Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Fly (Austin) ; 18(1): 2308737, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38374657

RESUMEN

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.


Asunto(s)
Aminoacil-ARNt Sintetasas , Fenilalanina-ARNt Ligasa , Animales , Humanos , Apetito/genética , Drosophila/genética , Drosophila/metabolismo , Hormonas , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/metabolismo , ARN de Transferencia
2.
Mol Genet Metab ; 140(3): 107657, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37523899

RESUMEN

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.


Asunto(s)
Epilepsia , Enfermedades Mitocondriales , Fenilalanina-ARNt Ligasa , Humanos , Lactante , Recién Nacido , Epilepsia/patología , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Mutación , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/química , ARN de Transferencia/genética , ARN de Transferencia de Fenilalanina/metabolismo
3.
J Phys Chem Lett ; 14(14): 3452-3460, 2023 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-37010935

RESUMEN

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.


Asunto(s)
Aminoacil-ARNt Sintetasas , Fenilalanina-ARNt Ligasa , Humanos , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/metabolismo , Entropía , Aminoacil-ARNt Sintetasas/química , Aminoacil-ARNt Sintetasas/genética , Aminoacil-ARNt Sintetasas/metabolismo , Anticodón , Dominio Catalítico
4.
Neuropediatrics ; 54(5): 351-355, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36603837

RESUMEN

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.


Asunto(s)
Fenilalanina-ARNt Ligasa , Niño , Femenino , Humanos , Preescolar , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/metabolismo , Fenilalanina/metabolismo , Calidad de Vida , Mitocondrias/genética , Mitocondrias/metabolismo , ARN de Transferencia/metabolismo , Proteínas Mitocondriales/genética
5.
Hum Cell ; 36(1): 121-131, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36205831

RESUMEN

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.


Asunto(s)
Drosophila melanogaster , Proteínas Hedgehog , Fenilalanina-ARNt Ligasa , Animales , Secuencia de Bases , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas Mitocondriales/genética , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/metabolismo
6.
Orphanet J Rare Dis ; 17(1): 306, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35918773

RESUMEN

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.


Asunto(s)
Fenilalanina-ARNt Ligasa , Humanos , Recién Nacido , Mutación/genética , Fenilalanina , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/genética , ARN de Transferencia/genética , Secuenciación del Exoma
7.
PLoS Genet ; 18(4): e1010185, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35486661

RESUMEN

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.


Asunto(s)
Fenilalanina-ARNt Ligasa , Animales , Drosophila/genética , Fenilalanina , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/metabolismo , ARN de Transferencia de Fenilalanina/química , ARN de Transferencia de Fenilalanina/metabolismo
8.
FEBS Lett ; 596(7): 947-957, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35038769

RESUMEN

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.


Asunto(s)
Aminoacil-ARNt Sintetasas , Mycoplasma/enzimología , Fenilalanina-ARNt Ligasa , Aminoácidos , Aminoacil-ARNt Sintetasas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/metabolismo
9.
Nat Commun ; 13(1): 459, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35075105

RESUMEN

Toxoplasma gondii commonly infects humans and while most infections are controlled by the immune response, currently approved drugs are not capable of clearing chronic infection in humans. Hence, approximately one third of the world's human population is at risk of reactivation, potentially leading to severe sequelae. To identify new candidates for treating chronic infection, we investigated a series of compounds derived from diversity-oriented synthesis. Bicyclic azetidines are potent low nanomolar inhibitors of phenylalanine tRNA synthetase (PheRS) in T. gondii, with excellent selectivity. Biochemical and genetic studies validate PheRS as the primary target of bicyclic azetidines in T. gondii, providing a structural basis for rational design of improved analogs. Favorable pharmacokinetic properties of a lead compound provide excellent protection from acute infection and partial protection from chronic infection in an immunocompromised mouse model of toxoplasmosis. Collectively, PheRS inhibitors of the bicyclic azetidine series offer promise for treatment of chronic toxoplasmosis.


