Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
1.
Fly (Austin) ; 18(1): 2308737, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38374657

ABSTRACT

Amino acyl-tRNA synthetases perform diverse non-canonical functions aside from their essential role in charging tRNAs with their cognate amino acid. The phenylalanyl-tRNA synthetase (PheRS/FARS) is an α2ß2 tetramer that is needed for charging the tRNAPhe for its translation activity. Fragments of the α-subunit have been shown to display an additional, translation-independent, function that activates growth and proliferation and counteracts Notch signalling. Here we show in Drosophila that overexpressing the ß-subunit in the context of the complete PheRS leads to larval roaming, food avoidance, slow growth, and a developmental delay that can last several days and even prevents pupation. These behavioural and developmental phenotypes are induced by PheRS expression in CCHa2+ and Pros+ cells. Simultaneous expression of ß-PheRS, α-PheRS, and the appetite-inducing CCHa2 peptide rescued these phenotypes, linking this ß-PheRS activity to the appetite-controlling pathway. The fragmentation dynamic of the excessive ß-PheRS points to ß-PheRS fragments as possible candidate inducers of these phenotypes. Because fragmentation of human FARS has also been observed in human cells and mutations in human ß-PheRS (FARSB) can lead to problems in gaining weight, Drosophila ß-PheRS can also serve as a model for the human phenotype and possibly also for obesity.


Subject(s)
Amino Acyl-tRNA Synthetases , Phenylalanine-tRNA Ligase , Animals , Humans , Appetite/genetics , Drosophila/genetics , Drosophila/metabolism , Hormones , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism , RNA, Transfer
2.
Circulation ; 149(16): 1268-1284, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38362779

ABSTRACT

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a common heritable heart disease. Although HCM has been reported to be associated with many variants of genes involved in sarcomeric protein biomechanics, pathogenic genes have not been identified in patients with partial HCM. FARS2 (the mitochondrial phenylalanyl-tRNA synthetase), a type of mitochondrial aminoacyl-tRNA synthetase, plays a role in the mitochondrial translation machinery. Several variants of FARS2 have been suggested to cause neurological disorders; however, FARS2-associated diseases involving other organs have not been reported. We identified FARS2 as a potential novel pathogenic gene in cardiomyopathy and investigated its effects on mitochondrial homeostasis and the cardiomyopathy phenotype. METHODS: FARS2 variants in patients with HCM were identified using whole-exome sequencing, Sanger sequencing, molecular docking analyses, and cell model investigation. Fars2 conditional mutant (p.R415L) or knockout mice, fars2-knockdown zebrafish, and Fars2-knockdown neonatal rat ventricular myocytes were engineered to construct FARS2 deficiency models both in vivo and in vitro. The effects of FARS2 and its role in mitochondrial homeostasis were subsequently evaluated using RNA sequencing and mitochondrial functional analyses. Myocardial tissues from patients were used for further verification. RESULTS: We identified 7 unreported FARS2 variants in patients with HCM. Heart-specific Fars2-deficient mice presented cardiac hypertrophy, left ventricular dilation, progressive heart failure accompanied by myocardial and mitochondrial dysfunction, and a short life span. Heterozygous cardiac-specific Fars2R415L mice displayed a tendency to cardiac hypertrophy at age 4 weeks, accompanied by myocardial dysfunction. In addition, fars2-knockdown zebrafish presented pericardial edema and heart failure. FARS2 deficiency impaired mitochondrial homeostasis by directly blocking the aminoacylation of mt-tRNAPhe and inhibiting the synthesis of mitochondrial proteins, ultimately contributing to an imbalanced mitochondrial quality control system by accelerating mitochondrial hyperfragmentation and disrupting mitochondrion-related autophagy. Interfering with the mitochondrial quality control system using adeno-associated virus 9 or specific inhibitors mitigated the cardiac and mitochondrial dysfunction triggered by FARS2 deficiency by restoring mitochondrial homeostasis. CONCLUSIONS: Our findings unveil the previously unrecognized role of FARS2 in heart and mitochondrial homeostasis. This study may provide new insights into the molecular diagnosis and prevention of heritable cardiomyopathy as well as therapeutic options for FARS2-associated cardiomyopathy.


