Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(12): 7096-7111, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38783009

RESUMEN

Aminoacyl-tRNA synthetases (AARS) and tRNAs translate the genetic code in all living cells. Little is known about how their molecular ancestors began to enforce the coding rules for the expression of their own genes. Schimmel et al. proposed in 1993 that AARS catalytic domains began by reading an 'operational' code in the acceptor stems of tRNA minihelices. We show here that the enzymology of an AARS urzyme•TΨC-minihelix cognate pair is a rich in vitro realization of that idea. The TΨC-minihelixLeu is a very poor substrate for full-length Leucyl-tRNA synthetase. It is a superior RNA substrate for the corresponding urzyme, LeuAC. LeuAC active-site mutations shift the choice of both amino acid and RNA substrates. AARS urzyme•minihelix cognate pairs are thus small, pliant models for the ancestral decoding hardware. They are thus an ideal platform for detailed experimental study of the operational RNA code.


Asunto(s)
Aminoacil-ARNt Sintetasas , Conformación de Ácido Nucleico , ARN de Transferencia , ARN de Transferencia/metabolismo , ARN de Transferencia/química , ARN de Transferencia/genética , Aminoacil-ARNt Sintetasas/metabolismo , Aminoacil-ARNt Sintetasas/química , Aminoacil-ARNt Sintetasas/genética , Dominio Catalítico , Código Genético , ARN Catalítico/química , ARN Catalítico/metabolismo , Especificidad por Sustrato , Leucina-ARNt Ligasa/metabolismo , Leucina-ARNt Ligasa/química , Leucina-ARNt Ligasa/genética
2.
Commun Biol ; 5(1): 883, 2022 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-36038645

RESUMEN

To correctly aminoacylate tRNALeu, leucyl-tRNA synthetase (LeuRS) catalyzes three reactions: activation of leucine by ATP to form leucyl-adenylate (Leu-AMP), transfer of this amino acid to tRNALeu and post-transfer editing of any mischarged product. Although LeuRS has been well characterized biochemically, detailed structural information is currently only available for the latter two stages of catalysis. We have solved crystal structures for all enzymatic states of Neisseria gonorrhoeae LeuRS during Leu-AMP formation. These show a cycle of dramatic conformational changes, involving multiple domains, and correlate with an energetically unfavorable peptide-plane flip observed in the active site of the pre-transition state structure. Biochemical analyses, combined with mutant structural studies, reveal that this backbone distortion acts as a trigger, temporally compartmentalizing the first two catalytic steps. These results unveil the remarkable effect of this small structural alteration on the global dynamics and activity of the enzyme.


Asunto(s)
Leucina-ARNt Ligasa , ARN de Transferencia de Leucina , Catálisis , Dominio Catalítico , Leucina-ARNt Ligasa/química , Leucina-ARNt Ligasa/genética , Leucina-ARNt Ligasa/metabolismo , Péptidos , ARN de Transferencia de Leucina/metabolismo
3.
J Biosci Bioeng ; 133(5): 436-443, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35216933

RESUMEN

Isoleucyl-tRNA synthetase (IleRS), leucyl-tRNA synthetase (LeuRS), and valyl-tRNA synthetase (ValRS) are enzymes that have potential for the determination of l-isoleucine, l-leucine, and l-valine in food products and plasma. However, the disadvantages of these enzymes are their specificity and sensitivity. Here, we examined the substrate specificity of IleRS, LeuRS, and ValRS under various conditions of pyrophosphate amplification to improve their specificity and sensitivity. The amount of pyrophosphate produced in IleRS, LeuRS, and ValRS reactions was amplified after the addition of excess adenosine-5'-triphosphate and magnesium ions, and was approximately 9-, 8-, and 7-fold higher, respectively, for each of the initial l-amino acid substrates (50 µM). However, in addition to their target amino acids, IleRS, LeuRS, and ValRS also reacted with l-valine, l-lysine, and l-threonine, respectively. This substrate misrecognition was overcome by making the reaction pH more acidic and by increasing the magnesium ion concentration. The pyrophosphate amplification in IleRS, LeuRS, and ValRS reactions resulted in the production of p1, p4-di (adenosine) 5'-tetraphosphate. We also observed a strong positive correlation (R = 0.99) between the amount of pyrophosphate produced and the initial concentration of l-amino acid with 5 and 50 µM l-isoleucine, l-leucine, and l-valine. Our results suggest that amino acid assays using IleRS, LeuRS, and ValRS are promising methods to accurately measure l-valine, l-isoleucine, and l-leucine in food products and plasma.


