Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 105
Filtrer
1.
Neurobiol Dis ; 195: 106501, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38583640

RÉSUMÉ

Charcot-Marie-Tooth disease (CMT) is a genetic peripheral neuropathy caused by mutations in many functionally diverse genes. The aminoacyl-tRNA synthetase (ARS) enzymes, which transfer amino acids to partner tRNAs for protein synthesis, represent the largest protein family genetically linked to CMT aetiology, suggesting pathomechanistic commonalities. Dominant intermediate CMT type C (DI-CMTC) is caused by YARS1 mutations driving a toxic gain-of-function in the encoded tyrosyl-tRNA synthetase (TyrRS), which is mediated by exposure of consensus neomorphic surfaces through conformational changes of the mutant protein. In this study, we first showed that human DI-CMTC-causing TyrRSE196K mis-interacts with the extracellular domain of the BDNF receptor TrkB, an aberrant association we have previously characterised for several mutant glycyl-tRNA synthetases linked to CMT type 2D (CMT2D). We then performed temporal neuromuscular assessments of YarsE196K mice modelling DI-CMT. We determined that YarsE196K homozygotes display a selective, age-dependent impairment in in vivo axonal transport of neurotrophin-containing signalling endosomes, phenocopying CMT2D mice. This impairment is replicated by injection of recombinant TyrRSE196K, but not TyrRSWT, into muscles of wild-type mice. Augmenting BDNF in DI-CMTC muscles, through injection of recombinant protein or muscle-specific gene therapy, resulted in complete axonal transport correction. Therefore, this work identifies a non-cell autonomous pathomechanism common to ARS-related neuropathies, and highlights the potential of boosting BDNF levels in muscles as a therapeutic strategy.


Sujet(s)
Transport axonal , Facteur neurotrophique dérivé du cerveau , Maladie de Charcot-Marie-Tooth , Modèles animaux de maladie humaine , Animaux , Maladie de Charcot-Marie-Tooth/génétique , Maladie de Charcot-Marie-Tooth/métabolisme , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/génétique , Souris , Tyrosine-tRNA ligase/génétique , Tyrosine-tRNA ligase/métabolisme , Humains , Souris transgéniques , Muscles squelettiques/métabolisme , Récepteur trkB/métabolisme , Récepteur trkB/génétique , Mutation
2.
bioRxiv ; 2024 Mar 11.
Article de Anglais | MEDLINE | ID: mdl-38559020

RÉSUMÉ

Charcot-Marie-Tooth disease (CMT) is a genetic peripheral neuropathy caused by mutations in many functionally diverse genes. The aminoacyl-tRNA synthetase (ARS) enzymes, which transfer amino acids to partner tRNAs for protein synthesis, represent the largest protein family genetically linked to CMT aetiology, suggesting pathomechanistic commonalities. Dominant intermediate CMT type C (DI-CMTC) is caused by YARS1 mutations driving a toxic gain-of-function in the encoded tyrosyl-tRNA synthetase (TyrRS), which is mediated by exposure of consensus neomorphic surfaces through conformational changes of the mutant protein. In this study, we first showed that human DI-CMTC-causing TyrRSE196K mis-interacts with the extracellular domain of the BDNF receptor TrkB, an aberrant association we have previously characterised for several mutant glycyl-tRNA synthetases linked to CMT type 2D (CMT2D). We then performed temporal neuromuscular assessments of YarsE196K mice modelling DI-CMT. We determined that YarsE196K homozygotes display a selective, age-dependent impairment in in vivo axonal transport of neurotrophin-containing signalling endosomes, phenocopying CMT2D mice. This impairment is replicated by injection of recombinant TyrRSE196K, but not TyrRSWT, into muscles of wild-type mice. Augmenting BDNF in DI-CMTC muscles, through injection of recombinant protein or muscle-specific gene therapy, resulted in complete axonal transport correction. Therefore, this work identifies a non-cell autonomous pathomechanism common to ARS-related neuropathies, and highlights the potential of boosting BDNF levels in muscles as a therapeutic strategy.

