Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
EMBO Rep ; 24(10): e56808, 2023 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-37642556

RESUMO

Nervous system function rests on the formation of functional synapses between neurons. We have identified TRMT9B as a new regulator of synapse formation and function in Drosophila. TRMT9B has been studied for its role as a tumor suppressor and is one of two metazoan homologs of yeast tRNA methyltransferase 9 (Trm9), which methylates tRNA wobble uridines. Whereas Trm9 homolog ALKBH8 is ubiquitously expressed, TRMT9B is enriched in the nervous system. However, in the absence of animal models, TRMT9B's role in the nervous system has remained unstudied. Here, we generate null alleles of TRMT9B and find it acts postsynaptically to regulate synaptogenesis and promote neurotransmission. Through liquid chromatography-mass spectrometry, we find that ALKBH8 catalyzes canonical tRNA wobble uridine methylation, raising the question of whether TRMT9B is a methyltransferase. Structural modeling studies suggest TRMT9B retains methyltransferase function and, in vivo, disruption of key methyltransferase residues blocks TRMT9B's ability to rescue synaptic overgrowth, but not neurotransmitter release. These findings reveal distinct roles for TRMT9B in the nervous system and highlight the significance of tRNA methyltransferase family diversification in metazoans.


Assuntos
Saccharomyces cerevisiae , tRNA Metiltransferases , Animais , tRNA Metiltransferases/genética , tRNA Metiltransferases/metabolismo , Metilação , Saccharomyces cerevisiae/genética , Uridina/química , Uridina/genética , Uridina/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo
2.
J Mol Biol ; 435(24)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38708190

RESUMO

Cytidine (C) to Uridine (U) RNA editing is a post-transcription modification that is involved in diverse biological processes. APOBEC1 (A1) catalyzes the conversion of C-to-U in RNA, which is important in regulating cholesterol metabolism through its editing activity on ApoB mRNA. However, A1 requires a cofactor to form an "editosome" for RNA editing activity. A1CF and RBM47, both RNA-binding proteins, have been identified as cofactors that pair with A1 to form editosomes and edit ApoB mRNA and other cellular RNAs. SYNCRIP is another RNA-binding protein that has been reported as a potential regulator of A1, although it is not directly involved in A1 RNA editing activity. Here, we describe the identification and characterization of a novel cofactor, RBM46 (RNA-Binding-Motif-protein-46), that can facilitate A1 to perform C-to-U editing on ApoB mRNA. Additionally, using the low-error circular RNA sequencing technique, we identified novel cellular RNA targets for the A1/RBM46 editosome. Our findings provide further insight into the complex regulatory network of RNA editing and the potential new function of A1 with its cofactors.


Assuntos
Desaminase APOBEC-1 , Edição de RNA , Proteínas de Ligação a RNA , Uridina , Desaminase APOBEC-1/metabolismo , Desaminase APOBEC-1/genética , Humanos , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Uridina/metabolismo , Uridina/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células HEK293 , Apolipoproteínas B/metabolismo , Apolipoproteínas B/genética , Citidina/metabolismo , Citidina/genética
3.
PLoS Pathog ; 18(11): e1010976, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36374932

RESUMO

The eukaryotic multisubunit Elongator complex has been shown to perform multiple functions in transcriptional elongation, histone acetylation and tRNA modification. However, the Elongator complex plays different roles in different organisms, and the underlying mechanisms remain unexplored. Moreover, the biological functions of the Elongator complex in human fungal pathogens remain unknown. In this study, we verified that the Elongator complex of the opportunistic fungal pathogen Aspergillus fumigatus consists of six subunits (Elp1-6), and the loss of any subunit results in similarly defective colony phenotypes with impaired hyphal growth and reduced conidiation. The catalytic subunit-Elp3 of the Elongator complex includes a S-adenosyl methionine binding (rSAM) domain and a lysine acetyltransferase (KAT) domain, and it plays key roles in the hyphal growth, biofilm-associated exopolysaccharide galactosaminogalactan (GAG) production, adhesion and virulence of A. fumigatus; however, Elp3 does not affect H3K14 acetylation levels in vivo. LC-MS/MS chromatograms revealed that loss of Elp3 abolished the 5-methoxycarbonylmethyl-2-thiouridine (mcm5s2U) modification of tRNA wobble uridine (U34), and the overexpression of tRNAGlnUUG and tRNAGluUUC, which normally harbor mcm5s2U modifications, mainly rescues the defects of the Δelp3 mutant, suggesting that tRNA modification rather than lysine acetyltransferase is responsible for the primary function of Elp3 in A. fumigatus. Strikingly, global proteomic comparison analyses showed significantly upregulated expression of genes related to amino acid metabolism in the Δelp3 mutant strain compared to the wild-type strain. Western blotting showed that deletion of elp3 resulted in overexpression of the amino acid starvation-responsive transcription factor CpcA, and deletion of CpcA markedly reversed the defective phenotypes of the Δelp3 mutant, including attenuated virulence. Therefore, the findings of this study demonstrate that A. fumigatus Elp3 functions as a tRNA-modifying enzyme in the regulation of growth, GAG production, adhesion and virulence by maintaining intracellular amino acid homeostasis. More broadly, our study highlights the importance of U34 tRNA modification in regulating cellular metabolic states and virulence traits of fungal pathogens.