Asunto(s)
Antiprotozoarios/administración & dosificación , Azetidinas/administración & dosificación , Inhibidores Enzimáticos/administración & dosificación , Fenilalanina-ARNt Ligasa/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Toxoplasma/efectos de los fármacos , Toxoplasma/enzimología , Toxoplasmosis/tratamiento farmacológico , Animales , Antiprotozoarios/química , Azetidinas/química , Inhibidores Enzimáticos/química , Femenino , Humanos , Cinética , Masculino , Ratones , Ratones Endogámicos CBA , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Toxoplasma/genética , Toxoplasma/crecimiento & desarrollo , Toxoplasmosis/parasitología
10.
Nucleic Acids Res ; 49(20): 11800-11809, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34581811

RESUMEN

High fidelity during protein synthesis is accomplished by aminoacyl-tRNA synthetases (aaRSs). These enzymes ligate an amino acid to a cognate tRNA and have proofreading and editing capabilities that ensure high fidelity. Phenylalanyl-tRNA synthetase (PheRS) preferentially ligates a phenylalanine to a tRNAPhe over the chemically similar tyrosine, which differs from phenylalanine by a single hydroxyl group. In bacteria that undergo exposure to oxidative stress such as Salmonella enterica serovar Typhimurium, tyrosine isomer levels increase due to phenylalanine oxidation. Several residues are oxidized in PheRS and contribute to hyperactive editing, including against mischarged Tyr-tRNAPhe, despite these oxidized residues not being directly implicated in PheRS activity. Here, we solve a 3.6 Å cryo-electron microscopy structure of oxidized S. Typhimurium PheRS. We find that oxidation results in widespread structural rearrangements in the ß-subunit editing domain and enlargement of its editing domain. Oxidization also enlarges the phenylalanyl-adenylate binding pocket but to a lesser extent. Together, these changes likely explain why oxidation leads to hyperaccurate editing and decreased misincorporation of tyrosine. Taken together, these results help increase our understanding of the survival of S. Typhimurium during human infection.


Asunto(s)
Proteínas Bacterianas/química , Estrés Oxidativo , Fenilalanina-ARNt Ligasa/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Microscopía por Crioelectrón , Fenilalanina-ARNt Ligasa/metabolismo , Salmonella typhimurium/enzimología , Salmonella typhimurium/ultraestructura
11.
Protein Sci ; 30(9): 1793-1803, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34184352

RESUMEN

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.


Asunto(s)
Aminoacil-ARNt Sintetasas/química , Antimaláricos/química , Inhibidores Enzimáticos/química , Lisina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/química , Plasmodium falciparum/efectos de los fármacos , Proteínas Protozoarias/química , Aminoacil-ARNt Sintetasas/antagonistas & inhibidores , Aminoacil-ARNt Sintetasas/genética , Aminoacil-ARNt Sintetasas/metabolismo , Antimaláricos/farmacología , Dominio Catalítico , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Expresión Génica , Humanos , Lisina-ARNt Ligasa/antagonistas & inhibidores , Lisina-ARNt Ligasa/genética , Lisina-ARNt Ligasa/metabolismo , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Modelos Moleculares , Fenilalanina-ARNt Ligasa/antagonistas & inhibidores , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/metabolismo , Plasmodium falciparum/química , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
12.
Nucleic Acids Res ; 49(9): 5351-5368, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33885823

RESUMEN

Tuberculosis, caused by Mycobacterium tuberculosis, responsible for ∼1.5 million fatalities in 2018, is the deadliest infectious disease. Global spread of multidrug resistant strains is a public health threat, requiring new treatments. Aminoacyl-tRNA synthetases are plausible candidates as potential drug targets, because they play an essential role in translating the DNA code into protein sequence by attaching a specific amino acid to their cognate tRNAs. We report structures of M. tuberculosis Phe-tRNA synthetase complexed with an unmodified tRNAPhe transcript and either L-Phe or a nonhydrolyzable phenylalanine adenylate analog. High-resolution models reveal details of two modes of tRNA interaction with the enzyme: an initial recognition via indirect readout of anticodon stem-loop and aminoacylation ready state involving interactions of the 3' end of tRNAPhe with the adenylate site. For the first time, we observe the protein gate controlling access to the active site and detailed geometry of the acyl donor and tRNA acceptor consistent with accepted mechanism. We biochemically validated the inhibitory potency of the adenylate analog and provide the most complete view of the Phe-tRNA synthetase/tRNAPhe system to date. The presented topography of amino adenylate-binding and editing sites at different stages of tRNA binding to the enzyme provide insights for the rational design of anti-tuberculosis drugs.


