Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 10213, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37353550

ABSTRACT

Novel small RNAs derived from tRNAs are continuously identified, however, their biological functions are rarely reported. Here, we accidentally found the reads peak at 32nt during statistical analysis on the miRNA-seq data of lamb skin tissue, and found that it was related to the wool type of lambs. This 32nt peak was composed of small tRNA fragments. The main component sequence of this peak was a novel small tRNA derived from Glycyl tRNA (tRNAGly), the expression level of tRNAGly-derived tRNA fragments (tRFGly) was 5.77 folds higher in the coarse wool lambs than that in the fine wool lambs. However, in contrast, the expression of tRNAGly in the skin of fine wool lambs is 6.28 folds more than that in coarse wool lambs. tRNAGly promoted the synthesis of high glycine protein including KAP6 in fine wool lamb skin. These proteins were reported as the major genes for fine curly wool. Integrative analysis of target gene prediction, proteomics and metabolomics results revealed that tRFGly reduced the level of reactive oxygen species (ROS) in the skin of coarse wool lambs by targeted inhibition of the Metabolic signal and the corresponding Glutathione metabolic pathway, on the contrary, the level of oxidative stress in the skin of fine wool lambs was significantly higher. This study revealed for the first time the relationship between tRNAGly and its derived tRFGly and animal traits. tRFGly has the function of targeting and regulating protein synthesis. At the same time, tRFGly can reduce the expression of its resource complete tRNA, thereby reducing its ability to transport specific amino acid and affecting the expression of corresponding proteins.


Subject(s)
RNA, Transfer, Gly , Wool , Sheep/genetics , Animals , Wool/metabolism , RNA, Transfer, Gly/metabolism , RNA, Transfer/metabolism , Sheep, Domestic/genetics , Sheep, Domestic/metabolism , Oxidative Stress/genetics
2.
Philos Trans R Soc Lond B Biol Sci ; 378(1871): 20220029, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36633285

ABSTRACT

By linking amino acids to their codon assignments, transfer RNAs (tRNAs) are essential for protein synthesis and translation fidelity. Some human tRNA variants cause amino acid mis-incorporation at a codon or set of codons. We recently found that a naturally occurring tRNASer variant decodes phenylalanine codons with serine and inhibits protein synthesis. Here, we hypothesized that human tRNA variants that misread glycine (Gly) codons with alanine (Ala) will also disrupt protein homeostasis. The A3G mutation occurs naturally in tRNAGly variants (tRNAGlyCCC, tRNAGlyGCC) and creates an alanyl-tRNA synthetase (AlaRS) identity element (G3 : U70). Because AlaRS does not recognize the anticodon, the human tRNAAlaAGC G35C (tRNAAlaACC) variant may function similarly to mis-incorporate Ala at Gly codons. The tRNAGly and tRNAAla variants had no effect on protein synthesis in mammalian cells under normal growth conditions; however, tRNAGlyGCC A3G depressed protein synthesis in the context of proteasome inhibition. Mass spectrometry confirmed Ala mistranslation at multiple Gly codons caused by the tRNAGlyGCC A3G and tRNAAlaAGC G35C mutants, and in some cases, we observed multiple mistranslation events in the same peptide. The data reveal mistranslation of Ala at Gly codons and defects in protein homeostasis generated by natural human tRNA variants that are tolerated under normal conditions. This article is part of the theme issue 'Reactivity and mechanism in chemical and synthetic biology'.


Subject(s)
Alanine-tRNA Ligase , Protein Biosynthesis , Humans , Alanine/genetics , Alanine/chemistry , Alanine/metabolism , Alanine-tRNA Ligase/chemistry , Alanine-tRNA Ligase/genetics , Alanine-tRNA Ligase/metabolism , Codon/genetics , Glycine/genetics , Glycine/metabolism , Proteostasis , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Transfer, Ala/chemistry , RNA, Transfer, Ala/genetics , RNA, Transfer, Ala/metabolism , RNA, Transfer, Gly/metabolism
3.
FEBS Lett ; 596(13): 1615-1627, 2022 07.
Article in English | MEDLINE | ID: mdl-35662005

ABSTRACT

Homochirality of biomacromolecules is a prerequisite for their proper functioning and hence essential for all life forms. This underscores the role of cellular chiral checkpoints in enforcing homochirality during protein biosynthesis. d-Aminoacyl-tRNA deacylase (DTD) is an enzyme that performs 'chirality-based proofreading' to remove d-amino acids mistakenly attached to tRNAs, thus recycling them for further rounds of translation. Paradoxically, owing to its l-chiral rejection mode of action, DTD can remove glycine as well, which is an achiral amino acid. However, this activity is modulated by discriminator base (N73) in tRNA, a unique element that protects the cognate Gly-tRNAGly . Here, we review our recent work showing various aspects of DTD and tRNAGly coevolution and its key role in maintaining proper translation surveillance in both bacteria and eukaryotes. Moreover, we also discuss two major optimization events on DTD and tRNA that resolved compatibility issues among the archaeal and the bacterial translation apparatuses. Importantly, such optimizations are necessary for the emergence of mitochondria and successful eukaryogenesis.


