Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
PLoS One ; 19(4): e0296995, 2024.
Article in English | MEDLINE | ID: mdl-38558084

ABSTRACT

Emerging resistance to existing antimalarial drugs drives the search for new antimalarials, and protein translation is a promising pathway to target. Threonyl t-RNA synthetase (ThrRS) is one of the enzymes involved in this pathway, and it has been validated as an anti-malarial drug target. Here, we present 9 structurally diverse low micromolar Plasmodium falciparum ThrRS inhibitors that were identified using high-throughput virtual screening (HTVS) and were verified in a FRET enzymatic assay. Salicylic acid-based compound (LE = 0.34) was selected as a most perspective hit and was subjected to hit-to-lead optimisation. A total of 146 hit analogues were synthesised or obtained from commercial vendors and were tested. Structure-activity relationship study was supported by the crystal structure of the complex of a salicylic acid analogue with a close homologue of the plasmodium target, E. coli ThrRS (EcThrRS). Despite the availability of structural information, the hit identified via virtual screening remained one of the most potent PfThrRS inhibitors within this series. However, the compounds presented herein provide novel scaffolds for ThrRS inhibitors, which could serve as starting points for further medicinal chemistry projects targeting ThrRSs or structurally similar enzymes.


Subject(s)
Antimalarials , Malaria , Threonine-tRNA Ligase , Humans , Threonine-tRNA Ligase/chemistry , Threonine-tRNA Ligase/genetics , Threonine-tRNA Ligase/metabolism , Escherichia coli/genetics , Structure-Activity Relationship , Plasmodium falciparum/genetics , Antimalarials/pharmacology , Salicylic Acid/pharmacology , RNA, Transfer
2.
Biochem Soc Trans ; 52(2): 661-670, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38477373

ABSTRACT

Aminoacyl-tRNA synthetases (AARSs) play an indispensable role in the translation of mRNAs into proteins. It has become amply clear that AARSs also have non-canonical or non-translational, yet essential, functions in a myriad of cellular and developmental processes. In this mini-review we discuss the current understanding of the roles of threonyl-tRNA synthetase (TARS) beyond protein synthesis and the underlying mechanisms. The two proteins in eukaryotes - cytoplasmic TARS1 and mitochondrial TARS2 - exert their non-canonical functions in the regulation of gene expression, cell signaling, angiogenesis, inflammatory responses, and tumorigenesis. The TARS proteins utilize a range of biochemical mechanisms, including assembly of a translation initiation complex, unexpected protein-protein interactions that lead to activation or inhibition of intracellular signaling pathways, and cytokine-like signaling through cell surface receptors in inflammation and angiogenesis. It is likely that new functions and novel mechanisms will continue to emerge for these multi-talented proteins.


Subject(s)
Protein Biosynthesis , Signal Transduction , Threonine-tRNA Ligase , Humans , Threonine-tRNA Ligase/metabolism , Animals , Inflammation/metabolism , Mitochondria/metabolism
3.
Cell Host Microbe ; 31(6): 1021-1037.e10, 2023 06 14.
Article in English | MEDLINE | ID: mdl-37269833

ABSTRACT

Commensal bacteria are critically involved in the establishment of tolerance against inflammatory challenges, the molecular mechanisms of which are just being uncovered. All kingdoms of life produce aminoacyl-tRNA synthetases (ARSs). Thus far, the non-translational roles of ARSs have largely been reported in eukaryotes. Here, we report that the threonyl-tRNA synthetase (AmTARS) of the gut-associated bacterium Akkermansia muciniphila is secreted and functions to monitor and modulate immune homeostasis. Secreted AmTARS triggers M2 macrophage polarization and orchestrates the production of anti-inflammatory IL-10 via its unique, evolutionary-acquired regions, which mediates specific interactions with TLR2. This interaction activates the MAPK and PI3K/AKT signaling pathways, which converge on CREB, leading to an efficient production of IL-10 and suppression of the central inflammatory mediator NF-κB. AmTARS restores IL-10-positive macrophages, increases IL-10 levels in the serum, and attenuates the pathological effects in colitis mice. Thus, commensal tRNA synthetases can act as intrinsic mediators that maintain homeostasis.