3.
Nucleic Acids Res ; 51(19): 10768-10781, 2023 10 27.
Article de Anglais | MEDLINE | ID: mdl-37739431

RÉSUMÉ

Translational readthrough of UGA stop codons by selenocysteine-specific tRNA (tRNASec) enables the synthesis of selenoproteins. Seryl-tRNA synthetase (SerRS) charges tRNASec with serine, which is modified into selenocysteine and delivered to the ribosome by a designated elongation factor (eEFSec in eukaryotes). Here we found that components of the human selenocysteine incorporation machinery (SerRS, tRNASec, and eEFSec) also increased translational readthrough of non-selenocysteine genes, including VEGFA, to create C-terminally extended isoforms. SerRS recognizes target mRNAs through a stem-loop structure that resembles the variable loop of its cognate tRNAs. This function of SerRS depends on both its enzymatic activity and a vertebrate-specific domain. Through eCLIP-seq, we identified additional SerRS-interacting mRNAs as potential readthrough genes. Moreover, SerRS overexpression was sufficient to reverse premature termination caused by a pathogenic nonsense mutation. Our findings expand the repertoire of selenoprotein biosynthesis machinery and suggest an avenue for therapeutic targeting of nonsense mutations using endogenous factors.


Sujet(s)
Biosynthèse des protéines , Serine-tRNA ligase , Humains , Codon non-sens , Codon stop , ARN messager/métabolisme , Sélénocystéine/génétique , Sélénocystéine/métabolisme , Sélénoprotéines/génétique , Serine-tRNA ligase/génétique
4.
Nucleic Acids Res ; 51(18): 10001-10010, 2023 Oct 13.
Article de Anglais | MEDLINE | ID: mdl-37638745

RÉSUMÉ

Through their aminoacylation reactions, aminoacyl tRNA-synthetases (aaRS) establish the rules of the genetic code throughout all of nature. During their long evolution in eukaryotes, additional domains and splice variants were added to what is commonly a homodimeric or monomeric structure. These changes confer orthogonal functions in cellular activities that have recently been uncovered. An unusual exception to the familiar architecture of aaRSs is the heterodimeric metazoan mitochondrial SerRS. In contrast to domain additions or alternative splicing, here we show that heterodimeric metazoan mitochondrial SerRS arose from its homodimeric ancestor not by domain additions, but rather by collapse of an entire domain (in one subunit) and an active site ablation (in the other). The collapse/ablation retains aminoacylation activity while creating a new surface, which is necessary for its orthogonal function. The results highlight a new paradigm for repurposing a member of the ancient tRNA synthetase family.


Sujet(s)
Serine-tRNA ligase , Animaux , Amino acyl-tRNA synthetases/métabolisme , Domaine catalytique , Serine-tRNA ligase/composition chimique , Serine-tRNA ligase/métabolisme
5.
Cell Rep ; 42(6): 112632, 2023 06 27.
Article de Anglais | MEDLINE | ID: mdl-37314928

RÉSUMÉ

Various stress conditions are signaled through phosphorylation of translation initiation factor eukaryotic initiation factor 2α (eIF2α) to inhibit global translation while selectively activating transcription factor ATF4 to aid cell survival and recovery. However, this integrated stress response is acute and cannot resolve lasting stress. Here, we report that tyrosyl-tRNA synthetase (TyrRS), a member of the aminoacyl-tRNA synthetase family that responds to diverse stress conditions through cytosol-nucleus translocation to activate stress-response genes, also inhibits global translation. However, it occurs at a later stage than eIF2α/ATF4 and mammalian target of rapamycin (mTOR) responses. Excluding TyrRS from the nucleus over-activates translation and increases apoptosis in cells under prolonged oxidative stress. Nuclear TyrRS transcriptionally represses translation genes by recruiting TRIM28 and/or NuRD complex. We propose that TyrRS, possibly along with other family members, can sense a variety of stress signals through intrinsic properties of this enzyme and strategically located nuclear localization signal and integrate them by nucleus translocation to effect protective responses against chronic stress.