Assuntos
Aspergillus fumigatus , Histona Acetiltransferases , Humanos , Uridina/genética , Aspergillus fumigatus/genética , Aspergillus fumigatus/metabolismo , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Virulência , Proteômica , Cromatografia Líquida , Espectrometria de Massas em Tandem , RNA de Transferência/genética , RNA de Transferência/química , RNA de Transferência/metabolismo , Aminoácidos
4.
Nucleic Acids Res ; 50(17): 9966-9983, 2022 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-36107771

RESUMO

RNA editing processes are strikingly different in animals and plants. Up to thousands of specific cytidines are converted into uridines in plant chloroplasts and mitochondria whereas up to millions of adenosines are converted into inosines in animal nucleo-cytosolic RNAs. It is unknown whether these two different RNA editing machineries are mutually incompatible. RNA-binding pentatricopeptide repeat (PPR) proteins are the key factors of plant organelle cytidine-to-uridine RNA editing. The complete absence of PPR mediated editing of cytosolic RNAs might be due to a yet unknown barrier that prevents its activity in the cytosol. Here, we transferred two plant mitochondrial PPR-type editing factors into human cell lines to explore whether they could operate in the nucleo-cytosolic environment. PPR56 and PPR65 not only faithfully edited their native, co-transcribed targets but also different sets of off-targets in the human background transcriptome. More than 900 of such off-targets with editing efficiencies up to 91%, largely explained by known PPR-RNA binding properties, were identified for PPR56. Engineering two crucial amino acid positions in its PPR array led to predictable shifts in target recognition. We conclude that plant PPR editing factors can operate in the entirely different genetic environment of the human nucleo-cytosol and can be intentionally re-engineered towards new targets.


Assuntos
Proteínas de Plantas , Proteínas de Ligação a RNA , Aminoácidos , Citidina , Humanos , Proteínas de Plantas/genética , RNA/genética , RNA Mitocondrial/genética , RNA de Plantas/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Uridina/genética
5.
Nat Commun ; 12(1): 5201, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34465779

RESUMO

N6-methyladenosine (m6A) is a post-transcriptional modification that controls gene expression by recruiting proteins to RNA sites. The modification also slows biochemical processes through mechanisms that are not understood. Using temperature-dependent (20°C-65°C) NMR relaxation dispersion, we show that m6A pairs with uridine with the methylamino group in the anti conformation to form a Watson-Crick base pair that transiently exchanges on the millisecond timescale with a singly hydrogen-bonded low-populated (1%) mismatch-like conformation in which the methylamino group is syn. This ability to rapidly interchange between Watson-Crick or mismatch-like forms, combined with different syn:anti isomer preferences when paired (~1:100) versus unpaired (~10:1), explains how m6A robustly slows duplex annealing without affecting melting at elevated temperatures via two pathways in which isomerization occurs before or after duplex annealing. Our model quantitatively predicts how m6A reshapes the kinetic landscape of nucleic acid hybridization and conformational transitions, and provides an explanation for why the modification robustly slows diverse cellular processes.


