4,116 research outputs found

    Role of the Bifunctional Aminoacyl-tRNA Synthetase EPRS in Human Disease

    Get PDF
    Aminoacyl-tRNA synthetases (AARS) are a class of enzymes that catalyze the charging of tRNAs with cognate amino acids, a critical step that contributes to the fidelity of protein synthesis. Many AARSs also possess noncanonical functions such as regulation of apoptosis, mRNA translation, and RNA splicing. Some AARSs have evolved new domains with no apparent connection to their charging functions. For example, WHEP domains were originally identified in tryptophanyl-tRNA synthetase (WRS), histidyl-tRNA synthetase (HRS), and glutamyl-prolyl-tRNA synthetase (EPRS). EPRS is a unique bifunctional AARS, found only in higher eukaryotes, and consists of glutamyl-tRNA synthetase (ERS) and prolyl-tRNA synthetase (PRS) joined by a non-catalytic linker containing three WHEP domains in humans. Two compound heterozygous point mutations within human ERS (P14R and E205G) have been identified in the genomes of two patients with type 1 diabetes and bone disease. However, the mechanism by which these mutations contribute to disease is unknown. Our goal is to determine whether the point mutations affect the canonical catalytic activity of EPRS responsible for tRNA charging or noncanonical functions. Both P14 and E205 are highly conserved residues located in the GST and catalytic domain, respectively. An ERS variant appended to 2.5 WHEP domains (ERS 2.5W) has been purified and shown to display robust tRNA binding and aminoacylation activity in vitro. The P14R and E205G single mutants display the same binding affinity for tRNAGlu as WT ERS 2.5W, suggesting that the observed defect is at the catalytic step. Whereas the ERS 2.5W P14R mutant has near wild-type (WT) aminoacylation activity, the ERS 2.5W E205G variant has a severe aminoacylation defect. Both mutations, however, lead to reduced amino acid activation. Together with a collaborator, we are currently characterizing the effect of these two mutations on cell proliferation and the integrated stress response. Taken together, this work has important implications for the understanding of AARS-related human disease mechanisms and development of new therapeutics.College of Arts & SciencesOffice of Undergraduate Research & Creative InquiryNo embargoAcademic Major: Biochemistr

    Aminoacyl tRNA synthetase complex interacting multifunctional protein 1 simultaneously binds Glutamyl-Prolyl-tRNA synthetase and scaffold protein aminoacyl tRNA synthetase complex interacting multifunctional protein 3 of the multi-tRNA synthetase complex

    Get PDF
    Higher eukaryotes have developed extensive compartmentalization of amino acid (aa) - tRNA coupling through the formation of a multi-synthetase complex (MSC) that is composed of eight aa-tRNA synthetases (ARS) and three scaffold proteins: aminoacyl tRNA synthetase complex interacting multifunctional proteins (AIMP1, 2 and 3). Lower eukaryotes have a much smaller complex while yeast MSC consists of only two ARS (MetRS and GluRS) and one ARS cofactor 1 protein, Arc1p (Simos et al., 1996), the homolog of the mammalian AIMP1. Arc1p is reported to form a tripartite complex with GluRS and MetRS through association of the N-terminus GST-like domains (GST-L) of the three proteins (Koehler et al., 2013). Mammalian AIMP1 has no GST-L domain corresponding to Arc1p N-terminus. Instead, AIMP3, another scaffold protein of 18โ€ฏkDa composed entirely of a GST-L domain, interacts with Methionyl-tRNA synthetase (MARS) (Quevillon et al., 1999) and Glutamyl-Prolyl-tRNA Synthetase (EPRS) (Cho et al., 2015). Here we report two new interactions between MSC members: AIMP1 binds to EPRS and AIMP1 binds to AIMP3. Interestingly, the interaction between AIMP1 and AIMP3 complex makes it the functional equivalent of a single Arc1p polypeptide in yeast. This interaction is not mapped to AIMP1 N-terminal coiled-coil domain, but rather requires an intact tertiary structure of the entire protein. Since AIMP1 also interacts with AIMP2, all three proteins appear to compose a core docking structure for the eight ARS in the MSC complex

    Nuclear Photosynthetic Gene Expression Is Synergistically Modulated by Rates of Protein Synthesis in Chloroplasts and Mitochondria