Subject(s)
Protein Biosynthesis , RNA, Transfer, Gly , Amino Acids/metabolism , Glycine/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Gly/metabolism
4.
Nucleic Acids Res ; 50(10): 5834-5849, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35580054

ABSTRACT

T-box riboswitches (T-boxes) are essential RNA regulatory elements with a remarkable structural diversity, especially among bacterial pathogens. In staphylococci, all glyS T-boxes synchronize glycine supply during synthesis of nascent polypeptides and cell wall formation and are characterized by a conserved and unique insertion in their antiterminator/terminator domain, termed stem Sa. Interestingly, in Staphylococcus aureus the stem Sa can accommodate binding of specific antibiotics, which in turn induce robust and diverse effects on T-box-mediated transcription. In the present study, domain swap mutagenesis and probing analysis were performed to decipher the role of stem Sa. Deletion of stem Sa significantly reduces both the S. aureus glyS T-box-mediated transcription readthrough levels and the ability to discriminate among tRNAGly isoacceptors, both in vitro and in vivo. Moreover, the deletion inverted the previously reported stimulatory effects of specific antibiotics. Interestingly, stem Sa insertion in the terminator/antiterminator domain of Geobacillus kaustophilus glyS T-box, which lacks this domain, resulted in elevated transcription in the presence of tigecycline and facilitated discrimination among proteinogenic and nonproteinogenic tRNAGly isoacceptors. Overall, stem Sa represents a lineage-specific structural feature required for efficient staphylococcal glyS T-box-mediated transcription and it could serve as a species-selective druggable target through its ability to modulate antibiotic binding.


Subject(s)
Riboswitch , Anti-Bacterial Agents/pharmacology , RNA , RNA, Transfer, Gly/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
5.
J Nutr Biochem ; 99: 108866, 2022 01.
Article in English | MEDLINE | ID: mdl-34563666

ABSTRACT

Palmitic acid (PA) induces apoptosis in the human trophoblast cell line HTR8/SVneo. However, the molecular mechanism underlying this effect remains unclear. Although small noncoding RNAs are involved in trophoblast growth and invasion during early pregnancy, the functional roles of tRNA-derived species are currently unknown. Therefore, the purpose of this study was to examine the involvement of tRNA-derived species in PA-induced apoptosis in human trophoblasts. In this study, we investigate the expression and function of tRNA-derived stress-induced RNAs (tiRNAs) in HTR8/SVneo. We determined the expression of tiRNAs in HTR8/SVneo cells in response to PA. Then, we transfected inhibitor of target tiRNA in HTR8/SVneo with or without PA to examine the tRNA-derived species-regulated intracellular signal transduction by detecting calcium homeostasis, mitochondrial membrane potential, and signaling proteins. We found that the expression of tRNAGly-derived tiRNAs decreased in PA-treated human trophoblasts. Moreover, inhibition of tiRNAGlyCCC/GCC enhanced the PA-induced apoptosis along with the induction of DNA fragmentation and mitochondrial depolarization. Inhibition of tiRNAGlyCCC/GCC enhanced the expression of endoplasmic reticulum stress-related proteins and increased Ca2+ levels in the cytoplasm and mitochondria. Moreover, the levels of cytochrome c released from the mitochondria were synergistically affected by tiRNAGlyCCC/GCC inhibitor and PA. Furthermore, artificial regulation of ANG inhibited the expression of tiRNAGlyCCC/GCC and similar effects were observed upon the inhibition of tiRNAGlyCCC/GCC in human trophoblasts. These results suggest that tiRNAGlyCCC/GCC might be the molecule via which PA induces its effects in human trophoblasts.