Subject(s)
Cardiomyopathy, Hypertrophic , Heart Failure , Mitochondrial Diseases , Phenylalanine-tRNA Ligase , Animals , Humans , Infant, Newborn , Mice , Rats , Cardiomyopathy, Hypertrophic/pathology , Heart Failure/pathology , Homeostasis , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/metabolism , Molecular Docking Simulation , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism , Zebrafish/genetics , Mutation
3.
BMC Neurol ; 24(1): 15, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38166857

ABSTRACT

BACKGROUND: Combined oxidative phosphorylation deficiency (COXPD) is a severe disorder with early onset and autosomal recessive inheritance, and has been divided into 51 types (COXPD1-COXPD51). COXPD14 is caused by a mutation in the FARS2 gene, which encodes mitochondrial phenylalanyl-tRNA synthetase (mt-PheRS), an enzyme that transfers phenylalanine to its cognate tRNA in mitochondria. Since the first case was reported in 2012, an increasing number of FARS2 variations have been subsequently identified, which present three main phenotypic manifestations: early onset epileptic encephalopathy, hereditary spastic paraplegia, and juvenile-onset epilepsy. To our knowledge, no adult cases have been reported in the literature. METHODS: We report in detail a case of genetically confirmed COXPD14 and review the relevant literature. RESULTS: Approximately 58 subjects with disease-causing variants of FARS2 have been reported, including 31 cases of early onset epileptic encephalopathy, 16 cases of hereditary spastic paraplegia, 3 cases of juvenile-onset epilepsy, and 8 cases of unknown phenotype. We report a case of autosomal recessive COXPD14 in an adult with status epilepticus as the only manifestation with a good prognosis, which is different from that in neonatal or infant patients reported in the literature. c.467C > T (p.T156M) has been previously reported, while c.119_120del (p.E40Vfs*87) is novel, and, both mutations are pathogenic. CONCLUSIONS: This case of autosomal recessive COXPD14 in an adult only presented as status epilepticus, which is different from the patients reported previously. Our study expands the mutation spectrum of FARS2, and we tended to define the phenotypes based on the clinical manifestation rather than the age of onset.


Subject(s)
Epilepsy , Mitochondrial Diseases , Phenylalanine-tRNA Ligase , Spastic Paraplegia, Hereditary , Status Epilepticus , Infant , Adult , Infant, Newborn , Humans , Spastic Paraplegia, Hereditary/genetics , Epilepsy/genetics , Mitochondrial Diseases/genetics , Mutation/genetics , Phenotype , Phenylalanine-tRNA Ligase/genetics , Mitochondrial Proteins/genetics
4.
BMC Med Genomics ; 16(1): 245, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37833669

ABSTRACT

BACKGROUND: Aminoacyl-tRNA synthetases (ARSs) are indispensable enzymes for protein biosynthesis in cells. The phenylalanyl-tRNA synthetase (FARS1) located in cytoplasm which consists of two FARS alpha subunits (FARSA) and two FARS beta subunits (FARSB). Autosomal recessive inheritance of pathogenic variants of FARSA or FARSB can result in defective FARS1 which are characterized by interstitial lung disease, liver disease, brain abnormalities, facial dysmorphism and growth restriction. METHODS: Exome sequencing was used to detect the candidate variants. The in silico prediction and expressional level analysis were performed to evaluate the pathogenicity of the variations. Additionally, we presented the patient's detailed clinical information and compared the clinical feature with other previously reported patients with FARSA-deficiency. RESULTS: We identified compound heterozygous rare missense variants (c.1172 T > C/ p.Leu391Pro and c.1211G > A/ p.Arg404His) in FARSA gene in a Chinese male patient. The protein structure prediction and the analysis of levels of FARSA and FARSB subunits indicated both variants pathogenic. Clinical feature review indicated inflammatory symptoms in young infants may be an additional key feature. Thyroid dysfunction should be considered as a phenotype with variable penetrance. CONCLUSIONS: Our results expanded the current phenotypic and genetic spectrum of FARSA-deficiency.


Subject(s)
Amino Acyl-tRNA Synthetases , Neurodevelopmental Disorders , Phenylalanine-tRNA Ligase , Infant , Humans , Male , Phenylalanine-tRNA Ligase/genetics , Amino Acyl-tRNA Synthetases/genetics , Mutation, Missense , Phenotype , Mutation
5.
Mol Genet Metab ; 140(3): 107657, 2023 11.
Article in English | MEDLINE | ID: mdl-37523899