    Get PDF
    Arabidopsis thaliana mutants prors1-1 and -2 were identified on the basis of a decrease in effective photosystem II quantum yield. Mutations were localized to the 5'-untranslated region of the nuclear gene PROLYL-tRNA SYNTHETASE1 (PRORS1), which acts in both plastids and mitochondria. In prors1-1 and -2, PRORS1 expression is reduced, along with protein synthesis in both organelles. PRORS1 null alleles (prors1-3 and -4) result in embryo sac and embryo development arrest. In mutants with the leaky prors1-1 and -2 alleles, transcription of nuclear genes for proteins involved in photosynthetic light reactions is downregulated, whereas genes for other chloroplast proteins are upregulated. Downregulation of nuclear photosynthetic genes is not associated with a marked increase in the level of reactive oxygen species in leaves and persists in the dark, suggesting that the transcriptional response is light and photooxidative stress independent. The mrpl11 and prpl11 mutants are impaired in the mitochondrial and plastid ribosomal L11 proteins, respectively. The prpl11 mrpl11 double mutant, but neither of the single mutants, resulted in strong downregulation of nuclear photosynthetic genes, like that seen in leaky mutants for PRORS1, implying that, when organellar translation is perturbed, signals derived from both types of organelles cooperate in the regulation of nuclear photosynthetic gene expression

    The Zinc-binding Domain of Mammalian prolyl-tRNA synthetase is Indispensable for Catalytic Activity and Organism Viability

    Get PDF
    Aminoacyl-tRNA synthetases (AARS) participate in decoding the genome by catalyzing conjugation of amino acids to their cognate tRNAs. During evolution, biochemical and environmental conditions markedly influenced the sequence and structure of the 20 AARSs, revealing adaptations dictating canonical and orthogonal activities. Here, we investigate the function of the appended Zn2+-binding domain (ZBD) in the bifunctional AARS, glutamyl-prolyl-tRNA synthetase (GluProRS). We developed GluProRS mutant mice by CRISPR-Cas9 with a deletion of 29 C-terminal amino acids, including two of four Zn2+-coordinating cysteines. Homozygous ZBD mutant mice die before embryonic day 12.5, but heterozygous mice are healthy. ZBD disruption profoundly reduces GluProRS canonical function by dual mechanisms: it induces rapid proteasomal degradation of the protein and inhibits ProRS aminoacylation activity, likely by suboptimal positioning of ATP in the spatially adjacent catalytic domain. Collectively, our studies reveal the ZBD as a critical determinant of ProRS activity and GluProRS stability in vitro and in vivo

    Segmentally Variable Genes: A New Perspective on Adaptation

    Get PDF
    Genomic sequence variation is the hallmark of life and is key to understanding diversity and adaptation among the numerous microorganisms on earth. Analysis of the sequenced microbial genomes suggests that genes are evolving at many different rates. We have attempted to derive a new classification of genes into three broad categories: lineage-specific genes that evolve rapidly and appear unique to individual species or strains; highly conserved genes that frequently perform housekeeping functions; and partially variable genes that contain highly variable regions, at least 70 amino acids long, interspersed among well-conserved regions. The latter we term segmentally variable genes (SVGs), and we suggest that they are especially interesting targets for biochemical studies. Among these genes are ones necessary to deal with the environment, including genes involved in hostโ€“pathogen interactions, defense mechanisms, and intracellular responses to internal and environmental changes. For the most part, the detailed function of these variable regions remains unknown. We propose that they are likely to perform important binding functions responsible for proteinโ€“protein, proteinโ€“nucleic acid, or proteinโ€“small molecule interactions. Discerning their function and identifying their binding partners may offer biologists new insights into the basic mechanisms of adaptation, context-dependent evolution, and the interaction between microbes and their environment. Segmentally variable genes show a mosaic pattern of one or more rapidly evolving, variable regions. Discerning their function may provide new insights into the forces that shape genome diversity and adaptationNational Science Foundation (998088, 0239435

    ๊ธ€๋ฃจํƒ€๋ฐ€-ํ”„๋กค๋ฆด tRNA ํ•ฉ์„ฑํšจ์†Œ์˜ ํ ๋ฐ ๊ฐ„ ์„ฌ์œ ํ™”์—์„œ STAT Signaling์„ ํ†ตํ•œ Collagen๊ณผ Fibronectin์˜ ๋ฐœํ˜„ ์กฐ์ ˆ ์—ฐ๊ตฌ