Sujet(s)
Tyrosine-tRNA ligase , Tyrosine-tRNA ligase/génétique , Tyrosine-tRNA ligase/métabolisme , Transport des protéines , Phosphorylation , Signaux de localisation nucléaire , Stress oxydatif
7.
JCI Insight ; 8(9)2023 05 08.
Article de Anglais | MEDLINE | ID: mdl-36928301

RÉSUMÉ

Gain-of-function mutations in the housekeeping gene GARS1, which lead to the expression of toxic versions of glycyl-tRNA synthetase (GlyRS), cause the selective motor and sensory pathology characterizing Charcot-Marie-Tooth disease (CMT). Aberrant interactions between GlyRS mutants and different proteins, including neurotrophin receptor tropomyosin receptor kinase receptor B (TrkB), underlie CMT type 2D (CMT2D); however, our pathomechanistic understanding of this untreatable peripheral neuropathy remains incomplete. Through intravital imaging of the sciatic nerve, we show that CMT2D mice displayed early and persistent disturbances in axonal transport of neurotrophin-containing signaling endosomes in vivo. We discovered that brain-derived neurotrophic factor (BDNF)/TrkB impairments correlated with transport disruption and overall CMT2D neuropathology and that inhibition of this pathway at the nerve-muscle interface perturbed endosome transport in wild-type axons. Accordingly, supplementation of muscles with BDNF, but not other neurotrophins, completely restored physiological axonal transport in neuropathic mice. Together, these findings suggest that selectively targeting muscles with BDNF-boosting therapies could represent a viable therapeutic strategy for CMT2D.


Sujet(s)
Maladie de Charcot-Marie-Tooth , Souris , Animaux , Maladie de Charcot-Marie-Tooth/génétique , Maladie de Charcot-Marie-Tooth/anatomopathologie , Transport axonal/génétique , Facteur neurotrophique dérivé du cerveau/génétique , Mutation
8.
Nat Commun ; 14(1): 999, 2023 03 08.
Article de Anglais | MEDLINE | ID: mdl-36890170

RÉSUMÉ

Dominant mutations in tyrosyl-tRNA synthetase (YARS1) and six other tRNA ligases cause Charcot-Marie-Tooth peripheral neuropathy (CMT). Loss of aminoacylation is not required for their pathogenicity, suggesting a gain-of-function disease mechanism. By an unbiased genetic screen in Drosophila, we link YARS1 dysfunction to actin cytoskeleton organization. Biochemical studies uncover yet unknown actin-bundling property of YARS1 to be enhanced by a CMT mutation, leading to actin disorganization in the Drosophila nervous system, human SH-SY5Y neuroblastoma cells, and patient-derived fibroblasts. Genetic modulation of F-actin organization improves hallmark electrophysiological and morphological features in neurons of flies expressing CMT-causing YARS1 mutations. Similar beneficial effects are observed in flies expressing a neuropathy-causing glycyl-tRNA synthetase. Hence, in this work, we show that YARS1 is an evolutionary-conserved F-actin organizer which links the actin cytoskeleton to tRNA-synthetase-induced neurodegeneration.


Sujet(s)
Actines , Tyrosine-tRNA ligase , Animaux , Humains , Actines/métabolisme , Maladie de Charcot-Marie-Tooth/génétique , Drosophila/génétique , Glycine-tRNA ligase/génétique , Mutation , ARN de transfert , Tyrosine-tRNA ligase/génétique , Tyrosine-tRNA ligase/métabolisme , Lignée cellulaire tumorale
9.
Sci Adv ; 9(6): eadf1027, 2023 02 10.
Article de Anglais | MEDLINE | ID: mdl-36753552

RÉSUMÉ

As a class of essential enzymes in protein translation, aminoacyl-transfer RNA (tRNA) synthetases (aaRSs) are organized into two classes of 10 enzymes each, based on two conserved active site architectures. The (αß)2 glycyl-tRNA synthetase (GlyRS) in many bacteria is an orphan aaRS whose sequence and unprecedented X-shaped structure are distinct from those of all other aaRSs, including many other bacterial and all eukaryotic GlyRSs. Here, we report a cocrystal structure to elucidate how the orphan GlyRS kingdom specifically recognizes its substrate tRNA. This structure is sharply different from those of other aaRS-tRNA complexes but conforms to the clash-free, cross-class aaRS-tRNA docking found with conventional structures and reinforces the class-reconstruction paradigm. In addition, noteworthy, the X shape of orphan GlyRS is condensed with the largest known spatial rearrangement needed by aaRSs to capture tRNAs, which suggests potential nonactive site targets for aaRS-directed antibiotics, instead of less differentiated hard-to-drug active site locations.