Asunto(s)
Mycobacterium tuberculosis/enzimología , Fenilalanina-ARNt Ligasa/química , ARN de Transferencia de Fenilalanina/química , Aminoacilación de ARN de Transferencia , Adenosina/análogos & derivados , Adenosina/química , Adenosina/metabolismo , Humanos , Ligandos , Modelos Moleculares , Mycobacterium tuberculosis/genética , Fenilalanina/análogos & derivados , Fenilalanina/química , Fenilalanina/metabolismo , Fenilalanina-ARNt Ligasa/metabolismo , Unión Proteica , ARN de Transferencia de Fenilalanina/metabolismo , Thermus thermophilus/enzimología
13.
J Biol Chem ; 296: 100257, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33837735

RESUMEN

Mycobacterium tuberculosis (Mtb) remains the deadliest pathogenic bacteria worldwide. The search for new antibiotics to treat drug-sensitive as well as drug-resistant tuberculosis has become a priority. The essential enzyme phenylalanyl-tRNA synthetase (PheRS) is an antibacterial drug target because of the large differences between bacterial and human PheRS counterparts. In a high-throughput screening of 2148 bioactive compounds, PF-3845, which is a known inhibitor of human fatty acid amide hydrolase, was identified inhibiting Mtb PheRS at Ki ∼ 0.73 ± 0.06 µM. The inhibition mechanism was studied with enzyme kinetics, protein structural modeling, and crystallography, in comparison to a PheRS inhibitor of the noted phenyl-thiazolylurea-sulfonamide class. The 2.3-Å crystal structure of Mtb PheRS in complex with PF-3845 revealed its novel binding mode, in which a trifluoromethyl-pyridinylphenyl group occupies the phenylalanine pocket, whereas a piperidine-piperazine urea group binds into the ATP pocket through an interaction network enforced by a sulfate ion. It represents the first non-nucleoside bisubstrate competitive inhibitor of bacterial PheRS. PF-3845 inhibits the in vitro growth of Mtb H37Rv at ∼24 µM, and the potency of PF-3845 increased against an engineered strain Mtb pheS-FDAS, suggesting on target activity in mycobacterial whole cells. PF-3845 does not inhibit human cytoplasmic or mitochondrial PheRS in biochemical assay, which can be explained from the crystal structures. Further medicinal chemistry efforts focused on the piperidine-piperazine urea moiety may result in the identification of a selective antibacterial lead compound.


Asunto(s)
Mycobacterium tuberculosis/enzimología , Fenilalanina-ARNt Ligasa/ultraestructura , Conformación Proteica , Tuberculosis Resistente a Múltiples Medicamentos/tratamiento farmacológico , Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/química , Secuencia de Aminoácidos/genética , Antibacterianos/química , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Cinética , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/patogenicidad , Fenilalanina-ARNt Ligasa/antagonistas & inhibidores , Fenilalanina-ARNt Ligasa/química , Piperidinas/química , Piperidinas/farmacología , Piridinas/química , Piridinas/farmacología , Tuberculosis Resistente a Múltiples Medicamentos/enzimología , Tuberculosis Resistente a Múltiples Medicamentos/genética
14.
Nat Commun ; 12(1): 343, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436639

RESUMEN

The inhibition of Plasmodium cytosolic phenylalanine tRNA-synthetase (cFRS) by a novel series of bicyclic azetidines has shown the potential to prevent malaria transmission, provide prophylaxis, and offer single-dose cure in animal models of malaria. To date, however, the molecular basis of Plasmodium cFRS inhibition by bicyclic azetidines has remained unknown. Here, we present structural and biochemical evidence that bicyclic azetidines are competitive inhibitors of L-Phe, one of three substrates required for the cFRS-catalyzed aminoacylation reaction that underpins protein synthesis in the parasite. Critically, our co-crystal structure of a PvcFRS-BRD1389 complex shows that the bicyclic azetidine ligand binds to two distinct sub-sites within the PvcFRS catalytic site. The ligand occupies the L-Phe site along with an auxiliary cavity and traverses past the ATP binding site. Given that BRD1389 recognition residues are conserved amongst apicomplexan FRSs, this work lays a structural framework for the development of drugs against both Plasmodium and related apicomplexans.