    Get PDF
    ํ•™์œ„๋…ผ๋ฌธ (๋ฐ•์‚ฌ)-- ์„œ์šธ๋Œ€ํ•™๊ต ๋Œ€ํ•™์› : ์•ฝํ•™๋Œ€ํ•™ ์•ฝํ•™๊ณผ, 2019. 2. ์ด์ •์›.์„ฌ์œ ํ™”๋ž€ ์—ผ์ฆ์„ฑ ๋ฐ ๋Œ€์‚ฌ์„ฑ ์งˆํ™˜์— ์˜ํ•ด ์„ธํฌ์˜ ๊ณผ๋ถ„์—ด๊ณผ ์žฅ๊ธฐ์˜ ๊ธฐ๋Šฅ๋ถ€์ „์ด ์ผ์–ด๋‚˜ ์„ธํฌ์™ธ๊ธฐ์งˆ (extracellular matrix, ECM) ์ด ๋น„์ •์ƒ์ ์œผ๋กœ ๊ณผ๋‹คํ•˜๊ฒŒ ์ถ•์ ๋˜๋Š” ๊ณผ์ •์„ ๋งํ•œ๋‹ค. ์ตœ๊ทผ ์—ฐ๊ตฌ์— ๋”ฐ๋ฅด๋ฉด ์ƒ์‚ฐ(ๅธธๅฑฑ, Dichroa febrifuga) ์œผ๋กœ๋ถ€ํ„ฐ ๋ถ„๋ฆฌ๋œ ํ™”ํ•ฉ๋ฌผ febrifugine์˜ ํ• ๋กœ๊ฒํ™” ์œ ๋„์ฒด์ธ halofuginone์€ glutamyl-prolyly tRNA synthetase (EPRS)์˜ ํ™œ์„ฑ์„ ์–ต์ œํ•จ์œผ๋กœ์จ ์„ฌ์œ ํ™”๋ฅผ ์ €ํ•ดํ•œ๋‹ค๊ณ  ๋ฐํ˜€์กŒ๋‹ค. ํ•˜์ง€๋งŒ ๊ธฐ์กด ์—ฐ๊ตฌ์—์„œ๋Š” halofuginone์ด EPRS์˜ ํ™œ์„ฑ์„ ์ €ํ•ดํ•จ์— ์žˆ์–ด์„œ EPRS์˜ ๋ฒˆ์—ญ์  ํ™œ์„ฑ์—๋งŒ ์ดˆ์ ์„ ๋งž์ถ”์—ˆ์œผ๋ฉฐ in vivo ํ™˜๊ฒฝ์—์„œ๋Š” ํ™•์ธํ•œ ๋ฐ” ์—†์œผ๋ฏ€๋กœ EPRS๋ฅผ ํƒ€๊ฒŸ์œผ๋กœํ•œ ์„ฌ์œ ํ™” ์น˜๋ฃŒ์ œ ๊ฐœ๋ฐœ์„ ์œ„ํ•ด์„œ๋Š” ์ถ”๊ฐ€ ์—ฐ๊ตฌ๊ฐ€ ํ•„์š”ํ•œ ์ƒํ™ฉ์ด๋‹ค. ๋ณธ ์—ฐ๊ตฌ์—์„œ๋Š” ํ ๋ฐ ๊ฐ„์˜ ์„ฌ์œ ํ™” ๊ณผ์ •์—์„œ ECM์„ ํ•ฉ์„ฑํ•˜๋Š” EPRS์˜ ๋น„-๋ฒˆ์—ญ์  ๊ธฐ๋Šฅ์„ ์—ฐ๊ตฌํ•˜๊ณ  EPRS์˜ hetero knock-out ๋งˆ์šฐ์Šค๋ฅผ ์ด์šฉํ•˜์—ฌ EPRS์˜ in vivo์—์„œ์˜ ์—ญํ• ์„ ๊ฒ€์ฆํ•˜๊ณ ์ž ํ•˜์˜€๋‹ค. ์šฐ์„  ์„ฌ์œ ํ™” ๊ณผ์ • ์ค‘ ECM ํ•ฉ์„ฑ์— ์žˆ์–ด์„œ EPRS์˜ ๊ธฐ๋Šฅ์„ ํ™•์ธํ•˜๊ณ ์ž ํ•˜์˜€๋‹ค. EPRS๊ฐ€ knock-down(KD) ๋˜๊ฑฐ๋‚˜ overexpression ๋œ hepatic stellate cell (LX2 cell) ๋ฐ alveolar epithelial cell (A549 cell)์— TGFฮฒ1์„ ์ฒ˜๋ฆฌํ•˜์—ฌ ์„ฌ์œ ํ™”๋ฅผ ์œ ๋„์‹œํ‚จ ํ›„ ECM์˜ ๋ฐœํ˜„์„ ์‚ดํŽด๋ณด์•˜๋‹ค. ๊ทธ ๊ฒฐ๊ณผ EPRS๊ฐ€ KD ๋œ ์„ธํฌ์—์„œ๋Š” Collagen I, Fibronectin, Laminin ๊ณผ ๊ฐ™์€ ECM์˜ ๋ฐœํ˜„์ด ๊ฐ์†Œํ–ˆ์ง€๋งŒ PRS๊ฐ€ ๊ณผ๋ฐœํ˜„๋œ ์„ธํฌ์—์„œ๋Š” ECM์˜ ๋ฐœํ˜„์ด ์ฆ๊ฐ€ํ•œ ๊ฒƒ์„ ๊ด€์ฐฐํ•  ์ˆ˜ ์žˆ์—ˆ๋‹ค. ์ด๋Š” western blotting์„ ํ†ตํ•œ ๋‹จ๋ฐฑ์งˆ ์ˆ˜์ค€๋ฟ๋งŒ ์•„๋‹ˆ๋ผ qPCR ๋ฐฉ์‹์„ ์ด์šฉํ•œ mRNA level ๋ชจ๋‘ ๋น„์Šทํ•œ ์–‘์ƒ์„ ๊ด€์ฐฐํ•  ์ˆ˜ ์žˆ์—ˆ๋‹ค. ๋˜ํ•œ, promoter luciferase assay๋ฅผ ํ†ตํ•ด EPRS๊ฐ€ KD๋œ ์„ธํฌ์—์„œ collagen I ๋ฐ laminin ฮณ2์˜ promoter๊ฐ€ down-regulation ๋˜๋Š” ๊ฒƒ์„ ํ™•์ธํ•  ์ˆ˜ ์žˆ์—ˆ์œผ๋ฏ€๋กœ EPRS๋Š” ECM ๋ฐœํ˜„์˜ ์ „์‚ฌ์ ์ธ ์ˆ˜์ค€์—์„œ ์ˆ˜ํ–‰ํ•จ์„ ์•Œ ์ˆ˜ ์žˆ์—ˆ๋‹ค. ๋‹ค์Œ์œผ๋กœ EPRS ๊ฐ€ ๋งค๊ฐœ๋œ ECM ์˜ ํ•ฉ์„ฑ ์กฐ์ ˆ์˜ ์‹ ํ˜ธ์ „๋‹ฌ์„ ๊ด€์ฐฐํ•˜๊ธฐ ์œ„ํ•˜์—ฌ ์„ฌ์œ ํ™”์™€ ๋ฐ€์ ‘ํ•œ ๊ด€๊ณ„๊ฐ€ ์žˆ๋‹ค๊ณ  ์•Œ๋ ค์ง„ STAT ๋ถ„์ž๋“ค์˜ ์ธ์‚ฐํ™” ์ •๋„๋ฅผ ๋ถ„์„ํ•˜์˜€๋‹ค. ๊ทธ ๊ฒฐ๊ณผ STAT6๊ฐ€ EPRS์˜ ๋ฐœํ˜„๊ณผ ํ•จ๊ป˜ EPRS๊ฐ€ ๊ณผ๋ฐœํ˜„ ๋˜์—ˆ์„ ๋•Œ ์ธ์‚ฐํ™”๊ฐ€ ์ฆ๊ฐ€๋˜๊ณ  EPRS๊ฐ€ KD ๋  ๋•Œ ์ธ์‚ฐํ™”๊ฐ€ ์ €ํ•ด ๋˜๋Š” ๊ฒƒ์œผ๋กœ ๊ด€์ฐฐ๋˜์—ˆ๋‹ค. ๋˜ํ•œ SMAD2 ๋‹จ๋ฐฑ์งˆ์ด ๊ณผ๋ฐœํ˜„ ๋  ๊ฒฝ์šฐ STAT์ด activation๋˜์–ด SMAD๊ฐ€ STAT์˜ ์ƒ์œ„ ์‹ ํ˜ธ์ „๋‹ฌ์— ๊ด€์—ฌํ•จ์„ ์•Œ ์ˆ˜ ์žˆ์—ˆ๋‹ค. Immunoprecipitation ๋ฐฉ๋ฒ•์„ ์ด์šฉํ•œ ์‹คํ—˜ ๊ฒฐ๊ณผ TGFฮฒ1 receptor, EPRS, Janus kinase ๊ทธ๋ฆฌ๊ณ  STAT6 ์‚ฌ์ด์— ๋‹จ๋ฐฑ์งˆ-๋‹จ๋ฐฑ์งˆ ๊ฒฐํ•ฉ์ด ์ด๋ฃจ์–ด ์ง์œผ๋กœ์จ ์‹ ํ˜ธ์ „๋‹ฌ์ด ์ด๋ฃจ์–ด ์ง์„ ํ™•์ธํ•  ์ˆ˜ ์žˆ์—ˆ๋‹ค. CCl4 ์ฒ˜๋ฆฌ ํ˜น์€ bile duct ligation ๋ฐฉ๋ฒ•์„ ํ†ตํ•ด ๊ฐ„ ์„ฌ์œ ํ™”๋ฅผ ์œ ๋„ํ•˜๊ฑฐ๋‚˜ bleomycin์„ ์ฒ˜๋ฆฌํ•˜์—ฌ ํ ์„ฌ์œ ํ™”๋ฅผ ์œ ๋„ํ•œ ๋™๋ฌผ ์‹คํ—˜ ๊ฒฐ๊ณผ fibrotic septa ์ฃผ๋ณ€์— STAT6์˜ ์ธ์‚ฐํ™”์™€ ECM์˜ ๋ฐœํ˜„์ด ๋™์‹œ์— ์ฆ๊ฐ€ ํ•œ ๋ฐ˜๋ฉด, ๊ทธ ํ˜„์ƒ์ด Eprs-/+ ๋งˆ์šฐ์Šค์—์„œ๋Š” ์ •๋„๊ฐ€ ๋‚ฎ์Œ์„ ์•Œ ์ˆ˜ ์žˆ์—ˆ๋‹ค. ๋”ฐ๋ผ์„œ ๋ณธ ์—ฐ๊ตฌ ๊ฒฐ๊ณผ ์„ฌ์œ ํ™” ๊ณผ์ •์—์„œ EPRS๋Š” TGFฮฒ1์˜์กด์ ์œผ๋กœ ์„ธํฌ์‹ ํ˜ธ์ „๋‹ฌ์— ์˜ํ•œ ๋น„-๋ฒˆ์—ญ์  ๊ณผ์ •์„ ๊ฑฐ์ฒ˜ ECM์˜ ๋ฐœํ˜„์„ ์กฐ์ ˆํ•˜๋Š” ๊ฒƒ์„ ํ™•์ธ ํ•  ์ˆ˜ ์žˆ์—ˆ๋‹ค. EPRS ๋‹จ๋ฐฑ์งˆ์€ ํ–ฅํ›„ ๊ฐ„ ๋ฐ ํ ์„ฌ์œ ํ™” ์–ต์ œ ์น˜๋ฃŒ์ œ์˜ ํƒ€๊ฒŸ์œผ๋กœ ํ™œ์šฉ ๋  ์ˆ˜ ์žˆ์„ ๊ฒƒ์œผ๋กœ ๊ธฐ๋Œ€๋œ๋‹ค.Background: Fibrosis is characterized by the increased accumulation of extracellular matrix (ECM), which drives abnormal cell proliferation and progressive organ dysfunction in many inflammatory and metabolic diseases. Studies have shown that halofuginone, a racemic halogenated derivative of febrifugine, purified from Dichroa febrifuga, inhibits glutamyl-prolyl-tRNA-synthetase (EPRS)-mediated fibrosis. However, the mechanism by which this occurs was only focused on the translational function of EPRS and in vivo efficacies were not studied. Thus, in order to develop efficacious drugs targeting EPRS, more studies are needed. Methods: In this study, I explored the mechanistic aspects of how EPRS could develop hepatic and pulmonary fibrotic phenotypes in cells and animal models. CCl4 administration, bile duct ligation operation, and bleomycin administration were used in order to induce fibrosis in wild type (Eprs+/+) or Eprs-/+ C57B/L6 mice. Results: Treatment of transforming growth factor 1 (TGF1) up-regulated extracellular matrix proteins, including fibronectin and collagen I, in LX2 hepatic stellate cells and A549 alveolar epithelial cells. This effect was inhibited in EPRS-suppressed cells and enhanced in PRS-overexpressed cells. Using the promoter luciferase assay, TGF1-mediated COL1A1 (collagen I, 1 chain) and LAMC2 (laminin 2) transcription in LX2 and A549 cells were down-regulated by EPRS suppression, suggesting that EPRS may play roles in ECM production at transcriptional levels. Furthermore, signal transducer and activator of transcription (STAT) signaling activation was involved in the effects of TGF1 on ECM expression in an EPRS-dependent manner. This was mediated via a protein-protein complex formation consisting of TGF1 receptor, EPRS, Janus kinases, and STATs. Additionally, ECM expression in fibrotic livers and lungs were overlapped with EPRS expression along fibrotic septa regions and was positively correlated with STAT6 activation in fibrosis mouse models. This was less