Sujet(s)
Amino acyl-tRNA synthetases , Glycine-tRNA ligase , Glycine-tRNA ligase/génétique , Glycine-tRNA ligase/composition chimique , Glycine-tRNA ligase/métabolisme , Amino acyl-tRNA synthetases/composition chimique , Amino acyl-tRNA synthetases/génétique , Amino acyl-tRNA synthetases/métabolisme , Ligases/métabolisme , ARN de transfert , Domaine catalytique
10.
Trends Biochem Sci ; 48(3): 288-302, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36280495

RÉSUMÉ

Antisynthetase syndrome (ASSD) is an autoimmune disease characterized by circulating autoantibodies against one of eight aminoacyl-tRNA synthetases (aaRSs). Although these autoantibodies are believed to play critical roles in ASSD pathogenesis, the nature of their roles remains unclear. Here we describe ASSD pathogenesis and discuss ASSD-linked aaRSs - from the WHEP domain that may impart immunogenicity to the role of tRNA in eliciting the innate immune response and the secretion of aaRSs from cells. Through these explorations, we propose that ASSD pathogenesis involves the tissue-specific secretion of aaRSs and that extracellular tRNAs or tRNA fragments and their ability to engage Toll-like receptor signaling may be important disease factors.


Sujet(s)
Amino acyl-tRNA synthetases , Myosite , Humains , Amino acyl-tRNA synthetases/génétique , ARN de transfert/génétique , Autoanticorps
11.
Proc Natl Acad Sci U S A ; 119(48): e2212659119, 2022 11 29.
Article de Anglais | MEDLINE | ID: mdl-36409883

RÉSUMÉ

Platelets play a role not only in hemostasis and thrombosis, but also in inflammation and innate immunity. We previously reported that an activated form of tyrosyl-tRNA synthetase (YRSACT) has an extratranslational activity that enhances megakaryopoiesis and platelet production in mice. Here, we report that YRSACT mimics inflammatory stress inducing a unique megakaryocyte (MK) population with stem cell (Sca1) and myeloid (F4/80) markers through a mechanism dependent on Toll-like receptor (TLR) activation and type I interferon (IFN-I) signaling. This mimicry of inflammatory stress by YRSACT was studied in mice infected by lymphocytic choriomeningitis virus (LCMV). Using Sca1/EGFP transgenic mice, we demonstrated that IFN-I induced by YRSACT or LCMV infection suppressed normal hematopoiesis while activating an alternative pathway of thrombopoiesis. Platelets of inflammatory origin (Sca1/EGFP+) were a relevant proportion of those circulating during recovery from thrombocytopenia. Analysis of these "inflammatory" MKs and platelets suggested their origin in myeloid/MK-biased hematopoietic stem cells (HSCs) that bypassed the classical MK-erythroid progenitor (MEP) pathway to replenish platelets and promote recovery from thrombocytopenia. Notably, inflammatory platelets displayed enhanced agonist-induced activation and procoagulant activities. Moreover, myeloid/MK-biased progenitors and MKs were mobilized from the bone marrow, as evidenced by their presence in the lung microvasculature within fibrin-containing microthrombi. Our results define the function of YRSACT in platelet generation and contribute to elucidate platelet alterations in number and function during viral infection.


Sujet(s)
Ataxies spinocérébelleuses , Thrombopénie , Thrombose , Tyrosine-tRNA ligase , Maladies virales , Souris , Animaux , Thrombopoïèse , Souris transgéniques
12.
Nat Commun ; 13(1): 4188, 2022 07 20.
Article de Anglais | MEDLINE | ID: mdl-35858913