Subject(s)
Threonine-tRNA Ligase , Animals , Mice , Threonine-tRNA Ligase/metabolism , Interleukin-10/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Verrucomicrobia/metabolism , Homeostasis , RNA, Transfer/metabolism
4.
J Biol Chem ; 299(5): 104704, 2023 05.
Article in English | MEDLINE | ID: mdl-37059185

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) are essential components for mRNA translation. Two sets of aaRSs are required for cytoplasmic and mitochondrial translation in vertebrates. Interestingly, TARSL2 is a recently evolved duplicated gene of TARS1 (encoding cytoplasmic threonyl-tRNA synthetase) and represents the only duplicated aaRS gene in vertebrates. Although TARSL2 retains the canonical aminoacylation and editing activities in vitro, whether it is a true tRNA synthetase for mRNA translation in vivo is unclear. In this study, we showed that Tars1 is an essential gene since homozygous Tars1 KO mice were lethal. In contrast, when Tarsl2 was deleted in mice and zebrafish, neither the abundance nor the charging levels of tRNAThrs were changed, indicating that cells relied on Tars1 but not on Tarsl2 for mRNA translation. Furthermore, Tarsl2 deletion did not influence the integrity of the multiple tRNA synthetase complex, suggesting that Tarsl2 is a peripheral member of the multiple tRNA synthetase complex. Finally, we observed that Tarsl2-deleted mice exhibited severe developmental retardation, elevated metabolic capacity, and abnormal bone and muscle development after 3 weeks. Collectively, these data suggest that, despite its intrinsic activity, loss of Tarsl2 has little influence on protein synthesis but does affect mouse development.


Subject(s)
Amino Acyl-tRNA Synthetases , Protein Biosynthesis , Threonine-tRNA Ligase , Animals , Mice , Amino Acyl-tRNA Synthetases/metabolism , RNA, Transfer/metabolism , Threonine-tRNA Ligase/genetics , Threonine-tRNA Ligase/metabolism , Zebrafish/genetics , Zebrafish/metabolism
5.
Genes (Basel) ; 14(2)2023 02 10.
Article in English | MEDLINE | ID: mdl-36833389

ABSTRACT

Aminoacyl tRNA synthetases (aaRSs) are a well-studied family of enzymes with a canonical role in charging tRNAs with a specific amino acid. These proteins appear to also have non-canonical roles, including post-transcriptional regulation of mRNA expression. Many aaRSs were found to bind mRNAs and regulate their translation into proteins. However, the mRNA targets, mechanism of interaction, and regulatory consequences of this binding are not fully resolved. Here, we focused on yeast cytosolic threonine tRNA synthetase (ThrRS) to decipher its impact on mRNA binding. Affinity purification of ThrRS with its associated mRNAs followed by transcriptome analysis revealed a preference for mRNAs encoding RNA polymerase subunits. An mRNA that was significantly bound compared to all others was the mRNA encoding RPC10, a small subunit of RNA polymerase III. Structural modeling suggested that this mRNA includes a stem-loop element that is similar to the anti-codon stem loop (ASL) structure of ThrRS cognate tRNA (tRNAThr). We introduced random mutations within this element and found that almost every change from the normal sequence leads to reduced binding by ThrRS. Furthermore, point mutations at six key positions that abolish the predicted ASL-like structure showed a significant decrease in ThrRS binding with a decrease in RPC10 protein levels. Concomitantly, tRNAThr levels were reduced in the mutated strain. These data suggest a novel regulatory mechanism in which cellular tRNA levels are regulated through a mimicking element within an RNA polymerase III subunit in a manner that involves the tRNA cognate aaRS.


Subject(s)
RNA Polymerase III , Amino Acyl-tRNA Synthetases/genetics , Codon , Ligases/genetics , RNA Polymerase III/genetics , RNA, Messenger/genetics , RNA, Transfer/metabolism , RNA, Transfer, Thr/metabolism , Saccharomyces cerevisiae/genetics , Threonine/genetics , Threonine/metabolism , Threonine-tRNA Ligase/chemistry , Threonine-tRNA Ligase/genetics , Threonine-tRNA Ligase/metabolism
6.
Commun Biol ; 6(1): 107, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36707692

ABSTRACT

Aminoacyl-tRNA synthetases (AARSs), a family of essential protein synthesis enzymes, are attractive targets for drug development. Although several different types of AARS inhibitors have been identified, AARS covalent inhibitors have not been reported. Here we present five unusual crystal structures showing that threonyl-tRNA synthetase (ThrRS) is covalently inhibited by a natural product, obafluorin (OB). The residue forming a covalent bond with OB is a tyrosine in ThrRS active center, which is not commonly modified by covalent inhibitors. The two hydroxyl groups on the o-diphenol moiety of OB form two coordination bonds with the conserved zinc ion in the active center of ThrRS. Therefore, the ß-lactone structure of OB can undergo ester exchange reaction with the phenolic group of the adjacent tyrosine to form a covalent bond between the compound and the enzyme, and allow its nitrobenzene structure to occupy the binding site of tRNA. In addition, when this tyrosine was replaced by a lysine or even a weakly nucleophilic arginine, similar bonds could also be formed. Our report of the mechanism of a class of AARS covalent inhibitor targeting multiple amino acid residues could facilitate approaches to drug discovery for cancer and infectious diseases.