obvious in livers and lungs of Eprs-/+ mice. Conclusion: These findings suggest that during fibrosis development, EPRS plays roles in non-translational processes of ECM expression via the TGF1/STAT signaling pathway. Therefore, EPRS can be used as a potential target to develop anti-fibrosis treatments.ABSTRACT ii TABLE OF CONTENTS iv LIST OF FIGURES vii LIST OF TABLES xi LIST OF ABBREBIATIONS xii BACKGROUND 1 โ…ฐ. Fibrosis 2 โ…ฑ. Hepatic fibrosis 4 โ…ฒ. Pulmonary fibrosis 8 โ…ณ. TGFฮฒ1 Pathway 11 โ…ด. EPRS 13 โ…ต. Halofuginone 22 CHAPTER 1. GLUTAMYL-PROLYL-TRNA SYNTHETASE INDUCES FIBROTIC EXTRACELLULAR MATRIX VIA BOTH TRANSCRIPTIONAL AND TRANSLATIONAL MECHANISMS 26 1.1. ABSTRACT 27 1.2. INTRODUCTION 28 1.3. MATERIALS AND METHODS 31 Reagents and plasmids 31 Cell culture 31 Western blot analysis 32 ECM deposition assay and collagen footprint assay 32 Quantitative reverse-transcription (qRT) PCR 33 Co-immunoprecipitation 33 Luciferase assay 34 Animal experiments 34 Liver organoid culture 35 Immunohistochemistry and staining 35 1.4. RESULTS 39 Inhibition or suppression of EPRS in LX2 HSCs decreased the production and deposition of ECMs under TGF1 signaling 39 EPRS-mediated transcriptional regulation of ECMs involved STAT6 activation 42 EPRS-mediated signaling occurred downstream of TGF1 44 In vivo liver tissues from fibrotic mice showed EPRS-dependent STAT6 phosphorylation and ECM production 45 Liver organoids in a three-dimensional (3D) Matrigel system revealed EPRS-dependent regulation of ECM induction 47 1.5. DISCUSSION 65 CHAPTER 2. GLUTAMYL-PROLYL-TRNA SYNTHETASE REGULATES EPITHELIAL EXPRESSION OF MESENCHYMAL MARKERS AND EXTRACELLULAR MATRIX PROTEINS: IMPLICATIONS FOR IDIOPATHIC PULMONARY FIBROSIS 69 2.1. ABSTRACT 70 2.2. INTRODUCTION 71 2.3. MATERIALS AND METHODS 73 Reagents and plasmids 73 Cell culture 73 Western blot analysis 74 qRT-PCR 74 Co-immunoprecipitation 74 Luciferase assay 75 Animal experiments 75 Immunohistochemistry and staining 76 Statistics 76 2.4. RESULTS 79 EPRS expression regulated ECM production in A549 alveolar type II cells upon TGF1 stimulation 79 Regulation of TGFฮฒ1-induced ECM protein synthesis by EPRS occurred via STAT activation 80 TGFฮฒ1-mediated SMAD3 phosphorylation upregulated phosphorylation of STAT6 depending on EPRS expression. 81 EPRS-mediated signaling in TGF1-treated cells involved the formation of a multi-protein complex consisting of STAT6 and TGF1R 81 Lung tissues from bleomycin-treated mice showed EPRS-dependent STAT6 phosphorylation and ECM protein production in vivo 82 2.5. DISCUSSION 93 CONCLUSIONS & PERSPECTIVES 97 REFERENCES 101 ABSTRACT IN KOREAN 112Docto