RÉSUMÉ

The formation of a functional blood vessel network relies on the ability of endothelial cells (ECs) to dynamically rearrange their adhesive contacts in response to blood flow and guidance cues, such as vascular endothelial growth factor-A (VEGF-A) and class 3 semaphorins (SEMA3s). Neuropilin 1 (NRP1) is essential for blood vessel development, independently of its ligands VEGF-A and SEMA3, through poorly understood mechanisms. Grounding on unbiased proteomic analysis, we report here that NRP1 acts as an endocytic chaperone primarily for adhesion receptors on the surface of unstimulated ECs. NRP1 localizes at adherens junctions (AJs) where, interacting with VE-cadherin, promotes its basal internalization-dependent turnover and favors vascular permeability initiated by histamine in both cultured ECs and mice. We identify a splice variant of tryptophanyl-tRNA synthetase (mini-WARS) as an unconventionally secreted extracellular inhibitory ligand of NRP1 that, by stabilizing it at the AJs, slows down both VE-cadherin turnover and histamine-elicited endothelial leakage. Thus, our work shows a role for NRP1 as a major regulator of AJs plasticity and reveals how mini-WARS acts as a physiological NRP1 inhibitory ligand in the control of VE-cadherin endocytic turnover and vascular permeability.


Sujet(s)
Neuropiline 1 , Tryptophane-tRNA ligase , Jonctions adhérentes/métabolisme , Animaux , Antigènes CD , Cadhérines/génétique , Perméabilité capillaire , Cellules endothéliales/métabolisme , Histamine , Ligands , Souris , Neuropiline 1/génétique , Neuropiline 1/métabolisme , Protéomique , Tryptophane-tRNA ligase/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme
13.
Signal Transduct Target Ther ; 6(1): 303, 2021 08 16.
Article de Anglais | MEDLINE | ID: mdl-34400610

RÉSUMÉ

Abnormally enhanced de novo lipid biosynthesis has been increasingly realized to play crucial roles in the initiation and progression of varieties of cancers including breast cancer. However, the mechanisms underlying the dysregulation of lipid biosynthesis in breast cancer remain largely unknown. Here, we reported that seryl tRNA synthetase (SerRS), a key enzyme for protein biosynthesis, could translocate into the nucleus in a glucose-dependent manner to suppress key genes involved in the de novo lipid biosynthesis. In normal mammary gland epithelial cells glucose can promote the nuclear translocation of SerRS by increasing the acetylation of SerRS at lysine 323. In SerRS knock-in mice bearing acetylation-defective lysine to arginine mutation, we observed increased body weight and adipose tissue mass. In breast cancer cells the acetylation and nuclear translocation of SerRS are greatly inhibited. Overexpression of SerRS, in particularly the acetylation-mimetic lysine to glutamine mutant, dramatically inhibits the de novo lipid synthesis and hence greatly suppresses the proliferation of breast cancer cells and the growth of breast cancer xenografts in mice. We further identified that HDAC4 and HDAC5 regulated the acetylation and nuclear translocation of SerRS. Thus, we identified a SerRS-meditated inhibitory pathway in glucose-induced lipid biosynthesis, which is dysregulated in breast cancer.


Sujet(s)
Tumeurs du sein/métabolisme , Glucose/génétique , Lipides/génétique , Serine-tRNA ligase/génétique , Acétylation , Transport nucléaire actif/génétique , Tissu adipeux/métabolisme , Séquence d'acides aminés/génétique , Animaux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Femelle , Régulation de l'expression des gènes tumoraux/génétique , Techniques de knock-in de gènes , Glucose/métabolisme , Hétérogreffes , Histone deacetylases/génétique , Humains , Lipides/biosynthèse , Souris , Serine-tRNA ligase/métabolisme , Spécificité du substrat/génétique
14.
Nucleic Acids Res ; 49(17): 10106-10119, 2021 09 27.
Article de Anglais | MEDLINE | ID: mdl-34390350

RÉSUMÉ

AaRSs (aminoacyl-tRNA synthetases) group into two ten-member classes throughout evolution, with unique active site architectures defining each class. Most are monomers or homodimers but, for no apparent reason, many bacterial GlyRSs are heterotetramers consisting of two catalytic α-subunits and two tRNA-binding ß-subunits. The heterotetrameric GlyRS from Escherichia coli (EcGlyRS) was historically tested whether its α- and ß-polypeptides, which are encoded by a single mRNA with a gap of three in-frame codons, are replaceable by a single chain. Here, an unprecedented X-shaped structure of EcGlyRS shows wide separation of the abutting chain termini seen in the coding sequences, suggesting strong pressure to avoid a single polypeptide format. The structure of the five-domain ß-subunit is unique across all aaRSs in current databases, and structural analyses suggest these domains play different functions on α-subunit binding, ATP coordination and tRNA recognition. Moreover, the X-shaped architecture of EcGlyRS largely fits with a model for how two classes of tRNA synthetases arose, according to whether enzymes from opposite classes can simultaneously co-dock onto separate faces of the same tRNA acceptor stem. While heterotetrameric GlyRS remains the last structurally uncharacterized member of aaRSs, our study contributes to a better understanding of this ancient and essential enzyme family.