Subject(s)
Apoptosis/drug effects , Palmitic Acid/adverse effects , RNA, Transfer, Gly/metabolism , Trophoblasts/cytology , Calcium/metabolism , DNA Fragmentation/drug effects , Humans , RNA, Transfer, Gly/genetics , Trophoblasts/drug effects , Trophoblasts/metabolism
6.
Cell Rep ; 37(12): 110130, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34936863

ABSTRACT

Bacterial toxin-antitoxin modules contribute to the stress adaptation, persistence, and dormancy of bacteria for survival under environmental stresses and are involved in bacterial pathogenesis. In Salmonella Typhimurium, the Gcn5-related N-acetyltransferase toxin TacT reportedly acetylates the α-amino groups of the aminoacyl moieties of several aminoacyl-tRNAs, inhibits protein synthesis, and promotes persister formation during the infection of macrophages. Here, we show that TacT exclusively acetylates Gly-tRNAGlyin vivo and in vitro. The crystal structure of the TacT:acetyl-Gly-tRNAGly complex and the biochemical analysis reveal that TacT specifically recognizes the discriminator U73 and G71 in tRNAGly, a combination that is only found in tRNAGly isoacceptors, and discriminates tRNAGly from other tRNA species. Thus, TacT is a Gly-tRNAGly-specific acetyltransferase toxin. The molecular basis of the specific aminoacyl-tRNA acetylation by TacT provides advanced information for the design of drugs targeting Salmonella.


Subject(s)
Acetyltransferases/metabolism , Bacterial Toxins/metabolism , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Gly/metabolism , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Acetylation , Acetyltransferases/chemistry , Antitoxins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , DNA, Bacterial , Protein Processing, Post-Translational , Salmonella Infections/microbiology , Salmonella typhimurium/chemistry
7.
Nucleic Acids Res ; 49(22): 13045-13061, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34871455

ABSTRACT

Dnmt2, a member of the DNA methyltransferase superfamily, catalyzes the formation of 5-methylcytosine at position 38 in the anticodon loop of tRNAs. Dnmt2 regulates many cellular biological processes, especially the production of tRNA-derived fragments and intergenerational transmission of paternal metabolic disorders to offspring. Moreover, Dnmt2 is closely related to human cancers. The tRNA substrates of mammalian Dnmt2s are mainly detected using bisulfite sequencing; however, we lack supporting biochemical data concerning their substrate specificity or recognition mechanism. Here, we deciphered the tRNA substrates of human DNMT2 (hDNMT2) as tRNAAsp(GUC), tRNAGly(GCC) and tRNAVal(AAC). Intriguingly, for tRNAAsp(GUC) and tRNAGly(GCC), G34 is the discriminator element; whereas for tRNAVal(AAC), the inosine modification at position 34 (I34), which is formed by the ADAT2/3 complex, is the prerequisite for hDNMT2 recognition. We showed that the C32U33(G/I)34N35 (C/U)36A37C38 motif in the anticodon loop, U11:A24 in the D stem, and the correct size of the variable loop are required for Dnmt2 recognition of substrate tRNAs. Furthermore, mammalian Dnmt2s possess a conserved tRNA recognition mechanism.


Subject(s)
5-Methylcytosine/metabolism , Anticodon/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , RNA, Transfer/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Anticodon/genetics , Base Sequence , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/genetics , HEK293 Cells , HeLa Cells , Humans , Inosine/metabolism , Mice , Models, Molecular , NIH 3T3 Cells , Nucleic Acid Conformation , Protein Binding , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer, Asp/chemistry , RNA, Transfer, Asp/genetics , RNA, Transfer, Asp/metabolism , RNA, Transfer, Gly/chemistry , RNA, Transfer, Gly/genetics , RNA, Transfer, Gly/metabolism , RNA, Transfer, Val/chemistry , RNA, Transfer, Val/genetics , RNA, Transfer, Val/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Substrate Specificity
8.
Science ; 373(6559): 1161-1166, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34516840

ABSTRACT

Heterozygous mutations in six transfer RNA (tRNA) synthetase genes cause Charcot-Marie-Tooth (CMT) peripheral neuropathy. CMT mutant tRNA synthetases inhibit protein synthesis by an unknown mechanism. We found that CMT mutant glycyl-tRNA synthetases bound tRNAGly but failed to release it, resulting in tRNAGly sequestration. This sequestration potentially depleted the cellular tRNAGly pool, leading to insufficient glycyl-tRNAGly supply to the ribosome. Accordingly, we found ribosome stalling at glycine codons and activation of the integrated stress response (ISR) in affected motor neurons. Moreover, transgenic overexpression of tRNAGly rescued protein synthesis, peripheral neuropathy, and ISR activation in Drosophila and mouse CMT disease type 2D (CMT2D) models. Conversely, inactivation of the ribosome rescue factor GTPBP2 exacerbated peripheral neuropathy. Our findings suggest a molecular mechanism for CMT2D, and elevating tRNAGly levels may thus have therapeutic potential.


