Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Mol Genet Metab ; 125(3): 281-291, 2018 11.
Article in English | MEDLINE | ID: mdl-30177229

ABSTRACT

An increasing number of mitochondrial diseases are found to be caused by pathogenic variants in nuclear encoded mitochondrial aminoacyl-tRNA synthetases. FARS2 encodes mitochondrial phenylalanyl-tRNA synthetase (mtPheRS) which transfers phenylalanine to its cognate tRNA in mitochondria. Since the first case was reported in 2012, a total of 21 subjects with FARS2 deficiency have been reported to date with a spectrum of disease severity that falls between two phenotypes; early onset epileptic encephalopathy and a less severe phenotype characterized by spastic paraplegia. In this report, we present an additional 15 individuals from 12 families who are mostly Arabs homozygous for the pathogenic variant Y144C, which is associated with the more severe early onset phenotype. The total number of unique pathogenic FARS2 variants known to date is 21 including three different partial gene deletions reported in four individuals. Except for the large deletions, all variants but two (one in-frame deletion of one amino acid and one splice-site variant) are missense. All large deletions and the single splice-site variant are in trans with a missense variant. This suggests that complete loss of function may be incompatible with life. In this report, we also review structural, functional, and evolutionary significance of select FARS2 pathogenic variants reported here.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Phenylalanine-tRNA Ligase/genetics , Adolescent , Adult , Amino Acyl-tRNA Synthetases/deficiency , Child , Child, Preschool , Female , Gene Deletion , Humans , Male , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/pathology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/deficiency , Mutation/genetics , Paraplegia/genetics , Paraplegia/pathology , Phenylalanine/genetics , Phenylalanine/metabolism , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/deficiency , Protein Isoforms/genetics , Structure-Activity Relationship , Young Adult
3.
Mol Genet Metab ; 122(4): 172-181, 2017 12.
Article in English | MEDLINE | ID: mdl-29126765

ABSTRACT

Mutations in FARS2 are known to cause dysfunction of mitochondrial translation due to deficient aminoacylation of the mitochondrial phenylalanine tRNA. Here, we report three novel mutations in FARS2 found in two patients in a compound heterozygous state. The missense mutation c.1082C>T (p.Pro361Leu) was detected in both patients. The mutations c.461C>T (p.Ala154Val) and c.521_523delTGG (p.Val174del) were each detected in one patient. We report abnormal in vitro aminoacylation assays as a functional validation of the molecular genetic findings. Based on the phenotypic data of previously reported subjects and the two subjects reported here, we conclude that FARS2 deficiency can be associated with two phenotypes: (i) an epileptic phenotype, and (ii) a spastic paraplegia phenotype.


Subject(s)
Epilepsy/genetics , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Phenotype , Phenylalanine-tRNA Ligase/deficiency , Phenylalanine-tRNA Ligase/genetics , Spastic Paraplegia, Hereditary/genetics , Adolescent , Amino Acyl-tRNA Synthetases/metabolism , Aminoacylation , Brain/diagnostic imaging , Cells, Cultured , Exome , Female , Fibroblasts/metabolism , Heterozygote , Humans , Infant , Magnetic Resonance Imaging , Male , Mitochondria/enzymology , Mitochondria/metabolism , Muscle, Skeletal/pathology , Mutation, Missense/genetics , Oxygen Consumption , RNA, Transfer/metabolism , Sequence Analysis, DNA
4.
Nat Commun ; 5: 5650, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-25427601

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) constitute a family of ubiquitously expressed essential enzymes that ligate amino acids to their cognate tRNAs for protein synthesis. Recently, aaRS mutations have been linked to various human diseases; however, how these mutations lead to diseases has remained unclear. In order to address the importance of aminoacylation fidelity in multicellular organisms, we generated an amino-acid double-sieving model in Drosophila melanogaster using phenylalanyl-tRNA synthetase (PheRS). Double-sieving-defective mutations dramatically misacylate non-cognate Tyr, induce protein mistranslation and cause endoplasmic reticulum stress in flies. Mutant adults exhibit many defects, including loss of neuronal cells, impaired locomotive performance, shortened lifespan and smaller organ size. At the cellular level, the mutations reduce cell proliferation and promote cell death. Our results also reveal the particular importance of the first amino-acid recognition sieve. Overall, these findings provide new mechanistic insights into how malfunctioning of aaRSs can cause diseases.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/growth & development , Phenylalanine-tRNA Ligase/genetics , Protein Biosynthesis , Animals , Cell Death , Cell Proliferation , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Endoplasmic Reticulum Stress , Gene Knockout Techniques , Mutation , Phenylalanine-tRNA Ligase/deficiency
5.
Microbiologyopen ; 3(3): 369-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24811065

ABSTRACT

The pheS5 Ts mutant of Escherichia coli defined by a G293 → A293 transition, which is responsible for thermosensitive Phenylalanyl-tRNA synthetase has been well studied at both biochemical and molecular level but genetic analyses pertaining to suppressors of pheS5 were hard to come by. Here we have systematically analyzed a spectrum of Temperature-insensitive derivatives isolated from pheS5 Ts mutant and identified two intragenic suppressors affecting the same base pair coordinate G673 (pheS19 defines G673 → T673 ; Gly225 → Cys225 and pheS28 defines G673 → C673 ; Gly225 → Arg225). In fact in the third derivative, the intragenic suppressor originally named pheS43 (G673 → C673 transversion) is virtually same as pheS28. In the fourth case, the very pheS5 lesion itself has got changed from A293 → T293 (named pheS40). Cloning of pheS(+), pheS5, pheS5-pheS19, pheS5-pheS28 alleles into pBR322 and introduction of these clones into pheS5 mutant revealed that excess of double mutant protein is not at all good for the survival of cells at 42°C. These results clearly indicate a pivotal role for Gly225 in the structural/functional integrity of alpha subunit of E. coli PheRS enzyme and it is proposed that G673 might define a hot spot for intragenic suppressors of pheS5.


Subject(s)
Escherichia coli/enzymology , Escherichia coli/genetics , Phenylalanine-tRNA Ligase/deficiency , Phenylalanine-tRNA Ligase/genetics , Suppression, Genetic , Escherichia coli/physiology , Genetic Complementation Test , Microbial Viability , Mutant Proteins/genetics , Mutant Proteins/metabolism , Point Mutation , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...