Sujet(s)
Domaine catalytique/génétique , Escherichia coli/génétique , Glycine-tRNA ligase/génétique , ARN de transfert de la glycine/composition chimique , Adénosine triphosphate/métabolisme , Cristallographie aux rayons X , Glycine/composition chimique , Modèles moléculaires , ARN de transfert de la glycine/génétique
15.
Nat Cell Biol ; 23(4): 424-436, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33820973

RÉSUMÉ

Although high-throughput RNA sequencing (RNA-seq) has greatly advanced small non-coding RNA (sncRNA) discovery, the currently widely used complementary DNA library construction protocol generates biased sequencing results. This is partially due to RNA modifications that interfere with adapter ligation and reverse transcription processes, which prevent the detection of sncRNAs bearing these modifications. Here, we present PANDORA-seq (panoramic RNA display by overcoming RNA modification aborted sequencing), employing a combinatorial enzymatic treatment to remove key RNA modifications that block adapter ligation and reverse transcription. PANDORA-seq identified abundant modified sncRNAs-mostly transfer RNA-derived small RNAs (tsRNAs) and ribosomal RNA-derived small RNAs (rsRNAs)-that were previously undetected, exhibiting tissue-specific expression across mouse brain, liver, spleen and sperm, as well as cell-specific expression across embryonic stem cells (ESCs) and HeLa cells. Using PANDORA-seq, we revealed unprecedented landscapes of microRNA, tsRNA and rsRNA dynamics during the generation of induced pluripotent stem cells. Importantly, tsRNAs and rsRNAs that are downregulated during somatic cell reprogramming impact cellular translation in ESCs, suggesting a role in lineage differentiation.


Sujet(s)
Maturation post-transcriptionnelle des ARN/génétique , Petit ARN non traduit/génétique , RNA-Seq , Transcriptome/génétique , ADN complémentaire/génétique , Cellules HeLa , Humains , microARN/génétique , ARN ribosomique/génétique
18.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article de Anglais | MEDLINE | ID: mdl-33753480

RÉSUMÉ

Through dominant mutations, aminoacyl-tRNA synthetases constitute the largest protein family linked to Charcot-Marie-Tooth disease (CMT). An example is CMT subtype 2N (CMT2N), caused by individual mutations spread out in AlaRS, including three in the aminoacylation domain, thereby suggesting a role for a tRNA-charging defect. However, here we found that two are aminoacylation defective but that the most widely distributed R329H is normal as a purified protein in vitro and in unfractionated patient cell samples. Remarkably, in contrast to wild-type (WT) AlaRS, all three mutant proteins gained the ability to interact with neuropilin 1 (Nrp1), the receptor previously linked to CMT pathogenesis in GlyRS. The aberrant AlaRS-Nrp1 interaction is further confirmed in patient samples carrying the R329H mutation. However, CMT2N mutations outside the aminoacylation domain do not induce the Nrp1 interaction. Detailed biochemical and biophysical investigations, including X-ray crystallography, small-angle X-ray scattering, hydrogen-deuterium exchange (HDX), switchSENSE hydrodynamic diameter determinations, and protease digestions reveal a mutation-induced structural loosening of the aminoacylation domain that correlates with the Nrp1 interaction. The b1b2 domains of Nrp1 are responsible for the interaction with R329H AlaRS. The results suggest Nrp1 is more broadly associated with CMT-associated members of the tRNA synthetase family. Moreover, we revealed a distinct structural loosening effect induced by a mutation in the editing domain and a lack of conformational impact with C-Ala domain mutations, indicating mutations in the same protein may cause neuropathy through different mechanisms. Our results show that, as with other CMT-associated tRNA synthetases, aminoacylation per se is not relevant to the pathology.