Asunto(s)
Azetidinas/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Malaria/enzimología , Parásitos/enzimología , Fenilalanina-ARNt Ligasa/antagonistas & inhibidores , Fenilalanina-ARNt Ligasa/química , Plasmodium falciparum/enzimología , Secuencia de Aminoácidos , Aminoacilación , Animales , Dominio Catalítico , Citosol/enzimología , Resistencia a Medicamentos/genética , Modelos Moleculares , Mutación/genética , Fenilalanina/metabolismo , Fenilalanina-ARNt Ligasa/metabolismo , Plasmodium falciparum/efectos de los fármacos
15.
FEBS J ; 287(17): 3814-3826, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32115907

RESUMEN

Various pathogenic variants in both mitochondrial tRNAPhe and Phenylalanyl-tRNA synthetase mitochondrial protein coding gene (FARS2) gene encoding for the human mitochondrial PheRS have been identified and associated with neurological and/or muscle-related pathologies. An important Guanine-34 (G34)A anticodon mutation associated with myoclonic epilepsy with ragged red fibers (MERRF) syndrome has been reported in hmit-tRNAPhe . The majority of G34 contacts in available aaRSs-tRNAs complexes specifically use that base as an important tRNA identity element. The network of intermolecular interactions providing its specific recognition also largely conserved. However, their conservation depends also on the invariance of the residues in the anticodon binding domain (ABD) of human mitochondrial Phenylalanyl-tRNA synthetase (hmit-PheRS). A defect in recognition of the anticodon of tRNAPhe may happen not only because of G34A mutation, but also due to mutations in the ABD. Indeed, a pathogenic mutation in FARS2 has been recently reported in a 9-year-old female patient harboring a p.Asp364Gly mutation. Asp364 is hydrogen bonded (HB) to G34 in WT hmit-PheRS. Thus, there are two pathogenic variants disrupting HB between G34 and Asp364: one is associated with G34A mutation, and the other with Asp364Gly mutation. We have measured the rates of tRNAPhe aminoacylation catalyzed by WT hmit-PheRS and mutant enzymes. These data ranked the residues making a HB with G34 according to their contribution to activity and the signal transduction pathway in the hmit-PheRS-tRNAPhe complex. Furthermore, we carried out extensive MD simulations to reveal the interdomain contact topology on the dynamic trajectories of the complex, and gaining insight into the structural and dynamic integrity effects of hmit-PheRS complexed with tRNAPhe . DATABASE: Structural data are available in PDB database under the accession number(s): 3CMQ, 3TUP, 5MGH, 5MGV.


Asunto(s)
Pleiotropía Genética , Proteínas Mitocondriales/química , Paraparesia Espástica/genética , Fenilalanina-ARNt Ligasa/química , ARN de Transferencia de Fenilalanina/química , Sustitución de Aminoácidos , Anticodón/química , Anticodón/metabolismo , Ácido Aspártico/química , Niño , Consanguinidad , ADN Mitocondrial/genética , Progresión de la Enfermedad , Femenino , Guanina/química , Humanos , Enlace de Hidrógeno , Síndrome MERRF/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Movimiento (Física) , Mutación Missense , Fenotipo , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/metabolismo , Mutación Puntual , Conformación Proteica , Dominios Proteicos
16.
Mol Genet Metab ; 125(3): 281-291, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30177229

RESUMEN

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.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Mitocondrias/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Fenilalanina-ARNt Ligasa/genética , Adolescente , Adulto , Aminoacil-ARNt Sintetasas/deficiencia , Niño , Preescolar , Femenino , Eliminación de Gen , Humanos , Masculino , Mitocondrias/enzimología , Mitocondrias/patología , Enfermedades Mitocondriales/enzimología , Enfermedades Mitocondriales/patología , Proteínas Mitocondriales/química , Proteínas Mitocondriales/deficiencia , Mutación/genética , Paraplejía/genética , Paraplejía/patología , Fenilalanina/genética , Fenilalanina/metabolismo , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/deficiencia , Isoformas de Proteínas/genética , Relación Estructura-Actividad , Adulto Joven
17.
Protein Sci ; 26(8): 1505-1516, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28419689