ABSTRACT

FARS2 encodes the mitochondrial phenylalanyl-tRNA synthetase (mtPheRS), which is essential for charging mitochondrial (mt-) tRNAPhe with phenylalanine for use in intramitochondrial translation. Many biallelic, pathogenic FARS2 variants have been described previously, which are mostly associated with two distinct clinical phenotypes; an early onset epileptic mitochondrial encephalomyopathy or a later onset spastic paraplegia. In this study, we report on a patient who presented at 3 weeks of age with tachypnoea and poor feeding, which progressed to severe metabolic decompensation with lactic acidosis and seizure activity followed by death at 9 weeks of age. Rapid trio whole exome sequencing identified compound heterozygous FARS2 variants including a pathogenic exon 2 deletion on one allele and a rare missense variant (c.593G > T, p.(Arg198Leu)) on the other allele, necessitating further work to aid variant classification. Assessment of patient fibroblasts demonstrated severely decreased steady-state levels of mtPheRS, but no obvious defect in any components of the oxidative phosphorylation system. To investigate the potential pathogenicity of the missense variant, we determined its high-resolution crystal structure, demonstrating a local structural destabilization in the catalytic domain. Moreover, the R198L mutation reduced the thermal stability and impaired the enzymatic activity of mtPheRS due to a lower binding affinity for tRNAPhe and a slower turnover rate. Together these data confirm the pathogenicity of this FARS2 variant in causing early-onset mitochondrial epilepsy.


Subject(s)
Epilepsy , Mitochondrial Diseases , Phenylalanine-tRNA Ligase , Humans , Infant , Infant, Newborn , Epilepsy/pathology , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/genetics , Mutation , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/chemistry , RNA, Transfer/genetics , RNA, Transfer, Phe/metabolism
6.
J Phys Chem Lett ; 14(14): 3452-3460, 2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37010935

ABSTRACT

We propose an improved transfer entropy approach called the dynamic version of the force constant fitted Gaussian network model based on molecular dynamics ensemble (dfcfGNMMD) to explore the allosteric mechanism of human mitochondrial phenylalanyl-tRNA synthetase (hmPheRS), one of the aminoacyl-tRNA synthetases that play a crucial role in translation of the genetic code. The dfcfGNMMD method can provide reliable estimates of the transfer entropy and give new insights into the role of the anticodon binding domain in driving the catalytic domain in aminoacylation activity and into the effects of tRNA binding and residue mutation on the enzyme activity, revealing the causal mechanism of the allosteric communication in hmPheRS. In addition, we incorporate the residue dynamic and co-evolutionary information to further investigate the key residues in hmPheRS allostery. This study sheds light on the mechanisms of hmPheRS allostery and can provide important information for related drug design.


Subject(s)
Amino Acyl-tRNA Synthetases , Phenylalanine-tRNA Ligase , Humans , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism , Entropy , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Anticodon , Catalytic Domain
7.
Neuropediatrics ; 54(5): 351-355, 2023 10.
Article in English | MEDLINE | ID: mdl-36603837

ABSTRACT

OBJECTIVE: By loading transfer RNAs with their cognate amino acids, aminoacyl-tRNA synthetases (ARS) are essential for protein translation. Both cytosolic ARS1-deficiencies and mitochondrial ARS2 deficiencies can cause severe diseases. Amino acid supplementation has shown to positively influence the clinical course of four individuals with cytosolic ARS1 deficiencies. We hypothesize that this intervention could also benefit individuals with mitochondrial ARS2 deficiencies. METHODS: This study was designed as a N-of-1 trial. Daily oral L-phenylalanine supplementation was used in a 3-year-old girl with FARS2 deficiency. A period without supplementation was implemented to discriminate the effects of treatment from age-related developments and continuing physiotherapy. Treatment effects were measured through a physiotherapeutic testing battery, including movement assessment battery for children, dynamic gait index, gross motor function measure 66, and quality of life questionnaires. RESULTS: The individual showed clear improvement in all areas tested, especially in gross motor skills, movement abilities, and postural stability. In the period without supplementation, she lost newly acquired motor skills but regained these upon restarting supplementation. No adverse effects and good tolerance of treatment were observed. INTERPRETATION AND CONCLUSION: Our positive results encourage further studies both on L-phenylalanine for other individuals with FARS2 deficiency and the exploration of this treatment rationale for other ARS2 deficiencies. Additionally, treatment costs were relatively low at 1.10 €/day.


Subject(s)
Phenylalanine-tRNA Ligase , Child , Female , Humans , Child, Preschool , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/metabolism , Phenylalanine/metabolism , Quality of Life , Mitochondria/genetics , Mitochondria/metabolism , RNA, Transfer/metabolism , Mitochondrial Proteins/genetics
8.
Hum Cell ; 36(1): 121-131, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36205831