Subject(s)
Charcot-Marie-Tooth Disease/metabolism , Glycine-tRNA Ligase/metabolism , RNA, Transfer, Gly/metabolism , Animals , Charcot-Marie-Tooth Disease/genetics , Disease Models, Animal , Drosophila melanogaster , Female , Glycine-tRNA Ligase/genetics , Humans , Male , Mice , Mice, Transgenic , Motor Neurons/physiology , RNA, Transfer, Gly/genetics
9.
J Exp Clin Cancer Res ; 40(1): 222, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34225773

ABSTRACT

BACKGROUND: tRNA-derived small noncoding RNAs (sncRNAs) are mainly categorized into tRNA halves (tiRNAs) and fragments (tRFs). Biological functions of tiRNAs in human solid tumor are attracting more and more attention, but researches concerning the mechanisms in tiRNAs-mediated tumorigenesis are rarely. The direct regulatory relationship between tiRNAs and splicing-related proteins remain elusive. METHODS: Papillary thyroid carcinoma (PTC) associated tRNA fragments were screened by tRNA fragments deep sequencing and validated by qRT-PCR and Northern Blot in PTC tissues. The biological function of tRNA fragments were assessed by cell counting kit, transwells and subcutaneous transplantation tumor of nude mice. For mechanistic study, tRNA fragments pull-down, RNA immunoprecipitation, Western Blot, Immunofluorescence, Immunohistochemical staining were performed. RESULTS: Herein, we have identified a 33 nt tiRNA-Gly significantly increases in papillary thyroid cancer (PTC) based on tRFs & tiRNAs sequencing. The ectopic expression of tiRNA-Gly promotes cell proliferation and migration, whereas down-regulation of tiRNA-Gly exhibits reverse effects. Mechanistic investigations reveal tiRNA-Gly directly bind the UHM domain of a splicing-related RNA-binding protein RBM17. The interaction with tiRNA-Gly could translocate RBM17 from cytoplasm into nucleus. In addition, tiRNA-Gly increases RBM17 protein expression via inhibiting its degradation in a ubiquitin/proteasome-dependent way. Moreover, RBM17 level in tiRNA-Gly high-expressing human PTC tissues is upregulated. In vivo mouse model shows that suppression of tiRNA-Gly decreases RBM17 expression. Importantly, tiRNA-Gly can induce exon 16 splicing of MAP4K4 mRNA leading to phosphorylation of downstream signaling pathway, which is RBM17 dependent. CONCLUSIONS: Our study firstly illustrates tiRNA-Gly can directly bind to RBM17 and display oncogenic effect via RBM17-mediated alternative splicing. This fully novel model broadens our understanding of molecular mechanism in which tRNA fragment in tumor cells directly bind RNA binding protein and play a role in alternative splicing.


Subject(s)
RNA Splicing Factors/metabolism , RNA, Transfer, Gly/metabolism , RNA, Transfer/metabolism , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/metabolism , Alternative Splicing , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Female , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Splicing Factors/genetics , RNA, Transfer/genetics , RNA, Transfer, Gly/genetics , Signal Transduction , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
10.
Nucleic Acids Res ; 49(2): 684-699, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33367813

ABSTRACT

The sequence of tRNAs is submitted to evolutionary constraints imposed by their multiple interactions with aminoacyl-tRNA synthetases, translation elongation factor Tu in complex with GTP (EF-Tu•GTP), and the ribosome, each being essential for accurate and effective decoding of messenger RNAs. In Staphylococcus aureus, an additional constraint is imposed by the participation of tRNAGly isoacceptors in the addition of a pentaglycine side chain to cell-wall peptidoglycan precursors by transferases FmhB, FemA and FemB. Three tRNAGly isoacceptors poorly interacting with EF-Tu•GTP and the ribosome were previously identified. Here, we show that these 'non-proteogenic' tRNAs are preferentially recognized by FmhB based on kinetic analyses and on synthesis of stable aminoacyl-tRNA analogues acting as inhibitors. Synthesis of chimeric tRNAs and of helices mimicking the tRNA acceptor arms revealed that this discrimination involves identity determinants exclusively present in the D and T stems and loops of non-proteogenic tRNAs, which belong to an evolutionary lineage only present in the staphylococci. EF-Tu•GTP competitively inhibited FmhB by sequestration of 'proteogenic' aminoacyl-tRNAs in vitro. Together, these results indicate that competition for the Gly-tRNAGly pool is restricted by both limited recognition of non-proteogenic tRNAs by EF-Tu•GTP and limited recognition of proteogenic tRNAs by FmhB.