Assuntos
Adenosina/análogos & derivados , DNA/química , DNA/metabolismo , Adenosina/química , Adenosina/genética , Adenosina/metabolismo , Pareamento de Bases , DNA/genética , Ligação de Hidrogênio , Cinética , Modelos Moleculares , Conformação de Ácido Nucleico , Hibridização de Ácido Nucleico , Processamento Pós-Transcricional do RNA , Uridina/química , Uridina/genética , Uridina/metabolismo
6.
Cell Commun Signal ; 19(1): 84, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34380502

RESUMO

RNA base editing is potential for cellular function research and genetic diseases treating. There are two main RNA base editors, REPAIR and RESCUE, for in vitro use. REPAIR was developed by fusing inactivated Cas13 (dCas13) with the adenine deaminase domain of ADAR2, which efficiently performs adenosine-to-inosine (A-to-I) RNA editing. RESCUE, which performs both cytidine-to-uridine (C-to-U) and A-to-I RNA editing, was developed by fusing inactivated Cas13 (dCas13) with the evolved ADAR2. However, the relatively low editing efficiency of the RESCUE system limits its broad application. Here, we constructed an enhanced RESCUE (eRESCUE) system; this dPspCas13b-RESCUE-NES system was generated by fusing inactivated PspCas13b with the evolved ADAR2. We determined the endogenous mRNA A-to-I and C-to-U editing efficiency mediated by the dPspCas13b-RESCUE-NES system in HEK-293T cells. This new RNA base editor was then used to induce 177Ser/Gly conversion of inhibitor kappa B kinase ß (IKKß) by changing the genetic code from AGU to GGU. The results showed that the eRESCUE editor mediates more efficient A-to-I and C-to-U RNA editing than the RESCUE RNA editor, as was previously reported. The 177Ser/Gly conversion of IKKß, accomplished by converting the genetic code from AGU to GGU, resulted in a decrease in the phosphorylation of IKKß and downregulation of downstream IKKß-related genes. In summary, we developed a more efficient RNA base editor, eRESCUE, which may provide a useful tool for biomedical research and genetic disease treatment. Video Abstract.


Assuntos
Adenosina Desaminase/genética , Doenças Genéticas Inatas/genética , Edição de RNA/genética , Proteínas de Ligação a RNA/genética , RNA/genética , Adenosina/genética , Sistemas CRISPR-Cas/genética , Citidina/genética , Doenças Genéticas Inatas/terapia , Células HEK293 , Humanos , Quinase I-kappa B/genética , Inosina/genética , Uridina/genética
7.
Virol J ; 18(1): 37, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33602251

RESUMO

BACKGROUND: Hepatitis B virus (HBV) infection is difficult to cure. HBV-specific immune tolerance plays a key role in HBV persistence, and enhancing cellular and humoral immunity will improve the control of HBV infection. The purpose of the study was to explore the anti-HBV and immunostimulatory effects of msiRNAs that introduce unpaired uridine bulges in the passenger strand. METHODS: msiRNAs targeting the HBV S and X genes were designed and named msiHBs and msiHBx, respectively. HepG2 cells were cotransfected with siRNA or msiRNA and the HBV replication-competent plasmid pHY106-wta or pHY106-X15. HepG2.215 cells were transfected with siRNA or msiRNA. The levels of HBsAg, HBeAg, and the cytokines TNF-α, IFN-α, IFN-ß, IL-1α, and IL-6 in the culture supernatant was detected by ELISA. The levels of intracellular HBV RNA, nuclear HBV replication intermediates, and HBV DNA in the supernatant were measured by quantitative RT-PCR and PCR. The levels of HBV replication intermediates were detected by Southern blotting. Peripheral blood mononuclear cells were transfected with siRNA or msiRNA, and the levels of secreted cytokines IFN-α and IFN-ß were detected by ELISA. The bioactivity of type I interferons in the supernatants was detected by the virus protection assay. RESULTS: msiHBx treatment led to a significant decrease in HBsAg (to a negative level) and HBV DNA (95.5%) in the supernatant and intrahepatocellular HBV replication intermediates (89.8%) in HepG2 cells with transient HBV replication and in HepG2.2.15 cells. There was no significant difference between msiHBx and siHBx in terms of the reduction in HBV proteins and HBV replication (P > 0.05). Compared with siHBx, msiHBx treatment of HepG2 cells transfected with the HBV replication-competent plasmid led to a significant increase in the levels of the antiviral cytokines TNF-α (3.3-fold), IFN-α (1.4-fold), and IFN-ß (2.5-fold) (P < 0.01), without upregulation of the proinflammatory cytokines IL-1α and IL-6. The virus protection assay results showed msiHBx-mediated type I interferons effectively protected L929 cells against ECMV infection. CONCLUSIONS: msiHBx could effectively inhibit HBV expression and replication and induce an antiviral innate immune response without proinflammatory activation. The dual RNAi and immunostimulatory activity of msiRNAs may play an important role in the control of HBV infection.