RESUMEN

Mutations in the mitochondrial aminoacyl-tRNA synthetases (mtaaRSs) can cause profound clinical presentations, and have manifested as diseases with very selective tissue specificity. To date most of the mtaaRS mutations could be phenotypically recognized, such that clinicians could identify the affected mtaaRS from the symptoms alone. Among the recently reported pathogenic variants are point mutations in FARS2 gene, encoding the human mitochondrial PheRS. Patient symptoms range from spastic paraplegia to fatal infantile Alpers encephalopathy. How clinical manifestations of these mutations relate to the changes in three-dimensional structures and kinetic characteristics remains unclear, although impaired aminoacylation has been proposed as possible etiology of diseases. Here, we report four crystal structures of HsmtPheRS mutants, and extensive MD simulations for wild-type and nine mutants to reveal the structural changes on dynamic trajectories of HsmtPheRS. Using steady-state kinetic measurements of phenylalanine activation and tRNAPhe aminoacylation, we gained insight into the structural and kinetic effects of mitochondrial disease-related mutations in FARS2 gene.


Asunto(s)
Esclerosis Cerebral Difusa de Schilder/genética , Proteínas Mitocondriales/química , Mutación , Paraplejía/genética , Fenilalanina-ARNt Ligasa/química , ARN de Transferencia de Fenilalanina/química , Adolescente , Secuencias de Aminoácidos , Aminoacilación , Sitios de Unión , Preescolar , Cristalografía por Rayos X , Esclerosis Cerebral Difusa de Schilder/diagnóstico , Esclerosis Cerebral Difusa de Schilder/metabolismo , Esclerosis Cerebral Difusa de Schilder/patología , Femenino , Humanos , Cinética , Masculino , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Simulación de Dinámica Molecular , Paraplejía/diagnóstico , Paraplejía/metabolismo , Paraplejía/patología , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , ARN de Transferencia de Fenilalanina/metabolismo , Alineación de Secuencia , Especificidad por Sustrato , Termodinámica
18.
J Mol Graph Model ; 73: 36-47, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28235746

RESUMEN

Increased resistance of MRSA (multidrug resistance Staphylococcus aureus) to anti-infective drugs is a threat to global health necessitating the development of anti-infectives with novel mechanisms of action. Phenylalanine tRNA synthetase (PheRS) is a unique enzyme of the aminoacyl-tRNA synthetases (aaRSs), which are essential enzymes for protein biosynthesis. PheRS is an (αb)2 tetrameric enzyme composed of two alpha subunits (PheS) and two larger beta subunits (PheT). Our potential target in the drug development for the treatment of MRSA infections is the phenylalanine tRNA synthetase alpha subunit that contains the binding site for the natural substrate. There is no crystal structure available for S. aureus PheRS, therefore comparative structure modeling is required to establish a putative 3D structure for the required enzyme enabling development of new inhibitors with greater selectivity. The S. aureus PheRS alpha subunit homology model was constructed using Molecular Operating Environment (MOE) software. Staphylococcus haemolyticus PheRS was the main template while Thermus thermophilus PheRS was utilised to predict the enzyme binding with tRNAphe. The model has been evaluated and compared with the main template through Ramachandran plots, Verify 3D and Protein Statistical Analysis (ProSA). The query protein active site was predicted from its sequence using a conservation analysis tool. Docking suitable ligands using MOE into the constructed model were used to assess the predicted active sites. The docked ligands involved the PheRS natural substrate (phenylalanine), phenylalanyl-adenylate and several described S. aureus PheRS inhibitors.