Asunto(s)
Aminoacil-ARNt Sintetasas , Leucina-ARNt Ligasa , Adenosina/metabolismo , Aminoácidos/metabolismo , Aminoacil-ARNt Sintetasas/metabolismo , Difosfatos , Escherichia coli/metabolismo , Isoleucina , Leucina/metabolismo , Leucina-ARNt Ligasa/química , Leucina-ARNt Ligasa/genética , Leucina-ARNt Ligasa/metabolismo , Magnesio/metabolismo , ARN de Transferencia , Especificidad por Sustrato , Valina/metabolismo , Valina-ARNt Ligasa/química , Valina-ARNt Ligasa/genética , Valina-ARNt Ligasa/metabolismo
4.
STAR Protoc ; 2(3): 100642, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34258600

RESUMEN

Leucyl-tRNA synthetase 1 (LARS1) synthesizes Leu-tRNALeu for protein synthesis and plays an important role in mTORC1 activation by sensing intracellular leucine concentrations. Here, we describe a protocol for the purification, reductive methylation, binding affinity measurement by microscale thermophoresis, T i value measurement by Tycho, and post-crystallization soaking and cooling in cryoprotectants to improve crystallization of LARS1. Collectively, this allowed us to build the RagD binding domain, which was shown to be a dynamic region of LARS1 refractory to crystallization. For complete details on the use and execution of this protocol, please refer to Kim et al. (2021).


Asunto(s)
Frío , Crioprotectores/química , Cristalografía por Rayos X/métodos , Leucina-ARNt Ligasa/química , Cristalización , Leucina-ARNt Ligasa/metabolismo , Metilación , Unión Proteica
5.
Biochim Biophys Acta Mol Cell Res ; 1868(1): 118889, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33091505

RESUMEN

Aminoacyl-tRNA synthetases (ARSs) are a family of evolutionarily conserved housekeeping enzymes used for protein synthesis that have pivotal roles in the ligation of tRNA with their cognate amino acids. Recent advances in the structural and functional studies of ARSs have revealed many previously unknown biological functions beyond the classical catalytic roles. Sensing the sufficiency of intracellular nutrients such as amino acids, ATP, and fatty acids is a crucial aspect for every living organism, and it is closely connected to the regulation of diverse cellular physiologies. Notably, among ARSs, leucyl-tRNA synthetase 1 (LARS1) has been identified to perform specifically as a leucine sensor upstream of the amino acid-sensing pathway and thus participates in the coordinated control of protein synthesis and autophagy for cell growth. In addition to LARS1, other types of ARSs are also likely involved in the sensing and signaling of their cognate amino acids inside cells. Collectively, this review focuses on the mechanisms of ARSs interacting within amino acid signaling and proposes the possible role of ARSs as general intracellular amino acid sensors.


Asunto(s)
Aminoácidos/genética , Aminoacil-ARNt Sintetasas/genética , Leucina-ARNt Ligasa/genética , Leucina/genética , Aminoácidos/química , Aminoacil-ARNt Sintetasas/química , Humanos , Leucina/química , Leucina-ARNt Ligasa/química , Biosíntesis de Proteínas/genética , ARN de Transferencia/genética , Transducción de Señal/genética
6.
J Chem Phys ; 153(13): 134104, 2020 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-33032427

RESUMEN

Molecular interactions are essential for regulation of cellular processes from the formation of multi-protein complexes to the allosteric activation of enzymes. Identifying the essential residues and molecular features that regulate such interactions is paramount for understanding the biochemical process in question, allowing for suppression of a reaction through drug interventions or optimization of a chemical process using bioengineered molecules. In order to identify important residues and information pathways within molecular complexes, the dynamical network analysis method was developed and has since been broadly applied in the literature. However, in the dawn of exascale computing, this method is frequently limited to relatively small biomolecular systems. In this work, we provide an evolution of the method, application, and interface. All data processing and analysis are conducted through Jupyter notebooks, providing automatic detection of important solvent and ion residues, an optimized and parallel generalized correlation implementation that is linear with respect to the number of nodes in the system, and subsequent community clustering, calculation of betweenness of contacts, and determination of optimal paths. Using the popular visualization program visual molecular dynamics (VMD), high-quality renderings of the networks over the biomolecular structures can be produced. Our new implementation was employed to investigate three different systems, with up to 2.5M atoms, namely, the OMP-decarboxylase, the leucyl-tRNA synthetase complexed with its cognate tRNA and adenylate, and respiratory complex I in a membrane environment. Our enhanced and updated protocol provides the community with an intuitive and interactive interface, which can be easily applied to large macromolecular complexes.