ABSTRACT

Hereditary spastic paraplegia (HSP) is a neurodegeneration disease, one of the reasons is caused by autosomal recessive missense mutation of the karyogene that encodes phenylalanyl-tRNA synthetase 2, mitochondrial (FARS2). However, the molecular mechanism underlying FARS2-mediated HSP progression is unknown. Mitochondrial phenylalanyl-tRNA synthetase gene (PheRS-m) is the Drosophila melanogaster homolog gene of human FARS2. This study constructed a Drosophila HSP missense mutation model and a PheRS-m knockout model. Some of the mutant fly phenotypes included developmental delay, shortened lifespan, wing-structure abnormalities and decreased mobility. RNA-sequencing results revealed a relationship between abnormal phenotypes and the hedgehog (Hh) pathway. A qRT-PCR assay was used to determine the key genes (ptc, hib, and slmb) of the Hh pathway that exhibited increased expression during different developmental stages. We demonstrated that Hh signaling transduction is negatively regulated during the developmental stages of PheRS-m mutants but positively regulated during adulthood. By inducing the agonist and inhibitor of Hh pathway in PheRS-m larvae, the developmental delay in mutants can be partly salvaged or postponed. Collectively, our findings indicate that Hh signaling negatively regulates the development of PheRS-m mutants, subsequently leading to developmental delay.


Subject(s)
Drosophila melanogaster , Hedgehog Proteins , Phenylalanine-tRNA Ligase , Animals , Base Sequence , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mitochondrial Proteins/genetics , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism
9.
Eur J Med Res ; 27(1): 184, 2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36155627

ABSTRACT

BACKGROUND: As a rare mitochondrial disease, combined oxidative phosphorylation deficiency 14 (COXPD14) is caused by biallelic variants in the phenylalanyl-tRNA synthetase 2, mitochondrial gene (FARS2) with clinical features of developmental delay, an elevated lactate level, early-onset encephalopathy, liver failure, and hypotonia. The objectives of this study were to analyze the clinical and molecular features of two Chinese siblings affected with COXPD14, and to review relevant literature. METHODS: Mutation screening was performed by whole exome sequencing (WES) in combination with Sanger sequencing validation to identify the disease-causing variants of the two patients. RESULTS: The two siblings presented with severe clinical features and both progressed aggressively and failed to survive after treatment abandonment. We identified two compound heterozygous FARS2 variants c.925G>A p.Gly309Ser and c.943G>C p.Gly315Arg in this proband, which were inherited from the unaffected father and mother, respectively. In addition, Sanger sequencing confirmed that the elder affected sister carried the same compound heterozygous variants. The c.925G>A p.Gly309Ser variant is known and commonly reported in COXPD14 patients, while c.943G>C p.Gly315Arg is a novel one. Neither of the variants was found in 100 Chinese healthy controls. Both variants were classified as "deleterious" and were located in the highly conserved regions of the protein. The above results suggested that the two variants were likely causative in this COXPD14-affected pedigree. CONCLUSIONS: Our study expands the mutation spectrum of FARS2 and highlights the importance of genetic testing in the diagnosis of diseases with a wide variety of phenotypes, especially in the differential diagnosis of diseases.


Subject(s)
Mitochondrial Diseases , Phenylalanine-tRNA Ligase , China , Humans , Lactates , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Mutation/genetics , Pedigree , Phenylalanine-tRNA Ligase/genetics , Siblings
10.
Orphanet J Rare Dis ; 17(1): 306, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35918773

ABSTRACT

BACKGROUND: Aminoacyl tRNA transferases play an essential role in protein biosynthesis, and variants of these enzymes result in various human diseases. FARSA, which encodes the α subunit of cytosolic phenylalanyl-tRNA synthetase, was recently reported as a suspected causal gene for multiorgan disorder. This study aimed to validate the pathogenicity of variants in the FARSA gene. RESULTS: Exome sequencing revealed novel compound heterozygous variants in FARSA, P347L and R475Q, from a patient who initially presented neonatal-onset failure to thrive, liver dysfunction, and frequent respiratory infections. His developmental milestones were nearly arrested, and the patient died at 28 months of age as a result of progressive hepatic and respiratory failure. The P347L variant was predicted to disrupt heterodimer interaction and failed to form a functional heterotetramer by structural and biochemical analyses. R475 is located at a highly conserved site and is reported to be involved in phenylalanine activation and transfer to tRNA. The R475Q mutant FARSA were co-purified with FARSB, but the mutant enzyme showed an approximately 36% reduction in activity in our assay relative to the wild-type protein. Additional functional analyses on variants from previous reports (N410K, F256L, R404C, E418D, and F277V) were conducted. The R404C variant from a patient waiting for organ transplantation also failed to form tetramers but the E418D, N410K, F256L, and F277V variants did not affect tetramer formation. In the functional assay, the N410K located at the phenylalanine-binding site exhibited no catalytic activity, whereas other variants (E418D, F256L and F277V) exhibited lower ATPase activity than wild-type FARSA at low phenylalanine concentrations. CONCLUSIONS: Our data demonstrated the pathogenicity of biallelic variants in FARSA and suggested the implication of hypomorphic variants in severe phenotypes.