Assuntos
Vírus da Hepatite B/genética , Vírus da Hepatite B/imunologia , Hepatite B/imunologia , Imunidade Inata , RNA Interferente Pequeno/química , RNA Interferente Pequeno/imunologia , Uridina/metabolismo , Genes Virais , Células Hep G2 , Humanos , Imunização , Leucócitos Mononucleares/metabolismo , RNA Interferente Pequeno/síntese química , RNA Interferente Pequeno/genética , Transfecção , Uridina/genética , Replicação Viral
8.
Int J Mol Sci ; 23(1)2021 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-35008519

RESUMO

Genome integrity must be tightly preserved to ensure cellular survival and to deter the genesis of disease. Endogenous and exogenous stressors that impose threats to genomic stability through DNA damage are counteracted by a tightly regulated DNA damage response (DDR). RNA binding proteins (RBPs) are emerging as regulators and mediators of diverse biological processes. Specifically, RBPs that bind to adenine uridine (AU)-rich elements (AREs) in the 3' untranslated region (UTR) of mRNAs (AU-RBPs) have emerged as key players in regulating the DDR and preserving genome integrity. Here we review eight established AU-RBPs (AUF1, HuR, KHSRP, TIA-1, TIAR, ZFP36, ZFP36L1, ZFP36L2) and their ability to maintain genome integrity through various interactions. We have reviewed canonical roles of AU-RBPs in regulating the fate of mRNA transcripts encoding DDR genes at multiple post-transcriptional levels. We have also attempted to shed light on non-canonical roles of AU-RBPs exploring their post-translational modifications (PTMs) and sub-cellular localization in response to genotoxic stresses by various factors involved in DDR and genome maintenance. Dysfunctional AU-RBPs have been increasingly found to be associated with many human cancers. Further understanding of the roles of AU-RBPS in maintaining genomic integrity may uncover novel therapeutic strategies for cancer.


Assuntos
Adenina/metabolismo , Genoma/genética , Processamento Pós-Transcricional do RNA/genética , Proteínas de Ligação a RNA/genética , Uridina/genética , Regiões 3' não Traduzidas/genética , Animais , Humanos
9.
Methods Mol Biol ; 2181: 13-34, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32729072

RESUMO

Computers are able to systematically exploit RNA-seq data allowing us to efficiently detect RNA editing sites in a genome-wide scale. This chapter introduces a very flexible computational framework for detecting RNA editing sites in plant organelles. This framework comprises three major steps: RNA-seq data processing, RNA read alignment, and RNA editing site detection. Each step is discussed in sufficient detail to be implemented by the reader. As a study case, the framework will be used with publicly available sequencing data to detect C-to-U RNA editing sites in the coding sequences of the mitochondrial genome of Nicotiana tabacum.


Assuntos
Biologia Computacional/métodos , Genoma Mitocondrial , Mitocôndrias/genética , Nicotiana/genética , Edição de RNA/genética , RNA Mitocondrial/genética , Citidina/química , Citidina/genética , Sequenciamento de Nucleotídeos em Larga Escala , Mitocôndrias/metabolismo , RNA Mitocondrial/metabolismo , Software , Nicotiana/metabolismo , Transcriptoma , Uridina/química , Uridina/genética
10.
Methods Mol Biol ; 2181: 51-67, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32729074

RESUMO

The AID/APOBEC family of enzymes are cytidine deaminases that act upon DNA and RNA. Among APOBECs, the best characterized family member to act on RNA is the enzyme APOBEC1. APOBEC1-mediated RNA editing plays a key role in lipid metabolism and in maintenance of brain homeostasis. Editing can be easily detected in RNA-seq data as a cytosine to thymine (C-to-T) change with regard to the reference. However, there are many other sources of base conversions relative to reference, such as PCR errors, SNPs, and even DNA editing by mutator APOBECs. Furthermore, APOBEC1 exhibits disparate activity in different cell types, with respect to which transcripts are edited and the level to which they are edited. When considering these potential sources of error and variability, an RNA-seq comparison between wild-type APOBEC1 sample and a matched control with an APOBEC1 knockout is a reliable method for the discrimination of true sites edited by APOBEC1. Here we present a detailed description of a method for studying APOBEC1 RNA editing, specifically in the murine macrophage cell line RAW 264.7. Our method covers the production of an APOBEC1 knockout cell line using the CRISPR/Cas9 system, through to experimental validation and quantification of editing sites (where we discuss a recently published algorithm (termed MultiEditR) which allows for the detection and quantification of RNA editing from Sanger sequencing). Importantly, this same protocol can be adapted to any RNA modification detectable by RNA-seq analysis for which the responsible protein is known.