Asunto(s)
Complejo I de Transporte de Electrón/química , Leucina-ARNt Ligasa/química , Orotidina-5'-Fosfato Descarboxilasa/química , Regulación Alostérica , Dominio Catalítico , Escherichia coli/enzimología , Methanobacteriaceae/enzimología , Simulación de Dinámica Molecular , Dominios Proteicos , Programas Informáticos , Thermus thermophilus/enzimología
7.
Bioorg Chem ; 105: 104354, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33091672

RESUMEN

Three series of nanosized-formazan analogues were synthesized from the reaction of dithiazone with various types of α-haloketones (ester and acetyl substituted hydrazonoyl chlorides and phenacyl bromides) in sodium ethoxide solution. The structure and the crystal size of the new synthesized derivatives were assured based on the spectral analyses, XRD and SEM data. The antibacterial and antifungal activities were evaluated by agar diffusion technique. The results showed mild to moderate antibacterial activities and moderate to potent antifungal activities. Significant antifungal activities were observed for four derivatives 3a, 3d, 5a and 5g on the pathogenic fungal strains; Aspergillus flavus and Candida albicans with inhibition zone ranging from 16 to 20 mm. Molecular docking simulations of the synthesized compounds into leucyl-tRNA synthetase editing domain of Candida albicans suggested that most formazan analogues can fit deeply forming stable complexes in the active site. Furthermore, we utilized the docking approach to examine the potential of these compounds to inhibit SARS-CoV-2 3CLpro. The results were very promising verifying these formazan analogues as a hopeful antiviral agents.


Asunto(s)
Antiinfecciosos/síntesis química , Proteasas 3C de Coronavirus/metabolismo , Formazáns/química , Simulación del Acoplamiento Molecular , Nanoestructuras/química , SARS-CoV-2/metabolismo , Antiinfecciosos/metabolismo , Antiinfecciosos/farmacología , Aspergillus flavus/efectos de los fármacos , Sitios de Unión , COVID-19/patología , COVID-19/virología , Candida albicans/efectos de los fármacos , Dominio Catalítico , Proteasas 3C de Coronavirus/química , Formazáns/metabolismo , Formazáns/farmacología , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Humanos , Leucina-ARNt Ligasa/química , Leucina-ARNt Ligasa/metabolismo , SARS-CoV-2/aislamiento & purificación
8.
J Biosci ; 452020.
Artículo en Inglés | MEDLINE | ID: mdl-32385222

RESUMEN

Leucyl-tRNA synthetases (LRS) catalyze the linkage of leucine with tRNALeu. A large insertion CP1 domain (Connective Polypeptide 1) in LRS is responsible for post-transfer editing of mis-charged aminoacyl-tRNAs. Here, we characterized the CP1 domain of Leishmania donovani, a protozoan parasite, and its role in editing activity and interaction with broad spectrum anti-fungal, AN2690. The deletion mutant of LRS, devoid of CP1 domain (LRS-CP1Δ) was constructed, followed by determination of its role in editing and aminoacylation. Binding of AN2690 and different amino acids with CP1 deletion mutant and full length LRS was evaluated using isothermal titration calorimetry (ITC) and molecular dynamics simulations. The recombinant LRS-CP1Δ protein did not catalyze the aminoacylation and the editing reaction when compared to full-length LRS. Thus, indicating that CP1 domain was imperative for both aminoacylation and editing activities of LRS. Binding studies with different amino acids indicated selectivity of isoleucine by CP1 domain over other amino acids. These studies also indicated high affinity of AN2690 with the editing domain. Molecular docking studies indicated that AN2690-CP1 domain complex was stabilized by hydrogen bonding and hydrophobic interactions resulting in high binding affinity between the two. Our data suggests CP1 is crucial for the function of L.donovani LRS.


Asunto(s)
Antiprotozoarios/farmacología , Compuestos de Boro/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Leishmania donovani/química , Leucina-ARNt Ligasa/antagonistas & inhibidores , Péptidos/química , Procesamiento Proteico-Postraduccional , Proteínas Protozoarias/antagonistas & inhibidores , Secuencia de Aminoácidos , Antifúngicos/química , Antifúngicos/farmacología , Antiprotozoarios/química , Sitios de Unión , Compuestos de Boro/química , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Reposicionamiento de Medicamentos , Expresión Génica , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Leishmania donovani/enzimología , Leishmania donovani/genética , Leucina-ARNt Ligasa/química , Leucina-ARNt Ligasa/genética , Leucina-ARNt Ligasa/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Péptidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , ARN de Transferencia de Leucina/química , ARN de Transferencia de Leucina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Eliminación de Secuencia , Homología de Secuencia de Aminoácido , Aminoacilación de ARN de Transferencia/genética
9.
Nucleic Acids Res ; 48(9): 4946-4959, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32232361