Subject(s)
Phenylalanine-tRNA Ligase , Humans , Infant, Newborn , Mutation/genetics , Phenylalanine , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/genetics , RNA, Transfer/genetics , Exome Sequencing
11.
Nat Commun ; 13(1): 2593, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35546551

ABSTRACT

Migraine is a common complex disorder with a significant polygenic SNP heritability ([Formula: see text]). Here we utilise genome-wide association study (GWAS) summary statistics to study pleiotropy between blood proteins and migraine under the polygenic model. We estimate [Formula: see text] for 4625 blood protein GWASs and identify 325 unique proteins with a significant [Formula: see text] for use in subsequent genetic analyses. Pleiotropy analyses link 58 blood proteins to migraine risk at genome-wide, gene and/or single-nucleotide polymorphism levels-suggesting shared genetic influences or causal relationships. Notably, the identified proteins are largely distinct from migraine GWAS loci. We show that higher levels of DKK1 and PDGFB, and lower levels of FARS2, GSTA4 and CHIC2 proteins have a significant causal effect on migraine. The risk-increasing effect of DKK1 is particularly interesting-indicating a role for downregulation of ß-catenin-dependent Wnt signalling in migraine risk, suggesting Wnt activators that restore Wnt/ß-catenin signalling in brain could represent therapeutic tools against migraine.


Subject(s)
Migraine Disorders , Phenylalanine-tRNA Ligase , Wnt Signaling Pathway , beta Catenin , Blood Proteins/genetics , Genetic Pleiotropy , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Migraine Disorders/genetics , Migraine Disorders/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism , Polymorphism, Single Nucleotide , beta Catenin/genetics , beta Catenin/metabolism
12.
PLoS Genet ; 18(4): e1010185, 2022 04.
Article in English | MEDLINE | ID: mdl-35486661

ABSTRACT

The alpha subunit of the cytoplasmic Phenylalanyl tRNA synthetase (α-PheRS, FARSA in humans) displays cell growth and proliferation activities and its elevated levels can induce cell fate changes and tumor-like phenotypes that are neither dependent on the canonical function of charging tRNAPhe with phenylalanine nor on stimulating general translation. In intestinal stem cells of Drosophila midguts, α-PheRS levels are naturally slightly elevated and human FARSA mRNA levels are elevated in multiple cancers. In the Drosophila midgut model, elevated α-PheRS levels caused the accumulation of many additional proliferating cells resembling intestinal stem cells (ISCs) and enteroblasts (EBs). This phenotype partially resembles the tumor-like phenotype described as Notch RNAi phenotype for the same cells. Genetic interactions between α-PheRS and Notch suggest that their activities neutralize each other and that elevated α-PheRS levels attenuate Notch signaling when Notch induces differentiation into enterocytes, type II neuroblast stem cell proliferation, or transcription of a Notch reporter. These non-canonical functions all map to the N-terminal part of α-PheRS which accumulates naturally in the intestine. This truncated version of α-PheRS (α-S) also localizes to nuclei and displays weak sequence similarity to the Notch intracellular domain (NICD), suggesting that α-S might compete with the NICD for binding to a common target. Supporting this hypothesis, the tryptophan (W) residue reported to be key for the interaction between the NICD and the Su(H) BTD domain is not only conserved in α-PheRS and α-S, but also essential for attenuating Notch signaling.


Subject(s)
Phenylalanine-tRNA Ligase , Animals , Drosophila/genetics , Phenylalanine , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism , RNA, Transfer, Phe/chemistry , RNA, Transfer, Phe/metabolism
13.
FEBS Lett ; 596(7): 947-957, 2022 04.
Article in English | MEDLINE | ID: mdl-35038769

ABSTRACT

To ensure that correct amino acids are incorporated during protein synthesis, aminoacyl-tRNA synthetases (aaRSs) use proofreading mechanisms collectively referred to as editing. Although editing is important for viability, editing-deficient aaRSs have been identified in host-dependent organisms. In Mycoplasma mobile, editing-deficient PheRS and LeuRS have been identified. We characterized the amino acid activation site of MmPheRS and identified a previously unknown hyperaccurate mutation, L287F. Additionally, we report that m-Tyr, an oxidation byproduct of Phe which is toxic to editing-deficient cells, is poorly discriminated by MmPheRS activation and is not subjected to editing. Furthermore, expressing MmPheRS and the hyperaccurate variants renders Escherichia coli susceptible to m-Tyr stress, indicating that active site discrimination is insufficient in tolerating excess m-Tyr.