Assuntos
Desaminase APOBEC-1/genética , Sistemas CRISPR-Cas , Biologia Computacional/métodos , Citidina/genética , Macrófagos/metabolismo , Edição de RNA/genética , Uridina/genética , Desaminase APOBEC-1/antagonistas & inibidores , Animais , Citidina/química , Sequenciamento de Nucleotídeos em Larga Escala , Macrófagos/citologia , Camundongos , Células RAW 264.7 , RNA Mensageiro/genética , Uridina/química
11.
Methods Mol Biol ; 2181: 69-81, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32729075

RESUMO

APOBEC1 is a member of the AID/APOBECs, a group of deaminases responsible for the editing of C>U in both DNA and RNA. APOBEC1 is physiologically involved in C>U RNA editing: while hundreds of targets have been discovered in mice, in humans the only well-characterized target of APOBEC1 is the apolipoprotein B (ApoB) transcript. APOBEC1 edits a CAA codon into a stop codon, which causes the translation of a truncated form of ApoB. A number of assays have been developed to investigate this process. Early assays, poisoned primer extension and Sanger sequencing, have focused on accuracy and sensitivity but rely on extraction of the RNA from tissues and cells. More recently, the need to visualize the RNA editing process directly in live cells have led to the development of fluorescence-based tools. These assays detect RNA editing through reporters whose editing causes a change in cellular localization or a change in fluorescent properties. Here we review the available assays to quantify RNA editing, and we present the protocol for cytofluorimetric analysis using a double-fluorescent reporter.


Assuntos
Desaminase APOBEC-1/genética , Biologia Computacional/métodos , Citidina/genética , Edição de RNA/genética , RNA Mensageiro/genética , Frações Subcelulares/metabolismo , Uridina/genética , Desaminase APOBEC-1/metabolismo , Citidina/química , Genes Reporter , Células HEK293 , Células Hep G2 , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , RNA Mensageiro/metabolismo , Uridina/química
12.
Plant Physiol ; 184(4): 2182-2198, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32972981

RESUMO

Mitochondria and chloroplasts play key roles in plant-pathogen interactions. Cytidine-to-uridine (C-to-U) RNA editing is a critical posttranscriptional modification in mitochondria and chloroplasts that is specific to flowering plants. Multiple organellar RNA-editing factors (MORFs) form a protein family that participates in C-to-U RNA editing, but little is known regarding their immune functions. Here, we report the identification of NbMORF8, a negative regulator of plant immunity to Phytophthora pathogens. Using virus-induced gene silencing and transient expression in Nicotiana benthamiana, we show that NbMORF8 functions through the regulation of reactive oxygen species production, salicylic acid signaling, and accumulation of multiple Arg-X-Leu-Arg effectors of Phytophthora pathogens. NbMORF8 is localized to mitochondria and chloroplasts, and its immune function requires mitochondrial targeting. The conserved MORF box domain is not required for its immune function. Furthermore, we show that the preferentially mitochondrion-localized NbMORF proteins negatively regulate plant resistance against Phytophthora, whereas the preferentially chloroplast-localized ones are positive immune regulators. Our study reveals that the C-to-U RNA-editing factor NbMORF8 negatively regulates plant immunity to the oomycete pathogen Phytophthora and that mitochondrion- and chloroplast-localized NbMORF family members exert opposing effects on immune regulation.