    Analysis of the Role of Aminoacyl tRNA Synthetase Genes in Global Protein Synthesis and mRNA Specific Regulation of Translation in Cancer Cells

    Get PDF
    Analysis of the Role of Aminoacyl tRNA Synthetase Genes in Global Protein Synthesis and mRNA Specific Regulation of Translation in Cancer Cells Elyse Nguyen, Depts. of Biology and Chemistry, Dipak Poria, & Esta Sterneck, with Dr. Sarah Williams, Dept. of Forensic Science Coordinated control of transcription and translation of gene expression impels cellular fate decision under different microenvironmental stresses. Cancer cells often usurp these regulatory machineries to adapt under microenvironmental stress or under therapeutic intervention. The transcription factor CEBPฮด is induced by various stressors and mediates cellular adaptation and survival. RNA-seq analysis of a CEBPD-silenced human melanoma cell line, MB-435s, showed decreased expression of 12 aminoacyl-tRNA synthetase (aaRS) genes. Our group recently found that deletion of CEBPD by CRISPR/Cas9 (CEBPD-KO) compromised aminoacyl tRNA synthetase (aaRS) expression and global protein synthesis. However, despite this decrease in global protein production, the synthesis of certain proteins, such as ATF4, which promotes survival and/or death under stress conditions, is increased. Aminoacyl tRNA synthetases are essential enzymes in the process of protein synthesis which catalyze the addition of appropriate amino acid to its corresponding tRNA, and therefore act as a rate limiting step in cellular protein synthesis. In the current project, we sought to investigate the effect of silencing of specific aaRS genes, glutamyl-prolyl-tRNA synthetase (EPRS) and valyl-tRNA synthetase (VARS) on global protein translation and ATF4 expression. To address this question, we silenced the EPRS and VARS gene expression using two independent short-hairpin-RNA (shRNA) targeting two different regions of EPRS and VARS mRNAs in MB-435s cells. Silencing of EPRS gene showed compensatory upregulation of VARS and vice versa. Interestingly, our preliminary data suggested an upregulation of global protein synthesis after EPRS and VARS silencing in MB435s cells measured by puromycin pulse labelling. Ongoing experiments to validate the preliminary data and ATF4 expression will be discussed.https://scholarscompass.vcu.edu/uresposters/1326/thumbnail.jp