Subject(s)
Amino Acyl-tRNA Synthetases , Mycoplasma/enzymology , Phenylalanine-tRNA Ligase , Amino Acids , Amino Acyl-tRNA Synthetases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism
14.
Nucleic Acids Res ; 49(22): 13108-13121, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34878141

ABSTRACT

Mutations in genes encoding mitochondrial aminoacyl-tRNA synthetases are linked to diverse diseases. However, the precise mechanisms by which these mutations affect mitochondrial function and disease development are not fully understood. Here, we develop a Drosophila model to study the function of dFARS2, the Drosophila homologue of the mitochondrial phenylalanyl-tRNA synthetase, and further characterize human disease-associated FARS2 variants. Inactivation of dFARS2 in Drosophila leads to developmental delay and seizure. Biochemical studies reveal that dFARS2 is required for mitochondrial tRNA aminoacylation, mitochondrial protein stability, and assembly and enzyme activities of OXPHOS complexes. Interestingly, by modeling FARS2 mutations associated with human disease in Drosophila, we provide evidence that expression of two human FARS2 variants, p.G309S and p.D142Y, induces seizure behaviors and locomotion defects, respectively. Together, our results not only show the relationship between dysfunction of mitochondrial aminoacylation system and pathologies, but also illustrate the application of Drosophila model for functional analysis of human disease-causing variants.


Subject(s)
Developmental Disabilities/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Mitochondrial Proteins/genetics , Mutation , Phenylalanine-tRNA Ligase/genetics , RNA, Transfer/genetics , Seizures/genetics , Animals , Cell Line , Developmental Disabilities/enzymology , Disease Models, Animal , Drosophila Proteins/deficiency , Drosophila melanogaster/enzymology , Gene Knockdown Techniques , Humans , Microscopy, Electron, Transmission , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/deficiency , Oxidative Phosphorylation , Phenylalanine-tRNA Ligase/deficiency , RNA, Transfer/metabolism , Seizures/enzymology , Transfer RNA Aminoacylation
15.
Protein Sci ; 30(9): 1793-1803, 2021 09.
Article in English | MEDLINE | ID: mdl-34184352

ABSTRACT

Malaria is a parasitic illness caused by the genus Plasmodium from the apicomplexan phylum. Five plasmodial species of P. falciparum (Pf), P. knowlesi, P. malariae, P. ovale, and P. vivax (Pv) are responsible for causing malaria in humans. According to the World Malaria Report 2020, there were 229 million cases and ~ 0.04 million deaths of which 67% were in children below 5 years of age. While more than 3 billion people are at risk of malaria infection globally, antimalarial drugs are their only option for treatment. Antimalarial drug resistance keeps arising periodically and thus threatens the main line of malaria treatment, emphasizing the need to find new alternatives. The availability of whole genomes of P. falciparum and P. vivax has allowed targeting their unexplored plasmodial enzymes for inhibitor development with a focus on multistage targets that are crucial for parasite viability in both the blood and liver stages. Over the past decades, aminoacyl-tRNA synthetases (aaRSs) have been explored as anti-bacterial and anti-fungal drug targets, and more recently (since 2009) aaRSs are also the focus of antimalarial drug targeting. Here, we dissect the structure-based knowledge of the most advanced three aaRSs-lysyl- (KRS), prolyl- (PRS), and phenylalanyl- (FRS) synthetases in terms of development of antimalarial drugs. These examples showcase the promising potential of this family of enzymes to provide druggable targets that stall protein synthesis upon inhibition and thereby kill malaria parasites selectively.


Subject(s)
Amino Acyl-tRNA Synthetases/chemistry , Antimalarials/chemistry , Enzyme Inhibitors/chemistry , Lysine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/chemistry , Plasmodium falciparum/drug effects , Protozoan Proteins/chemistry , Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Antimalarials/pharmacology , Catalytic Domain , Drug Discovery , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , Lysine-tRNA Ligase/antagonists & inhibitors , Lysine-tRNA Ligase/genetics , Lysine-tRNA Ligase/metabolism , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Models, Molecular , Phenylalanine-tRNA Ligase/antagonists & inhibitors , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism , Plasmodium falciparum/chemistry , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
16.
J Gastroenterol Hepatol ; 36(11): 3113-3126, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34159625