Assuntos
Citidina/genética , Citidina/metabolismo , Interações Hospedeiro-Patógeno/genética , Nicotiana/genética , Phytophthora/patogenicidade , Imunidade Vegetal/genética , Uridina/genética , Uridina/metabolismo , Regulação da Expressão Gênica de Plantas , Genes de Plantas , Interações Hospedeiro-Patógeno/fisiologia , Doenças das Plantas/imunologia , Plantas Geneticamente Modificadas , Edição de RNA , Nicotiana/microbiologia
13.
Biochim Biophys Acta Biomembr ; 1862(7): 183247, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32126230

RESUMO

Humans possess three members of the cation-coupled concentrative nucleoside transporter CNT (SLC 28) family, hCNT1-3: hCNT1 is selective for pyrimidine nucleosides but also transports adenosine, hCNT2 transports purine nucleosides and uridine, and hCNT3 transports both pyrimidine and purine nucleosides. hCNT1/2 transport nucleosides using the transmembrane Na+ electrochemical gradient, while hCNT3 is both Na+- and H+-coupled. By producing recombinant hCNT3 in Xenopus laevis oocytes, we have used radiochemical high performance liquid chromatography (HPLC) analysis to investigate the metabolic fate of transported [3H] or [14C] pyrimidine and purine nucleosides once inside cells. With the exception of adenosine, transported nucleosides were generally subject to minimal intracellular metabolism. We also used radiochemical HPLC analysis to study the mechanism by which adenosine functions as a low Km, low Vmax permeant of hCNT1. hCNT1-producing oocytes were pre-loaded with [3H] uridine, after which efflux of accumulated radioactivity was measured in transport medium alone, or in the presence of extracellular non-radiolabelled adenosine or uridine. hCNT1-mediated [3H]-efflux was stimulated by extracellular uridine, but inhibited by extracellular adenosine, with >95% of the radioactivity exiting cells being unmetabolized uridine, consistent with a low transmembrane mobility of the hCNT1/adenosine complex. Humans also possess four members of the equilibrative nucleoside transporter ENT (SLC 29) family, hENT1-4. Of these, hENT1 and hENT2 transport both nucleosides and nucleobases into and out of cells, but their relative contributions to nucleoside and nucleobase homeostasis and, in particular, to adenosine signaling via purinoreceptors, are not known. We therefore used HPLC to determine plasma nucleoside and nucleobase concentrations in wild-type, mENT1-, mENT2- and mENT1/mENT2-knockout (KO) mice, and to compare the findings with knockout of mCNT3. Results demonstrated that ENT1 was more important than ENT2 or CNT3 in determining plasma adenosine concentrations, indicated modest roles of ENT1 in the homeostasis of other nucleosides, and suggested that none of the transporters is a major participant in handling of nucleobases.


Assuntos
Homeostase , Proteínas de Transporte de Nucleosídeos/genética , Nucleosídeos/química , Adenosina/genética , Sequência de Aminoácidos/genética , Animais , Transporte Biológico , Cromatografia Líquida de Alta Pressão , Humanos , Camundongos , Proteínas de Transporte de Nucleosídeos/química , Oócitos/química , Oócitos/metabolismo , Sódio/química , Uridina/genética , Xenopus laevis/genética
14.
Nucleic Acids Res ; 48(6): 3304-3314, 2020 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-31930330

RESUMO

Three-dimensional structures have been solved for several naturally occurring RNA triple helices, although all are limited to six or fewer consecutive base triples, hindering accurate estimation of global and local structural parameters. We present an X-ray crystal structure of a right-handed, U•A-U-rich RNA triple helix with 11 continuous base triples. Due to helical unwinding, the RNA triple helix spans an average of 12 base triples per turn. The double helix portion of the RNA triple helix is more similar to both the helical and base step structural parameters of A'-RNA rather than A-RNA. Its most striking features are its wide and deep major groove, a smaller inclination angle and all three strands favoring a C3'-endo sugar pucker. Despite the presence of a third strand, the diameter of an RNA triple helix remains nearly identical to those of DNA and RNA double helices. Contrary to our previous modeling predictions, this structure demonstrates that an RNA triple helix is not limited in length to six consecutive base triples and that longer RNA triple helices may exist in nature. Our structure provides a starting point to establish structural parameters of the so-called 'ideal' RNA triple helix, analogous to A-RNA and B-DNA double helices.