RESUMEN

Human cytosolic leucyl-tRNA synthetase (hcLRS) is an essential and multifunctional enzyme. Its canonical function is to catalyze the covalent ligation of leucine to tRNALeu, and it may also hydrolyze mischarged tRNAs through an editing mechanism. Together with eight other aminoacyl-tRNA synthetases (AaRSs) and three auxiliary proteins, it forms a large multi-synthetase complex (MSC). Beyond its role in translation, hcLRS has an important moonlight function as a leucine sensor in the rapamycin complex 1 (mTORC1) pathway. Since this pathway is active in cancer development, hcLRS is a potential target for anti-tumor drug development. Moreover, LRS from pathogenic microbes are proven drug targets for developing antibiotics, which however should not inhibit hcLRS. Here we present the crystal structure of hcLRS at a 2.5 Å resolution, the first complete structure of a eukaryotic LRS, and analyze the binding of various compounds that target different sites of hcLRS. We also deduce the assembly mechanism of hcLRS into the MSC through reconstitution of the entire mega complex in vitro. Overall, our study provides the molecular basis for understanding both the multifaceted functions of hcLRS and for drug development targeting these functions.


Asunto(s)
Leucina-ARNt Ligasa/química , Antiinfecciosos/química , Biocatálisis , Dominio Catalítico , Diseño de Fármacos , Humanos , Leucina-ARNt Ligasa/efectos de los fármacos , Leucina-ARNt Ligasa/metabolismo , Modelos Moleculares , Proteínas de Unión al GTP Monoméricas/metabolismo , Dominios Proteicos , ARN de Transferencia de Leucina/metabolismo , Aminoacilación de ARN de Transferencia
10.
ACS Chem Biol ; 15(6): 1535-1540, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32330002

RESUMEN

Selenoproteins contain the amino acid selenocysteine (Sec) and are found in all domains of life. The functions of many selenoproteins are poorly understood, partly due to difficulties in producing recombinant selenoproteins for cell-biological evaluation. Endogenous mammalian selenoproteins are produced through a noncanonical translation mechanism requiring suppression of the UGA stop codon and a Sec insertion sequence (SECIS) element in the 3' untranslated region of the mRNA. Here, recombinant selenoproteins are generated in mammalian cells through genetic code expansion, circumventing the requirement for the SECIS element and selenium availability. An engineered orthogonal E. coli leucyl-tRNA synthetase/tRNA pair is used to incorporate a photocaged Sec (DMNB-Sec) at the UAG amber stop codon. DMNB-Sec is successfully incorporated into GFP and uncaged by irradiation of living cells. Furthermore, DMNB-Sec is used to generate the native selenoprotein methionine-R-sulfoxide reductase B1 (MsrB1). Importantly, MsrB1 is shown to be catalytically active after uncaging, constituting the first use of genetic code expansion to generate a functional selenoprotein in mammalian systems. The ability to site-specifically introduce Sec directly in mammalian cells, and temporally modulate selenoprotein activity, will aid in the characterization of mammalian selenoprotein function.


Asunto(s)
Código Genético , Selenocisteína/química , Selenoproteínas/genética , Codón de Terminación , Escherichia coli/genética , Proteínas Fluorescentes Verdes/química , Células HEK293 , Humanos , Leucina-ARNt Ligasa/química , Ingeniería de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Selenoproteínas/química
11.
J Biol Chem ; 295(14): 4563-4576, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32102848

RESUMEN

Aminoacyl-tRNA synthetases (aaRSs) are ancient enzymes that play a fundamental role in protein synthesis. They catalyze the esterification of specific amino acids to the 3'-end of their cognate tRNAs and therefore play a pivotal role in protein synthesis. Although previous studies suggest that aaRS-dependent errors in protein synthesis can be beneficial to some microbial species, evidence that reduced aaRS fidelity can be adaptive is limited. Using bioinformatics analyses, we identified two distinct leucyl-tRNA synthetase (LeuRS) genes within all genomes of the archaeal family Sulfolobaceae. Remarkably, one copy, designated LeuRS-I, had key amino acid substitutions within its editing domain that would be expected to disrupt hydrolytic editing of mischarged tRNALeu and to result in variation within the proteome of these extremophiles. We found that another copy, LeuRS-F, contains canonical active sites for aminoacylation and editing. Biochemical and genetic analyses of the paralogs within Sulfolobus islandicus supported the hypothesis that LeuRS-F, but not LeuRS-I, functions as an essential tRNA synthetase that accurately charges leucine to tRNALeu for protein translation. Although LeuRS-I was not essential, its expression clearly supported optimal S. islandicus growth. We conclude that LeuRS-I may have evolved to confer a selective advantage under the extreme and fluctuating environmental conditions characteristic of the volcanic hot springs in which these archaeal extremophiles reside.