ABSTRACT

BACKGROUND AND AIM: Metabolic reprogramming is characterized by dysregulated levels of metabolites and metabolic enzymes. Integrated metabolomic and transcriptomic data analysis can help to elucidate changes in the levels of metabolites and metabolic enzymes, screen the core metabolic pathways, and develop novel therapeutic strategies for cancer. METHODS: Here, the metabolome of gastric cancer tissues was determined using liquid chromatography-mass spectrometry. The transcriptome data from The Cancer Genome Atlas dataset were integrated with the liquid chromatography-mass spectrometry data to identify the common dysregulated gastric cancer-specific metabolic pathways. Additionally, the protein expression and clinical significance of key metabolic enzymes were examined using a gastric cancer tissue array. RESULTS: Metabolomic analysis of 16 gastric cancer tissues revealed that among the 15 dysregulated metabolomic pathways, the aminoacyl-tRNA biosynthesis pathway in the gastric tissues was markedly upregulated relative to that in the adjacent noncancerous tissues, which was consistent with the results of transcriptome analysis. Bioinformatic analysis revealed that among the key regulators in the aminoacyl-tRNA biosynthesis pathway, the expression levels of threonyl-tRNA synthetase (TARS) and phenylalanyl-tRNA synthetase (FARSB) were correlated with tumor grade and poor survival, respectively. Additionally, gastric tissue array data analysis indicated that TARS and FARSB were upregulated in gastric cancer tissues and were correlated with poor prognosis and tumor metastasis. CONCLUSIONS: This study demonstrated that the aminoacyl-tRNA biosynthesis pathway is upregulated in gastric cancer and both TARS and FARSB play key roles in the progression of gastric cancer. Additionally, a novel therapeutic strategy for gastric cancer was proposed that involves targeting the aminoacyl-tRNA biosynthesis pathway.


Subject(s)
Phenylalanine-tRNA Ligase , Stomach Neoplasms , Threonine-tRNA Ligase , Amino Acyl-tRNA Synthetases/biosynthesis , Amino Acyl-tRNA Synthetases/genetics , Humans , Metabolome , Phenylalanine-tRNA Ligase/biosynthesis , Phenylalanine-tRNA Ligase/genetics , RNA, Transfer/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Threonine-tRNA Ligase/biosynthesis , Threonine-tRNA Ligase/genetics , Transcriptome , Up-Regulation
17.
mBio ; 12(3): e0113221, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34126764

ABSTRACT

Bacterial antibiotic persistence occurs when bacteria are treated with an antibiotic and the majority of the population rapidly dies off, but a small subpopulation enters into a dormant, persistent state and evades death. Diverse pathways leading to nucleoside triphosphate (NTP) depletion and restricted translation have been implicated in persistence, suggesting alternative redundant routes may exist to initiate persister formation. To investigate the molecular mechanism of one such pathway, functional variants of an essential component of translation (phenylalanyl-tRNA synthetase [PheRS]) were used to study the effects of quality control on antibiotic persistence. Upon amino acid limitation, elevated PheRS quality control led to significant decreases in aminoacylated tRNAPhe accumulation and increased antibiotic persistence. This increase in antibiotic persistence was most pronounced (65-fold higher) when the relA-encoded tRNA-dependent stringent response was inactivated. The increase in persistence with elevated quality control correlated with ∼2-fold increases in the levels of the RNase MazF and the NTPase MazG and a 3-fold reduction in cellular NTP pools. These data reveal a mechanism for persister formation independent of the stringent response where reduced translation capacity, as indicated by reduced levels of aminoacylated tRNA, is accompanied by active reduction of cellular NTP pools which in turn triggers antibiotic persistence. IMPORTANCE Bacterial antibiotic persistence is a transient physiological state wherein cells become dormant and thereby evade being killed by antibiotics. Once the antibiotic is removed, bacterial persisters are able to resuscitate and repopulate. It is thought that antibiotic bacterial persisters may cause reoccurring infections in the clinical setting. The molecular triggers and pathways that cause bacteria to enter into the persister state are not fully understood. Our results suggest that accumulation of deacylated tRNA is a trigger for antibiotic persistence independent of the RelA-dependent stringent response, a pathway thought to be required for persistence in many organisms. Overall, this provides a mechanism where changes in translation quality control in response to physiological cues can directly modulate bacterial persistence.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Escherichia coli Proteins/metabolism , Escherichia coli/drug effects , Escherichia coli/metabolism , RNA, Transfer/metabolism , Transfer RNA Aminoacylation/drug effects , Amino Acids/metabolism , Drug Resistance, Bacterial/genetics , Drug Resistance, Bacterial/physiology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Microbial Viability , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/metabolism
18.
Mol Genet Metab ; 133(2): 222-229, 2021 06.
Article in English | MEDLINE | ID: mdl-33972171