    Evolution of acceptor stem tRNA recognition by class II prolyl-tRNA synthetase

    Get PDF
    Aminoacyl-tRNA synthetases (AARS) are an essential family of enzymes that catalyze the attachment of amino acids to specific tRNAs during translation. Previously, we showed that base-specific recognition of the tRNAPro acceptor stem is critical for recognition by Escherichia coli prolyl-tRNA synthetase (ProRS), but not for human ProRS. To further delineate species-specific differences in acceptor stem recognition, atomic group mutagenesis was used to probe the role of sugarโ€“phosphate backbone interactions in recognition of human tRNAPro. Incorporation of site-specific 2โ€ฒ-deoxynucleotides, as well as phosphorothioate and methylphosphonate modifications within the tRNA acceptor stem revealed an extensive network of interactions with specific functional groups proximal to the first base pair and the discriminator base. Backbone functional groups located at the base of the acceptor stem, especially the 2โ€ฒ-hydroxyl of A66, are also critical for aminoacylation catalytic efficiency by human ProRS. Therefore, in contrast to the bacterial system, backbone-specific interactions contribute significantly more to tRNA recognition by the human enzyme than base-specific interactions. Taken together with previous studies, these data show that ProRS-tRNA acceptor stem interactions have co-adapted through evolution from a mechanism involving โ€˜direct readoutโ€™ of nucleotide bases to one relying primarily on backbone-specific โ€˜indirect readoutโ€™

    ์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