Asunto(s)
Proteínas Arqueales/metabolismo , Leucina-ARNt Ligasa/metabolismo , Sulfolobus/enzimología , Secuencia de Aminoácidos , Aminoacilación , Proteínas Arqueales/química , Proteínas Arqueales/clasificación , Proteínas Arqueales/genética , Dominio Catalítico , Extremófilos/metabolismo , Edición Génica , Concentración de Iones de Hidrógeno , Leucina/metabolismo , Leucina-ARNt Ligasa/química , Leucina-ARNt Ligasa/clasificación , Leucina-ARNt Ligasa/genética , Mutagénesis Sitio-Dirigida , Filogenia , Biosíntesis de Proteínas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia , Sulfolobus/crecimiento & desarrollo , Temperatura
12.
FEBS J ; 287(4): 800-813, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31486189

RESUMEN

Isoleucyl-tRNA synthetase (IleRS) is a paradigm for understanding how specificity against smaller hydrophobic substrates evolved in both the synthetic and editing reactions. IleRS misactivates nonproteinogenic norvaline (Nva) and proteinogenic valine (Val), with a 200-fold lower efficiency than the cognate isoleucine (Ile). Translational errors are, however, prevented by IleRS hydrolytic editing. Nva and Val are both smaller than Ile by a single methylene group. How does the removal of one additional methylene group affects IleRS specificity? We found that the nonproteinogenic α-aminobutyrate (Abu) is activated 30-fold less efficiently than Nva and Val, indicating that the removal of the second methylene group comes with a lower penalty. As with Nva and Val, discrimination against Abu predominantly originated from a higher KM . To examine whether increased hydrophobicity could compensate for the loss of van der Waals interactions, we tested fluorinated Abu analogues. We found that fluorination further hampered activation by IleRS, and even more so by the evolutionary-related ValRS. This suggests that hydrophobicity is not a main driving force of substrate binding in these enzymes. Finally, a discrimination factor of 7100 suggests that IleRS is not expected to edit Abu. However, we found that the IleRS editing domain hydrolyzes Abu-tRNAIle with a rate of 40 s-1 and the introduction of fluorine did not slow down the hydrolysis. This raises interesting questions regarding the mechanism of specificity of the editing domain and its evolution. Understanding what shapes IleRS specificity is also of importance for reengineering translation to accommodate artificial substrates including fluorinated amino acids. ENZYMES: Isoleucyl-tRNA synthetase (EC 6.1.1.5), leucyl-tRNA synthetase (EC 6.1.1.4), valyl-tRNA synthetase (EC 6.1.1.9).


Asunto(s)
Aminobutiratos/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Isoleucina-ARNt Ligasa/química , Leucina-ARNt Ligasa/química , Valina-ARNt Ligasa/química , Aminobutiratos/metabolismo , Sitios de Unión , Clonación Molecular , Escherichia coli/química , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Expresión Génica , Halogenación , Isoleucina-ARNt Ligasa/genética , Isoleucina-ARNt Ligasa/metabolismo , Cinética , Leucina-ARNt Ligasa/genética , Leucina-ARNt Ligasa/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Termodinámica , Valina-ARNt Ligasa/genética , Valina-ARNt Ligasa/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-31791947

RESUMEN

In this study, we aimed to assess the in vitro susceptibility to GSK656 among multiple mycobacterial species and to investigate the correlation between leucyl-tRNA synthetase (LeuRS) sequence variations and in vitro susceptibility to GSK656 among mycobacterial species. A total of 187 mycobacterial isolates, comprising 105 Mycobacterium tuberculosis isolates and 82 nontuberculous mycobacteria (NTM) isolates, were randomly selected for the determination of in vitro susceptibility. For M. tuberculosis, 102 of 105 isolates had MICs of ≤0.5 mg/liter, demonstrating a MIC50 of 0.063 mg/liter and a MIC90 of 0.25 mg/liter. An epidemiological cutoff value of 0.5 mg/liter was proposed for identification of GSK656-resistant M. tuberculosis strains. For NTM, the MIC50 and MIC90 values were >8.0 mg/liter for both Mycobacterium intracellulare and Mycobacterium avium In contrast, all Mycobacterium abscessus isolates had MICs of ≤0.25 mg/liter, yielding a MIC90 of 0.063 mg/liter. LeuRS from M. abscessus showed greater sequence similarity to M. tuberculosis LeuRS than to LeuRSs from M. avium and M. intracellulare Sequence alignment revealed 28 residues differing between LeuRSs from M. avium and M. intracellulare and LeuRSs from M. tuberculosis and M. abscessus; among them, 15 residues were in the drug binding domain. Structure modeling revealed that several different residues were close to the tRNA-LeuRS interface or the entrance of the drug-tRNA binding pocket. In conclusion, our data demonstrate significant species diversity in in vitro susceptibility to GSK656 among various mycobacterial species. GSK656 has potent efficacy against M. tuberculosis and M. abscessus, whereas inherent resistance was noted for M. intracellulare and M. avium.


