Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Aminoacyl-tRNA synthetases as therapeutic targets

Abstract

Aminoacyl-tRNA synthetases (ARSs) are essential enzymes for protein synthesis with evolutionarily conserved enzymatic mechanisms. Despite their similarity across organisms, scientists have been able to generate effective anti-infective agents based on the structural differences in the catalytic clefts of ARSs from pathogens and humans. However, recent genomic, proteomic and functionomic advances have unveiled unexpected disease-associated mutations and altered expression, secretion and interactions in human ARSs, revealing hidden biological functions beyond their catalytic roles in protein synthesis. These studies have also brought to light their potential as a rich and unexplored source for new therapeutic targets and agents through multiple avenues, including direct targeting of the catalytic sites, controlling disease-associated protein–protein interactions and developing novel biologics from the secreted ARS proteins or their parts. This Review addresses the emerging biology and therapeutic applications of human ARSs in diseases including autoimmune and rare diseases, and cancer.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Catalysis and architecture of the ARS catalytic site.
Fig. 2: The metamorphosis of ARSs for functional expansion.
Fig. 3: Disease specificity of ARSN.
Fig. 4: Multiple routes for drug development from unique ARS activities.
Fig. 5: ARSN in human body fluids.

Similar content being viewed by others

References

  1. Kim, S., You, S. & Hwang, D. Aminoacyl-tRNA synthetases and tumorigenesis: more than housekeeping. Nat. Rev. Cancer 11, 708–718 (2011). This is a comprehensive and analytical Review on the relationship between ARSs and cancer.

    CAS  PubMed  Google Scholar 

  2. Yao, P. & Fox, P. L. Aminoacyl-tRNA synthetases in medicine and disease. EMBO Mol. Med. 5, 332–343 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Fang, P. & Guo, M. Evolutionary limitation and opportunities for developing tRNA synthetase inhibitors with 5-binding-mode classification. Life (Basel) 5, (1703–1725 (2015).

    Google Scholar 

  4. Hurdle, J. G., O’Neill, A. J. & Chopra, I. Prospects for aminoacyl-tRNA synthetase inhibitors as new antimicrobial agents. Antimicrob. Agents Chemother. 49, 4821–4833 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Schimmel, P. The emerging complexity of the tRNA world: mammalian tRNAs beyond protein synthesis. Nat. Rev. Mol. Cell Biol. 19, 45–58 (2018).

    CAS  PubMed  Google Scholar 

  6. Bullwinkle, T. J. & Ibba, M. Emergence and evolution. Top. Curr. Chem. 344, 43–87 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Perona, J. J. & Gruic-Sovulj, I. Synthetic and editing mechanisms of aminoacyl-tRNA synthetases. Top. Curr. Chem. 344, 1–41 (2014).

    CAS  PubMed  Google Scholar 

  8. Giege, R. & Springer, M. Aminoacyl-tRNA synthetases in the bacterial world. EcoSal Plus https://doi.org/10.1128/ecosalplus.ESP-0002-2016 (2016).

    Article  PubMed  Google Scholar 

  9. Lee, J. W. et al. Editing-defective tRNA synthetase causes protein misfolding and neurodegeneration. Nature 443, 50–55 (2006).

    CAS  PubMed  Google Scholar 

  10. Ribas de Pouplana, L. & Schimmel, P. Aminoacyl-tRNA synthetases: potential markers of genetic code development. Trends Biochem. Sci. 26, 591–596 (2001). This study presents the route of catalytic evolution of ARSs.

    CAS  PubMed  Google Scholar 

  11. Eriani, G., Delarue, M., Poch, O., Gangloff, J. & Moras, D. Partition of tRNA synthetases into two classes based on mutually exclusive sets of sequence motifs. Nature 347, 203–206 (1990).

    CAS  PubMed  Google Scholar 

  12. Newberry, K. J., Hou, Y. M. & Perona, J. J. Structural origins of amino acid selection without editing by cysteinyl-tRNA synthetase. EMBO J. 21, 2778–2787 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Nureki, O. et al. Architectures of class-defining and specific domains of glutamyl-tRNA synthetase. Science 267, 1958–1965 (1995).

    CAS  PubMed  Google Scholar 

  14. Brick, P., Bhat, T. N. & Blow, D. M. Structure of tyrosyl-tRNA synthetase refined at 2.3 A resolution. Interaction of the enzyme with the tyrosyl adenylate intermediate. J. Mol. Biol. 208, 83–98 (1989).

    CAS  PubMed  Google Scholar 

  15. Schmidt, E. & Schimmel, P. Residues in a class I tRNA synthetase which determine selectivity of amino acid recognition in the context of tRNA. Biochemistry 34, 11204–11210 (1995).

    CAS  PubMed  Google Scholar 

  16. Palencia, A. et al. Structural dynamics of the aminoacylation and proofreading functional cycle of bacterial leucyl-tRNA synthetase. Nat. Struct. Mol. Biol. 19, 677–684 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Guo, M. et al. The C-Ala domain brings together editing and aminoacylation functions on one tRNA. Science 325, 744–747 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Delagoutte, B., Moras, D. & Cavarelli, J. tRNA aminoacylation by arginyl-tRNA synthetase: induced conformations during substrates binding. EMBO J. 19, 5599–5610 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Beuning, P. J. & Musier-Forsyth, K. Species-specific differences in amino acid editing by class II prolyl-tRNA synthetase. J. Biol. Chem. 276, 30779–30785 (2001).

    CAS  PubMed  Google Scholar 

  20. Guo, M., Yang, X. L. & Schimmel, P. New functions of aminoacyl-tRNA synthetases beyond translation. Nat. Rev. Mol. Cell Biol. 11, 668–674 (2010). This Review discusses the non-catalytic evolution of ARSs and AIMPs.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Fournier, G. P., Andam, C. P., Alm, E. J. & Gogarten, J. P. Molecular evolution of aminoacyl tRNA synthetase proteins in the early history of life. Orig. Life Evol. Biosph. 41, 621–632 (2011).

    CAS  PubMed  Google Scholar 

  22. Beebe, K., Ribas De Pouplana, L. & Schimmel, P. Elucidation of tRNA-dependent editing by a class II tRNA synthetase and significance for cell viability. EMBO J. 22, 668–675 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Sasaki, H. M. et al. Structural and mutational studies of the amino acid-editing domain from archaeal/eukaryal phenylalanyl-tRNA synthetase. Proc. Natl Acad. Sci. USA 103, 14744–14749 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Guo, M. & Yang, X. L. Architecture and metamorphosis. Top. Curr. Chem. 344, 89–118 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Schimmel, P. & Ribas De Pouplana, L. Footprints of aminoacyl-tRNA synthetases are everywhere. Trends Biochem. Sci. 25, 207–209 (2000).

    CAS  PubMed  Google Scholar 

  26. Cen, S., Javanbakht, H., Niu, M. & Kleiman, L. Ability of wild-type and mutant lysyl-tRNA synthetase to facilitate tRNA(Lys) incorporation into human immunodeficiency virus type 1. J. Virol. 78, 1595–1601 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Kim, D. G. et al. Interaction of two translational components, lysyl-tRNA synthetase and p40/37LRP, in plasma membrane promotes laminin-dependent cell migration. FASEB J. 26, 4142–4159 (2012).

    CAS  PubMed  Google Scholar 

  28. Kim, D. G. et al. Chemical inhibition of prometastatic lysyl-tRNA synthetase-laminin receptor interaction. Nat. Chem. Biol. 10, 29–34 (2014).

    CAS  PubMed  Google Scholar 

  29. Fu, Y. et al. Structure of the ArgRS-GlnRS-AIMP1 complex and its implications for mammalian translation. Proc. Natl Acad. Sci. USA 111, 15084–15089 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Wakasugi, K. & Schimmel, P. Two distinct cytokines released from a human aminoacyl-tRNA synthetase. Science 284, 147–151 (1999). This study demonstrates the function of secreted YRSs working as cytokines.

    CAS  PubMed  Google Scholar 

  31. Park, S. G., Choi, E. C. & Kim, S. Aminoacyl-tRNA synthetase-interacting multifunctional proteins (AIMPs): a triad for cellular homeostasis. IUBMB Life 62, 296–302 (2010).

    CAS  PubMed  Google Scholar 

  32. Kim, D., Kwon, N. H. & Kim, S. Association of aminoacyl-tRNA synthetases with cancer. Top. Curr. Chem. 344, 207–245 (2014).

    CAS  PubMed  Google Scholar 

  33. Cho, H. Y. et al. Assembly of multi-tRNA synthetase complex via heterotetrameric glutathione transferase-homology domains. J. Biol. Chem. 290, 29313–29328 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Arif, A. et al. Two-site phosphorylation of EPRS coordinates multimodal regulation of noncanonical translational control activity. Mol. Cell 35, 164–180 (2009). This study demonstrates the role of phosphorylation on the relocalization and novel function of EPRS.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Jia, J., Arif, A., Ray, P. S. & Fox, P. L. WHEP domains direct noncanonical function of glutamyl-Prolyl tRNA synthetase in translational control of gene expression. Mol. Cell 29, 679–690 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Sajish, M. et al. Trp-tRNA synthetase bridges DNA-PKcs to PARP-1 to link IFN-gamma and p53 signaling. Nat. Chem. Biol. 8, 547–554 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Ahn, Y. H. et al. Secreted tryptophanyl-tRNA synthetase as a primary defence system against infection. Nat. Microbiol. 2, 16191 (2016).

    CAS  PubMed  Google Scholar 

  38. Han, J. M. et al. Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway. Cell 149, 410–424 (2012).

    CAS  PubMed  Google Scholar 

  39. Bonfils, G. et al. Leucyl-tRNA synthetase controls TORC1 via the EGO complex. Mol. Cell. 46, 105–110 (2012).

    CAS  PubMed  Google Scholar 

  40. Xu, X. et al. Unique domain appended to vertebrate tRNA synthetase is essential for vascular development. Nat. Commun. 3, 681 (2012).

    PubMed  Google Scholar 

  41. Lo, W. S. et al. Human tRNA synthetase catalytic nulls with diverse functions. Science 345, 328–332 (2014). This study presents the diverse splicing variants of ARSs identified by omics studies.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Choi, J. W. et al. Cancer-associated splicing variant of tumor suppressor AIMP2/p38: pathological implication in tumorigenesis. PLOS Genet. 7, e1001351 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Xu, Z. et al. Internally deleted human tRNA synthetase suggests evolutionary pressure for repurposing. Structure 20, 1470–1477 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Kanaji, T. et al. Tyrosyl-tRNA synthetase stimulates thrombopoietin-independent hematopoiesis accelerating recovery from thrombocytopenia. Proc. Natl Acad. Sci. USA 115, E8228–E8235 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Tolstrup, A. B., Bejder, A., Fleckner, J. & Justesen, J. Transcriptional regulation of the interferon-gamma-inducible tryptophanyl-tRNA synthetase includes alternative splicing. J. Biol. Chem. 270, 397–403 (1995).

    CAS  PubMed  Google Scholar 

  46. Kim, J. E. et al. An elongation factor-associating domain is inserted into human cysteinyl-tRNA synthetase by alternative splicing. Nucleic Acids Res. 28, 2866–2872 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Yao, P. et al. Coding region polyadenylation generates a truncated tRNA synthetase that counters translation repression. Cell 149, 88–100 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Kim, D. G. et al. Oncogenic mutation of AIMP2/p38 inhibits its tumor-suppressive interaction with Smurf2. Cancer Res. 76, 3422–3436 (2016).

    CAS  PubMed  Google Scholar 

  49. Ofir-Birin, Y. et al. Structural switch of lysyl-tRNA synthetase between translation and transcription. Mol. Cell. 49, 30–42 (2013).

    CAS  PubMed  Google Scholar 

  50. Nam, S. H. et al. Lysyl-tRNA synthetase-expressing colon spheroids induce M2 macrophage polarization to promote metastasis. J. Clin. Invest. 128, 5034–5055 (2018).

    PubMed  PubMed Central  Google Scholar 

  51. Arif, A. et al. EPRS is a critical mTORC1-S6K1 effector that influences adiposity in mice. Nature 542, 357–361 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Lee, E. Y. et al. Infection-specific phosphorylation of glutamyl-prolyl tRNA synthetase induces antiviral immunity. Nat. Immunol. 17, 1252–1262 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Kwon, N. H. et al. Dual role of methionyl-tRNA synthetase in the regulation of translation and tumor suppressor activity of aminoacyl-tRNA synthetase-interacting multifunctional protein-3. Proc. Natl Acad. Sci. USA 108, 19635–19640 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Lee, J. Y. et al. Promiscuous methionyl-tRNA synthetase mediates adaptive mistranslation to protect cells against oxidative stress. J. Cell Sci. 127, 4234–4245 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Luo, S. & Levine, R. L. Methionine in proteins defends against oxidative stress. FASEB J. 23, 464–472 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Otani, A. et al. A fragment of human TrpRS as a potent antagonist of ocular angiogenesis. Proc. Natl Acad. Sci. USA 99, 178–183 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Tzima, E. et al. VE-cadherin links tRNA synthetase cytokine to anti-angiogenic function. J. Biol. Chem. 280, 2405–2408 (2005).

    CAS  PubMed  Google Scholar 

  58. Vo, M. N., Yang, X. L. & Schimmel, P. Dissociating quaternary structure regulates cell-signaling functions of a secreted human tRNA synthetase. J. Biol. Chem. 286, 11563–11568 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Kim, S. B. et al. Caspase-8 controls the secretion of inflammatory lysyl-tRNA synthetase in exosomes from cancer cells. J. Cell. Biol. 216, 2201–2216 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Vo, M. N. et al. ANKRD16 prevents neuron loss caused by an editing-defective tRNA synthetase. Nature 557, 510–515 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhou, Q. et al. Orthogonal use of a human tRNA synthetase active site to achieve multifunctionality. Nat. Struct. Mol. Biol. 17, 57–61 (2010).

    CAS  PubMed  Google Scholar 

  62. Sajish, M. & Schimmel, P. A human tRNA synthetase is a potent PARP1-activating effector target for resveratrol. Nature 519, 370–373 (2015).

    CAS  PubMed  Google Scholar 

  63. Uhlen, M. et al. A pathology atlas of the human cancer transcriptome. Science 357, eaan2507 (2017).

    PubMed  Google Scholar 

  64. Park, B. J. et al. The haploinsufficient tumor suppressor p18 upregulates p53 via interactions with ATM/ATR. Cell 120, 209–221 (2005).

    CAS  PubMed  Google Scholar 

  65. Park, B. J. et al. AIMP3 haploinsufficiency disrupts oncogene-induced p53 activation and genomic stability. Cancer Res. 66, 6913–6918 (2006).

    CAS  PubMed  Google Scholar 

  66. Choi, J. W., Um, J. Y., Kundu, J. K., Surh, Y. J. & Kim,  S. Multidirectional tumor-suppressive activity of AIMP2/p38 and the enhanced susceptibility of AIMP2 heterozygous mice to carcinogenesis. Carcinogenesis 30, 1638–1644 (2009).

    CAS  PubMed  Google Scholar 

  67. Thul, P. J. & Lindskog, C. The human protein atlas: a spatial map of the human proteome. Protein Sci. 27, 233–244 (2018).

    CAS  PubMed  Google Scholar 

  68. Kim, E. Y., Jung, J. Y., Kim, A., Kim, K. & Chang, Y. S. Methionyl-tRNA synthetase overexpression is associated with poor clinical outcomes in non-small cell lung cancer. BMC Cancer 17, 467 (2017).

    PubMed  PubMed Central  Google Scholar 

  69. Forus, A., Florenes, V. A., Maelandsmo, G. M., Fodstad, O. & Myklebost, O. The protooncogene CHOP/GADD153, involved in growth arrest and DNA damage response, is amplified in a subset of human sarcomas. Cancer Genet. Cytogenet. 78, 165–171 (1994).

    CAS  PubMed  Google Scholar 

  70. Nilbert, M., Rydholm, A., Mitelman, F., Meltzer, P. S. & Mandahl, N. Characterization of the 12q13-15 amplicon in soft tissue tumors. Cancer Genet. Cytogenet. 83, 32–36 (1995).

    CAS  PubMed  Google Scholar 

  71. Palmer, J. L., Masui, S., Pritchard, S., Kalousek, D. K. & Sorensen, P. H. Cytogenetic and molecular genetic analysis of a pediatric pleomorphic sarcoma reveals similarities to adult malignant fibrous histiocytoma. Cancer Genet. Cytogenet. 95, 141–147 (1997).

    CAS  PubMed  Google Scholar 

  72. Reifenberger, G. et al. Refined mapping of 12q13-q15 amplicons in human malignant gliomas suggests CDK4/SAS and MDM2 as independent amplification targets. Cancer Res. 56, 5141–5145 (1996).

    CAS  PubMed  Google Scholar 

  73. Vellaichamy, A. et al. Proteomic interrogation of androgen action in prostate cancer cells reveals roles of aminoacyl tRNA synthetases. PLOS ONE 4, e7075 (2009).

    PubMed  PubMed Central  Google Scholar 

  74. Wellman, T. L. et al. Threonyl-tRNA synthetase overexpression correlates with angiogenic markers and progression of human ovarian cancer. BMC Cancer 14, 620 (2014).

    PubMed  PubMed Central  Google Scholar 

  75. Jeong, S. J. et al. Inhibition of MUC1 biosynthesis via threonyl-tRNA synthetase suppresses pancreatic cancer cell migration. Exp. Mol. Med. 50, e424 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Lee, C. W. et al. Overexpressed tryptophanyl-tRNA synthetase, an angiostatic protein, enhances oral cancer cell invasiveness. Oncotarget 6, 21979–21992 (2015).

    PubMed  PubMed Central  Google Scholar 

  77. Chi, L. M. et al. Enhanced interferon signaling pathway in oral cancer revealed by quantitative proteome analysis of microdissected specimens using 16O/18O labeling and integrated two-dimensional LC-ESI-MALDI tandem MS. Mol. Cell. Proteomics 8, 1453–1474 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Liu, J., Shue, E., Ewalt, K. L. & Schimmel, P. A new gamma-interferon-inducible promoter and splice variants of an anti-angiogenic human tRNA synthetase. Nucleic Acids Res. 32, 719–727 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Turpaev, K. T. et al. Alternative processing of the tryptophanyl-tRNA synthetase mRNA from interferon-treated human cells. Eur. J. Biochem. 240, 732–737 (1996).

    CAS  PubMed  Google Scholar 

  80. Koscielny, G. et al. Open Targets: a platform for therapeutic target identification and validation. Nucleic Acids Res. 45, D985–D994 (2017).

    CAS  PubMed  Google Scholar 

  81. Santos-Cortez, R. L. et al. Mutations in KARS, encoding lysyl-tRNA synthetase, cause autosomal-recessive nonsyndromic hearing impairment DFNB89. Am. J. Hum. Genet. 93, 132–140 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Garbern, J. Y. Pelizaeus-Merzbacher disease: genetic and cellular pathogenesis. Cell. Mol. Life Sci. 64, 50–65 (2007).

    CAS  PubMed  Google Scholar 

  83. Nafisinia, M. et al. Mutations in RARS cause a hypomyelination disorder akin to Pelizaeus-Merzbacher disease. Eur. J. Hum. Genet. 25, 1134–1141 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Mendes, M. I. et al. Bi-allelic mutations in EPRS, encoding the glutamyl-prolyl-aminoacyl-tRNA Synthetase, cause a hypomyelinating leukodystrophy. Am. J. Hum. Genet. 102, 676–684 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Wolf, N. I. et al. Mutations in RARS cause hypomyelination. Ann. Neurol. 76, 134–139 (2014).

    CAS  PubMed  Google Scholar 

  86. Shukla, A. et al. Homozygosity for a nonsense variant in AIMP2 is associated with a progressive neurodevelopmental disorder with microcephaly, seizures, and spastic quadriparesis. J. Hum. Genet. 63, 19–25 (2018).

    CAS  PubMed  Google Scholar 

  87. Iqbal, Z. et al. Missense variants in AIMP1 gene are implicated in autosomal recessive intellectual disability without neurodegeneration. Eur. J. Hum. Genet. 24, 392–399 (2016).

    CAS  PubMed  Google Scholar 

  88. Zhu, X. et al. MSC p43 required for axonal development in motor neurons. Proc. Natl Acad. Sci. USA 106, 15944–15949 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Xu, H., Malinin, N. L., Awasthi, N., Schwarz, R. E. & Schwarz, M. A. The N terminus of pro-endothelial monocyte-activating polypeptide II (EMAP II) regulates its binding with the C terminus, arginyl-tRNA synthetase, and neurofilament light protein. J. Biol. Chem. 290, 9753–9766 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Simons, C. et al. Loss-of-function alanyl-tRNA synthetase mutations cause an autosomal-recessive early-onset epileptic encephalopathy with persistent myelination defect. Am. J. Hum. Genet. 96, 675–681 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Casey, J. P. et al. Clinical and genetic characterisation of infantile liver failure syndrome type 1, due to recessive mutations in LARS. J. Inherit. Metab. Dis. 38, 1085–1092 (2015).

    CAS  PubMed  Google Scholar 

  92. van Meel, E. et al. Rare recessive loss-of-function methionyl-tRNA synthetase mutations presenting as a multi-organ phenotype. BMC Med. Genet. 14, 106 (2013).

    PubMed  PubMed Central  Google Scholar 

  93. Kopajtich, R. et al. Biallelic IARS mutations cause growth retardation with prenatal onset, intellectual disability, muscular hypotonia, and infantile hepatopathy. Am. J. Hum. Genet. 99, 414–422 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Puffenberger, E. G. et al. Genetic mapping and exome sequencing identify variants associated with five novel diseases. PLOS ONE 7, e28936 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Zhang, X. et al. Mutations in QARS, encoding glutaminyl-tRNA synthetase, cause progressive microcephaly, cerebral-cerebellar atrophy, and intractable seizures. Am. J. Hum. Genet. 94, 547–558 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Xu, Z. et al. Bi-allelic mutations in phe-tRNA synthetase associated with a multi-system pulmonary disease support non-translational function. Am. J. Hum. Genet. 103, 100–114 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Antonellis, A. et al. Compound heterozygosity for loss-of-function FARSB variants in a patient with classic features of recessive aminoacyl-tRNA synthetase-related disease. Hum. Mutat. 39, 834–840 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Sissler, M., Gonzalez-Serrano, L. E. & Westhof, E. Recent advances in mitochondrial aminoacyl-tRNA synthetases and disease. Trends Mol. Med. 23, 693–708 (2017).

    CAS  PubMed  Google Scholar 

  99. Schwenzer, H., Zoll, J., Florentz, C. & Sissler, M. Pathogenic implications of human mitochondrial aminoacyl-tRNA synthetases. Top. Curr. Chem. 344, 247–292 (2014).

    CAS  PubMed  Google Scholar 

  100. Datt, M. & Sharma, A. Evolutionary and structural annotation of disease-associated mutations in human aminoacyl-tRNA synthetases. BMC Genomics 15, 1063 (2014).

    PubMed  PubMed Central  Google Scholar 

  101. Motley, W. W., Talbot, K. & Fischbeck, K. H. GARS axonopathy: not every neuron’s cup of tRNA. Trends Neurosci. 33, 59–66 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Storkebaum, E. Peripheral neuropathy via mutant tRNA synthetases: Inhibition of protein translation provides a possible explanation. Bioessays 38, 818–829 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. He, W. et al. CMT2D neuropathy is linked to the neomorphic binding activity of glycyl-tRNA synthetase. Nature 526, 710–714 (2015). This study demonstrates the gain-of-function mutation in GRS and its role in disease development.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Schwarz, Q. et al. Vascular endothelial growth factor controls neuronal migration and cooperates with Sema3A to pattern distinct compartments of the facial nerve. Genes Dev. 18, 2822–2834 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Mo, Z. et al. Aberrant GlyRS-HDAC6 interaction linked to axonal transport deficits in Charcot-Marie-Tooth neuropathy. Nat. Commun. 9, 1007 (2018).

    PubMed  PubMed Central  Google Scholar 

  106. Sleigh, J. N. et al. Trk receptor signaling and sensory neuron fate are perturbed in human neuropathy caused by Gars mutations. Proc. Natl Acad. Sci. USA 114, E3324–E3333 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Kunst, C. B., Mezey, E., Brownstein, M. J. & Patterson, D. Mutations in SOD1 associated with amyotrophic lateral sclerosis cause novel protein interactions. Nat. Genet. 15, 91–94 (1997).

    CAS  PubMed  Google Scholar 

  108. Kawamata, H., Magrane, J., Kunst, C., King, M. P. & Manfredi, G. Lysyl-tRNA synthetase is a target for mutant SOD1 toxicity in mitochondria. J. Biol. Chem. 283, 28321–28328 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Kwon, N. H. et al. Stabilization of cyclin-dependent kinase 4 by methionyl-tRNA synthetase in p16INK4a-negative cancer. ACS Pharmacol. Transl Sci. 1, 21–31 (2018). This study describes the little effect of reduced level of MRS on translation under normal conditions and the novel function of MRS in cell cycle regulation.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Kitada, T. et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 392, 605–608 (1998).

    CAS  PubMed  Google Scholar 

  111. Lee, Y. et al. Parthanatos mediates AIMP2-activated age-dependent dopaminergic neuronal loss. Nat. Neurosci. 16, 1392–1400 (2013). This study demonstrates the gain of function of AIMP2 mediated by the mutation in its binding partners and its relationship to disease phenotype.

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Ko, H. S. et al. Accumulation of the authentic parkin substrate aminoacyl-tRNA synthetase cofactor, p38/JTV-1, leads to catecholaminergic cell death. J. Neurosci. 25, 7968–7978 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. David, K. K., Andrabi, S. A., Dawson, T. M. & Dawson, V. L. Parthanatos, a messenger of death. Front. Biosci. (Landmark Ed.) 14, 1116–1128 (2009).

    CAS  Google Scholar 

  114. Choi, J. W. et al. AIMP2 promotes TNFalpha-dependent apoptosis via ubiquitin-mediated degradation of TRAF2. J. Cell Sci. 122, 2710–2715 (2009).

    CAS  PubMed  Google Scholar 

  115. Choi, J. W. et al. Splicing variant of AIMP2 as an effective target against chemoresistant ovarian cancer. J. Mol. Cell. Biol. 4, 164–173 (2012).

    PubMed  Google Scholar 

  116. Oh, A. Y. et al. Inhibiting DX2-p14/ARF interaction exerts antitumor effects in lung cancer and delays tumor progression. Cancer Res. 76, 4791–4804 (2016).

    CAS  PubMed  Google Scholar 

  117. Lega, J. C. et al. The clinical phenotype associated with myositis-specific and associated autoantibodies: a meta-analysis revisiting the so-called antisynthetase syndrome. Autoimmun. Rev. 13, 883–891 (2014).

    CAS  PubMed  Google Scholar 

  118. Cavagna, L. et al. Serum Jo-1 autoantibody and isolated arthritis in the antisynthetase syndrome: review of the literature and report of the experience of AENEAS Collaborative Group. Clin. Rev. Allergy Immunol. 52, 71–80 (2017).

    CAS  PubMed  Google Scholar 

  119. Zhou, J. J. et al. Secreted histidyl-tRNA synthetase splice variants elaborate major epitopes for autoantibodies in inflammatory myositis. J. Biol. Chem. 289, 19269–19275 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Park, M. C. et al. Secreted human glycyl-tRNA synthetase implicated in defense against ERK-activated tumorigenesis. Proc. Natl Acad. Sci. USA 109, E640–E647 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Fischer, A. et al. Anti-synthetase syndrome in ANA and anti-Jo-1 negative patients presenting with idiopathic interstitial pneumonia. Respir. Med. 103, 1719–1724 (2009).

    PubMed  PubMed Central  Google Scholar 

  122. Hughes, J. & Mellows, G. Interaction of pseudomonic acid A with Escherichia coli B isoleucyl-tRNA synthetase. Biochem. J. 191, 209–219 (1980).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Elewski, B. E. et al. Efficacy and safety of tavaborole topical solution, 5%, a novel boron-based antifungal agent, for the treatment of toenail onychomycosis: results from 2 randomized phase-III studies. J. Am. Acad. Dermatol. 73, 62–69 (2015). This study presents the results from two clinical trials assessing AN2690.

    CAS  PubMed  Google Scholar 

  124. Hui, X. et al. In vitro penetration of a novel oxaborole antifungal (AN2690) into the human nail plate. J. Pharm. Sci. 96, 2622–2631 (2007).

    CAS  PubMed  Google Scholar 

  125. Rock, F. L. et al. An antifungal agent inhibits an aminoacyl-tRNA synthetase by trapping tRNA in the editing site. Science 316, 1759–1761 (2007).

    CAS  PubMed  Google Scholar 

  126. Yao, P. et al. Unique residues crucial for optimal editing in yeast cytoplasmic Leucyl-tRNA synthetase are revealed by using a novel knockout yeast strain. J. Biol. Chem. 283, 22591–22600 (2008).

    CAS  PubMed  Google Scholar 

  127. Pang, Y. L. & Martinis, S. A. A paradigm shift for the amino acid editing mechanism of human cytoplasmic leucyl-tRNA synthetase. Biochemistry 48, 8958–8964 (2009).

    CAS  PubMed  Google Scholar 

  128. Palencia, A. et al. Cryptosporidium and toxoplasma parasites are inhibited by a benzoxaborole targeting leucyl-tRNA synthetase. Antimicrob. Agents Chemother. 60, 5817–5827 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Li, X. et al. Discovery of a potent and specific M. tuberculosis leucyl-tRNA synthetase inhibitor: (S)-3-(aminomethyl)-4-chloro-7-(2-hydroxyethoxy)benzo[c][1,2]oxaborol-1(3 H)-ol (GSK656). J. Med. Chem. 60, 8011–8026 (2017).

    CAS  PubMed  Google Scholar 

  130. Hernandez, V. et al. Discovery of a novel class of boron-based antibacterials with activity against gram-negative bacteria. Antimicrob. Agents Chemother. 57, 1394–1403 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Kato, N. et al. Diversity-oriented synthesis yields novel multistage antimalarial inhibitors. Nature 538, 344–349 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Keller, T. L. et al. Halofuginone and other febrifugine derivatives inhibit prolyl-tRNA synthetase. Nat. Chem. Biol. 8, 311–317 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Zhou, H., Sun, L., Yang, X. L. & Schimmel, P. ATP-directed capture of bioactive herbal-based medicine on human tRNA synthetase. Nature 494, 121–124 (2013). This study demonstrates the binding mode of halofuginone in PRS.

    CAS  PubMed  Google Scholar 

  134. Sundrud, M. S. et al. Halofuginone inhibits TH17 cell differentiation by activating the amino acid starvation response. Science 324, 1334–1338 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Park, J. S. et al. Inhibition of prolyl-tRNA Synthetase as a novel mediator of cardiac fibrosis [abstract]. Am. Heart Associ. 136 (Suppl. 1), A24036 (2017).

    Google Scholar 

  136. Fang, P. et al. Structural basis for full-spectrum inhibition of translational functions on a tRNA synthetase. Nat. Commun. 6, 6402 (2015). This study shows the structure-based interaction between borrelidin and TRS.

    CAS  PubMed  Google Scholar 

  137. Wang, X., Lan, H., Li, J., Su, Y. & Xu, L. Muc1 promotes migration and lung metastasis of melanoma cells. Am. J. Cancer Res. 5, 2590–2604 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Funahashi, Y. et al. Establishment of a quantitative mouse dorsal air sac model and its application to evaluate a new angiogenesis inhibitor. Oncol. Res. 11, 319–329 (1999).

    CAS  PubMed  Google Scholar 

  139. Taft, R. J. et al. Mutations in DARS cause hypomyelination with brain stem and spinal cord involvement and leg spasticity. Am. J. Hum. Genet. 92, 774–780 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Hu, J. et al. Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat. Biotechnol. 31, 522–529 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Dobbelstein, M. & Moll, U. Targeting tumour-supportive cellular machineries in anticancer drug development. Nat. Rev. Drug Discov. 13, 179–196 (2014).

    CAS  PubMed  Google Scholar 

  142. Luo, J., Solimini, N. L. & Elledge, S. J. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 136, 823–837 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Kim, J. H. et al. Control of leucine-dependent mTORC1 pathway through chemical intervention of leucyl-tRNA synthetase and RagD interaction. Nat. Commun. 8, 732 (2017). This study demonstrates how LRS inhibitors regulate mTORC1 signalling.

    PubMed  PubMed Central  Google Scholar 

  144. Bae, S. et al. in 2018 Fall International Convention of The Pharmaceutical Society of Korea P6-72 (The Pharmaceutical Society of Korea, 2018).

  145. Son, S. H., Park, M. C. & Kim, S. Extracellular activities of aminoacyl-tRNA synthetases: new mediators for cell-cell communication. Top. Curr. Chem. 344, 145–166 (2014).

    CAS  PubMed  Google Scholar 

  146. aTyr Pharma. ATYR1923: about ATYR1923. aTyrPharma https://www.atyrpharma.com/programs/atyr1923/ (2019).

  147. aTyr Pharma. Interstitial lung disease and the immune system: introduction to the iMod.Fc program. aTyrPharma https://investors.atyrpharma.com/static-files/f5cf2a36-e7a2-4bcb-8cac-189b08dc5f89 (2017).

  148. Australian New Zealand Clinical Trials Registry. A randomized, double-blind, placebo-controlled study to investigate the safety, tolerability, immunogenicity, pharmacokinetics and pharmacodynamics of single doses of intravenous ATYR1923 in healthy volunteers (registration number: ACTRN12617001446358). ANZCTR https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=373652 (2018).

  149. Albericio, F. & Kruger, H. G. Therapeutic peptides. Future Med. Chem. 4, 1527–1531 (2012).

    CAS  PubMed  Google Scholar 

  150. Fosgerau, K. & Hoffmann, T. Peptide therapeutics: current status and future directions. Drug. Discov. Today 20, 122–128 (2015).

    CAS  PubMed  Google Scholar 

  151. Han, J. M., Myung, H. & Kim, S. Antitumor activity and pharmacokinetic properties of ARS-interacting multi-functional protein 1 (AIMP1/p43). Cancer Lett. 287, 157–164 (2010).

    CAS  PubMed  Google Scholar 

  152. Lee, Y. S. et al. Antitumor activity of the novel human cytokine AIMP1 in an in vivo tumor model. Mol. Cells 21, 213–217 (2006).

    CAS  PubMed  Google Scholar 

  153. Park, S. G. et al. Dose-dependent biphasic activity of tRNA synthetase-associating factor, p43, in angiogenesis. J. Biol. Chem. 277, 45243–45248 (2002).

    CAS  PubMed  Google Scholar 

  154. Park, S. G. et al. Hormonal activity of AIMP1/p43 for glucose homeostasis. Proc. Natl Acad. Sci. USA 103, 14913–14918 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Park, S. G. et al. The novel cytokine p43 stimulates dermal fibroblast proliferation and wound repair. Am. J. Pathol. 166, 387–398 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Kim, S. Y. et al. ARS-interacting multi-functional protein 1 induces proliferation of human bone marrow-derived mesenchymal stem cells by accumulation of beta-catenin via fibroblast growth factor receptor 2-mediated activation of Akt. Stem Cells Dev. 22, 2630–2640 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Kwon, H. S. et al. Identification of CD23 as a functional receptor for the proinflammatory cytokine AIMP1/p43. J. Cell Sci. 125, 4620–4629 (2012).

    CAS  PubMed  Google Scholar 

  158. Hong, S. H. et al. The antibody atliximab attenuates collagen-induced arthritis by neutralizing AIMP1, an inflammatory cytokine that enhances osteoclastogenesis. Biomaterials 44, 45–54 (2015).

    CAS  PubMed  Google Scholar 

  159. Pines, M. & Spector, I. Halofuginone - the multifaceted molecule. Molecules 20, 573–594 (2015).

    PubMed  PubMed Central  Google Scholar 

  160. Neenan, T. X., Burrier, R. E. & Kim, S. Biocon’s target factory. Nat. Biotechnol. 36, 791–797 (2018).

    CAS  PubMed  Google Scholar 

  161. Beebe, K., Waas, W., Druzina, Z., Guo, M. & Schimmel, P. A universal plate format for increased throughput of assays that monitor multiple aminoacyl transfer RNA synthetase activities. Anal. Biochem. 368, 111–121 (2007).

    CAS  PubMed  Google Scholar 

  162. Cestari, I. & Stuart, K. A spectrophotometric assay for quantitative measurement of aminoacyl-tRNA synthetase activity. J. Biomol. Screen. 18, 490–497 (2013).

    PubMed  Google Scholar 

  163. Lloyd, A. J., Thomann, H. U., Ibba, M. & Soll, D. A broadly applicable continuous spectrophotometric assay for measuring aminoacyl-tRNA synthetase activity. Nucleic Acids Res. 23, 2886–2892 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Wu, M. X. & Hill, K. A. A continuous spectrophotometric assay for the aminoacylation of transfer RNA by alanyl-transfer RNA synthetase. Anal. Biochem. 211, 320–323 (1993).

    CAS  PubMed  Google Scholar 

  165. Brennan, J. D., Hogue, C. W., Rajendran, B., Willis, K. J. & Szabo, A. G. Preparation of enantiomerically pure L-7-azatryptophan by an enzymatic method and its application to the development of a fluorimetric activity assay for tryptophanyl-tRNA synthetase. Anal. Biochem. 252, 260–270 (1997).

    CAS  PubMed  Google Scholar 

  166. Kong, J. et al. High-throughput screening for protein synthesis inhibitors targeting aminoacyl-tRNA synthetases. SLAS Discov. 23, 174–182 (2018).

    CAS  PubMed  Google Scholar 

  167. Cochrane, R. V. K., Norquay, A. K. & Vederas, J. C. Natural products and their derivatives as tRNA synthetase inhibitors and antimicrobial agents. Medchemcomm 7, 1535–1545 (2016).

    CAS  Google Scholar 

  168. Han, J. M. et al. Identification of gp96 as a novel target for treatment of autoimmune disease in mice. PLOS ONE 5, e9792 (2010).

    PubMed  PubMed Central  Google Scholar 

  169. Kong, J., Kim, D. G., Ahn, H., Kwon, N. H. & Kim, S. in 26th tRNA Conference P52 (Biocon, 2016).

  170. Arkin, M. R., Tang, Y. & Wells, J. A. Small-molecule inhibitors of protein-protein interactions: progressing toward the reality. Chem. Biol. 21, 1102–1114 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Shin, S. M. et al. Antibody targeting intracellular oncogenic Ras mutants exerts anti-tumour effects after systemic administration. Nat. Commun. 8, 15090 (2017).

    PubMed  PubMed Central  Google Scholar 

  172. Che Nordin, M. A. & Teow, S. Y. Review of current cell-penetrating antibody developments for HIV-1 therapy. Molecules 23, 335 (2018).

    PubMed Central  Google Scholar 

  173. Irwin, M. J., Nyborg, J., Reid, B. R. & Blow, D. M. The crystal structure of tyrosyl-transfer RNA synthetase at 2–7 A resolution. J. Mol. Biol. 105, 577–586 (1976).

    CAS  PubMed  Google Scholar 

  174. Lai, A. C. & Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discov. 16, 101–114 (2017).

    CAS  PubMed  Google Scholar 

  175. Jia, J. et al. Mechanisms of drug combinations: interaction and network perspectives. Nat. Rev. Drug Discov. 8, 111–128 (2009).

    CAS  PubMed  Google Scholar 

  176. Holohan, C., Van Schaeybroeck, S., Longley, D. B. & Johnston, P. G. Cancer drug resistance: an evolving paradigm. Nat. Rev. Cancer 13, 714–726 (2013).

    CAS  PubMed  Google Scholar 

  177. Marston, H. D., Dixon, D. M., Knisely, J. M., Palmore, T. N. & Fauci, A. S. Antimicrobial resistance. JAMA 316, 1193–1204 (2016).

    CAS  PubMed  Google Scholar 

  178. O’Dwyer, K. et al. Bacterial resistance to leucyl-tRNA synthetase inhibitor GSK2251052 develops during treatment of complicated urinary tract infections. Antimicrob. Agents Chemother. 59, 289–298 (2015).

    PubMed  Google Scholar 

  179. Zeng, R. et al. Inhibition of mini-TyrRS-induced angiogenesis response in endothelial cells by VE-cadherin-dependent mini-TrpRS. Heart Vessels 27, 193–201 (2012).

    PubMed  Google Scholar 

  180. Dewan, V., Reader, J. & Forsyth, K. M. Role of aminoacyl-tRNA synthetases in infectious diseases and targets for therapeutic development. Top. Curr. Chem. 344, 293–329 (2014).

    CAS  PubMed  Google Scholar 

  181. Nakama, T., Nureki, O. & Yokoyama, S. Structural basis for the recognition of isoleucyl-adenylate and an antibiotic, mupirocin, by isoleucyl-tRNA synthetase. J. Biol. Chem. 276, 47387–47393 (2001).

    CAS  PubMed  Google Scholar 

  182. Hoepfner, D. et al. Selective and specific inhibition of the plasmodium falciparum lysyl-tRNA synthetase by the fungal secondary metabolite cladosporin. Cell Host Microbe 11, 654–663 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Fang, P. et al. Structural basis for specific inhibition of tRNA synthetase by an ATP competitive inhibitor. Chem. Biol. 22, 734–744 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Mirando, A. C. et al. Aminoacyl-tRNA synthetase dependent angiogenesis revealed by a bioengineered macrolide inhibitor. Sci. Rep. 5, 13160 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Woolard, J. et al. Borrelidin modulates the alternative splicing of VEGF in favour of anti-angiogenic isoforms. Chem. Sci. 2011, 273–278 (2011).

    PubMed  Google Scholar 

  186. Novoa, E. M. et al. Analogs of natural aminoacyl-tRNA synthetase inhibitors clear malaria in vivo. Proc. Natl Acad. Sci. USA 111, E5508–E5517 (2014). This study demonstrates the optimization process of borrelidin with reduced toxicity and enhanced efficacy.

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Sugawara, A. et al. Borrelidin analogues with antimalarial activity: design, synthesis and biological evaluation against Plasmodium falciparum parasites. Bioorg. Med. Chem. Lett. 23, 2302–2305 (2013).

    CAS  PubMed  Google Scholar 

  188. Kim, J. H., Han, J. M. & Kim, S. Protein-protein interactions and multi-component complexes of aminoacyl-tRNA synthetases. Top. Curr. Chem. 344, 119–144 (2014).

    CAS  PubMed  Google Scholar 

  189. Lee, S. W., Cho, B. H., Park, S. G. & Kim, S. Aminoacyl-tRNA synthetase complexes: beyond translation. J. Cell Sci. 117, 3725–3734 (2004).

    CAS  PubMed  Google Scholar 

  190. McLaughlin, H. M. et al. A recurrent loss-of-function alanyl-tRNA synthetase (AARS) mutation in patients with Charcot-Marie-Tooth disease type 2N (CMT2N). Hum. Mutat. 33, 244–253 (2012).

    CAS  PubMed  Google Scholar 

  191. Zhao, Z. et al. Alanyl-tRNA synthetase mutation in a family with dominant distal hereditary motor neuropathy. Neurology 78, 1644–1649 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Motley, W. W. et al. A novel AARS mutation in a family with dominant myeloneuropathy. Neurology 84, 2040–2047 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Nakayama, T. et al. Deficient activity of alanyl-tRNA synthetase underlies an autosomal recessive syndrome of progressive microcephaly, hypomyelination, and epileptic encephalopathy. Hum. Mutat. 38, 1348–1354 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Vester, A. et al. A loss-of-function variant in the human histidyl-tRNA synthetase (HARS) gene is neurotoxic in vivo. Hum. Mutat. 34, 191–199 (2013).

    CAS  PubMed  Google Scholar 

  195. McLaughlin, H. M. et al. Compound heterozygosity for loss-of-function lysyl-tRNA synthetase mutations in a patient with peripheral neuropathy. Am. J. Hum. Genet. 87, 560–566 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Hadchouel, A. et al. Biallelic mutations of methionyl-tRNA synthetase cause a specific type of pulmonary alveolar proteinosis prevalent on reunion island. Am. J. Hum. Genet. 96, 826–831 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Musante, L. et al. Mutations of the aminoacyl-tRNA-synthetases SARS and WARS2 are implicated in the etiology of autosomal recessive intellectual disability. Hum. Mutat. 38, 621–636 (2017).

    CAS  PubMed  Google Scholar 

  198. Stephen, J. et al. Loss of function mutations in VARS encoding cytoplasmic valyl-tRNA synthetase cause microcephaly, seizures, and progressive cerebral atrophy. Hum. Genet. 137, 293–303 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Khan, S. Recent advances in the biology and drug targeting of malaria parasite aminoacyl-tRNA synthetases. Malar. J. 15, 203 (2016).

    PubMed  PubMed Central  Google Scholar 

  200. Van de Vijver, P. et al. Synthetic microcin C analogs targeting different aminoacyl-tRNA synthetases. J. Bacteriol. 191, 6273–6280 (2009).

    PubMed  PubMed Central  Google Scholar 

  201. Petraitis, V. et al. Efficacy of PLD-118, a novel inhibitor of candida isoleucyl-tRNA synthetase, against experimental oropharyngeal and esophageal candidiasis caused by fluconazole-resistant C. albicans. Antimicrob. Agents Chemother. 48, 3959–3967 (2004).

    CAS  PubMed  Google Scholar 

  202. Cochrane, R. V. et al. Production of new cladosporin analogues by reconstitution of the polyketide synthases responsible for the biosynthesis of this antimalarial agent. Angew. Chem. Int. Ed. Engl. 55, 664–668 (2016).

    CAS  PubMed  Google Scholar 

  203. Yoon, S. et al. Discovery of leucyladenylate sulfamates as novel leucyl-tRNA synthetase (LRS)-targeted mammalian target of rapamycin complex 1 (mTORC1) inhibitors. J. Med. Chem. 59, 10322–10328 (2016).

    CAS  PubMed  Google Scholar 

  204. Sonoiki, E. et al. Antimalarial benzoxaboroles target Plasmodium falciparum leucyl-tRNA synthetase. Antimicrob. Agents Chemother. 60, 4886–4895 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Bharathkumar, H. et al. Screening of quinoline, 1,3-benzoxazine, and 1,3-oxazine-based small molecules against isolated methionyl-tRNA synthetase and A549 and HCT116 cancer cells including an in silico binding mode analysis. Org. Biomol. Chem. 13, 9381–9387 (2015).

    CAS  PubMed  Google Scholar 

  206. Nayak, S. U. et al. Safety, tolerability, systemic exposure, and metabolism of CRS3123, a methionyl-tRNA synthetase inhibitor developed for treatment of Clostridium difficile, in a phase 1 study. Antimicrob. Agents Chemother. 61, e02760-16 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. US National Library of Medicine. ClinicalTrials.gov https://www.clinicaltrials.gov/ct2/show/NCT02106338 (2016).

  208. US National Library of Medicine. ClinicalTrials.gov https://www.clinicaltrials.gov/ct2/show/NCT01551004 (2017).

  209. Yu, Z., Vodanovic-Jankovic, S., Kron, M. & Shen, B. New WS9326A congeners from Streptomyces sp. 9078 inhibiting Brugia malayi asparaginyl-tRNA synthetase. Org. Lett. 14, 4946–4949 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Shibata, A. et al. Discovery and pharmacological characterization of a new class of prolyl-tRNA synthetase inhibitor for anti-fibrosis therapy. PLOS ONE 12, e0186587 (2017).

    PubMed  PubMed Central  Google Scholar 

  211. Lin, Z. et al. Total synthesis and antimicrobial evaluation of natural albomycins against clinical pathogens. Nat. Commun. 9, 3445 (2018).

    PubMed  PubMed Central  Google Scholar 

  212. Brown, P. et al. Synthetic analogues of SB-219383. Novel C-glycosyl peptides as inhibitors of tyrosyl tRNA synthetase. Bioorg. Med. Chem. Lett. 11, 711–714 (2001).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by NRF-M3A6A4-2010-0029785 (S.K.), NRF-2015M3A6A4065724 (N.H.K.) and NRF-2017M3A9F7079378 (N.H.K.) from the National Research Foundation, the Ministry of Science and ICT (MSIT) of Korea and by the US National Institutes of Health (NIH) P01 HL029582 (P.L.F.). The authors thank B. S. Kang (Kyungbuk University) for drawing the architecture of class I and class II catalytic sites. They also thank J. Y. Lee (Buck Institute) for collecting data for secreted ARSN in human body fluids.

Author information

Authors and Affiliations

Authors

Contributions

P.L.F. provided substantial contribution to discussion of the content and reviewed and edited the manuscript before submission. N.H.K and S.K. contributed equally to all aspects of the article.

Corresponding author

Correspondence to Sunghoon Kim.

Ethics declarations

Competing interests

S.K. has financial interest in aTyr and Curebio, and N.H.K. has financial interest in Oncotag Diagnostics, although none specifically related to this Review.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

ANZCTR: http://www.anzctr.org.au

aTyr Pharma: http://www.atyrpharma.com

Biocon: http://biocon.re.kr

Bioxiness Pharmaceuticals: http://bioxiness.com/en

Clinical Trials website: www.clinicaltrials.gov

Curebio: http://www.cure-bio.com/

Exosome protein, RNA and lipid database: http://exocarta.org

Max-Planck Unified Proteome Database: http://www.mapuproteome.com/

Open Targets Platform: https://www.targetvalidation.org

The Human Protein Atlas: https://www.proteinatlas.org

The Online Mendelian Inheritance in Man (OMIM) database: https://omim.org/

Urinary Protein Biomarker Database: http://upbd.bmicc.cn

Urine Proteomics: http://www.urineproteomics.org

Supplementary information

Glossary

Polyketide

A type of secondary metabolite that either contains alternating carbonyl and methylene groups or is derived from precursors that contain such alternating groups.

Rossmann fold

A super-secondary structure composed of a series of alternating β-strand and α-helical segments that commonly appears in a variety of nucleotide binding proteins.

WHEP domains

Helix-turn-helix domains whose name comes from the first letters of tryptophanyl-tRNA synthetase (WRS), histidyl-tRNA synthetase (HRS) and glutamyl-prolyl-tRNA synthetase (EPRS), in which WHEP domains were first discovered.

Isoacceptors

In the context of this review, the different tRNA species that bind to alternate codons for the same amino acid residue.

Angiostatic factor

A substance that inhibits angiogenesis.

Megakaryopoiesis

A complex process in the bone marrow that ends with platelet formation from commitment of pluripotent haematopoietic stem cells.

PDZ-binding motif

A specific C-terminal motif that is usually approximately four or five residues in length and interacts with PDZ domains that are found in anchoring proteins.

Hypomyelinating leukodystrophy

(HLD). An autosomal recessive neurodegenerative disorder characterized by infant or childhood onset of progressive motor decline.

Frameshift

A shift of translation from one reading frame to another, generally caused by an addition or deletion in the nucleic acid sequence.

Early infantile epileptic encephalopathy

A debilitating progressive neurological disorder that involves intractable seizures and severe mental retardation.

Compound heterozygous missense mutation

A condition in which a gene has two different point mutations resulting in single amino acid change in both alleles.

Charcot–Marie–Tooth (CMT) disease

One of the hereditary motor and sensory neuropathies, a group of varied inherited disorders of the peripheral nervous system characterized by progressive loss of muscle tissue and touch sensation across various parts of the body.

Rhombomere

Any of the nine segments of the embryonic neural tube.

Lewy-body inclusions

Abnormal aggregates of protein that develop inside nerve cells contributing to disorders including Parkinson disease.

Scleroderma

A rare autoimmune connective tissue disease that can affect skin, joints, tendons and internal organs.

Coefficient of variation

A standardized measure of dispersion of a probability distribution or frequency distribution, defined as the ratio of the standard deviation to the mean.

Z′ factor

A measure of statistical effect size proposed for use in high-throughput screening.

NanoLuc luciferase

An engineered small luciferase derived from a deep sea luminous shrimp, which reveals stable, bright and sustained luminescence.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kwon, N.H., Fox, P.L. & Kim, S. Aminoacyl-tRNA synthetases as therapeutic targets. Nat Rev Drug Discov 18, 629–650 (2019). https://doi.org/10.1038/s41573-019-0026-3

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41573-019-0026-3

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research