Subject(s)
Peptidoglycan/biosynthesis , RNA, Bacterial/metabolism , RNA, Transfer, Gly/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/metabolism , Base Sequence , Binding, Competitive , Cell Wall/metabolism , Guanosine Triphosphate/metabolism , Models, Molecular , Nucleic Acid Conformation , Peptide Elongation Factor Tu/metabolism , Protein Binding
11.
Nat Commun ; 9(1): 4865, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451861

ABSTRACT

The precise interplay between the mRNA codon and the tRNA anticodon is crucial for ensuring efficient and accurate translation by the ribosome. The insertion of RNA nucleobase derivatives in the mRNA allowed us to modulate the stability of the codon-anticodon interaction in the decoding site of bacterial and eukaryotic ribosomes, allowing an in-depth analysis of codon recognition. We found the hydrogen bond between the N1 of purines and the N3 of pyrimidines to be sufficient for decoding of the first two codon nucleotides, whereas adequate stacking between the RNA bases is critical at the wobble position. Inosine, found in eukaryotic mRNAs, is an important example of destabilization of the codon-anticodon interaction. Whereas single inosines are efficiently translated, multiple inosines, e.g., in the serotonin receptor 5-HT2C mRNA, inhibit translation. Thus, our results indicate that despite the robustness of the decoding process, its tolerance toward the weakening of codon-anticodon interactions is limited.


Subject(s)
2-Aminopurine/analogs & derivatives , Anticodon/chemistry , Codon/chemistry , Inosine/metabolism , Protein Biosynthesis , Receptor, Serotonin, 5-HT2C/genetics , 2-Aminopurine/chemistry , 2-Aminopurine/metabolism , Anticodon/metabolism , Bacteriophage T7/genetics , Bacteriophage T7/metabolism , Base Sequence , Codon/metabolism , Cytidine/analogs & derivatives , Cytidine/genetics , Cytidine/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , HEK293 Cells , Humans , Hydrogen Bonding , Inosine/genetics , Pyridones/chemistry , Pyridones/metabolism , RNA, Transfer, Gly/genetics , RNA, Transfer, Gly/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Elife ; 72018 08 09.
Article in English | MEDLINE | ID: mdl-30091703

ABSTRACT

D-aminoacyl-tRNA deacylase (DTD) acts on achiral glycine, in addition to D-amino acids, attached to tRNA. We have recently shown that this activity enables DTD to clear non-cognate Gly-tRNAAla with 1000-fold higher efficiency than its activity on Gly-tRNAGly, indicating tRNA-based modulation of DTD (Pawar et al., 2017). Here, we show that tRNA's discriminator base predominantly accounts for this activity difference and is the key to selection by DTD. Accordingly, the uracil discriminator base, serving as a negative determinant, prevents Gly-tRNAGly misediting by DTD and this protection is augmented by EF-Tu. Intriguingly, eukaryotic DTD has inverted discriminator base specificity and uses only G3•U70 for tRNAGly/Ala discrimination. Moreover, DTD prevents alanine-to-glycine misincorporation in proteins rather than only recycling mischarged tRNAAla. Overall, the study reveals the unique co-evolution of DTD and discriminator base, and suggests DTD's strong selection pressure on bacterial tRNAGlys to retain a pyrimidine discriminator code.


Subject(s)
Aminoacyltransferases/metabolism , Escherichia coli/metabolism , Glycine/metabolism , Protein Biosynthesis , RNA, Transfer, Ala/metabolism , RNA, Transfer, Gly/metabolism , Animals , Escherichia coli/enzymology , Mice
13.
Nucleic Acids Res ; 46(17): 9081-9093, 2018 09 28.
Article in English | MEDLINE | ID: mdl-29893896