Subject(s)
Amino Acyl-tRNA Synthetases , Threonine-tRNA Ligase , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Tyrosine , Zinc , Threonine-tRNA Ligase/metabolism , Binding Sites
7.
J Mol Graph Model ; 115: 108224, 2022 09.
Article in English | MEDLINE | ID: mdl-35636339

ABSTRACT

ß-Hydroxynorvaline (ßHNV) is unnatural amino acid structurally identical to the threonine amino acid with branched ethyl group instead of threonine's methyl. It is a known competitive inhibitor that readily bind to Threonyl-tRNA synthetase's (ThrRS) catalytic site and blocks its function. In this work, we utilized a combination of Molecular Dynamics simulation (MD) and Quantum Mechanics/Molecular Mechanics (QM/MM) methodologies to provide mechanistic insights into its inhibition reaction for ThrRS. Due to the presence of Zn(II) with its Lewis acidity character, only the ionized form of ßHNV gives an enzymatically feasible energy barrier. Furthermore, in consistence with the homochirality behavior of this active site, we observed only one conformation of ßHNV that could be acylated in the active site of ThrRS. Considering these new findings together with the recent search for new antibacterial agents, our findings should guide pharmaceutical scientists with further knowledge regarding the chemical nature of this drug. Moreover, benchmarking analysis of the utilized DFT functional has also been performed to identify the impact of various DFT functionals on representing the geometry and kinetics of our system. Notably, our Zn(II) containing chemical models are found to be responsive to the %HF contribution included together with the dispersion correction. Importantly, the BP86(0%HF)-D3 functional is found to display the greatest impact on the rate-limiting step kinetically. The crucial role played by Zn(II) is further enriched when its mutation with the chemically similar Cd(II) led to dramatic difference via obtaining less feasible reaction mechanism from thermodynamic and kinetic perspectives.


Subject(s)
Threonine-tRNA Ligase , Amino Acids , Catalytic Domain , Molecular Dynamics Simulation , RNA, Transfer/chemistry , Threonine/analogs & derivatives , Threonine/chemistry , Threonine-tRNA Ligase/chemistry , Threonine-tRNA Ligase/metabolism
8.
Bioengineered ; 13(3): 5190-5204, 2022 03.
Article in English | MEDLINE | ID: mdl-35184682

ABSTRACT

Lung cancer is a significant global burden. Aminoacyl-tRNA synthetases (aaRSs) can be reliably identified by the occurrence and improvement of tumors. Threonyl-tRNA synthetase (TARS) and mitochondrial threonyl-tRNA synthetase 2 (TARS2) are both aaRSs. Many studies have shown that TARS are involved in tumor angiogenesis and metastasis. However, TARS2 has not yet been reported in tumors. This study explored the role of TARS2 in the proliferation and apoptosis of lung adenocarcinoma (LUAD). TARS2 expression in lung adenocarcinoma and non-cancerous lung tissues was detected via immunohistochemistry. Cell proliferation was detected using MTS, clone formation, and EdU staining assays. Flow cytometry was used to detect cell cycle, mitochondria reactive oxygen species (mROS) production, and apoptosis. Mitochondrial membrane potential (MMP ΔΨm) was detected using JC-1 fluorescent probes. Cell cycle, apoptosis-related pathway, and mitochondrial DNA (mtDNA) -encoded protein expression was detected via Western blotting. Finally, the effect of TARS2 on tumor growth was examined using a xenotransplanted tumor model in nude mice. We found that TARS2 was highly expressed in lung adenocarcinoma tissues and associated with poor overall survival (OS). Mechanistic analysis showed that knockdown of TARS2 inhibited proliferation through the retinoblastoma protein (RB) pathway and promoted mROS-induced apoptosis. Knockdown of TARS2 inhibits tumor growth in a xenotransplanted tumor model. TARS2 plays an important role in LUAD cell proliferation and apoptosis and may be a new therapeutic target.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Threonine-tRNA Ligase , Adenocarcinoma of Lung/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Tars , Threonine-tRNA Ligase/genetics , Threonine-tRNA Ligase/metabolism
9.
FASEB J ; 35(10): e21948, 2021 10.
Article in English | MEDLINE | ID: mdl-34569098

