Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Cold Spring Harb Mol Case Stud. 2022 Dec; 8(7): a006246.
PMCID: PMC9808560
PMID: 36307205

A missense, loss-of-function YARS1 variant in a patient with proximal-predominant motor neuropathy

Associated Data

Supplementary Materials

Abstract

Aminoacyl-tRNA synthetases (ARSs) are essential enzymes with a critical role in protein synthesis: charging tRNA molecules with cognate amino acids. Heterozygosity for variants in five genes (AARS1, GARS1, HARS1, WARS1, and YARS1) encoding cytoplasmic, dimeric ARSs have been associated with autosomal dominant neurological phenotypes, including axonal Charcot–Marie–Tooth disease (CMT). Missense variants in the catalytic domain of YARS1 were previously linked to dominant intermediate CMT type C (DI-CMTC). Here, we report a patient with a missense variant of unknown significance predicted to modify residue 308 in the anticodon binding domain of YARS1 (p.Asp308Tyr). Interestingly, p.Asp308Tyr is associated with proximal-predominant motor neuropathy, which has not been reported in patients with pathogenic YARS1 variants. We demonstrate that this allele causes a loss-of-function effect in yeast complementation assays when modeled in YARS1 and the yeast ortholog TYS1; structural modeling of this variant further supports a loss-of-function effect. Taken together, this study raises the possibility that certain YARS1 variants cause proximal-prominent motor neuropathy and indicates that patients with this phenotype should be screened for genetic lesions in YARS1.

Keywords: areflexia of upper limbs, generalized limb muscle atrophy, lower limb muscle weakness, upper limb muscle weakness

CASE PRESENTATION

Aminoacyl-tRNA synthetases (ARSs) are a group of ubiquitously expressed, essential enzymes that are responsible for charging transfer RNAs (tRNAs) with cognate amino acids in the cytoplasm and mitochondria, a process critical for establishing fidelity in protein synthesis. Variants in the 37 ARS genes (all of which are encoded in the nucleus) have previously been associated with a spectrum of genetic disorders (Meyer-Schuman and Antonellis 2017; Kuo and Antonellis 2020). Tyrosyl-tRNA synthetase (encoded by YARS1) is responsible for charging tyrosyl-tRNA with tyrosine in the cytoplasm via a two-step aminoacylation reaction. Importantly, two YARS1 subunits must form an asymmetric homodimeric complex with two tyrosyl tRNAs for the aminoacylation reaction to occur (Ward and Fersht 1988). Previously, homozygosity or compound heterozygosity for loss-of-function YARS1 variants has been associated with severe, early-onset, multisystem disease, with phenotypes including liver dysfunction, developmental delay, brain anomalies, and sensorineural hearing loss (Nowaczyk et al. 2017; Williams et al. 2019; Averdunk et al. 2021; Estève et al. 2021). Furthermore, heterozygosity for missense variants or a small, in-frame deletion in YARS1 have been associated with dominant intermediate Charcot–Marie–Tooth neuropathy type C (DI-CMTC; MIM #608323), which is characterized by progressive weakness and sensory loss in the distal upper and lower extremities, with intermediate motor nerve conduction velocities between 25 and 45 m/sec (Jordanova et al. 2003, 2006; Hyun et al. 2013).

Here, we present a 47-yr-old female proband of European ancestry. She first noted symptoms at 18 yr of age when she developed tremors, weakness, and cramping of her hands. At 22, neuromuscular examination revealed asymmetric weakness in the distal upper extremities and proximal lower extremities. Reflexes were absent at the triceps, but were otherwise normal. Serial examinations have demonstrated slow progression of weakness. At 47, her exam showed asymmetric upper extremity weakness affecting both proximal and distal muscles and proximal > distal lower extremity weakness (Fig. 1A–C). Hip flexion and extension measured the weakest at 2/5 bilaterally on manual muscle testing. Atrophy was prominent in her forearms and hands (Fig. 1A). Deep tendon reflexes were absent except at the ankles, where hyporeflexia was noted. Sensory exam was normal to touch, vibration, position, and pinprick. Additional findings included rotary nystagmus (present since 18 yr of age), kyphoscoliosis, and tongue fasciculations. Creatine kinase was mildly elevated (947–1692 units/L) at symptom onset, but was reduced to 300 units/L at last measurement (47 yr of age). Electrodiagnostic studies performed at 23 yr of age demonstrated signs of widespread denervation in the upper and lower extremities and increased amplitude of motor unit potentials, suggesting abnormalities in motor neurons of the spinal cord. The motor and sensory nerve conduction velocities were normal. A left deltoid biopsy obtained at 21 yr of age showed grouped angular fibers, consistent with a neurogenic process. Clinical findings are summarized in Table 1.

An external file that holds a picture, illustration, etc.
Object name is MCS006246For_F1.jpg

A loss-of-function YARS1 variant in a patient with proximal-prominent neuropathy. (AC) Clinical findings noted on the patient's physical exam, including atrophy of the distal upper extremities and hand muscles (A) and finger extensor weakness demonstrated by finger drop (B,C). (D) Pedigree of the proband's (arrow) family. Genotypes at p.Asp308 YARS1 are indicated below each individual (??/?? indicates an unknown genotype). Filled objects indicate that the individual is affected, whereas unfilled objects indicate that the individual is unaffected. A slash through an individual indicates that they are deceased. (E) YARS1 protein sequence alignment, including six evolutionarily diverse species centered around the affected p.Asp308 residue (indicated in red). (F) Haploid yeast lacking endogenous TYS1 were transformed with a LEU2-bearing pRS315 vector to express wild type, p.Gly41Arg, p.Asp308Tyr, or p.Pro167Thr TYS1, or with a vector containing no TYS1 insert (“No Insert”). Resultant yeast cultures were plated undiluted or diluted (1:10, 1:100, or 1:1000) on solid media without leucine or uracil (−leu −ura) (see Supplemental Fig. 1A) or on solid media containing 5-FOA and grown at 30°C. Replicates A and B indicate two separate colonies from a single transformation. Image shown is representative of two independent transformations using two different plasmid DNA preparations (p.Asp308Tyr TYS1 total n = 8). (G) Haploid yeast lacking endogenous TYS1 were transformed with a LEU2-bearing pAG425-GPD vector to express wild-type TYS1, p.Asp308Tyr TYS1, wild-type YARS1, or p.Asp308Tyr YARS1, or with a vector containing no TYS1 insert (“No Insert”). Resultant yeast cultures were plated undiluted or diluted (1:10, 1:100, or 1:1000) on solid media without leucine or uracil (−leu −ura) (see Supplemental Fig. 1B) or on solid media containing 5-FOA and grown at 30°C. Replicates A and B indicate colonies from transformations using two different plasmid DNA preparations. Image shown is representative of two independent transformations using two different plasmid DNA preparations (p.Asp308Tyr YARS1 total n = 6).

Table 1.

Summary of clinical findings in the proband described in this study compared with other patients with YARS1-associated dominant intermediate Charcot–Marie–Tooth neuropathy type C (DI-CMTC)

c.922G > T (p.Asp308Tyr) current studyc. 121G > A (p.Gly41Arg) (Jordanova et al. 2003; Thomas et al. 2016)c. 586G > A (p.Glu196Lys) (Jordanova et al. 2003; Thomas et al. 2016)c.586G > C p.Glu196Gln (Gonzaga-Jauregui et al. 2015)c. 458-469del (p.153-156del) (Jordanova et al. 2006)c.241_242delGAinsAT (p.Asp81Ile) (Nam et al. 2022; Hyun et al. 2013)c.497A > G (p.Tyr166Cys) (Nam et al. 2022)
Age of onset18 yr5–20 yr7–59 yr16 yrNR23 yr17–27 yr
CountryUnited StatesUnited StatesBulgariaNRBelgiumKoreaKorea
InheritanceUnknown (possible de novo)Autosomal dominantAutosomal dominantAutosomal dominantDe novoUnknownAutosomal dominant (2/3); unknown (presumed de novo) (1/3)
Presenting symptomDistal upper extremity weaknessAnkle dorsiflexion weaknessAnkle dorsiflexion weaknessNRNRDistal muscle weaknessDistal muscle weakness and atrophy
Pes cavus (HP:0001761)/hammertoe (HP:0001765)aNo/noYes (13/15)/Yes (3/15)Yes (22/27)/Yes (3/27)Yes/yesNRYesYes (3/3)/NR
Upper limb distal weakness (HP:0008959)aYesYes (2/21)Yes (14/27)YesNRYesYes (3/3)
Upper limb proximal weakness (HP:0008997)aYesYes (a few, mild)NoNRNRNRNR
Lower limb distal weakness (HP:0009053)aYesYes (9/15)Yes (16/27)YesNRYesYes (3/3)
Lower limb proximal weakness (HP:0008994)aYesYes (2/15)NoNRNRYesNR
Muscle atrophy (HP:0008944; HP:0007149)aYes (distal UE)Yes (15/15 LE, 11/15 UE)Yes (27/27 LE, 14/27 UE)YesNRYesYes
Distal sensory impairment (HP:0002936)aNoYes (21/21)Yes (20/27)NoNRYesYes
Areflexia/hyporeflexia (HP:0001284)aYes (at most recent exam)Yes (15/15)Yes (26/26)YesNRNoYes (1/3)
Hyperreflexia (HP:0001347)aYes (knees/ankles, at disease onset)NRNRNRNRNoYes (3/3; knees)
Hand tremor (HP:0002378)aYesYes (13/15)NoNRNRNRNo
Other symptomsRotary nystagmus (16 y; HP:0001583), tongue fasciculations (HP:0001308), kyphoscoliosis (HP:0002751)aKyphoscoliosis (HP:0002751)a (2/27)NR
Median motor NCVs (m/sec)65.229.5–45.6 (25/25 abnormal)24.7–57.8 (15/18 abnormal)NRNR29.1 (left), 45.1 (right) (abnormal)37.4–41.5 (3/3 abnormal)

(NR) Not reported, (UE) upper extremities, (LE) lower extremities, (NCV) nerve conduction velocity.

aHuman Phenotype Ontology (HP) terms for peripheral neuropathy phenotypes.

Based on the observations above, the patient was thought to have symptoms consistent with spinal muscular atrophy type III; however, genetic testing of SMN1 was normal (further genetic testing information provided below). Family history was negative for similar disease, and the patient's mother had no symptoms of neuropathy; however, the patient's father developed a sensory peripheral neuropathy in adulthood without symptoms of weakness. He did not undergo electrodiagnostic testing. The patient's two daughters (ages 20 and 22 yr) were examined by the proband's neuromuscular physician and had normal strength and reflexes.

TECHNICAL ANALYSIS

Based on clinical presentation, genetic testing was performed using the Invitae Comprehensive Neuropathies Gene Panel (Invitae), which evaluates 111 genes associated with hereditary neuropathies for (1) single-nucleotide variants and deletions/insertions <15 nt in length by targeted next-generation sequencing of coding exons plus 10–20 bp of flanking adjacent intronic sequence, and (2) duplications and deletions at single-exon resolution (Invitae 03200, https://www.invitae.com/en/providers/test-catalog/test-03200). This analysis identified a missense variant in YARS1 (NM_003680.4:c.922G > T [p.Asp308Tyr]) (see Table 2). Importantly, familial testing demonstrated that this variant was absent in the patient's mother and two daughters, who are not affected with the neuropathy phenotype present in the patient (Fig. 1D). The patient's father was not available for genetic testing. Given these findings, it is possible that the variant observed in the proband may have arisen de novo, but further haplotype analysis would be required to rule out paternal inheritance.

Table 2.

Summary of the identified YARS1 variant

GeneGenomic locationHGVSa cDNAHGVSa proteinZygosityParent of originVariant interpretation
YARS1 Chr 1:32,782,524 (GRCh38);
Chr 1:33,248,125 (GRCh37)
NM_003680.4: c.922G > TNP_003671.1: p.Asp308TyrHeterozygousPaternal or de novoUncertain significance, with an effect on gene function
aHuman Genome Variation Society accepted nomenclature.

VARIANT INTERPRETATION

The p.Asp308 YARS1 Residue Is Located in a Highly Conserved Structure Involved in tRNA-Tyr Anticodon Recognition

The p.Asp308 YARS1 residue is located within a stretch of conserved amino acid residues in the anticodon binding domain that show high sequence similarity among a range of species (Fig. 1E); importantly, this amino acid residue is conserved between yeast and humans. Consistent with this high degree of conservation, PolyPhen-2 analysis of sequence, phylogenetic, and structural data (Adzhubei et al. 2010) predicts that the p.Asp308Tyr allele is “probably damaging.” Previously determined crystal structures for tyrosyl-tRNA synthetases from multiple species indicate that the p.Asp308 residue is located at the beginning of a highly conserved α-helical structure that is present in archaea and bacteria tyrosyl-tRNA ligases as well as tyrosyl-tRNA synthetase homologs in yeast and higher eukaryotes (Qiu et al. 2001; Yaremchuk et al. 2002; Tsunoda et al. 2007). Although the structure of human YARS1 complexed with tRNA-Tyr has not been solved, the yeast p.Asp321 residue (equivalent to p.Asp308) (see Supplemental Table 1) forms direct hydrogen bonds with the guanine-34 residue of the tRNA-Tyr-GTA anticodon triplet for substrate discrimination (Tsunoda et al. 2007). Given the high degree of structural similarity between different tyrosyl-tRNA synthetase homologs (Tsunoda et al. 2007), it is possible that this residue is also important for tRNA-Tyr substrate discrimination in higher eukaryotes, including humans.

The p.Asp308Tyr YARS1 Allele Is Present as a Singleton in Population Sequence Databases

To assess the frequency of the p.Asp308Tyr YARS1 allele in the general population, we queried the 1000 Genomes and gnomAD databases at the appropriate chromosomal position (see Table 2). At the time of initial variant evaluation, the p.Asp308Tyr allele was not present in the 1000 Genomes database (Auton et al. 2015) or in the gnomAD database (Karczewski et al. 2020). However, during the preparation of this manuscript, a single observation of the p.Asp308Tyr allele in the heterozygous state was reported in the gnomAD database (1-33248125-C-A, GRCh37; allele frequency 1/251,452; 0.000003977). Importantly, this allele frequency is consistent with other pathogenic ARS variants associated with dominant neuropathy (e.g., p.Gly294Arg GARS1, 1-33248125-C-A, GRCh37; allele frequency 1/249,268; 0.000004012) (Antonellis et al. 2003). Furthermore, the individual reported in gnomAD is in the <30-yr age category; thus, it is possible that this individual may develop neuropathy symptoms at a later age.

The p.Asp308Tyr YARS1 Allele Does Not Support Viability in Yeast Complementation Assays

Previously reported neuropathy-associated ARS alleles demonstrated loss-of-function effects in biochemical, yeast, and mouse models; however, not all loss-of-function ARS alleles cause dominant neuropathy. Yeast is particularly useful as an in vivo model to assess ARS variant effects on gene function, and previous studies demonstrated successful modeling of human YARS1 variants using the yeast ortholog TYS1 via yeast complementation assays (Jordanova et al. 2006; Gonzaga-Jauregui et al. 2015; Williams et al. 2019). We therefore used this model to determine the ability of wild-type and p.Asp308Tyr TYS1 alleles to complement TYS1 deletion in a previously validated haploid yeast strain; viability of this strain is maintained via a pRS316 vector containing wild-type TYS1 and URA3 (Antonellis et al. 2006; Oprescu et al. 2017; Williams et al. 2019). To allow comparison with previously identified pathogenic alleles, we included p.Gly41Arg TYS1 (a loss-of-function allele identified in a patient with dominant neuropathy [Jordanova et al. 2006]) and p.Pro167Thr TYS1 (a hypomorphic allele identified in a patient with a recessive syndromic phenotype [Williams et al. 2019]) in these analyses. Yeast cells were transformed with wild-type, p.Asp308Tyr, p.Gly41Arg, or p.Pro167Thr TYS1 expression constructs or with a pRS315 construct with no TYS1 insert. Resultant yeast strains were grown on media without leucine or uracil as a positive growth control (Supplemental Fig. 1A) and on media containing 5-FOA, which selects for cells that have spontaneously lost the URA3-bearing maintenance vector (Fig. 1F; Boeke et al. 1987). Importantly, the pRS315 vector with no insert was not sufficient to support yeast growth on 5-FOA media, confirming that TYS1 is essential for yeast survival. Yeast cells transformed with wild-type TYS1 showed appreciable growth at 30°C. The previously characterized p.Gly41Arg and p.Pro167Thr TYS1 alleles show the expected loss-of-function effects: (1) The hypomorphic p.Pro167Thr allele results in reduced, but not ablated, yeast growth, and (2) the loss-of-function p.Gly41Arg allele is unable to support any yeast growth. Finally, the p.Asp308Tyr TYS1 variant reported in this study did not support any yeast growth, indicating that p.Asp308Tyr YARS1 is a loss-of-function allele in this assay, similar to previously reported pathogenic, neuropathy-associated ARS alleles. To further characterize the effects of the p.Asp308Tyr variant in the context of human YARS1, we repeated this assay using the same haploid yeast strain with TYS1 deleted and transformed it with wild-type YARS1 or p.Asp308Tyr YARS1 cDNA expression constructs (Fig. 1G; Supplemental Fig. 1B). Yeast cells transformed with wild-type human YARS1 demonstrated growth on 5-FOA, supporting conservation of function between yeast TYS1 and human YARS1. Furthermore, the human p.Asp308Tyr YARS1 allele was not able to support yeast growth in this assay, consistent with a loss-of-function effect. Importantly, yeast transformed with wild-type YARS1 or p.Asp308Tyr YARS1 showed stable expression of YARS1 (Supplemental Fig. 1C), suggesting that the loss-of-function effect is not due to protein instability.

Taken together with our findings regarding YARS1 structure and conservation as well as YARS1 population data, we conclude that the p.Asp308Tyr YARS1 allele is a variant of unknown significance according to current variant interpretation guidelines using evidence criteria PS3 and PP3 (Richards et al. 2015) However, the data presented in this study provide significant supporting evidence for pathogenicity, particularly with respect to the importance of the affected amino acid residue in tRNA binding and the observed loss-of-function effect in in vivo functional studies, which is similar to other neuropathy-associated ARS alleles. Although it is possible that this variant arose de novo in the proband, lack of availability of a paternal sample for genetic testing made it impossible to clarify the inheritance pattern. Further genetic evidence (e.g., reports of additional patients with variants at this residue) may allow for reclassification in the future.

DISCUSSION

In this study, we identified a previously unreported YARS1 variant that affects an amino acid residue in the anticodon binding domain. This variant was observed in a patient with a neuropathy phenotype that is clinically distinct from previously described YARS1-related dominant intermediate CMT (Table 1). If this variant is confirmed as pathogenic, this report will have expanded the allelic and phenotypic spectrum of YARS1-related dominant disease. Notably, a similar range of dominant neuropathy phenotypes have been described in association with loss-of-function missense variants and small, in-frame deletions in four other cytoplasmic, homodimeric ARSs (AARS1, GARS1, HARS1, and WARS1) (Antonellis et al. 2003; Latour et al. 2010; McLaughlin et al. 2012; Vester et al. 2013; Safka Brozkova et al. 2015; Meyer-Schuman and Antonellis 2017; Tsai et al. 2017).

As noted, the patient reported here has several phenotypic features not previously seen in patients with YARS1-associated DI-CMTC (Table 1). This case is unique in that the patient presented with upper extremity weakness, whereas all prior patients presented with lower extremity involvement prior to developing upper extremity symptoms; however, such upper limb phenotypes have been reported in patients with pathogenic GARS1 variants (Antonellis et al. 2003). The pattern of muscle weakness also differs from that seen in other patients with YARS1 variants. The patient presented here has predominant proximal lower extremity weakness with minimal distal weakness of these extremities. Although some patients with DI-CMTC have been reported to have proximal lower extremity weakness, it was reported as less severe than the distal weakness (Thomas et al. 2016). The patient reported here also lacks foot deformities and sensory abnormalities seen in most patients (Thomas et al. 2016; Nam et al. 2022). In addition, median motor nerve conduction velocities (NCVs) were normal on three separate electrodiagnostic studies in the third decade of life, whereas most previously reported adult DI-CMTC patients presented with reduced NCVs. The patient reported here was also identified to have persistent elevation of creatine kinase (CK) early in her disease course, which has not been reported in association with DI-CMTC and is rarely associated with CMT disease (Rudnik-Schöneborn et al. 2016).

To our knowledge, the p.Asp308Tyr variant described in this study is the first potentially pathogenic YARS1 variant identified in the anticodon binding domain; in contrast, all previously identified YARS1 variants map to the catalytic domain (Supplemental Fig. 1D). Importantly, variants in the anticodon binding domain of other ARSs have been associated with a more severe, atypical dominant neuropathy, including an infantile-onset spinal muscular atrophy caused by GARS1 variants (James et al. 2006; Eskuri et al. 2012; Markovitz et al. 2020). It will be interesting to test whether, and by what mechanism, mutations in the tRNA binding domain lead to differential clinical presentations.

In addition to aminoacylation activity, YARS1 has noncanonical functions that may be relevant for YARS1-related disease. First, a Drosophila model of YARS1-related CMT showed that YARS1 localizes to the nucleus and complexes with the scaffolding protein TRIM28 and histone deacetylase HDAC1 to enact a transcriptional regulation program, which is altered in the context of neurons expressing CMT-associated YARS1 variants (Bervoets et al. 2019). Second, noncanonical cytokine activity by secreted YARS1 cleavage products plays a role in angiogenesis and platelet biogenesis (Wakasugi et al. 2002; Yang et al. 2002; Greenberg et al. 2008). Finally, noncanonical cytoplasmic mRNA binding has been reported for many cytoplasmic ARSs in yeast, often with downstream consequences on post-transcriptional gene regulation (Levi and Arava 2019; Levi et al. 2020; Garin et al. 2021). Therefore, it is possible that dysfunction in noncanonical activities—in concert with any effects on tRNA charging—could explain the clinical heterogeneity observed in YARS1-related neuropathy. Further studies will be needed to fully understand the molecular mechanisms underlying the range of YARS1-related phenotypes.

ADDITIONAL INFORMATION

Database Deposition and Access

The p.Asp308Tyr YARS1 variant can be found in ClinVar (https://www.ncbii.nlm.nih.gov/clinvar/) under accession number VCV001681503.3.

Ethics Statement

Written consent was obtained from the patient and other family members ahead of manuscript preparation.

Acknowledgments

We thank the patient and their family for inspiring and supporting this study. We thank Jerry Mendell, M.D. (Nationwide Children's Hospital), for his support of this study and for clarification of the details of clinical presentation.

Author Contributions

M.E.F. and A.A. designed the yeast functional studies. M.E.F., A.P.M., and A.A. wrote the manuscript. M.E.F. and S.M.L.F. collected and analyzed the yeast functional assay data. A.P.M. provided clinical data. All authors reviewed the manuscript.

Funding

A.A. is supported by a grant from the National Institute of General Medical Sciences (GM136441). M.E.F. is supported by a grant from the National Institutes of Health (5T32NS007222-40). S.M.L.F. is supported by a grant from the National Institutes of Health (T32GM007863).

Competing Interest Statement

The authors declare no competing interests.

Supplementary Material

Supplemental Material:

Footnotes

[Supplemental material is available for this article.]

REFERENCES

  • Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS, Sunyaev SR. 2010. A method and server for predicting damaging missense mutations. Nat Methods 7: 248–249. 10.1038/nmeth0410-248 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Antonellis A, Ellsworth RE, Sambuughin N, Puls I, Abel A, Lee-Lin SQ, Jordanova A, Kremensky I, Christodoulou K, Middleton LT, et al. 2003. Glycyl tRNA synthetase mutations in Charcot–Marie–Tooth disease type 2D and distal spinal muscular atrophy type V. Am J Hum Genet 72: 1293–1299. 10.1086/375039 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Antonellis A, Lee-Lin SQ, Wasterlain A, Leo P, Quezado M, Goldfarb LG, Myung K, Burgess S, Fischbeck KH, Green ED. 2006. Functional analyses of glycyl-tRNA synthetase mutations suggest a key role for tRNA-charging enzymes in peripheral axons. J Neurosci 26: 10397–10406. 10.1523/JNEUROSCI.1671-06.2006 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Auton A, Abecasis GR, Altshuler DM, Durbin RM, Bentley DR, Chakravarti A, Clark AG, Donnelly P, Eichler EE, Flicek P, et al. 2015. A global reference for human genetic variation. Nature 526: 68–74. 10.1038/nature15393 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Averdunk L, Sticht H, Surowy H, Lüdecke HJ, Koch-Hogrebe M, Alsaif HS, Kahrizi K, Alzaidan H, Alawam BS, Tohary M, et al. 2021. The recurrent missense mutation p.(Arg367Trp) in YARS1 causes a distinct neurodevelopmental phenotype. J Mol Med (Berl) 99: 1755–1768. 10.1007/s00109-021-02124-9 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bervoets S, Wei N, Erfurth ML, Yusein-Myashkova S, Ermanoska B, Mateiu L, Asselbergh B, Blocquel D, Kakad P, Penserga T, et al. 2019. Transcriptional dysregulation by a nucleus-localized aminoacyl-tRNA synthetase associated with Charcot–Marie–Tooth neuropathy. Nat Commun 10: 5045. 10.1038/s41467-019-12909-9 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Boeke JD, Trueheart J, Natsoulis G, Fink GR. 1987. 5-Fluoroorotic acid as a selective agent in yeast molecular genetics. Methods Enzymol 154: 164–175. 10.1016/0076-6879(87)54076-9 [PubMed] [CrossRef] [Google Scholar]
  • Eskuri JM, Stanley CM, Moore SA, Mathews KD. 2012. Infantile onset CMT2D/dSMA V in monozygotic twins due to a mutation in the anticodon-binding domain of GARS. J Peripher Nerv Syst 17: 132–134. 10.1111/j.1529-8027.2012.00370.x [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Estève C, Roman C, DeLeusse C, Baravalle M, Bertaux K, Blanc F, Bourgeois P, Bresson V, Cano A, Coste ME, et al. 2021. Novel partial loss-of-function variants in the tyrosyl-tRNA synthetase 1 (YARS1) gene involved in multisystem disease. Eur J Med Genet 64: 104294. 10.1016/j.ejmg.2021.104294 [PubMed] [CrossRef] [Google Scholar]
  • Garin S, Levi O, Forrest ME, Antonellis A, Arava YS. 2021. Comprehensive characterization of mRNAs associated with yeast cytosolic aminoacyl-tRNA synthetases. RNA Biol 18: 2605–2616. 10.1080/15476286.2021.1935116 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Gonzaga-Jauregui C, Harel T, Gambin T, Kousi M, Griffin LB, Francescatto L, Ozes B, Karaca E, Jhangiani SN, Bainbridge MN, et al. 2015. Exome sequence analysis suggests that genetic burden contributes to phenotypic variability and complex neuropathy. Cell Rep 12: 1169–1183. 10.1016/j.celrep.2015.07.023 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Greenberg Y, King M, Kiosses WB, Ewalt K, Yang X, Schimmel P, Reader JS, Tzima E. 2008. The novel fragment of tyrosyl tRNA synthetase, mini-TyrRS, is secreted to induce an angiogenic response in endothelial cells. FASEB J 22: 1597–1605. 10.1096/fj.07-9973com [PubMed] [CrossRef] [Google Scholar]
  • Hyun YS, Park HJ, Heo SH, Yoon BR, Nam SH, Kim SB, Park CI, Choi BO, Chung KW. 2013. Rare variants in methionyl- and tyrosyl-tRNA synthetase genes in late-onset autosomal dominant Charcot–Marie–Tooth neuropathy. Clin Genet 86: 592–594. 10.1111/cge.12327 [PubMed] [CrossRef] [Google Scholar]
  • James PA, Cader MZ, Muntoni F, Childs AM, Crow YJ, Talbot K. 2006. Severe childhood SMA and axonal CMT due to anticodon binding domain mutations in the GARS gene. Neurology 67: 1710–1712. 10.1212/01.wnl.0000242619.52335.bc [PubMed] [CrossRef] [Google Scholar]
  • Jordanova A, Thomas FP, Guergueltcheva V, Tournev I, Gondim FAA, Ishpekova B, De Vriendt E, Jacobs A, Litvinenko I, Ivanova N, et al. 2003. Dominant intermediate Charcot–Marie–Tooth type C maps to chromosome 1p34-p35. Am J Hum Genet 73: 1423–1430. 10.1086/379792 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Jordanova A, Irobi J, Thomas FP, Van Dijck P, Meerschaert K, Dewil M, Dierick I, Jacobs A, De Vriendt E, Guergueltcheva V, et al. 2006. Disrupted function and axonal distribution of mutant tyrosyl-tRNA synthetase in dominant intermediate Charcot–Marie–Tooth neuropathy. Nat Genet 38: 197–202. 10.1038/ng1727 [PubMed] [CrossRef] [Google Scholar]
  • Karczewski KJ, Francioli LC, Tiao G, Cummings BB, Alföldi J, Wang Q, Collins RL, Laricchia KM, Ganna A, Birnbaum DP, et al. 2020. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581: 434–443. 10.1038/s41586-020-2308-7 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kuo ME, Antonellis A. 2020. Ubiquitously expressed proteins and restricted phenotypes: exploring cell-specific sensitivities to impaired tRNA charging. Trends Genet 36: 105–117. 10.1016/j.tig.2019.11.007 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Latour P, Thauvin-Robinet C, Baudelet-Méry C, Soichot P, Cusin V, Faivre L, Locatelli MC, Mayençon M, Sarcey A, Broussolle E, et al. 2010. A major determinant for binding and aminoacylation of tRNA(Ala) in cytoplasmic Alanyl-tRNA synthetase is mutated in dominant axonal Charcot–Marie–Tooth disease. Am J Hum Genet 86: 77–82. 10.1016/j.ajhg.2009.12.005 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Levi O, Arava Y. 2019. mRNA association by aminoacyl tRNA synthetase occurs at a putative anticodon mimic and autoregulates translation in response to tRNA levels. PLoS Biol 17: 1–24. 10.1371/journal.pbio.3000274 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Levi O, Garin S, Arava Y. 2020. RNA mimicry in post-transcriptional regulation by aminoacyl tRNA synthetases. Wiley Interdiscip Rev RNA 11: e1564. 10.1002/wrna.1564 [PubMed] [CrossRef] [Google Scholar]
  • Markovitz R, Ghosh R, Kuo ME, Hong W, Lim J, Bernes S, Manberg S, Crosby K, Tanpaiboon P, Bharucha-Goebel D, et al. 2020. GARS-related disease in infantile spinal muscular atrophy: implications for diagnosis and treatment. Am J Med Genet A 182: 1167–1176. 10.1002/ajmg.a.61544 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • McLaughlin HM, Sakaguchi R, Giblin W, Wilson TE, Biesecker L, Lupski JR, Talbot K, Vance JM, Züchner S, Lee YC, et al. 2012. 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. 10.1002/humu.21635 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Meyer-Schuman R, Antonellis A. 2017. Emerging mechanisms of aminoacyl-tRNA synthetase mutations in recessive and dominant human disease. Hum Mol Genet 26: R114–R127. 10.1093/hmg/ddx231 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nam DE, Park JH, Park CE, Jung NY, Nam SH, Kwon HM, Kim HS, Kim SB, Son WS, Choi BO, et al. 2022. Variants of aminoacyl-tRNA synthetase genes in Charcot–Marie–Tooth disease: a Korean cohort study. J Peripher Nerv Syst 27: 38–49. 10.1111/jns.12476 [PubMed] [CrossRef] [Google Scholar]
  • Nowaczyk MJM, Huang L, Tarnopolsky M, Schwartzentruber J, Majewski J, Bulman DE, Hartley T, Boycott KM. 2017. A novel multisystem disease associated with recessive mutations in the tyrosyl-tRNA synthetase (YARS) gene. Am J Med Genet A 173: 126–134. 10.1002/ajmg.a.37973 [PubMed] [CrossRef] [Google Scholar]
  • Oprescu SN, Griffin LB, Beg AA, Antonellis A. 2017. Predicting the pathogenicity of aminoacyl-tRNA synthetase mutations. Methods 113: 139–151. 10.1016/j.ymeth.2016.11.013 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Qiu X, Janson CA, Smith WW, Green SM, McDevitt P, Johanson K, Carter P, Hibbs M, Lewis C, Chalker A, et al. 2001. Crystal structure of Staphylococcus aureus tyrosyl-tRNA synthetase in complex with a class of potent and specific inhibitors. Protein Sci 10: 2008. 10.1110/ps.18001 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, et al. 2015. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 17: 405–424. 10.1038/gim.2015.30 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rudnik-Schöneborn S, Tölle D, Senderek J, Eggermann K, Elbracht M, Kornak U, von der Hagen M, Kirschner J, Leube B, Müller-Felber W, et al. 2016. Diagnostic algorithms in Charcot–Marie–Tooth neuropathies: experiences from a German genetic laboratory on the basis of 1206 index patients. Clin Genet 89: 34–43. 10.1111/cge.12594 [PubMed] [CrossRef] [Google Scholar]
  • Safka Brozkova D, Deconinck T, Beth Griffin L, Ferbert A, Haberlova J, Mazanec R, Lassuthova P, Roth C, Pilunthanakul T, Rautenstrauss B, et al. 2015. Loss of function mutations in HARS cause a spectrum of inherited peripheral neuropathies. Brain 138: 2161–2172. 10.1093/brain/awv158 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Thomas FP, Guergueltcheva V, Gondim FAA, Tournev I, Rao CV, Ishpekova B, Kinsella LJ, Pan Y, Geller TJ, Litvinenko I, et al. 2016. Clinical, neurophysiological and morphological study of dominant intermediate Charcot–Marie–Tooth type C neuropathy. J Neurol 263: 467–476. 10.1007/s00415-015-7989-8 [PubMed] [CrossRef] [Google Scholar]
  • Tsai PC, Soong BW, Mademan I, Huang YH, Liu CR, Hsiao CT, Wu HT, Liu TT, Liu YT, Tseng YT, et al. 2017. A recurrent WARS mutation is a novel cause of autosomal dominant distal hereditary motor neuropathy. Brain 140: 1252–1266. 10.1093/brain/awx058 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tsunoda M, Kusakabe Y, Tanaka N, Ohno S, Nakamura M, Senda T, Moriguchi T, Asai N, Sekine M, Yokogawa T, et al. 2007. Structural basis for recognition of cognate tRNA by tyrosyl-tRNA synthetase from three kingdoms. Nucl Acids Res 35: 4289. 10.1093/nar/gkm417 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Vester, A., Velez-Ruiz, G., McLaughlin, HM., NISC Comparative Sequencing Program, Lupski, JR., Talbot, K., Vance, JM., Züchner, S., Roda, RH., Fischbeck, KH., et al. 2013. A loss-of-function variant in the human histidyl-tRNA synthetase (HARS) gene is neurotoxic in vivo. Hum Mutat 34: 191–199. 10.1002/humu.22210 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wakasugi K, Slike BM, Hood J, Ewalt KL, Cheresh DA, Schimme P. 2002. Induction of angiogenesis by a fragment of human tyrosyl-tRNA synthetase. J Biol Chem 277: 20124–20126. 10.1074/jbc.C200126200 [PubMed] [CrossRef] [Google Scholar]
  • Ward WH, Fersht AR. 1988. Tyrosyl-tRNA synthetase acts as an asymmetric dimer in charging tRNA. A rationale for half-of-the-sites activity. Biochemistry 27: 14499–14509. 10.1021/bi00415a021 [PubMed] [CrossRef] [Google Scholar]
  • Williams KB, Brigatti KW, Puffenberger EG, Gonzaga-Jauregui C, Griffin LB, Martinez ED, Wenger OK, Yoder MA, Kandula VVR, Fox MD, et al. 2019. Homozygosity for a mutation affecting the catalytic domain of tyrosyl-tRNA synthetase (YARS) causes multisystem disease. Hum Mol Genet 28: 525–538. 10.1093/hmg/ddy344 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yang XL, Skene RJ, McRee DE, Schimmel P. 2002. Crystal structure of a human aminoacyl-tRNA synthetase cytokine. Proc Natl Acad Sci 99: 15369–15374. 10.1073/pnas.242611799 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yaremchuk A, Kriklivyi I, Tukalo M, Cusack S. 2002. Class I tyrosyl-tRNA synthetase has a class II mode of cognate tRNA recognition. EMBO J 21: 3829. 10.1093/emboj/cdf373 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

Articles from Cold Spring Harbor Molecular Case Studies are provided here courtesy of Cold Spring Harbor Laboratory Press

-