Asunto(s)
Compuestos de Boro/farmacología , Compuestos Heterocíclicos con 2 Anillos/farmacología , Leucina-ARNt Ligasa/genética , Infecciones por Mycobacterium/tratamiento farmacológico , Mycobacterium/efectos de los fármacos , Humanos , Leucina-ARNt Ligasa/química , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Mycobacterium/enzimología , Mycobacterium/genética , Infecciones por Mycobacterium/microbiología , Filogenia
14.
BMC Mol Cell Biol ; 20(1): 18, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31226926

RESUMEN

The manual classification of protein domains is approaching its 20th anniversary. ECOD is our mixed manual-automatic domain classification. Over time, the types of proteins which require manual curation has changed. Depositions with complex multidomain and multichain arrangements are commonplace. Transmembrane domains are regularly classified. Repeatedly, domains which are initially believed to be novel are found to have homologous links to existing classified domains. Here we present a brief summary of recent manual curation efforts in ECOD generally combined with specific case studies of transmembrane and multidomain proteins wherein manual curation was useful for discovering new homologous relationships. We present a new taxonomy for the classification of ABC transporter transmembrane domains. We examine alternate topologies of the leucine-specific (LS) domain of Leucine tRNA-synthetase. Finally, we elaborate on a distant homologous links between two helical dimerization domains.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/clasificación , Dominios Proteicos , Homología Estructural de Proteína , Proteínas Portadoras/química , Proteínas de Ciclo Celular/química , Cristalografía por Rayos X , Bases de Datos de Proteínas , Endopeptidasas/química , Escherichia coli/química , Humanos , Leucina-ARNt Ligasa/química , Proteínas de la Membrana/química , Proteínas de Transporte de Catión Orgánico/química , Multimerización de Proteína , Estructura Secundaria de Proteína , Proteínas ras/química
15.
Nucleic Acids Res ; 47(12): 6369-6385, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31114902

RESUMEN

Transfer RNAs (tRNAs) are divided into two types, type I with a short variable loop and type II with a long variable loop. Aminoacylation of type I or type II tRNALeu is catalyzed by their cognate leucyl-tRNA synthetases (LeuRSs). However, in Streptomyces coelicolor, there are two types of tRNALeu and only one LeuRS (ScoLeuRS). We found that the enzyme could leucylate both types of ScotRNALeu, and had a higher catalytic efficiency for type II ScotRNALeu(UAA) than for type I ScotRNALeu(CAA). The results from tRNA and enzyme mutagenesis showed that ScoLeuRS did not interact with the canonical discriminator A73. The number of nucleotides, rather than the type of base of the variable loop in the two types of ScotRNALeus, was determined as important for aminoacylation. In vitro and in vivo assays showed that the tertiary structure formed by the D-loop and TψC-loop is more important for ScotRNALeu(UAA). We showed that the leucine-specific domain (LSD) of ScoLeuRS could help LeuRS, which originally only leucylates type II tRNALeu, to aminoacylate type I ScotRNALeu(CAA) and identified the crucial amino acid residues at the C-terminus of the LSD to recognize type I ScotRNALeu(CAA). Overall, our findings identified a rare recognition mechanism of LeuRS to tRNALeu.


Asunto(s)
Leucina-ARNt Ligasa/metabolismo , ARN de Transferencia de Leucina/metabolismo , Streptomyces coelicolor/enzimología , Aminoacilación de ARN de Transferencia , Leucina-ARNt Ligasa/química , ARN Mitocondrial/metabolismo , ARN de Transferencia de Leucina/química , Streptomyces coelicolor/genética
16.
J Mol Graph Model ; 84: 74-81, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29935476