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) are house-keeping enzymes that are essential for protein synthesis. However, it has become increasingly evident that some aaRSs also have non-translational functions. Here we report the identification of a non-translational function of threonyl-tRNA synthetase (ThrRS) in myogenic differentiation. We find that ThrRS negatively regulates myoblast differentiation in vitro and injury-induced skeletal muscle regeneration in vivo. This function is independent of amino acid binding or aminoacylation activity of ThrRS, and knockdown of ThrRS leads to enhanced differentiation without affecting the global protein synthesis rate. Furthermore, we show that the non-catalytic new domains (UNE-T and TGS) of ThrRS are both necessary and sufficient for the myogenic function. In searching for a molecular mechanism of this new function, we find the kinase JNK to be a downstream target of ThrRS. Our data further reveal MEKK4 and MKK4 as upstream regulators of JNK in myogenesis and the MEKK4-MKK4-JNK pathway to be a mediator of the myogenic function of ThrRS. Finally, we show that ThrRS physically interacts with Axin1, disrupts Axin1-MEKK4 interaction and consequently inhibits JNK signaling. In conclusion, we uncover a non-translational function for ThrRS in the maintenance of homeostasis of skeletal myogenesis and identify the Axin1-MEKK4-MKK4-JNK signaling axis to be an immediate target of ThrRS action.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Muscle Development , Threonine-tRNA Ligase/metabolism , Animals , Axin Protein/metabolism , Female , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding , Protein Biosynthesis , Protein Domains , Threonine-tRNA Ligase/chemistry
10.
Hum Cell ; 34(5): 1504-1516, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34024034

ABSTRACT

Glioblastoma (GBM) is the most lethal type of primary brain tumor and is characterized by diffuse infiltrative growth. However, the mechanisms that control this phenotype remain largely unknown. Emerging evidence has demonstrated that the abnormal expression of microRNAs and their target genes are involved in the migration and invasion of glioma cells. In this study, we demonstrated that microRNA-720 (miR-720) was significantly upregulated in glioma tissues and cells. Functional experiments showed that overexpression of miR-720 promotes glioma migration and invasion, while downregulation of miR-720 inhibits glioma migration and invasion. Meanwhile, we found that threonyl-tRNA synthetase like-2 (TARSL2) was a direct and functional target of miR-720 in glioma. Reintroduction of TARSL2 into glioma cells repressed the invasion promoting function of miR-720, whereas downregulation of TARSL2 reversed the anti-invasion function of anti-miR-720. Furthermore, quantitative real-time polymerase chain reaction results showed that miR-720 was inversely correlated with TARSL2 expression in 40 GBM tissues. Finally, in vivo experiments showed that miR-720 promotes glioma growth and upregulates invasion-related genes in nude mice. Overall, our findings suggest increasing miR-720 enhances glioma migration and invasion through downregulation of TARSL2, which may provide novel insight into the treatment of glioma.


Subject(s)
Cell Movement/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Glioma/pathology , MicroRNAs/genetics , MicroRNAs/physiology , Neoplasm Invasiveness/genetics , Threonine-tRNA Ligase/genetics , Threonine-tRNA Ligase/metabolism , Humans , Tumor Cells, Cultured
11.
Mol Cell ; 81(2): 398-407.e4, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33340489

ABSTRACT

Mechanistic target of rapamycin complex 1 (mTORC1) controls cell growth and proliferation by sensing fluctuations in environmental cues such as nutrients, growth factors, and energy levels. The Rag GTPases (Rags) serve as a critical module that signals amino acid (AA) availability to modulate mTORC1 localization and activity. Recent studies have demonstrated how AAs regulate mTORC1 activity through Rags. Here, we uncover an unconventional pathway that activates mTORC1 in response to variations in threonine (Thr) levels via mitochondrial threonyl-tRNA synthetase TARS2. TARS2 interacts with inactive Rags, particularly GTP-RagC, leading to increased GTP loading of RagA. mTORC1 activity in cells lacking TARS2 is resistant to Thr repletion, showing that TARS2 is necessary for Thr-dependent mTORC1 activation. The requirement of TARS2, but not cytoplasmic threonyl-tRNA synthetase TARS, for this effect demonstrates an additional layer of complexity in the regulation of mTORC1 activity.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/genetics , Mitochondria/metabolism , Monomeric GTP-Binding Proteins/genetics , Threonine-tRNA Ligase/genetics , Threonine/metabolism , Gene Expression Regulation , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Monomeric GTP-Binding Proteins/metabolism , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction , Threonine-tRNA Ligase/antagonists & inhibitors , Threonine-tRNA Ligase/metabolism
12.
Front Immunol ; 11: 571959, 2020.
Article in English | MEDLINE | ID: mdl-33178197