Assuntos
DNA/genética , Modelos Moleculares , Conformação Molecular , RNA/ultraestrutura , Adenosina/química , Adenosina/genética , DNA/química , DNA/ultraestrutura , Ligação de Hidrogênio , Conformação de Ácido Nucleico , Conformação Proteica em alfa-Hélice/genética , RNA/genética , Uridina/química , Uridina/genética
15.
Nat Commun ; 10(1): 5542, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31804502

RESUMO

Transfer (t)RNAs contain a wide variety of post-transcriptional modifications, which play critical roles in tRNA stability and functions. 3-(3-amino-3-carboxypropyl)uridine (acp3U) is a highly conserved modification found in variable- and D-loops of tRNAs. Biogenesis and functions of acp3U have not been extensively investigated. Using a reverse-genetic approach supported by comparative genomics, we find here that the Escherichia coli yfiP gene, which we rename tapT (tRNA aminocarboxypropyltransferase), is responsible for acp3U formation in tRNA. Recombinant TapT synthesizes acp3U at position 47 of tRNAs in the presence of S-adenosylmethionine. Biochemical experiments reveal that acp3U47 confers thermal stability on tRNA. Curiously, the ΔtapT strain exhibits genome instability under continuous heat stress. We also find that the human homologs of tapT, DTWD1 and DTWD2, are responsible for acp3U formation at positions 20 and 20a of tRNAs, respectively. Double knockout cells of DTWD1 and DTWD2 exhibit growth retardation, indicating that acp3U is physiologically important in mammals.


Assuntos
Conformação de Ácido Nucleico , RNA Bacteriano/química , RNA de Transferência/química , Uridina/química , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Estrutura Molecular , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo , Uridina/genética , Uridina/metabolismo
16.
Hum Cell ; 32(4): 477-486, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31428943

RESUMO

MicroRNAs (miRNAs) are defined as small, non-coding RNAs that act as post-transcriptional regulators of gene expression. Dysfunction of miRNAs was involved in the initiation and progression of nasopharyngeal carcinoma (NPC). Here, we found that miR-543 was markedly overexpressed in NPC tissues and cell lines. Overexpression of miR-543 promoted the proliferation, cell cycle progression and invasion of NPC cells. Down-regulation of miR-543 inhibited the proliferation and induced apoptosis of NPC cells. Bioinformatics analysis suggested the junctional adhesion molecule A (JAM-A) as a potential target of miR-543. Furthermore, molecular study showed that the miR-543 bound the 3'-untranslated region (UTR) of JAM-A and decreased the expression of JAM-A in NPC cells. The expression of JAM-A in NPC tissues was decreased and negatively correlated with that of miR-543. Overexpression of JAM-A attenuated miR-543-induced proliferation of NPC cells. Collectively, these evidence indicated the important roles of miR-543/JAM-A signaling in the progression of NPC, highlighting the potential of miR-543 as a target in the treatment of NPC.


Assuntos
Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proliferação de Células/genética , MicroRNAs/fisiologia , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Regiões 3' não Traduzidas , Linhagem Celular Tumoral , Expressão Gênica/genética , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Terapia de Alvo Molecular , Carcinoma Nasofaríngeo/terapia , Neoplasias Nasofaríngeas/terapia , Invasividade Neoplásica/genética , Ácidos Fosfatídicos/genética , Transdução de Sinais , Uridina/análogos & derivados , Uridina/genética
17.
Science ; 365(6451): 382-386, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31296651

RESUMO

Programmable RNA editing enables reversible recoding of RNA information for research and disease treatment. Previously, we developed a programmable adenosine-to-inosine (A-to-I) RNA editing approach by fusing catalytically inactivate RNA-targeting CRISPR-Cas13 (dCas13) with the adenine deaminase domain of ADAR2. Here, we report a cytidine-to-uridine (C-to-U) RNA editor, referred to as RNA Editing for Specific C-to-U Exchange (RESCUE), by directly evolving ADAR2 into a cytidine deaminase. RESCUE doubles the number of mutations targetable by RNA editing and enables modulation of phosphosignaling-relevant residues. We apply RESCUE to drive ß-catenin activation and cellular growth. Furthermore, RESCUE retains A-to-I editing activity, enabling multiplexed C-to-U and A-to-I editing through the use of tailored guide RNAs.