Sujet(s)
Alanine-tRNA ligase/métabolisme , Maladie de Charcot-Marie-Tooth/génétique , Neuropiline 1/métabolisme , Alanine-tRNA ligase/composition chimique , Alanine-tRNA ligase/génétique , Aminoacylation/génétique , Cellules cultivées , Maladie de Charcot-Marie-Tooth/sang , Cristallographie aux rayons X , Mesure d'échange de deutérium , Humains , Lymphocytes , Mutation , Neuropiline 1/génétique , Culture de cellules primaires , Liaison aux protéines/génétique , Domaines protéiques/génétique , Protéines recombinantes/génétique , Protéines recombinantes/isolement et purification , Protéines recombinantes/métabolisme , Protéines recombinantes/ultrastructure , Diffusion aux petits angles
19.
Nat Commun ; 12(1): 1616, 2021 03 12.
Article de Anglais | MEDLINE | ID: mdl-33712620

RÉSUMÉ

The polyketide natural product reveromycin A (RM-A) exhibits antifungal, anticancer, anti-bone metastasis, anti-periodontitis and anti-osteoporosis activities by selectively inhibiting eukaryotic cytoplasmic isoleucyl-tRNA synthetase (IleRS). Herein, a co-crystal structure suggests that the RM-A molecule occupies the substrate tRNAIle binding site of Saccharomyces cerevisiae IleRS (ScIleRS), by partially mimicking the binding of tRNAIle. RM-A binding is facilitated by the copurified intermediate product isoleucyl-adenylate (Ile-AMP). The binding assays confirm that RM-A competes with tRNAIle while binding synergistically with L-isoleucine or intermediate analogue Ile-AMS to the aminoacylation pocket of ScIleRS. This study highlights that the vast tRNA binding site of the Rossmann-fold catalytic domain of class I aminoacyl-tRNA synthetases could be targeted by a small molecule. This finding will inform future rational drug design.


Sujet(s)
Sites de fixation/effets des médicaments et des substances chimiques , Ligases/composition chimique , Ligases/effets des médicaments et des substances chimiques , Pyrannes/antagonistes et inhibiteurs , ARN de transfert/effets des médicaments et des substances chimiques , Spiranes/antagonistes et inhibiteurs , Amino acyl-tRNA synthetases/composition chimique , Amino acyl-tRNA synthetases/effets des médicaments et des substances chimiques , Isoleucine , Isoleucine-tRNA ligase/composition chimique , Isoleucine-tRNA ligase/effets des médicaments et des substances chimiques , Ligands , Modèles moléculaires , Ostéoporose/traitement médicamenteux , ARN de transfert/composition chimique , Saccharomyces cerevisiae
20.
RNA Biol ; 18(11): 1501-1511, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-33317386

RÉSUMÉ

Among the 20 cytoplasmic aminoacyl-tRNA synthetases (aaRSs), alanyl-tRNA synthetase (AlaRS) has unique features. AlaRS is the only aaRS that exclusively recognizes a single G3:U70 wobble base pair in the acceptor stem of tRNA, which serves as the identity element for both the synthetic and the proofreading activities of the synthetase. The recognition is relaxed during evolution and eukaryotic AlaRS can mis-aminoacylate noncognate tRNAs with a G4:U69 base pair seemingly as a deliberate gain of function for unknown reasons. Unlike other class II aaRSs, dimerization of AlaRS is not necessarily required for aminoacylation possibly due to functional compensations from the C-terminal domain (C-Ala). In contrast to other 19 cytoplasmic aaRSs that append additional domains or motifs to acquire new functions during evolution, the functional expansion of AlaRS is likely achieved through transformations of the existing C-Ala. Given both essential canonical and diverse non-canonical roles of AlaRS, dysfunction of AlaRS leads to neurodegenerative disorders in human and various pathological phenotypes in mouse models. In this review, the uniqueness of AlaRS in both physiological and pathological events is systematically discussed, with a particular focus on its novel functions gained in evolution.


Sujet(s)
Alanine-tRNA ligase/métabolisme , Aminoacylation , Maladies neurodégénératives/anatomopathologie , Animaux , Humains , Maladies neurodégénératives/enzymologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...