ABSTRACT

We have previously shown that 5' halves from tRNAGlyGCC and tRNAGluCUC are the most enriched small RNAs in the extracellular space of human cell lines, and especially in the non-vesicular fraction. Extracellular RNAs are believed to require protection by either encapsulation in vesicles or ribonucleoprotein complex formation. However, deproteinization of non-vesicular tRNA halves does not affect their retention in size-exclusion chromatography. Thus, we considered alternative explanations for their extracellular stability. In-silico analysis of the sequence of these tRNA-derived fragments showed that tRNAGly 5' halves can form homodimers or heterodimers with tRNAGlu 5' halves. This capacity is virtually unique to glycine tRNAs. By analyzing synthetic oligonucleotides by size exclusion chromatography, we provide evidence that dimerization is possible in vitro. tRNA halves with single point substitutions preventing dimerization are degraded faster both in controlled nuclease digestion assays and after transfection in cells, showing that dimerization can stabilize tRNA halves against the action of cellular nucleases. Finally, we give evidence supporting dimerization of endogenous tRNAGlyGCC 5' halves inside cells. Considering recent reports have shown that 5' tRNA halves from Ala and Cys can form tetramers, our results highlight RNA intermolecular structures as a new layer of complexity in the biology of tRNA-derived fragments.


Subject(s)
Dimerization , RNA Stability , RNA, Transfer, Glu/metabolism , RNA, Transfer, Gly/metabolism , Ribonucleases/metabolism , 5' Flanking Region , Base Sequence , Glutamic Acid/metabolism , Glycine/metabolism , Humans , MCF-7 Cells , Nucleic Acid Conformation , RNA, Transfer, Glu/chemistry , RNA, Transfer, Gly/chemistry
14.
Nucleic Acids Res ; 45(17): 10242-10258, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28973457

ABSTRACT

Recently, it was discovered that exposure to mainstream antibiotics activate numerous bacterial riboregulators that control antibiotic resistance genes including metabolite-binding riboswitches and other transcription attenuators. However, the effects of commonly used antibiotics, many of which exhibit RNA-binding properties, on the widespread T-box riboswitches, remain unknown. In Staphylococcus aureus, a species-specific glyS T-box controls the supply of glycine for both ribosomal translation and cell wall synthesis, making it a promising target for next-generation antimicrobials. Here, we report that specific protein synthesis inhibitors could either significantly increase T-box-mediated transcription antitermination, while other compounds could suppress it, both in vitro and in vivo. In-line probing of the full-length T-box combined with molecular modelling and docking analyses suggest that the antibiotics that promote transcription antitermination stabilize the T-box:tRNA complex through binding specific positions on stem I and the Staphylococcal-specific stem Sa. By contrast, the antibiotics that attenuate T-box transcription bind to other positions on stem I and do not interact with stem Sa. Taken together, our results reveal that the transcription of essential genes controlled by T-box riboswitches can be directly modulated by commonly used protein synthesis inhibitors. These findings accentuate the regulatory complexities of bacterial response to antimicrobials that involve multiple riboregulators.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Bacterial/genetics , RNA, Messenger/genetics , RNA, Transfer/genetics , Riboswitch/drug effects , Transcription, Genetic/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Dose-Response Relationship, Drug , Glycine/metabolism , Glycine-tRNA Ligase/biosynthesis , Glycine-tRNA Ligase/genetics , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/metabolism , Models, Molecular , Molecular Docking Simulation , Nucleic Acid Conformation/drug effects , Phylogeny , Protein Binding , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , RNA, Transfer/metabolism , RNA, Transfer, Gly/metabolism , Recombinant Fusion Proteins/metabolism , T-Box Domain Proteins/metabolism
15.
Oncogene ; 36(47): 6640-6648, 2017 11 23.
Article in English | MEDLINE | ID: mdl-28783176

ABSTRACT

Two proteins comprising the ZEB family of zinc finger transcription factors, ZEB1 and ZEB2, execute EMT programs in embryonic development and cancer. By studying regulation of their expression, we describe a novel mechanism that limits ZEB2 protein synthesis. A protein motif located at the border of the SMAD-binding domain of ZEB2 protein induces ribosomal pausing and compromises protein synthesis. The function of this protein motif is dependent on stretches of rare codons, Leu(UUA)-Gly(GGU)-Val(GUA). Incorporation of these triplets in the homologous region of ZEB1 does not affect protein translation. Our data suggest that rare codons have a regulatory role only if they are present within appropriate protein structures. We speculate that pools of transfer RNA available for protein translation impact on the configuration of epithelial mesenchymal transition pathways in tumor cells.