Asunto(s)
Fenilalanina-ARNt Ligasa/química , Staphylococcus aureus/enzimología , Homología Estructural de Proteína , Secuencia de Aminoácidos , Anticodón/genética , Sitios de Unión , Metales/metabolismo , Simulación del Acoplamiento Molecular , Conformación de Ácido Nucleico , Filogenia , Subunidades de Proteína/metabolismo , ARN de Transferencia de Fenilalanina/química , ARN de Transferencia de Fenilalanina/metabolismo , Alineación de Secuencia , Thermus thermophilus/enzimología
19.
Protein Sci ; 25(3): 618-26, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26645192

RESUMEN

Mitochondria are considered as the primary source of reactive oxygen species (ROS) in nearly all eukaryotic cells during respiration. The harmful effects of these compounds range from direct neurotoxicity to incorporation into proteins producing aberrant molecules with multiple physiological problems. Phenylalanine exposure to ROS produces multiple oxidized isomers: tyrosine, Levodopa, ortho-Tyr, meta-Tyr (m-Tyr), and so on. Cytosolic phenylalanyl-tRNA synthetase (PheRS) exerts control over the translation accuracy, hydrolyzing misacylated products, while monomeric mitochondrial PheRS lacks the editing activity. Recently we showed that "teamwork" of cytosolic and mitochondrial PheRSs cannot prevent incorporation of m-Tyr and l-Dopa into proteins. Here, we present human mitochondrial chimeric PheRS with implanted editing module taken from EcPheRS. The monomeric mitochondrial chimera possesses editing activity, while in bacterial and cytosolic PheRSs this type of activity was detected for the (αß)2 architecture only. The fusion protein catalyzes aminoacylation of tRNA(Phe) with cognate phenylalanine and effectively hydrolyzes the noncognate aminoacyl-tRNAs: Tyr-tRNA(Phe) and m-Tyr-tRNA(Phe) .


Asunto(s)
Aminoácidos/metabolismo , Mitocondrias/enzimología , Fenilalanina-ARNt Ligasa/metabolismo , Acilación , Clonación Molecular , Humanos , Hidrólisis , Mitocondrias/química , Mitocondrias/genética , Mitocondrias/metabolismo , Modelos Moleculares , Fenilalanina/metabolismo , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/genética , Aminoacil-ARN de Transferencia/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
20.
Proc Natl Acad Sci U S A ; 112(19): 6038-43, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25918365

RESUMEN

The cellular translational machinery (TM) synthesizes proteins using exclusively L- or achiral aminoacyl-tRNAs (aa-tRNAs), despite the presence of D-amino acids in nature and their ability to be aminoacylated onto tRNAs by aa-tRNA synthetases. The ubiquity of L-amino acids in proteins has led to the hypothesis that D-amino acids are not substrates for the TM. Supporting this view, protein engineering efforts to incorporate D-amino acids into proteins using the TM have thus far been unsuccessful. Nonetheless, a mechanistic understanding of why D-aa-tRNAs are poor substrates for the TM is lacking. To address this deficiency, we have systematically tested the translation activity of D-aa-tRNAs using a series of biochemical assays. We find that the TM can effectively, albeit slowly, accept D-aa-tRNAs into the ribosomal aa-tRNA binding (A) site, use the A-site D-aa-tRNA as a peptidyl-transfer acceptor, and translocate the resulting peptidyl-D-aa-tRNA into the ribosomal peptidyl-tRNA binding (P) site. During the next round of continuous translation, however, we find that ribosomes carrying a P-site peptidyl-D-aa-tRNA partition into subpopulations that are either translationally arrested or that can continue translating. Consistent with its ability to arrest translation, chemical protection experiments and molecular dynamics simulations show that P site-bound peptidyl-D-aa-tRNA can trap the ribosomal peptidyl-transferase center in a conformation in which peptidyl transfer is impaired. Our results reveal a novel mechanism through which D-aa-tRNAs interfere with translation, provide insight into how the TM might be engineered to use D-aa-tRNAs, and increase our understanding of the physiological role of a widely distributed enzyme that clears D-aa-tRNAs from cells.


Asunto(s)
Aminoácidos/química , Peptidil Transferasas/química , ARN de Transferencia/química , Ribosomas/química , Sitios de Unión , Cromatografía en Capa Delgada , Escherichia coli/enzimología , Simulación de Dinámica Molecular , Péptidos/química , Fenilalanina-ARNt Ligasa/química , Unión Proteica , Biosíntesis de Proteínas , Ingeniería de Proteínas , Estructura Terciaria de Proteína , Aminoacil-ARN de Transferencia/química , Estereoisomerismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...