods , Tandem Mass Spectrometry , Chromatography, Liquid/methods , DNA, Complementary , Humans , Oligonucleotides/analysis , RNA, Messenger/genetics , Ribonuclease T1/metabolism , Tandem Mass Spectrometry/methods
17.
Structure ; 30(5): 721-732.e4, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35290794

ABSTRACT

Poxviruses encode decapping enzymes that remove the protective 5' cap from both host and viral mRNAs to commit transcripts for decay by the cellular exonuclease Xrn1. Decapping by these enzymes is critical for poxvirus pathogenicity by means of simultaneously suppressing host protein synthesis and limiting the accumulation of viral double-stranded RNA (dsRNA), a trigger for antiviral responses. Here we present a high-resolution structural view of the vaccinia virus decapping enzyme D9. This Nudix enzyme contains a domain organization different from other decapping enzymes in which a three-helix bundle is inserted into the catalytic Nudix domain. The 5' mRNA cap is positioned in a bipartite active site at the interface of the two domains. Specificity for the methylated guanosine cap is achieved by stacking between conserved aromatic residues in a manner similar to that observed in canonical cap-binding proteins VP39, eIF4E, and CBP20, and distinct from eukaryotic decapping enzyme Dcp2.


Subject(s)
Poxviridae , Viral Proteins , Catalysis , Endoribonucleases/chemistry , Endoribonucleases/genetics , Endoribonucleases/metabolism , Poxviridae/genetics , Poxviridae/metabolism , RNA Caps/metabolism , RNA, Double-Stranded , RNA, Messenger/metabolism , Vaccinia virus/genetics , Viral Proteins/metabolism
18.
Viruses ; 14(2)2022 01 27.
Article in English | MEDLINE | ID: mdl-35215858

ABSTRACT

The genus Pestivirus, family Flaviviridae, includes four historically accepted species, i.e., bovine viral diarrhea virus (BVDV)-1 and -2, classical swine fever virus (CSFV), and border disease virus (BDV). A large number of new pestivirus species were identified in recent years. A common feature of most members is the presence of two unique proteins, Npro and Erns, that pestiviruses evolved to regulate the host's innate immune response. In addition to its function as a structural envelope glycoprotein, Erns is also released in the extracellular space, where it is endocytosed by neighboring cells. As an endoribonuclease, Erns is able to cleave viral ss- and dsRNAs, thus preventing the stimulation of the host's interferon (IFN) response. Here, we characterize the basic features of soluble Erns of a large variety of classified and unassigned pestiviruses that have not yet been described. Its ability to form homodimers, its RNase activity, and the ability to inhibit dsRNA-induced IFN synthesis were investigated. Overall, we found large differences between the various Erns proteins that cannot be predicted solely based on their primary amino acid sequences, and that might be the consequence of different virus-host co-evolution histories. This provides valuable information to delineate the structure-function relationship of pestiviral endoribonucleases.


Subject(s)
Endoribonucleases/metabolism , Immune Evasion , Immunity, Innate , Pestivirus/immunology , Pestivirus/pathogenicity , Viral Envelope Proteins/metabolism , Animals , Cell Line , Endocytosis , Endoribonucleases/chemistry , Endoribonucleases/genetics , Interferons/antagonists & inhibitors , Interferons/biosynthesis , Mutation , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Pestivirus/metabolism , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
19.
Bioorg Med Chem ; 58: 116653, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35152173

ABSTRACT

Aminothiophene is a scaffold that is widely present in drugs and biologically active small molecules as chemical probes. In this study, 43 compounds sharing a 2-aminothiophenone-3-carboxylate (ATPC) scaffold, known to activate the ribonuclease L (RNase L), were synthesized and selected ATPCs showed enhancement of thermal stability of RNase L upon binding. Screening of antiproliferation activities against human cancer cell lines revealed that ATPCs represented by compounds 4l and 50 showed potent single-digit micromolar antiproliferation activity against human cancer cell lines. Compounds 4l and 50 exhibited time- and dose-dependent proliferation inhibition, induced cellular apoptosis measured by cleaved PARP and via flow cytometry, inhibited cell migration, and inhibited cell colony formation. Combining the results reported in this work, ATPCs were evaluated as potential anticancer agents mediated by RNase L-binding and apoptosis induction. The work contributes to the study on the polypharmacological properties of aminothiophene-containing small molecules.


Subject(s)
Antineoplastic Agents/pharmacology , Endoribonucleases/chemistry , Thiophenes/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Endoribonucleases/metabolism , Humans , Molecular Structure , Structure-Activity Relationship , Thiophenes/chemistry , Thiophenes/metabolism
20.
J Biomol Struct Dyn ; 40(11): 4879-4892, 2022 07.
Article in English | MEDLINE | ID: mdl-33357040

ABSTRACT

The World Health Organization has classified the COVID-19 outbreak a pandemic which is caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) and declared it a global health emergency. Repurposing drugs with minimum side effects are one approach to quickly respond in attempt to prevent the spread of COVID-19. SARS-CoV-2 encodes several RNA processing enzymes that are unusual and unique for single-stranded RNA viruses, including Nsp15, a hexameric endoribonuclease that discriminatory cleaves immediately 3' of uridines. The structure of SARS-CoV-2 Nsp15 is reported to be homologous to that of the Nsp15 endoribonucleases of SARS-CoV and MERS-CoV, but it exhibits differences that may contribute to the greater virulence of SARS-CoV-2. This study aimed to identify drugs that targeted SARS-COV-2 Nsp15 using a molecular docking-based virtual screening of a library containing 10,000 approved and experimental drugs. The molecular docking results revealed 19 medications that demonstrated a good ability to inhibit Nsp15. Among all the candidated 19 drugs only five FDA approved drugs were used for further investigation by molecular dynamics simulation, the stability of Nsp15-ligand system was evaluated by calculating the RMSD, RMSF, radius of gyration and hydrogen bond profile. Furthermore, MM-PBSA method was employed to validate the binding affinity. According to the obtained results of MD, the complex of Olaparib was showed more stability and lower binding free energy than the control inhibitor during MD simulation time. Finally, we suggest that Olaparib is a potential drug for treating patients infected with SARS-CoV-2 and provide insight into the host immune response to viral RNA.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antiviral Agents , Endoribonucleases , SARS-CoV-2 , Viral Nonstructural Proteins , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/chemistry , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , RNA, Viral , SARS-CoV-2/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL
...