Subject(s)
Codon/genetics , Neoplasms/metabolism , Protein Biosynthesis/genetics , RNA, Transfer, Gly/metabolism , RNA, Transfer, Leu/metabolism , RNA, Transfer, Val/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , Amino Acid Motifs/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Glycine/genetics , Humans , Leucine/genetics , Signal Transduction , Valine/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
16.
Nucleic Acids Res ; 45(13): 8079-8090, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28531275

ABSTRACT

A T-box regulator or riboswitch actively monitors the levels of charged/uncharged tRNA and participates in amino acid homeostasis by regulating genes involved in their utilization or biosynthesis. It has an aptamer domain for cognate tRNA recognition and an expression platform to sense the charge state and modulate gene expression. These two conserved domains are connected by a variable linker that harbors additional secondary structural elements, such as Stem III. The structural basis for specific tRNA binding is known, but the structural basis for charge sensing and the role of other elements remains elusive. To gain new structural insights on the T-box mechanism, a molecular envelope was calculated from small angle X-ray scattering data for the Bacillus subtilis glyQS T-box riboswitch in complex with an uncharged tRNAGly. A structural model of an anti-terminated glyQS T-box in complex with its cognate tRNAGly was derived based on the molecular envelope. It shows the location and relative orientation of various secondary structural elements. The model was validated by comparing the envelopes of the wild-type complex and two variants. The structural model suggests that in addition to a possible regulatory role, Stem III could aid in preferential stabilization of the T-box anti-terminated state allowing read-through of regulated genes.


Subject(s)
RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Transfer, Gly/chemistry , RNA, Transfer, Gly/metabolism , Riboswitch/genetics , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Base Sequence , Models, Molecular , Nucleic Acid Conformation , RNA, Bacterial/genetics , RNA, Transfer, Gly/genetics , Scattering, Small Angle , X-Ray Diffraction
17.
J Mol Biol ; 428(18): 3603-14, 2016 09 11.
Article in English | MEDLINE | ID: mdl-27261259

ABSTRACT

Aminoacyl-tRNA synthetases are essential components of the protein translational machinery in all living species, among which the human glycyl-tRNA synthetase (hGlyRS) is of great research interest because of its unique species-specific aminoacylation properties and noncanonical roles in the Charcot-Marie-Tooth neurological disease. However, the molecular mechanisms of how the enzyme carries out its classical and alternative functions are not well understood. Here, we report a complex structure of the wild-type hGlyRS bound with tRNA(Gly) at 2.95Å. In the complex, the flexible Whep-TRS domain is visible in one of the subunits of the enzyme dimer, and the tRNA molecule is also completely resolved. At the active site, a glycyl-AMP molecule is synthesized and is waiting for the transfer of the glycyl moiety to occur. This cocrystal structure provides us with new details about the recognition mechanism in the intermediate stage during glycylation, which was not well elucidated in the previous crystal structures where the inhibitor AMPPNP was used for crystallization. More importantly, the structural and biochemical work conducted in the current and previous studies allows us to build a model of the full-length hGlyRS in complex with tRNA(Gly), which greatly helps us to understand the roles that insertions and the Whep-TRS domain play in the tRNA-binding process. Finally, through structure comparison with other class II aminoacyl-tRNA synthetases bound with their tRNA substrates, we found some commonalities of the aminoacylation mechanism between these enzymes.


Subject(s)
Glycine-tRNA Ligase/chemistry , Glycine-tRNA Ligase/metabolism , RNA, Transfer, Gly/chemistry , RNA, Transfer, Gly/metabolism , Crystallography, X-Ray , Humans , Models, Biological , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Conformation
18.
PLoS Biol ; 14(5): e1002465, 2016 05.
Article in English | MEDLINE | ID: mdl-27224426

ABSTRACT

D-aminoacyl-tRNA deacylase (DTD) removes D-amino acids mischarged on tRNAs and is thus implicated in enforcing homochirality in proteins. Previously, we proposed that selective capture of D-aminoacyl-tRNA by DTD's invariant, cross-subunit Gly-cisPro motif forms the mechanistic basis for its enantioselectivity. We now show, using nuclear magnetic resonance (NMR) spectroscopy-based binding studies followed by biochemical assays with both bacterial and eukaryotic systems, that DTD effectively misedits Gly-tRNAGly. High-resolution crystal structure reveals that the architecture of DTD's chiral proofreading site is completely porous to achiral glycine. Hence, L-chiral rejection is the only design principle on which DTD functions, unlike other chiral-specific enzymes such as D-amino acid oxidases, which are specific for D-enantiomers. Competition assays with elongation factor thermo unstable (EF-Tu) and DTD demonstrate that EF-Tu precludes Gly-tRNAGly misediting at normal cellular concentrations. However, even slightly higher DTD levels overcome this protection conferred by EF-Tu, thus resulting in significant depletion of Gly-tRNAGly. Our in vitro observations are substantiated by cell-based studies in Escherichia coli that show that overexpression of DTD causes cellular toxicity, which is largely rescued upon glycine supplementation. Furthermore, we provide direct evidence that DTD is an RNA-based catalyst, since it uses only the terminal 2'-OH of tRNA for catalysis without the involvement of protein side chains. The study therefore provides a unique paradigm of enzyme action for substrate selection/specificity by DTD, and thus explains the underlying cause of DTD's activity on Gly-tRNAGly. It also gives a molecular and functional basis for the necessity and the observed tight regulation of DTD levels, thereby preventing cellular toxicity due to misediting.