ABSTRACT

BACKGROUND AND PURPOSE: Mitochondrial aminoacyl-tRNA synthetases-encoded by ARS2 genes-are evolutionarily conserved enzymes that catalyse the attachment of amino acids to their cognate tRNAs, ensuring the accuracy of the mitochondrial translation process. ARS2 gene mutations are associated with a wide range of clinical presentations affecting the CNS. METHODS: Two senior neuroradiologists analysed brain MRI of 25 patients (age range: 3 d-25 yrs.; 11 males; 14 females) with biallelic pathogenic variants of 11 ARS2 genes in a retrospective study conducted between 2002 and 2019. RESULTS: Though several combinations of brain MRI anomalies were highly suggestive of specific aetiologies (DARS2, EARS2, AARS2 and RARS2 mutations), our study detected no MRI pattern common to all patients. Stroke-like lesions were associated with pathogenic SARS2 and FARS2 variants. We also report early onset cerebellar atrophy and calcifications in AARS2 mutations, early white matter involvement in RARS2 mutations, and absent involvement of thalami in EARS2 mutations. Finally, our findings show that normal brain MRI results do not exclude the presence of ARS2 mutations: 5 patients with normal MRI images were carriers of pathogenic IARS2, YARS2, and FARS2 variants. CONCLUSION: Our study extends the spectrum of brain MRI anomalies associated with pathogenic ARS2 variants and suggests ARS2 mutations are largely underdiagnosed.


Subject(s)
Alanine-tRNA Ligase/genetics , Arginine-tRNA Ligase/genetics , Aspartate-tRNA Ligase/genetics , Brain/diagnostic imaging , Mitochondrial Proteins/genetics , Phenylalanine-tRNA Ligase/genetics , Adolescent , Adult , Amino Acyl-tRNA Synthetases/classification , Amino Acyl-tRNA Synthetases/genetics , Brain/pathology , Child , Child, Preschool , Female , Genetic Variation , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Mutation/genetics , Phenotype , Young Adult
19.
Clin Genet ; 99(6): 789-801, 2021 06.
Article in English | MEDLINE | ID: mdl-33598926

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) catalyze the first step of protein biosynthesis (canonical function) and have additional (non-canonical) functions outside of translation. Bi-allelic pathogenic variants in genes encoding ARSs are associated with various recessive mitochondrial and multisystem disorders. We describe here a multisystem clinical phenotype based on bi-allelic mutations in the two genes (FARSA, FARSB) encoding distinct subunits for tetrameric cytosolic phenylalanyl-tRNA synthetase (FARS1). Interstitial lung disease with cholesterol pneumonitis on histology emerged as an early characteristic feature and significantly determined disease burden. Additional clinical characteristics of the patients included neurological findings, liver dysfunction, and connective tissue, muscular and vascular abnormalities. Structural modeling of newly identified missense mutations in the alpha subunit of FARS1, FARSA, showed exclusive mapping to the enzyme's conserved catalytic domain. Patient-derived mutant cells displayed compromised aminoacylation activity in two cases, while remaining unaffected in another. Collectively, these findings expand current knowledge about the human ARS disease spectrum and support a loss of canonical and non-canonical function in FARS1-associated recessive disease.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Lung Diseases, Interstitial/genetics , Lung/pathology , Mutation/genetics , Phenylalanine-tRNA Ligase/genetics , Adolescent , Alleles , Child , Child, Preschool , Female , Genes, Recessive/genetics , Humans , Infant , Infant, Newborn , Male , Pedigree , Phenotype
20.
Eur J Hum Genet ; 29(3): 533-538, 2021 03.
Article in English | MEDLINE | ID: mdl-33168986

ABSTRACT

Mitochondrial translation is essential for the biogenesis of the mitochondrial oxidative phosphorylation system (OXPHOS) that synthesizes the bulk of ATP for the cell. Hypomorphic and loss-of-function variants in either mitochondrial DNA or in nuclear genes that encode mitochondrial translation factors can result in impaired OXPHOS biogenesis and mitochondrial diseases with variable clinical presentations. Compound heterozygous or homozygous missense and frameshift variants in the FARS2 gene, that encodes the mitochondrial phenylalanyl-tRNA synthetase, are commonly linked to either early-onset epileptic mitochondrial encephalopathy or spastic paraplegia. Here, we expand the genetic spectrum of FARS2-linked disease with three patients carrying novel compound heterozygous variants in the FARS2 gene and presenting with spastic tetraparesis, axial hypotonia and myoclonic epilepsy in two cases.


Subject(s)
Mitochondrial Proteins/genetics , Phenylalanine-tRNA Ligase/genetics , Spasms, Infantile/genetics , Adolescent , Female , Humans , Male , Mutation, Missense , Phenotype , Spasms, Infantile/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...