RESUMEN

The accuracy of protein synthesis is provided by the editing functions of aminoacyl-tRNA synthetases (aaRSs), a mechanism that eliminates misactivated amino acids or mischarged tRNAs. Despite research efforts, some molecular bases of these mechanisms are still unclear. The post-transfer editing pathway of leucyl-tRNA synthetase (LeuRS) carried out in a special insertion domain (the Connective Polypeptide 1 or CP1), as editing domain. Recently, it was shown by in vivo studies and was supported by mutagenesis, and the kinetics approaches that the CP1 domain of LeuRS has discriminatory power for different substrates. The goal of this work is to investigate the structural basis for amino acid recognition of LeuRS post-transfer editing processes with molecular dynamics (MD) simulation method. To pursue this aim, the molecular modeling studies on Thermus thermophiles LeuRS (LeuRSTT) with two post-transfer substrates (norvalyl-tRNALeu and isoleucyl-tRNALeu) was performed. Our results revealed that post-transfer substrate norvalyl-tRNALeu is more favorable. Moreover, the MD simulations show that branched side chain of Ile-A76 cannot allow water molecules to get close, which leads to a significant decrease in the rate of hydrolysis. Finally, the study showed that site mutation Asp347Ala has elucidated a number of fine structural differences in the binding mode of two post-transfer substrates to the active centre of LeuRS editing domain and two conserved threonines, namely Thr247 and Thr248, are responsible for the amino acid selection through the interaction with substrates.


Asunto(s)
Aminoácidos/química , Leucina-ARNt Ligasa/química , Simulación de Dinámica Molecular , Dominios y Motivos de Interacción de Proteínas , Thermus thermophilus/enzimología , Aminoácidos/metabolismo , Sitios de Unión , Enlace de Hidrógeno , Cinética , Leucina-ARNt Ligasa/genética , Leucina-ARNt Ligasa/metabolismo , Simulación del Acoplamiento Molecular , Unión Proteica , Especificidad por Sustrato
17.
J Mol Graph Model ; 76: 289-295, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28743072

RESUMEN

Aminoacyl-tRNA synthetases (aaRSs) play important roles in maintaining the accuracy of protein synthesis. Some aaRSs accomplish this via editing mechanisms, among which leucyl-tRNA synthetase (LeuRS) edits non-cognate amino acid norvaline mainly by post-transfer editing. However, the molecular basis for this pathway for eukaryotic and archaeal LeuRS remain unclear. In this study, a complex of archaeal P. horikoshii LeuRS (PhLeuRS) with misacylated tRNALeu was modeled wherever tRNA's acceptor stem was oriented directly into the editing site. To understand the distinctive features of organization we reconstructed a complex of PhLeuRS with tRNA and visualize post-transfer editing interactions mode by performing molecular dynamics (MD) simulation studies. To study molecular basis for substrate selectivity by PhLeuRS's editing site we utilized MD simulation of the entire LeuRS complexes using a diverse charged form of tRNAs, namely norvalyl-tRNALeu and isoleucyl-tRNALeu. In general, the editing site organization of LeuRS from P.horikoshii has much in common with bacterial LeuRS. The MD simulation results revealed that the post-transfer editing substrate norvalyl-A76, binds more strongly than isoleucyl-A76. Moreover, the branched side chain of isoleucine prevents water molecules from being closer and hence the hydrolysis reaction slows significantly. To investigate a possible mechanism of the post-transfer editing reaction, by PhLeuRS we have determined that two water molecules (the attacking and assisting water molecules) are localized near the carbonyl group of the amino acid to be cleaved off. These water molecules approach the substrate from the opposite side to that observed for Thermus thermophilus LeuRS (TtLeuRS). Based on the results obtained, it was suggested that the post-transfer editing mechanism of PhLeuRS differs from that of prokaryotic TtLeuRS.


Asunto(s)
Archaea/enzimología , Archaea/genética , Leucina-ARNt Ligasa/química , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , ARN de Transferencia/química , Hidrólisis , Leucina-ARNt Ligasa/metabolismo , Conformación de Ácido Nucleico , Unión Proteica , Edición de ARN , ARN de Transferencia/genética , Especificidad por Sustrato
18.
J Biol Chem ; 292(25): 10709-10722, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28455447

RESUMEN

Previous proteomic analyses have shown that aminoacyl-tRNA synthetases in many organisms can be modified by acetylation of Lys. In this present study, leucyl-tRNA synthetase and arginyl-tRNA synthetase from Escherichia coli (EcLeuRS and EcArgRS) were overexpressed and purified and found to be acetylated on Lys residues by MS. Gln scanning mutagenesis revealed that Lys619, Lys624, and Lys809 in EcLeuRS and Lys126 and Lys408 in EcArgRS might play important roles in enzyme activity. Furthermore, we utilized a novel protein expression system to obtain enzymes harboring acetylated Lys at specific sites and investigated their catalytic activity. Acetylation of these Lys residues could affect their aminoacylation activity by influencing amino acid activation and/or the affinity for tRNA. In vitro assays showed that acetyl-phosphate nonenzymatically acetylates EcLeuRS and EcArgRS and suggested that the sirtuin class deacetylase CobB might regulate acetylation of these two enzymes. These findings imply a potential regulatory role for Lys acetylation in controlling the activity of aminoacyl-tRNA synthetases and thus protein synthesis.


Asunto(s)
Arginino-ARNt Ligasa/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Leucina-ARNt Ligasa/química , Sirtuinas/química , Acetilación , Arginino-ARNt Ligasa/genética , Arginino-ARNt Ligasa/metabolismo , Activación Enzimática , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Leucina-ARNt Ligasa/genética , Leucina-ARNt Ligasa/metabolismo , Lisina/química , Lisina/genética , Lisina/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo
19.
Antimicrob Agents Chemother ; 60(10): 6271-80, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27503647

RESUMEN

The recent development and spread of extensively drug-resistant and totally drug-resistant resistant (TDR) strains of Mycobacterium tuberculosis highlight the need for new antitubercular drugs. Protein synthesis inhibitors have played an important role in the treatment of tuberculosis (TB) starting with the inclusion of streptomycin in the first combination therapies. Although parenteral aminoglycosides are a key component of therapy for multidrug-resistant TB, the oxazolidinone linezolid is the only orally available protein synthesis inhibitor that is effective against TB. Here, we show that small-molecule inhibitors of aminoacyl-tRNA synthetases (AARSs), which are known to be excellent antibacterial protein synthesis targets, are orally bioavailable and effective against M. tuberculosis in TB mouse infection models. We applied the oxaborole tRNA-trapping (OBORT) mechanism, which was first developed to target fungal cytoplasmic leucyl-tRNA synthetase (LeuRS), to M. tuberculosis LeuRS. X-ray crystallography was used to guide the design of LeuRS inhibitors that have good biochemical potency and excellent whole-cell activity against M. tuberculosis Importantly, their good oral bioavailability translates into in vivo efficacy in both the acute and chronic mouse models of TB with potency comparable to that of the frontline drug isoniazid.


Asunto(s)
Antituberculosos/farmacología , Leucina-ARNt Ligasa/antagonistas & inhibidores , Mycobacterium tuberculosis/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Administración Oral , Animales , Antituberculosos/administración & dosificación , Antituberculosos/química , Antituberculosos/farmacocinética , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Femenino , Humanos , Leucina-ARNt Ligasa/química , Leucina-ARNt Ligasa/genética , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos , Pruebas de Sensibilidad Microbiana , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/genética , Mycobacterium tuberculosis/genética , Inhibidores de la Síntesis de la Proteína/administración & dosificación , Inhibidores de la Síntesis de la Proteína/química , Inhibidores de la Síntesis de la Proteína/farmacocinética , Relación Estructura-Actividad , Tuberculosis/tratamiento farmacológico , Células Vero
20.
Antimicrob Agents Chemother ; 60(10): 5817-27, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27431220

RESUMEN

The apicomplexan parasites Cryptosporidium and Toxoplasma are serious threats to human health. Cryptosporidiosis is a severe diarrheal disease in malnourished children and immunocompromised individuals, with the only FDA-approved drug treatment currently being nitazoxanide. The existing therapies for toxoplasmosis, an important pathology in immunocompromised individuals and pregnant women, also have serious limitations. With the aim of developing alternative therapeutic options to address these health problems, we tested a number of benzoxaboroles, boron-containing compounds shown to be active against various infectious agents, for inhibition of the growth of Cryptosporidium parasites in mammalian cells. A 3-aminomethyl benzoxaborole, AN6426, with activity in the micromolar range and with activity comparable to that of nitazoxanide, was identified and further characterized using biophysical measurements of affinity and crystal structures of complexes with the editing domain of Cryptosporidium leucyl-tRNA synthetase (LeuRS). The same compound was shown to be active against Toxoplasma parasites, with the activity being enhanced in the presence of norvaline, an amino acid that can be mischarged by LeuRS. Our observations are consistent with AN6426 inhibiting protein synthesis in both Cryptosporidium and Toxoplasma by forming a covalent adduct with tRNA(Leu) in the LeuRS editing active site and suggest that further exploitation of the benzoxaborole scaffold is a valid strategy to develop novel, much needed antiparasitic agents.


Asunto(s)
Antiprotozoarios/farmacología , Compuestos de Boro/farmacología , Cryptosporidium parvum/efectos de los fármacos , Leucina-ARNt Ligasa/antagonistas & inhibidores , Leucina-ARNt Ligasa/química , Toxoplasma/efectos de los fármacos , Animales , Antiprotozoarios/química , Antiprotozoarios/metabolismo , Compuestos de Boro/química , Cristalografía por Rayos X , Perros , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/parasitología , Humanos , Leucina-ARNt Ligasa/metabolismo , Células de Riñón Canino Madin Darby/parasitología , Simulación del Acoplamiento Molecular , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...