Subject(s)
Aminoacyltransferases/chemistry , Aminoacyltransferases/metabolism , Peptide Elongation Factor Tu/metabolism , Alanine/chemistry , Alanine/metabolism , Aminoacyltransferases/genetics , Catalytic Domain , Crystallography, X-Ray , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Escherichia coli/cytology , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Glycine/chemistry , Glycine/metabolism , Hydrolysis , Magnetic Resonance Spectroscopy , Peptide Elongation Factor Tu/genetics , Plasmodium falciparum/enzymology , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Gly/chemistry , RNA, Transfer, Gly/metabolism , Ribosomes/metabolism , Substrate Specificity , Zebrafish Proteins/metabolism
19.
Sci Rep ; 6: 20850, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26865164

ABSTRACT

Ischemic injuries will lead to necrotic tissue damage, and post-ischemia angiogenesis plays critical roles in blood flow restoration and tissue recovery. Recently, several types of small RNAs have been reported to be involved in this process. In this study, we first generated a rat brain ischemic model to investigate the involvement of new types of small RNAs in ischemia. We utilized deep sequencing and bioinformatics analyses to demonstrate that the level of small RNA fragments derived from tRNAs strikingly increased in the ischemic rat brain. Among these sequences, tRNA(Val)- and tRNA(Gly)-derived small RNAs account for the most abundant segments. The up-regulation of tRNA(Val)- and tRNA(Gly)-derived fragments was verified through northern blot and quantitative PCR analyses. The levels of these two fragments also increased in a mouse hindlimb ischemia model and cellular hypoxia model. Importantly, up-regulation of the tRNA(Val)- and tRNA(Gly)-derived fragments in endothelial cells inhibited cell proliferation, migration and tube formation. Furthermore, we showed that these small RNAs are generated by angiogenin cleavage. Our results indicate that tRNA-derived fragments are involved in tissue ischemia, and we demonstrate for the first time that tRNA(Val)- and tRNA(Gly)-derived fragments inhibit angiogenesis by modulating the function of endothelial cells.


Subject(s)
Brain Ischemia/genetics , Neovascularization, Physiologic/genetics , RNA, Small Untranslated/genetics , RNA, Transfer, Gly/genetics , RNA, Transfer, Val/genetics , Animals , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Hypoxia , Cell Movement , Cell Proliferation , Computational Biology , Gene Expression Regulation , Hindlimb/blood supply , Hindlimb/pathology , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Proteolysis , RNA Cleavage , RNA, Small Untranslated/metabolism , RNA, Transfer, Gly/metabolism , RNA, Transfer, Val/metabolism , Rats , Rats, Sprague-Dawley , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Signal Transduction
20.
J Biol Chem ; 291(11): 5740-5752, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26797133

ABSTRACT

Glycyl-tRNA synthetase (GlyRS) is the enzyme that covalently links glycine to cognate tRNA for translation. It is of great research interest because of its nonconserved quaternary structures, unique species-specific aminoacylation properties, and noncanonical functions in neurological diseases, but none of these is fully understood. We report two crystal structures of human GlyRS variants, in the free form and in complex with tRNA(Gly) respectively, and reveal new aspects of the glycylation mechanism. We discover that insertion 3 differs considerably in conformation in catalysis and that it acts like a "switch" and fully opens to allow tRNA to bind in a cross-subunit fashion. The flexibility of the protein is supported by molecular dynamics simulation, as well as enzymatic activity assays. The biophysical and biochemical studies suggest that human GlyRS may utilize its flexibility for both the traditional function (regulate tRNA binding) and alternative functions (roles in diseases).


Subject(s)
Glycine-tRNA Ligase/chemistry , Glycine-tRNA Ligase/metabolism , RNA, Transfer, Gly/metabolism , Amino Acid Sequence , Aminoacylation , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Crystallography, X-Ray , Glycine-tRNA Ligase/genetics , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Mutation , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...