Assuntos
Adenosina Desaminase/genética , Citidina/genética , Citosina Desaminase/genética , Engenharia de Proteínas/métodos , Edição de RNA , Proteínas de Ligação a RNA/genética , Uridina/genética , Adenosina/genética , Adenosina Desaminase/química , Citosina Desaminase/química , Células HEK293 , Humanos , Inosina/genética , Domínios Proteicos , Proteínas de Ligação a RNA/química , beta Catenina/química , beta Catenina/genética , beta Catenina/metabolismo
18.
Mol Cell ; 75(3): 511-522.e4, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31178353

RESUMO

Many microRNAs (miRNAs) exist alongside abundant miRNA isoforms (isomiRs), most of which arise from post-maturation sequence modifications such as 3' uridylation. However, the ways in which these sequence modifications affect miRNA function remain poorly understood. Here, using human miR-27a in cell lines as a model, we discovered that a nonfunctional target site unable to base-pair extensively with the miRNA seed sequence can regain function when an upstream adenosine is able to base-pair with a post-transcriptionally added uridine in the miR-27a tail. This tail-U-mediated repression (TUMR) is abolished in cells lacking the uridylation enzymes TUT4 and TUT7, indicating that uridylation alters miRNA function by modulating target recognition. We identified a set of non-canonical targets in human cells that are specifically regulated by uridylated miR-27a. We provide evidence that TUMR expands the targets of other endogenous miRNAs. Our study reveals a function of uridylated isomiRs in regulating non-canonical miRNA targets.


Assuntos
Proteínas de Ligação a DNA/genética , MicroRNAs/genética , RNA Nucleotidiltransferases/genética , Uridina/genética , Adenosina/genética , Pareamento de Bases/genética , Células HeLa , Humanos , Estabilidade de RNA , Uridina/metabolismo
19.
Am J Hum Genet ; 104(6): 1202-1209, 2019 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-31079898

RESUMO

The wobble hypothesis was proposed to explain the presence of fewer tRNAs than possible codons. The wobble nucleoside position in the anticodon stem-loop undergoes a number of modifications that help maintain the efficiency and fidelity of translation. AlkB homolog 8 (ALKBH8) is an atypical member of the highly conserved AlkB family of dioxygenases and is involved in the formation of mcm5s2U, (S)-mchm5U, (R)-mchm5U, mcm5U, and mcm5Um at the anticodon wobble uridines of specific tRNAs. In two multiplex consanguineous families, we identified two homozygous truncating ALKBH8 mutations causing intellectual disability. Analysis of tRNA derived from affected individuals showed the complete absence of these modifications, consistent with the presumptive loss of function of the variants. Our results highlight the sensitivity of the brain to impaired wobble modification and expand the list of intellectual-disability syndromes caused by mutations in genes related to tRNA modification.


Assuntos
Homólogo AlkB 8 da RNAt Metiltransferase/genética , Códon/metabolismo , Genes Recessivos/genética , Deficiência Intelectual/etiologia , Mutação , RNA de Transferência/metabolismo , Uridina/metabolismo , Adolescente , Adulto , Criança , Pré-Escolar , Códon/genética , Feminino , Humanos , Deficiência Intelectual/patologia , Masculino , RNA de Transferência/genética , Uridina/química , Uridina/genética , Adulto Jovem , tRNA Metiltransferases/metabolismo
20.
Nucleic Acids Res ; 47(9): 4814-4830, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-30916349

RESUMO

Posttranscriptional RNA modifications occur in all domains of life. Modifications of anticodon bases are of particular importance for ribosomal decoding and proteome homeostasis. The Elongator complex modifies uridines in the wobble position and is highly conserved in eukaryotes. Despite recent insights into Elongator's architecture, the structure and function of its regulatory factor Kti12 have remained elusive. Here, we present the crystal structure of Kti12's nucleotide hydrolase domain trapped in a transition state of ATP hydrolysis. The structure reveals striking similarities to an O-phosphoseryl-tRNA kinase involved in the selenocysteine pathway. Both proteins employ similar mechanisms of tRNA binding and show tRNASec-dependent ATPase activity. In addition, we demonstrate that Kti12 binds directly to Elongator and that ATP hydrolysis is crucial for Elongator to maintain proper tRNA anticodon modification levels in vivo. In summary, our data reveal a hitherto uncharacterized link between two translational control pathways that regulate selenocysteine incorporation and affect ribosomal tRNA selection via specific tRNA modifications.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Adenosina Trifosfatases/genética , Processamento Pós-Transcricional do RNA/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Adenosina Trifosfatases/química , Anticódon/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Chaetomium/química , Chaetomium/enzimologia , Cristalografia por Raios X , Conformação Proteica , RNA de Transferência/genética , Ribossomos/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Uridina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA