69 research outputs found

    Modular pathways for editing non-cognate amino acids by human cytoplasmic leucyl-tRNA synthetase

    Get PDF
    To prevent potential errors in protein synthesis, some aminoacyl-transfer RNA (tRNA) synthetases have evolved editing mechanisms to hydrolyze misactivated amino acids (pre-transfer editing) or misacylated tRNAs (post-transfer editing). Class Ia leucyl-tRNA synthetase (LeuRS) may misactivate various natural and non-protein amino acids and then mischarge tRNALeu. It is known that the fidelity of prokaryotic LeuRS depends on multiple editing pathways to clear the incorrect intermediates and products in the every step of aminoacylation reaction. Here, we obtained human cytoplasmic LeuRS (hcLeuRS) and tRNALeu (hctRNALeu) with high activity from Escherichia coli overproducing strains to study the synthetic and editing properties of the enzyme. We revealed that hcLeuRS could adjust its editing strategy against different non-cognate amino acids. HcLeuRS edits norvaline predominantly by post-transfer editing; however, it uses mainly pre-transfer editing to edit ฮฑ-amino butyrate, although both amino acids can be charged to tRNALeu. Post-transfer editing as a final checkpoint of the reaction was very important to prevent mis-incorporation in vitro. These results provide insight into the modular editing pathways created to prevent genetic code ambiguity by evolution

    Multiple Quality Control Pathways Limit Non-Protein Amino Acid Use by Yeast Cytoplasmic Phenylalanyl-tRNA Synthetase

    Get PDF
    Non-protein amino acids, particularly isomers of the proteinogenic amino acids, present a threat to proteome integrity if they are mistakenly inserted into proteins. Quality control during aminoacyl-tRNA synthesis reduces non-protein amino acid incorporation by both substrate discrimination and proofreading. For example phenylalanyl-tRNA synthetase (PheRS) proofreads the non-protein hydroxylated phenylalanine derivative m-Tyr after its attachment to tRNAPhe. We now show in Saccharomyces cerevisiae that PheRS misacylation of tRNAPhe with the more abundant Phe oxidation product o-Tyr is limited by kinetic discrimination against o-Tyr-AMP in the transfer step followed by o-Tyr-AMP release from the synthetic active site. This selective rejection of a non-protein aminoacyl-adenylate is in addition to known kinetic discrimination against certain non-cognates in the activation step as well as catalytic hydrolysis of mispaired aminoacyl-tRNAPhe species. We also report an unexpected resistance to cytotoxicity by a S. cerevisiae mutant with ablated post-transfer editing activity when supplemented with o-Tyr, cognate Phe, or Ala, the latter of which is not a substrate for activation by this enzyme. Our phenotypic, metabolomic, and kinetic analyses indicate at least three modes of discrimination against non-protein amino acids by S. cerevisiae PheRS and support a non-canonical role for SccytoPheRS post-transfer editing in response to amino acid stress

    Nucleic Acids Res

    Get PDF
    Yeast mitochondria contain a minimalist threonyl-tRNA synthetase (ThrRS) composed only of the catalytic core and tRNA binding domain but lacking the entire editing domain. Besides the usual tRNAThr2, some budding yeasts, such as Saccharomyces cerevisiae, also contain a non-canonical tRNAThr1 with an enlarged 8-nucleotide anticodon loop, reprograming the usual leucine CUN codons to threonine. This raises interesting questions about the aminoacylation fidelity of such ThrRSs and the possible contribution of the two tRNAThrs during editing. Here, we found that, despite the absence of the editing domain, S. cerevisiae mitochondrial ThrRS (ScmtThrRS) harbors a tRNA-dependent pre-transfer editing activity. Remarkably, only the usual tRNAThr2 stimulated pre-transfer editing, thus, establishing the first example of a synthetase exhibiting tRNA-isoacceptor specificity during pre-transfer editing. We also showed that the failure of tRNAThr1 to stimulate tRNA-dependent pre-transfer editing was due to the lack of an editing domain. Using assays of the complementation of a ScmtThrRS gene knockout strain, we showed that the catalytic core and tRNA binding domain of ScmtThrRS co-evolved to recognize the unusual tRNAThr1. In combination, the results provide insights into the tRNA-dependent editing process and suggest that tRNA-dependent pre-transfer editing takes place in the aminoacylation catalytic core

    Crucial role of the Cterminal domain of Mycobacterium tuberculosis leucyl-tRNA synthetase in aminoacylation and editing

    Get PDF
    ABSTRACT The C-terminal extension of prokaryotic leucy

    Aminoacyl-tRNA Synthetases

    Get PDF
    The aminoacyl-tRNA synthetases are an essential and universally distributed family of enzymes that plays a critical role in protein synthesis, pairing tRNAs with their cognate amino acids for decoding mRNAs according to the genetic code. Synthetases help to ensure accurate translation of the genetic code by using both highly accurate cognate substrate recognition and stringent proofreading of noncognate products. While alterations in the quality control mechanisms of synthetases are generally detrimental to cellular viability, recent studies suggest that in some instances such changes facilitate adaption to stress conditions. Beyond their central role in translation, synthetases are also emerging as key players in an increasing number of other cellular processes, with far-reaching consequences in health and disease. The biochemical versatility of the synthetases has also proven pivotal in efforts to expand the genetic code, further emphasizing the wide-ranging roles of the aminoacyl-tRNA synthetase family in synthetic and natural biology

    Nucleic Acids Res

    Get PDF
    Aminoacyl-tRNA synthetases (aaRSs) are remarkable enzymes that are in charge of the accurate recognition and ligation of amino acids and tRNA molecules. The greatest difficulty in accurate aminoacylation appears to be in discriminating between highly similar amino acids. To reduce mischarging of tRNAs by non-cognate amino acids, aaRSs have evolved an editing activity in a second active site to cleave the incorrect aminoacyl-tRNAs. Editing occurs after translocation of the aminoacyl-CCA(76) end to the editing site, switching between a hairpin and a helical conformation for aminoacylation and editing. Here, we studied the consequence of nucleotide changes in the CCA(76) accepting end of tRNA(Leu) during the aminoacylation and editing reactions. The analysis showed that the terminal A(76) is essential for both reactions, suggesting that critical interactions occur in the two catalytic sites. Substitutions of C(74) and C(75) selectively decreased aminoacylation keeping nearly unaffected editing. These mutations might favor the regular helical conformation required to reach the editing site. Mutating the editing domain residues that contribute to CCA(76) binding reduced the aminoacylation fidelity leading to cell-toxicity in the presence of non-cognate amino acids. Collectively, the data show how protein synthesis quality is controlled by the CCA(76) homogeneity of tRNAs

    A genomic glimpse of aminoacyl-tRNA synthetases in malaria parasite Plasmodium falciparum

    Get PDF
    <p>Abstract</p> <p>Background</p> <p><it>Plasmodium </it>parasites are causative agents of malaria which affects >500 million people and claims ~2 million lives annually. The completion of <it>Plasmodium </it>genome sequencing and availability of PlasmoDB database has provided a platform for systematic study of parasite genome. Aminoacyl-tRNA synthetases (<it>aaRS</it>s) are pivotal enzymes for protein translation and other vital cellular processes. We report an extensive analysis of the <it>Plasmodium falciparum </it>genome to identify and classify <it>aaRSs </it>in this organism.</p> <p>Results</p> <p>Using various computational and bioinformatics tools, we have identified 37 <it>aaRS</it>s in <it>P. falciparum</it>. Our key observations are: (i) fraction of proteome dedicated to <it>aaRS</it>s in <it>P. falciparum </it>is very high compared to many other organisms; (ii) 23 out of 37 <it>Pf-aaRS </it>sequences contain signal peptides possibly directing them to different cellular organelles; (iii) expression profiles of <it>Pf-aaRSs </it>vary considerably at various life cycle stages of the parasite; (iv) several <it>PfaaRSs </it>posses very unusual domain architectures; (v) phylogenetic analyses reveal evolutionary relatedness of several parasite <it>aaRS</it>s to bacterial and plants <it>aaRSs</it>; (vi) three dimensional structural modelling has provided insights which could be exploited in inhibitor discovery against parasite <it>aaRSs</it>.</p> <p>Conclusion</p> <p>We have identified 37 <it>Pf-aaRSs </it>based on our bioinformatics analysis. Our data reveal several unique attributes in this protein family. We have annotated all 37 <it>Pf-aaRSs </it>based on predicted localization, phylogenetics, domain architectures and their overall protein expression profiles. The sets of distinct features elaborated in this work will provide a platform for experimental dissection of this family of enzymes, possibly for the discovery of novel drugs against malaria.</p