ABSTRACT

Threonyl-tRNA synthetase (TRS) is an aminoacyl-tRNA synthetase that catalyzes the aminoacylation of tRNA by transferring threonine. In addition to an essential role in translation, TRS was extracellularly detected in autoimmune diseases and also exhibited pro-angiogenetic activity. TRS is reported to be secreted into the extracellular space when vascular endothelial cells encounter tumor necrosis factor-α. As T helper (Th) type 1 response and IFN-γ levels are associated with autoimmunity and angiogenesis, in this study, we investigated the effects of TRS on dendritic cell (DC) activation and CD4 T cell polarization. TRS-treated DCs exhibited up-regulated expression of activation-related cell-surface molecules, including CD40, CD80, CD86, and MHC class II. Treatment of DCs with TRS resulted in a significant increase of IL-12 production. TRS triggered nuclear translocation of the NF-κB p65 subunit along with the degradation of IκB proteins and the phosphorylation of MAPKs in DCs. Additionally, MAPK inhibitors markedly recovered the degradation of IκB proteins and the increased IL-12 production in TRS-treated DCs, suggesting the involvement of MAPKs as the upstream regulators of NF-κB in TRS-induced DC maturation and activation. Importantly, TRS-stimulated DCs significantly increased the populations of IFN-γ+CD4 T cells, and the levels of IFN-γ when co-cultured with CD4+ T cells. The addition of a neutralizing anti-IL-12 mAb to the cell cultures of TRS-treated DCs and CD4+ T cells resulted in decreased IFN-γ production, indicating that TRS-stimulated DCs may enhance the Th1 response through DC-derived IL-12. Injection of OT-II mice with OVA-pulsed, TRS-treated DCs also enhanced Ag-specific Th1 responses in vivo. Importantly, injection with TRS-treated DC exhibited increased populations of IFN-γ+-CD4+ and -CD8+ T cells as well as secretion level of IFN-γ, resulting in viral clearance and increased survival periods in mice infected with influenza A virus (IAV), as the Th1 response is associated with the enhanced cellular immunity, including anti-viral activity. Taken together, these results indicate that TRS promotes the maturation and activation of DCs, DC-mediated Th1 responses, and anti-viral effect on IAV infection.


Subject(s)
Dendritic Cells/immunology , Influenza A virus/physiology , Interleukin-12/metabolism , NF-kappa B/metabolism , Orthomyxoviridae Infections/immunology , Th1 Cells/immunology , Threonine-tRNA Ligase/metabolism , Animals , Antibodies, Blocking/metabolism , Cell Differentiation , Cells, Cultured , Female , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Threonine-tRNA Ligase/immunology
13.
Eur J Med Chem ; 207: 112848, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32980741

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) are an attractive class of antibacterial drug targets due to their essential roles in protein translation. While most traditional aaRS inhibitors target the binding pockets of substrate amino acids and/or ATP, we recently developed a class of novel tRNA-amino acid dual-site inhibitors including inhibitor 3 ((2S,3R)-2-amino-N-((E)-4-(6,7-dichloro-4-oxoquinazolin-3(4H)-yl)but-2-en-1-yl)-3-hydroxybutanamide) against threonyl-tRNA synthetase (ThrRS). Here, the binding modes and structure-activity relationships (SARs) of these inhibitors were analyzed by the crystal structures of Salmonella enterica ThrRS (SeThrRS) in complex with three of them. Based on the cocrystal structures, twelve quinazolinone-threonine hybrids were designed and synthesized, and their affinities, enzymatic inhibitory activities, and cellular potencies were evaluated. The best derivative 8g achieved a Kd value of 0.40 µM, an IC50 value of 0.50 µM against SeThrRS and MIC values of 16-32 µg/mL against the tested bacterial strains. The cocrystal structure of the SeThrRS-8g complex revealed that 8g induced a bended conformation for Met332 by forming hydrophobic interactions, which better mimicked the binding of tRNAThr to ThrRS. Moreover, the inhibitory potency of 8g was less impaired than a reported ATP competitive inhibitor at high concentrations of ATP, supporting our hypothesis that tRNA site inhibitors are likely superior to ATP site inhibitors in vivo, where ATP typically reaches millimolar concentrations.


Subject(s)
Drug Design , Quinazolinones/chemistry , Salmonella enterica/enzymology , Threonine-tRNA Ligase/antagonists & inhibitors , Threonine/chemistry , Threonine/pharmacology , Adenosine Triphosphate/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding, Competitive , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Salmonella enterica/drug effects , Structure-Activity Relationship , Threonine-tRNA Ligase/metabolism
14.
PLoS Negl Trop Dis ; 14(2): e0007983, 2020 02.
Article in English | MEDLINE | ID: mdl-32106219

ABSTRACT

The development of chemotherapies against eukaryotic pathogens is especially challenging because of both the evolutionary conservation of drug targets between host and parasite, and the evolution of strain-dependent drug resistance. There is a strong need for new nontoxic drugs with broad-spectrum activity against trypanosome parasites such as Leishmania and Trypanosoma. A relatively untested approach is to target macromolecular interactions in parasites rather than small molecular interactions, under the hypothesis that the features specifying macromolecular interactions diverge more rapidly through coevolution. We computed tRNA Class-Informative Features in humans and independently in eight distinct clades of trypanosomes, identifying parasite-specific informative features, including base pairs and base mis-pairs, that are broadly conserved over approximately 250 million years of trypanosome evolution. Validating these observations, we demonstrated biochemically that tRNA:aminoacyl-tRNA synthetase (aaRS) interactions are a promising target for anti-trypanosomal drug discovery. From a marine natural products extract library, we identified several fractions with inhibitory activity toward Leishmania major alanyl-tRNA synthetase (AlaRS) but no activity against the human homolog. These marine natural products extracts showed cross-reactivity towards Trypanosoma cruzi AlaRS indicating the broad-spectrum potential of our network predictions. We also identified Leishmania major threonyl-tRNA synthetase (ThrRS) inhibitors from the same library. We discuss why chemotherapies targeting multiple aaRSs should be less prone to the evolution of resistance than monotherapeutic or synergistic combination chemotherapies targeting only one aaRS.


Subject(s)
Alanine-tRNA Ligase/antagonists & inhibitors , Antiprotozoal Agents/pharmacology , Enzyme Inhibitors/pharmacology , Leishmania/enzymology , Protozoan Proteins/antagonists & inhibitors , Threonine-tRNA Ligase/antagonists & inhibitors , Trypanosoma/drug effects , Alanine-tRNA Ligase/genetics , Alanine-tRNA Ligase/metabolism , Antiprotozoal Agents/chemistry , Enzyme Inhibitors/chemistry , Humans , Leishmania/drug effects , Leishmania/genetics , Leishmaniasis/parasitology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Threonine-tRNA Ligase/genetics , Threonine-tRNA Ligase/metabolism , Trypanosoma/enzymology , Trypanosoma/genetics , Trypanosomiasis/parasitology
15.
Eur J Med Chem ; 187: 111941, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31821989

ABSTRACT

Threonyl-tRNA synthetase (ThrRS) is a key member of the aminoacyl-tRNA synthetase (aaRS) family that plays essential roles in protein biosynthesis, and ThrRS inhibitors have potential in the therapy of multiple diseases, such as microbial infections and cancers. Based on a unique tRNA-amino acid dual-site inhibitory mechanism identified recently with the herb-derived prolyl-tRNA synthetase (ProRS) inhibitor halofuginone (HF), a series of compounds have been designed and synthesized by employing a fragment-based target hopping approach to simultaneously target the tRNAThr and l-threonine binding pockets of ThrRS. Among them, compound 30d showed an IC50 value of 1.4 µM against Salmonella enterica ThrRS (SeThrRS) and MIC values of 16-32 µg/mL against the tested bacterial strains. The cocrystal structure of SeThrRS in complex with 30d was determined at high resolution, revealing that 30d simultaneously occupies both binding pockets for the nucleotide A76 of tRNAThr and l-threonine in an ATP-independent manner, a novel mechanism compared to all other reported ThrRS inhibitors. Our study provides a new class of ThrRS inhibitors, and more importantly, it presents the first experimental evidence that the tRNA-amino acid dual-site inhibitory mechanism could apply to other aaRSs beyond ProRS, thus providing great opportunities for designing new mechanistic inhibitors for aaRS-based therapeutics.


Subject(s)
Drug Discovery , RNA, Transfer, Amino Acid-Specific/pharmacology , Threonine-tRNA Ligase/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , RNA, Transfer, Amino Acid-Specific/chemical synthesis , RNA, Transfer, Amino Acid-Specific/chemistry , Salmonella enterica/enzymology , Structure-Activity Relationship , Threonine-tRNA Ligase/metabolism
16.
ACS Chem Biol ; 14(12): 2663-2671, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31675206

ABSTRACT

To meet the ever-growing demands of antibiotic discovery, new chemical matter and antibiotic targets are urgently needed. Many potent natural product antibiotics which were previously discarded can also provide lead molecules and drug targets. One such example is the structurally unique ß-lactone obafluorin, produced by Pseudomonas fluorescens ATCC 39502. Obafluorin is active against both Gram-positive and -negative pathogens; however, the biological target was unknown. We now report that obafluorin targets threonyl-tRNA synthetase, and we identify a homologue, ObaO, which confers immunity to the obafluorin producer. Disruption of obaO in P. fluorescens ATCC 39502 results in obafluorin sensitivity, whereas expression in sensitive E. coli strains confers resistance. Enzyme assays demonstrate that E. coli threonyl-tRNA synthetase is fully inhibited by obafluorin, whereas ObaO is only partly susceptible, exhibiting a very unusual partial inhibition mechanism. Altogether, our data highlight the utility of an immunity-guided approach for the identification of an antibiotic target de novo and will ultimately enable the generation of improved obafluorin variants.


Subject(s)
Anti-Bacterial Agents/metabolism , Lactones/metabolism , Threonine-tRNA Ligase/metabolism , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Lactones/pharmacology , Microbial Sensitivity Tests
17.
Nucleic Acids Res ; 47(16): 8662-8674, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31287872

ABSTRACT

A typical feature of eukaryotic aminoacyl-tRNA synthetases (aaRSs) is the evolutionary gain of domains at either the N- or C-terminus, which frequently mediating protein-protein interaction. TARSL2 (mouse Tarsl2), encoding a threonyl-tRNA synthetase-like protein (ThrRS-L), is a recently identified aaRS-duplicated gene in higher eukaryotes, with canonical functions in vitro, which exhibits a different N-terminal extension (N-extension) from TARS (encoding ThrRS). We found the first half of the N-extension of human ThrRS-L (hThrRS-L) is homologous to that of human arginyl-tRNA synthetase. Using the N-extension as a probe in a yeast two-hybrid screening, AIMP1/p43 was identified as an interactor with hThrRS-L. We showed that ThrRS-L is a novel component of the mammalian multiple tRNA synthetase complex (MSC), and is reliant on two leucine zippers in the N-extension for MSC-incorporation in humans, and mouse cell lines and muscle tissue. The N-extension was sufficient to target a foreign protein into the MSC. The results from a Tarsl2-deleted cell line showed that it does not mediate MSC integrity. The effect of phosphorylation at various sites of hThrRS-L on its MSC-targeting is also explored. In summary, we revealed that ThrRS-L is a bona fide component of the MSC, which is mediated by a newly evolved N-extension domain.


Subject(s)
Arginine-tRNA Ligase/genetics , Cytokines/genetics , Multienzyme Complexes/genetics , Neoplasm Proteins/genetics , RNA-Binding Proteins/genetics , Threonine-tRNA Ligase/genetics , Amino Acid Sequence , Animals , Arginine-tRNA Ligase/metabolism , Cloning, Molecular , Cytokines/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HEK293 Cells , Humans , Leucine Zippers , Mice , Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Neoplasm Proteins/metabolism , Phosphorylation , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , RNA-Binding Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Threonine-tRNA Ligase/metabolism , Two-Hybrid System Techniques
18.
Nat Commun ; 10(1): 1357, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30902983

ABSTRACT

A fundamental question in biology is how vertebrates evolved and differ from invertebrates, and little is known about differences in the regulation of translation in the two systems. Herein, we identify a threonyl-tRNA synthetase (TRS)-mediated translation initiation machinery that specifically interacts with eIF4E homologous protein, and forms machinery that is structurally analogous to the eIF4F-mediated translation initiation machinery via the recruitment of other translation initiation components. Biochemical and RNA immunoprecipitation analyses coupled to sequencing suggest that this machinery emerged as a gain-of-function event in the vertebrate lineage, and it positively regulates the translation of mRNAs required for vertebrate development. Collectively, our findings demonstrate that TRS evolved to regulate vertebrate translation initiation via its dual role as a scaffold for the assembly of initiation components and as a selector of target mRNAs. This work highlights the functional significance of aminoacyl-tRNA synthetases in the emergence and control of higher order organisms.


Subject(s)
Peptide Chain Initiation, Translational , Threonine-tRNA Ligase/metabolism , Amino Acid Sequence , Animals , Blood Vessels/growth & development , Blood Vessels/metabolism , Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4F/metabolism , Eukaryotic Initiation Factor-4G/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Inbred C57BL , Protein Binding , RNA Cap-Binding Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Species Specificity , Threonine-tRNA Ligase/chemistry , Vertebrates/growth & development , Vertebrates/metabolism , Zebrafish
19.
PLoS Negl Trop Dis ; 12(6): e0006575, 2018 06.
Article in English | MEDLINE | ID: mdl-29897900

ABSTRACT

BACKGROUND: Aminoacyl tRNA synthetases are central enzymes required for protein synthesis. These enzymes are the known drug targets in bacteria and fungi. Here, we for the first time report the functional characterization of threonyl tRNA synthetase (LdThrRS) of Leishmania donovani, a protozoan parasite, the primary causative agent of visceral leishmaniasis. METHODOLOGY: Recombinant LdThrRS (rLdThrRS) was expressed in E. coli and purified. The kinetic parameters for rLdThrRS were determined. The subcellular localization of LdThrRS was done by immunofluorescence analysis. Heterozygous mutants of LdThrRS were generated in Leishmania promastigotes. These genetically manipulated parasites were checked for their proliferation, virulence, aminoacylation activity and sensitivity to the known ThrRS inhibitor, borrelidin. An in silico generated structural model of L. donovani ThrRS was compared to that of human. CONCLUSIONS: Recombinant LdThrRS displayed aminoacylation activity, and the protein is possibly localized to both the cytosol and mitochondria. The comparison of the 3D-model of LdThrRS to human ThrRS displayed considerable similarity. Heterozygous parasites showed restrictive growth phenotype and had attenuated infectivity. These heterozygous parasites were more susceptible to inhibition by borrelidin. Several attempts to obtain ThrRS homozygous null mutants were not successful, indicating its essentiality for the Leishmania parasite. Borrelidin showed a strong affinity for LdThrRS (KD: 0.04 µM) and was effective in inhibiting the aminoacylation activity of the rLdThrRS (IC50: 0.06 µM). Borrelidin inhibited the promastigotes (IC50: 21 µM) stage of parasites. Our data shows that LdThrRS is essential for L. donovani survival and is likely to bind with small drug-like molecules with strong affinity, thus making it a potential target for drug discovery efforts.


Subject(s)
Leishmania donovani/enzymology , Leishmaniasis, Visceral/parasitology , Threonine-tRNA Ligase/genetics , Drug Delivery Systems , Escherichia coli/enzymology , Escherichia coli/genetics , Fatty Alcohols/pharmacology , Gene Expression , Humans , Leishmania donovani/drug effects , Leishmania donovani/genetics , Leishmania donovani/pathogenicity , Organisms, Genetically Modified , Phylogeny , Protein Domains , Protein Transport , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Protozoan Proteins/isolation & purification , Protozoan Proteins/metabolism , Recombinant Proteins , Sequence Deletion , Threonine-tRNA Ligase/antagonists & inhibitors , Threonine-tRNA Ligase/isolation & purification , Threonine-tRNA Ligase/metabolism
20.
Nucleic Acids Res ; 46(9): 4662-4676, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29648639

ABSTRACT

Six pathogenic mutations have been reported in human mitochondrial tRNAThr (hmtRNAThr); however, the pathogenic molecular mechanism remains unclear. Previously, we established an activity assay system for human mitochondrial threonyl-tRNA synthetase (hmThrRS). In the present study, we surveyed the structural and enzymatic effects of pathogenic mutations in hmtRNAThr and then focused on m.15915 G > A (G30A) and m.15923A > G (A38G). The harmful evolutionary gain of non-Watson-Crick base pair A29/C41 caused hmtRNAThr to be highly susceptible to mutations disrupting the G30-C40 base pair in various ways; for example, structural integrity maintenance, modification and aminoacylation of tRNAThr, and editing mischarged tRNAThr. A similar phenomenon was observed for hmtRNATrp with an A29/C41 non-Watson-Crick base pair, but not in bovine mtRNAThr with a natural G29-C41 base pair. The A38G mutation caused a severe reduction in Thr-acceptance and editing of hmThrRS. Importantly, A38 is a nucleotide determinant for the t6A modification at A37, which is essential for the coding properties of hmtRNAThr. In summary, our results revealed the crucial role of the G30-C40 base pair in maintaining the proper structure and function of hmtRNAThr because of A29/C41 non-Watson-Crick base pair and explained the molecular outcome of pathogenic G30A and A38G mutations.


Subject(s)
Mutation , RNA, Mitochondrial/chemistry , RNA, Transfer, Thr/chemistry , Anticodon , Base Pairing , Humans , Mitochondria/enzymology , RNA Editing , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism , RNA, Transfer, Thr/genetics , RNA, Transfer, Thr/metabolism , Threonine-tRNA Ligase/metabolism , Transfer RNA Aminoacylation
SELECTION OF CITATIONS
SEARCH DETAIL
...