    Mechanisms of leucyl-tRNA synthetase dependent group I intron splicing

    Get PDF
    Leucyl-tRNA synthetase (LeuRS) plays dual roles within the yeast mitochondria. In addition to protein synthesis, it is also essential to RNA splicing of critical respiratory genes. The LeuRS collaborates with a maturase to excise the bI4 and aI4ฮฑ introns from the cob and cox1ฮฑ genes respectively. The LeuRS-based suppressor mutations have been isolated within the amino acid editing CP1 domain and restore native RNA splicing activity in the presence of an inactive maturase. Mutational analysis of these sites and the regions that surround them demonstrated that certain substitutions can also inactivate LeuRS-dependent splicing activity under in vivo and in vitro conditions. Binding measurements suggest that these suppressor sites are important in maintaining interaction between LeuRS and the group I intron RNA. Thus, CP1 domain binds specifically to the bI4 and aI4ฮฑ intron to promote RNA splicing. In addition to LeuRS from yeast mitochondria (ymLeuRS), diverse LeuRSs from varied origins such as M. tuberculosis and human mitochondria complement the ymLeuRS activities. Similarly, wild-type E. coli LeuRS (EcLeuRS) complemented a ymLeuRS null strain. Interestingly, at reduced levels of EcLeuRS expression in yeast mitochondria, the heterologous synthetase supported protein synthesis, but not intron splicing. Thus, it is a weak splicing suppressor. Surprisingly, a gain of splicing activity was exhibited by positive charge substitutions at the Ala293 position, suggesting that this Ala293 can be adapted for alternative activities. Preliminary footprinting data suggest that LeuRS binds to the P4-P6 core region of the bI4 intron that is cognate to LeuRS. The RNA duplex mimics of the P6 helix were designed and it was shown that LeuRS promotes their annealing in an ATP-independent manner. Domain analysis of LeuRS shows that the C-terminal domain is critical to the RNA annealing activity. Yeast mitochondrial tRNALeu (ymtRNALeu) competitively inhibit annealing. Also, an ymtRNALeu variable-stem-like region was identified on the P6 stem that is important for LeuRS-dependent annealing. These data support that the annealing and tRNA variable arm binding sites overlap on the C-terminal domain of LeuRS. It was shown that the overhang location and length of the duplexes are important features that LeuRS recognizes. It was hypothesized that LeuRS plays a key role in remodeling specific group I intron ribozymes so that they can productively self-splice

    Editing by leucyl-trna synthetase: Discrimination of norvaline and isoleucine

    Get PDF
    Aminoacyl tRNA-synthetases (AARS) are housekeeping enzymes that are tasked with accurate synthesis of aminoacylated tRNA for protein synthesis and other cellular functions. The specificity of amino acid attachment challenges the AARSs that need to distinguish between structurally similar amino acids. In such cases, AARSs have developed editing mechanisms to circumvent the issue of misaminoacylation. Leucyl-tRNA synthetase (LeuRS), for instance selectively edits misactivated and mischarged non-leucine amino acids via pre-transfer editing of misactivated adenylates in the synthetic site or by hydrolyzing mischarged amino acids in the CP1 editing domain. The enzymeโ€™s dependence between the two editing mechanisms can shift based on the origin from which the AARS is derived, the amino acid that is targeted for editing, or presence of a mutation in the enzyme. In the absence of the CP1 domain, E. coli LeuRS (LeuRS-ฮ”CP1) maintains fidelity by clearing non-leucine aminoacyl-adenylates in the enzymeโ€™s synthetic site. The intact tRNA 3โ€™-terminal adenosine (A76) residue is a prerequisite for aminoacylation. Leveraging A76 essentiality tRNA analogues were designed to investigate amino acid dependent specificity of editing by LeuRS. The tRNA analogues were synthesized by addition of a modified adenosine triphosphate to an in vitro transcribed E. coli tRNALeuUAA using the CCA-adding enzyme from E. coli. Incorporation of unchargeable tRNA analogues stimulated ATP hydrolysis by wild type LeuRS in the presence of norvaline. In contrast, pre-transfer editing occurs independent of the tRNA for LeuRS-ฮ”CP1, which lacks the CP1 domain. Therefore it is hypothesized that the CP1 domain of LeuRS plays a critical role for tRNA-dependent pre-transfer editing

    ์ธ๊ฐ„ ์„ธํฌ์งˆ aminoacyl-tRNA synthetases์˜ ๋™์ ์ธ ์ƒํ˜ธ์ž‘์šฉ๊ณผ ํšจ์†Œ ํ™œ์„ฑ์— ๋Œ€ํ•œ ๋ถ„์„๋ฒ• ๊ฐœ๋ฐœ

    Get PDF
    ํ•™์œ„๋…ผ๋ฌธ (๋ฐ•์‚ฌ) -- ์„œ์šธ๋Œ€ํ•™๊ต ๋Œ€ํ•™์› : ์•ฝํ•™๋Œ€ํ•™ ์•ฝํ•™๊ณผ, 2020. 8. ๊น€์„ฑํ›ˆ.To the best of our knowledge, protein synthesis (translation) is a universal process, which resides in all extant lifeforms. An aminoacyl-tRNA synthetase (ARS) takes a role in the very first step of the translation process; it catalyzes esterification (aminoacylation) of a specific amino acid on its cognate transfer RNAs (tRNAs) to make aminoacylated tRNAs (aminoacyl-tRNAs). The aminoacyl-tRNA delivers the amino acid to the ribosome which catalyzes the translation of a messenger RNA (mRNA) into a polypeptide chain. The cytoplasmic ARSs are differentially regulated in different species; they have gained additional domains and noncanonical functions throughout evolution, and the largest multi-tRNA synthetase complex (MSC) among the eukaryotes exists in higher eukaryotes, which is comprised of eight ARSs for eight or nine amino acids. Among them, the mammalian MSC is the most complexed one, which is composed of eight cytoplasmic ARSs for nine amino acids, and three scaffold proteins. Consequently, nearly half of the aminoacyl-tRNA efflux becomes concentrated at the MSC. Stable supply of the aminoacyl-tRNA to the ribosome is, therefore, considered to be a major role of the mammalian MSC. Furthermore, the mammalian MSC also serves as a reservoir for releasable ARSs or scaffold proteins to support the noncanonical functions of them. In part I, a split-luciferase complementation system was applied to investigate the configuration of the MSC in live mammalian cells. Multiplex interconnections between the components of the MSC were simplified into binary protein-protein interactions, and pairwise comparison of the interactions reconstituted a framework that is consistent with previous in vitro studies. Reversibility of the split-luciferase reporter binding demonstrated convertible organization of the mammalian MSC, including interferon gamma (IFNฮณ)-stimulated glutamyl-prolyl-tRNA synthetase 1 (EPRS1) release, as well as the cooperation with the ribosome bridged by the tRNAs. The cell-based analysis provided an improved understanding of the flexible framework of the mammalian MSC in physiological conditions. On the other hand, abnormality of the aminoacylation has been implicated in a wide variety of cancer pathologies. The ARSs exist in large excess in cancer cells due to their increased demand for the protein synthesis. Meanwhile, most other translation apparatuses are quantitatively limited. There has been no report for mutations of the ARSs that demonstrate constitutive activity of the aminoacylation; the hyperactivity of the ARSs may disrupt stable association of the MSC. Hence, interference of the aminoacylation activity is expected to be independent of genotype variation and may not develop drug resistance. In part II, a high-throughput screen (HTS) platform was established to find the mammalian ARS inhibitors. The ARSs of rabbit reticulocyte closely resemble both the individual and complexed structures of human ones. Therefore, an in vitro translation system with the rabbit-reticulocyte lysate may predispose active compounds to be readily applicable for mankind. The assay was further validated for identifying familiar translational inhibitors from a pilot screen, such as emetine, proving its suitability for the purpose. Having demonstrated excellent quality control (QC) parameters and reproducibility, it is proven ready for further HTS campaign with large molecular entities.๋‹จ๋ฐฑ์งˆ ํ•ฉ์„ฑ (๋ฒˆ์—ญ)์€ ๋ชจ๋“  ํ˜•ํƒœ์˜ ์ƒ๋ช…์ด ๊ฐ€์ง€๊ณ  ์žˆ๋Š” ๊ณตํ†ต์ ์ธ ํŠน์„ฑ์ด๋‹ค. ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ (aminoacyl-tRNA synthetase; ARS)๋Š” ๋‹จ๋ฐฑ์งˆ ํ•ฉ์„ฑ ๊ณผ์ •์—์„œ ๊ฐ€์žฅ ์ฒซ ๋ฒˆ์งธ ๋‹จ๊ณ„๋ฅผ ๋‹ด๋‹นํ•˜๊ณ  ์žˆ๋‹ค. ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ๋Š” ์šด๋ฐ˜RNA (transfer RNA; tRNA)์™€ ์ƒ๋ณด์ ์ธ ์•„๋ฏธ๋…ธ์‚ฐ (amino acid) ์‚ฌ์ด์˜ ์—์Šคํ„ฐํ™” ๋ฐ˜์‘ (esterification/aminoacylation)์„ ์ด‰๋งคํ•˜์—ฌ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์ค‘ํ•ฉ์ฒด (aminoacyl-tRNA)๋กœ ์—ฐ๊ฒฐํ•œ๋‹ค. ์ƒ์„ฑ๋œ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์ค‘ํ•ฉ์ฒด๋Š” ๋ฆฌ๋ณด์†œ (ribosome)์œผ๋กœ ์ „๋‹ฌ๋˜์–ด ์ „๋ นRNA (messenger RNA; mRNA)๋ฅผ ํŽฉํƒ€์ดํŠธ ์ค‘ํ•ฉ์ฒด (polypeptide)๋กœ ๋ฒˆ์—ญํ•˜๋Š” ๊ณผ์ •์˜ ์žฌ๋ฃŒ๋กœ ์‚ฌ์šฉ๋œ๋‹ค. ์„ธํฌ์งˆ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ (cytoplasmic ARS)๋Š” ์ข…์— ๋”ฐ๋ผ ์„ธํฌ๋‚ด์—์„œ ์ œ์–ด๋˜๋Š” ๋ฐฉ์‹์ด ๋‹ค๋ฅด๋‹ค. ์ด ํšจ์†Œ๋“ค์€ ์ง„ํ™”๊ณผ์ • ๋™์•ˆ ์ถ”๊ฐ€์ ์ธ ๋‹จ๋ฐฑ์งˆ ๋„๋ฉ”์ธ (protein domain)๊ณผ ์ƒˆ๋กœ์šด ๊ธฐ๋Šฅ๋“ค์„ ํš๋“ํ•ด์™”๋‹ค. ๋˜ํ•œ ๊ณ ๋“ฑ ์ง„ํ•ต์ƒ๋ฌผ (higher eukaryote)์—๋Š” ์ง„ํ•ต์ƒ๋ฌผ (eukaryote) ์ค‘์—์„œ ๊ฐ€์žฅ ํฐ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ ๋ณตํ•ฉ์ฒด (multi-tRNA synthetase complex; MSC)๊ฐ€ ์กด์žฌํ•˜๋ฉฐ, ์ด ๋ณตํ•ฉ์ฒด๋Š” 8์ข…๋ฅ˜์˜ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ๊ฐ€ 8 ๋‚ด์ง€๋Š” 9์ข…๋ฅ˜์˜ ์•„๋ฏธ๋…ธ์‚ฐ์„ ๋‹ด๋‹นํ•œ๋‹ค. ์ด ์ค‘์—์„œ ํฌ์œ ๋™๋ฌผ์˜ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ ๋ณตํ•ฉ์ฒด๊ฐ€ ๊ฐ€์žฅ ๋ณต์žกํ•œ ํ˜•ํƒœ์ธ๋ฐ, 8์ข…๋ฅ˜์˜ ์„ธํฌ์งˆ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ๊ฐ€ 9์ข…๋ฅ˜์˜ ์•„๋ฏธ๋…ธ์‚ฐ์„ ๋‹ด๋‹นํ•˜๋ฉฐ, ์ถ”๊ฐ€์ ์œผ๋กœ 3์ข…๋ฅ˜์˜ ๋ผˆ๋Œ€ ๋‹จ๋ฐฑ์งˆ (scaffold protein)์ด ์กด์žฌํ•œ๋‹ค. ์ด์ฒ˜๋Ÿผ ํฌ์œ ๋™๋ฌผ์˜ ์„ธํฌ ๋‚ด์—์„œ๋Š” ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์ค‘ํ•ฉ์ฒด ์ด๋™์˜ ์ ˆ๋ฐ˜ ์ •๋„๊ฐ€ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ ๋ณตํ•ฉ์ฒด์—์„œ ์‹œ์ž‘๋˜๊ณ  ์žˆ๋‹ค. ๋”ฐ๋ผ์„œ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์ค‘ํ•ฉ์ฒด๋ฅผ ๋ฆฌ๋ณด์†œ์œผ๋กœ ์•ˆ์ •์ ์œผ๋กœ ๊ณต๊ธ‰ํ•˜๋Š” ๊ฒƒ์ด ํฌ์œ ๋™๋ฌผ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ ๋ณตํ•ฉ์ฒด์˜ ๊ฐ€์žฅ ์ค‘์š”ํ•œ ์—ญํ•  ์ค‘์˜ ํ•˜๋‚˜์ผ ๊ฒƒ์ด๋ผ ์˜ˆ์ƒ๋œ๋‹ค. ํ•œํŽธ์œผ๋กœ, ๋ช‡๋ช‡์˜ ์„ธํฌ์งˆ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ๋“ค๊ณผ ๋ผˆ๋Œ€ ๋‹จ๋ฐฑ์งˆ๋“ค์€ ๋ณตํ•ฉ์ฒด์—์„œ ๋ฒ—์–ด๋‚˜ ์ƒˆ๋กœ์šด ๊ธฐ๋Šฅ์„ ์ˆ˜ํ–‰ํ•˜๊ธฐ ๋•Œ๋ฌธ์—, ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ ๋ณตํ•ฉ์ฒด๋Š” ์ด๋“ค์„ ์œ„ํ•œ ์ €์žฅ๊ณ  (reservoir)๊ฐ€ ๋˜๊ธฐ๋„ ํ•œ๋‹ค. ๋ณธ ๋…ผ๋ฌธ์˜ ์ฒซ ๋ฒˆ์งธ ๋ถ€๋ถ„์—์„œ๋Š” ๋ถ„ํ•  ๋ฃจ์‹œํผ๋ ˆ์ด์ฆˆ ์ƒ๋ณด ์‹œ์Šคํ…œ (split-luciferase complementation system)์„ ์‚ฌ์šฉํ•˜์—ฌ ํฌ์œ ๋™๋ฌผ ์„ธํฌ ๋‚ด์—์„œ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ ๋ณตํ•ฉ์ฒด์˜ ๊ตฌ์„ฑ์„ ํƒ์ƒ‰ํ•˜์˜€๋‹ค. ๊ตฌ์„ฑ์š”์†Œ๋“ค ๊ฐ„์˜ ๋ณตํ•ฉ์ ์ธ ์ƒํ˜ธ์—ฐ๊ฒฐ์€ ๋‘ ๋‹จ๋ฐฑ์งˆ ๊ฐ„์˜ ์ƒํ˜ธ์ž‘์šฉ (binary protein-protein interaction; binary PPI)์˜ ํ•ฉ์œผ๋กœ ๋‹จ์ˆœํ™”์‹œ์ผฐ๊ณ , ๊ทธ๋“ค๊ฐ„์˜ ์Œ๋ณ„ ๋น„๊ต (pairwise comparison; ๊ตฌ์„ฑ ์š”์†Œ๋“ค์˜ ์„œ๋กœ ๋‹ค๋ฅธ ๋ชจ๋“  ์กฐํ•ฉ์„ ๋น„๊ตํ•˜๋Š” ๋ฐฉ์‹)๋ฅผ ํ†ตํ•˜์—ฌ ๊ธฐ์กด์˜ ์ƒ์ฒด ์™ธ ์—ฐ๊ตฌ (in vitro studies)์— ์ƒ์‘ํ•˜๋Š” ๋ณตํ•ฉ์ฒด์˜ ๊ณจ์กฐ (framework)๋ฅผ ์œ ์ถ”ํ•ด๋‚ผ ์ˆ˜ ์žˆ์—ˆ๋‹ค. ๊ทธ๋ฆฌ๊ณ  ๋ถ„ํ•  ๋ฃจ์‹œํผ๋ ˆ์ด์ฆˆ ๋ฆฌํฌํ„ฐ (split-luciferase reporter) ๊ฐ„์˜ ๊ฒฐํ•ฉ์ด ๊ฐ€์—ญ์ ์ด๋ผ๋Š” ์ ์„ ์ด์šฉํ•˜์—ฌ ์ธํ„ฐํŽ˜๋ก ๊ฐ๋งˆ (interferon gamma; IFNฮณ)์— ์˜ํ•œ ๊ธ€๋ฃจํƒ€๋ฏผ-ํ”„๋กค๋ฆฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ (glutamyl-prolyl-tRNA synthetase 1; EPRS1)์˜ ๋ฐฉ์ถœ์ด๋‚˜, ์šด๋ฐ˜RNA๋ฅผ ๋งค๊ฐœ๋กœ ํ•œ ๋ฆฌ๋ณด์†œ๊ณผ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ ๋ณตํ•ฉ์ฒด์˜ ํ˜‘์—…๊ณผ ๊ฐ™์€ ์—ฌ๋Ÿฌ ์ž๊ทน๋“ค์— ์˜ํ•œ ๋ณตํ•ฉ์ฒด ๋‚ด์˜ ์—ญ๋™์ ์ธ ๊ตฌ์กฐ ๋ณ€ํ™”๋ฅผ ๊ด€์ฐฐํ•˜์˜€๋‹ค. ๋ณธ ์—ฐ๊ตฌ๋Š” ์ด์™€ ๊ฐ™์ด ์„ธํฌ๋ฅผ ๊ธฐ๋ฐ˜์œผ๋กœ ํ•œ ๋ถ„์„๋ฒ•์„ ๋ฐ”ํƒ•์œผ๋กœ ์ƒ๋ฆฌ์  ํ™˜๊ฒฝ (physiological condition)์—์„œ ํฌ์œ ๋™๋ฌผ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ ๋ณตํ•ฉ์ฒด์˜ ๊ณจ์กฐ๊ฐ€ ์œ ๋™์ ์œผ๋กœ ๋ณ€ํ™”ํ•˜๊ณ  ์žˆ์Œ์„ ๋ฐํ˜€๋‚ผ ์ˆ˜ ์žˆ์—ˆ๋‹ค. ํ•œํŽธ, ์ •์ƒ์ ์ด์ง€ ๋ชปํ•œ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐ๋ฐ˜์‘์˜ ์กด์žฌ๋Š” ์—ฌ๋Ÿฌ ์ข…๋ฅ˜์˜ ์•”์—์„œ ์ž˜ ์•Œ๋ ค์ ธ ์žˆ๋‹ค. ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ๋Š” ์•”์„ธํฌ์—์„œ ๊ณผ๋Ÿ‰์œผ๋กœ ์กด์žฌํ•˜๋ฉฐ, ์•”์ด ์ง„ํ–‰๋˜๋ฉด์„œ ๋Š˜์–ด๋‚œ ๋‹จ๋ฐฑ์งˆ ํ•ฉ์„ฑ ์š”๊ตฌ๋Ÿ‰์„ ์ถฉ์กฑ์‹œํ‚จ๋‹ค. ์ด๋Š” ๋Œ€๋ถ€๋ถ„์˜ ๋‹ค๋ฅธ ๋‹จ๋ฐฑ์งˆ ํ•ฉ์„ฑ ์š”์†Œ๋“ค์ด ์•”์„ธํฌ์—์„œ ๊ณผ๋Ÿ‰์œผ๋กœ ์กด์žฌํ•˜์ง€ ์•Š๋Š”๋‹ค๋Š” ์‚ฌ์‹ค๊ณผ ๋Œ€๋น„๋œ๋‹ค. ๋˜ํ•œ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ์˜ ํ™œ์„ฑ์„ ์ง€์†์ ์œผ๋กœ ์œ ์ง€์‹œํ‚ค๋Š” ๋Œ์—ฐ๋ณ€์ด๋Š” ์ง€๊ธˆ๊นŒ์ง€ ์•Œ๋ ค์ ธ ์žˆ์ง€ ์•Š๋‹ค. ์ด๋Š” ํšจ์†Œํ™œ์„ฑ์ด ๋น„์ •์ƒ์ ์œผ๋กœ ๋†’์•„์ง„ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ๋Š” ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ ๋ณตํ•ฉ์ฒด์˜ ํ˜•์„ฑ๊ณผ ์œ ์ง€๋ฅผ ์ €ํ•ดํ•˜๊ธฐ ๋•Œ๋ฌธ์ผ ๊ฒƒ์ด๋‹ค. ๋”ฐ๋ผ์„œ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐ๋ฐ˜์‘์„ ์ €ํ•ดํ•˜๋Š” ์น˜๋ฃŒ๋ฒ•์€ ํ™˜์ž ๊ฐœ๊ฐœ์ธ์˜ ์œ ์ „์ฒด ๋‹ค์–‘์„ฑ (genotype variation)์— ์ƒ๊ด€์—†์ด ํšจ๊ณผ๋ฅผ ๋‚ผ ์ˆ˜ ์žˆ๊ณ , ์•ฝ๋ฌผ ์ €ํ•ญ์„ฑ๋„ ๋‚˜ํƒ€๋‚˜์ง€ ์•Š์„ ๊ฒƒ์ด๋ผ ๊ธฐ๋Œ€๋œ๋‹ค. ๋ณธ ๋…ผ๋ฌธ์˜ ๋‘ ๋ฒˆ์งธ ๋ถ€๋ถ„์—์„œ๋Š” ๊ณ ์† ๋Œ€๋Ÿ‰ ์Šคํฌ๋ฆฌ๋‹ ํ”Œ๋žซํผ (high-throughput screening platform)์„ ๊ตฌ์ถ•ํ•˜์—ฌ ํฌ์œ ๋™๋ฌผ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ์˜ ์ €ํ•ด์ œ๋ฅผ ์ฐพ๊ณ ์ž ํ•˜์˜€๋‹ค. ์ด ์‹œ์Šคํ…œ์—์„œ ์‚ฌ์šฉ๋œ ํ† ๋ผ์˜ ๋ง์ƒ์ ํ˜ˆ๊ตฌ ์šฉํ•ด๋ฌผ (rabbit-reticulocyte lysate) ๋‚ด์˜ ์•„๋ฏธ๋…ธ์‚ฐ-์šด๋ฐ˜RNA ์—ฐ๊ฒฐํšจ์†Œ๋“ค์€ ๋‹จ๋… ๋˜๋Š” ๊ฒฐํ•ฉ ๊ตฌ์กฐ๊ฐ€ ์ธ๊ฐ„์˜ ํšจ์†Œ๋‚˜ ๊ทธ ๋ณตํ•ฉ์ฒด์™€ ๋งค์šฐ ๊ฐ€๊น๊ฒŒ ๋‹ฎ์•„์žˆ๊ธฐ ๋•Œ๋ฌธ์— ์ฐพ์•„๋‚ธ ํ™”ํ•ฉ๋ฌผ์ด ์ธ๊ฐ„์—๊ฒŒ ๋ฐ”๋กœ ์ ์šฉ๋  ์ˆ˜ ์žˆ๋Š” ๊ฐ€๋Šฅ์„ฑ์„ ๋†’์—ฌ์ค€๋‹ค. ์ด ์‹œ์Šคํ…œ์€ ๋ณธ ์—ฐ๊ตฌ์—์„œ ์ˆ˜ํ–‰๋œ ์„ ํ–‰ ์Šคํฌ๋ฆฌ๋‹ (pilot screening)์—์„œ ์—๋ฉ”ํ‹ด (emetine)๊ณผ ๊ฐ™์ด ์ž˜ ์•Œ๋ ค์ง„ ๋‹จ๋ฐฑ์งˆ ํ•ฉ์„ฑ ์ €ํ•ด์ œ๋ฅผ ์ฐพ์•„๋‚ด์—ˆ์„ ๋ฟ๋งŒ ์•„๋‹ˆ๋ผ, ํ›Œ๋ฅญํ•œ ํ’ˆ์งˆ๊ด€๋ฆฌ ๋งค๊ฐœ๋ณ€์ˆ˜ (quality control parameters; QC parameters)์™€ ๊ฒฐ๊ณผ์˜ ๋ฐ˜๋ณต์„ฑ์„ ๋ณด์—ฌ์ฃผ์—ˆ๋‹ค. ๋”ฐ๋ผ์„œ ์ด ์‹œ์Šคํ…œ์€ ์ถ”ํ›„ ๋Œ€๋Ÿ‰ ํ™”ํ•ฉ๋ฌผ ๋ผ์ด๋ธŒ๋Ÿฌ๋ฆฌ๋ฅผ ํƒ€๊ฒŸ์œผ๋กœ ํ•œ ๊ณ ์† ๋Œ€๋Ÿ‰ ์Šคํฌ๋ฆฌ๋‹์— ํ™œ์šฉ์ด ์šฉ์ดํ•  ๊ฒƒ์œผ๋กœ ๊ธฐ๋Œ€๋œ๋‹ค.Introduction 1 Part I. Cell-based analysis of pairwise interactions between the components of the multi-tRNA synthetase complex 11 Graphical abstract 11 Highlights 12 Introduction 13 Materials and methods 18 Results 24 Discussion 34 Figures and table 37 Part II. High-throughput screen for protein synthesis inhibitors targeting aminoacyl-tRNA synthetases 89 Graphical abstract 89 Highlights 90 Introduction 91 Materials and methods 95 Results 98 Discussion 106 Figures and tables 107 Glossary 123 Abbreviations 124 References 128 ๊ตญ๋ฌธ์ดˆ๋ก 150Docto
    • โ€ฆ
    corecore