Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Phys Chem B ; 125(42): 11606-11616, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34648705

RESUMEN

Catalytic fields representing the topology of the optimal molecular environment charge distribution that reduces the activation barrier have been used to examine alternative reaction variants and to determine the role of conserved catalytic residues for two consecutive reactions catalyzed by the same enzyme. Until now, most experimental and conventional top-down theoretical studies employing QM/MM or ONIOM methods have focused on the role of enzyme electric fields acting on broken bonds of reactants. In contrast, our bottom-up approach dealing with a small reactant and transition-state model allows the analysis of the opposite effects: how the catalytic field resulting from the charge redistribution during the enzyme reaction acts on conserved amino acid residues and contributes to the reduction of the activation barrier. This approach has been applied to the family of histidyl tRNA synthetases involved in the translation of the genetic code into the protein amino acid sequence. Activation energy changes related to conserved charged amino acid residues for 12 histidyl tRNA synthetases from different biological species allowed to compare on equal footing the catalytic residues involved in ATP aminoacylation and tRNA charging reactions and to analyze different reaction mechanisms proposed in the literature. A scan of the library of atomic multipoles for amino acid side-chain rotamers within the catalytic field pointed out the change in the Glu83 conformation as the critical catalytic effect, providing, at low computational cost, insight into the electrostatic preorganization of the enzyme catalytic site at a level of detail that has not yet been accessible in conventional experimental or theoretical methods. This opens the way for rational reverse biocatalyst design at a very limited computational cost without resorting to empirical methods.


Asunto(s)
Histidina-ARNt Ligasa , Aminoacilación , Catálisis , Dominio Catalítico , Histidina-ARNt Ligasa/metabolismo , Electricidad Estática
2.
FEBS J ; 288(1): 142-159, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32543048

RESUMEN

Charcot-Marie-Tooth disease (CMT) encompasses a set of genetically and clinically heterogeneous neuropathies characterized by length-dependent dysfunction of the peripheral nervous system. Mutations in over 80 diverse genes are associated with CMT, and aminoacyl-tRNA synthetases (ARS) constitute a large gene family implicated in the disease. Despite considerable efforts to elucidate the mechanistic link between ARS mutations and the CMT phenotype, the molecular basis of the pathology is unknown. In this work, we investigated the impact of three CMT-associated substitutions (V155G, Y330C, and R137Q) in the cytoplasmic histidyl-tRNA synthetase (HARS1) on neurite outgrowth and peripheral nervous system development. The model systems for this work included a nerve growth factor-stimulated neurite outgrowth model in rat pheochromocytoma cells (PC12), and a zebrafish line with GFP/red fluorescent protein reporters of sensory and motor neuron development. The expression of CMT-HARS1 mutations led to attenuation of protein synthesis and increased phosphorylation of eIF2α in PC12 cells and was accompanied by impaired neurite and axon outgrowth in both models. Notably, these effects were phenocopied by histidinol, a HARS1 inhibitor, and cycloheximide, a protein synthesis inhibitor. The mutant proteins also formed heterodimers with wild-type HARS1, raising the possibility that CMT-HARS1 mutations cause disease through a dominant-negative mechanism. Overall, these findings support the hypothesis that CMT-HARS1 alleles exert their toxic effect in a neuronal context, and lead to dysregulated protein synthesis. These studies demonstrate the value of zebrafish as a model for studying mutant alleles associated with CMT, and for characterizing the processes that lead to peripheral nervous system dysfunction.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Histidina-ARNt Ligasa/genética , Proyección Neuronal/genética , Neuronas/metabolismo , Sistema Nervioso Periférico/metabolismo , Biosíntesis de Proteínas , Animales , Animales Modificados Genéticamente , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedad de Charcot-Marie-Tooth/patología , Cicloheximida/farmacología , Modelos Animales de Enfermedad , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histidina-ARNt Ligasa/antagonistas & inhibidores , Histidina-ARNt Ligasa/metabolismo , Histidinol/farmacología , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mutación , Proyección Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Células PC12 , Sistema Nervioso Periférico/patología , Multimerización de Proteína , Ratas , Pez Cebra , Proteína Fluorescente Roja
3.
Genes (Basel) ; 11(9)2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32906706

RESUMEN

Aminoacyl-tRNA synthetases (aaRSs) play essential roles in protein translation. In addition, numerous aaRSs (mostly in vertebrates) have also been discovered to possess a range of non-canonical functions. Very few studies have been conducted to elucidate or characterize non-canonical functions of plant aaRSs. A genome-wide search for aaRS genes in Arabidopsis thaliana revealed a total of 59 aaRS genes. Among them, asparaginyl-tRNA synthetase (AsnRS) was found to possess a WHEP domain inserted into the catalytic domain in a plant-specific manner. This insertion was observed only in the cytosolic isoform. In addition, a long stretch of sequence that exhibited weak homology with histidine ammonia lyase (HAL) was found at the N-terminus of histidyl-tRNA synthetase (HisRS). This HAL-like domain has only been seen in plant HisRS, and only in cytosolic isoforms. Additionally, a number of genes lacking minor or major portions of the full-length aaRS sequence were found. These genes encode 14 aaRS fragments that lack key active site sequences and are likely catalytically null. These identified genes that encode plant-specific additional domains or aaRS fragment sequences are candidates for aaRSs possessing non-canonical functions.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Aspartato-ARNt Ligasa/metabolismo , Genoma de Planta , Histidina-ARNt Ligasa/metabolismo , Aminoacil-ARN de Transferencia/metabolismo , Aminoacil-ARNt Sintetasas/genética , Arabidopsis/genética , Arabidopsis/crecimiento & desarrollo , Proteínas de Arabidopsis/genética , Aspartato-ARNt Ligasa/genética , Dominio Catalítico , Histidina-ARNt Ligasa/genética , Biosíntesis de Proteínas , Aminoacil-ARN de Transferencia/genética
4.
Proc Natl Acad Sci U S A ; 116(39): 19440-19448, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31501329

RESUMEN

Aminoacyl-transfer RNA (tRNA) synthetases (aaRSs) are the largest protein family causatively linked to neurodegenerative Charcot-Marie-Tooth (CMT) disease. Dominant mutations cause the disease, and studies of CMT disease-causing mutant glycyl-tRNA synthetase (GlyRS) and tyrosyl-tRNA synthetase (TyrRS) showed their mutations create neomorphic structures consistent with a gain-of-function mechanism. In contrast, based on a haploid yeast model, loss of aminoacylation function was reported for CMT disease mutants in histidyl-tRNA synthetase (HisRS). However, neither that nor prior work of any CMT disease-causing aaRS investigated the aminoacylation status of tRNAs in the cellular milieu of actual patients. Using an assay that interrogated aminoacylation levels in patient cells, we investigated a HisRS-linked CMT disease family with the most severe disease phenotype. Strikingly, no difference in charged tRNA levels between normal and diseased family members was found. In confirmation, recombinant versions of 4 other HisRS CMT disease-causing mutants showed no correlation between activity loss in vitro and severity of phenotype in vivo. Indeed, a mutation having the most detrimental impact on activity was associated with a mild disease phenotype. In further work, using 3 independent biophysical analyses, structural opening (relaxation) of mutant HisRSs at the dimer interface best correlated with disease severity. In fact, the HisRS mutation in the severely afflicted patient family caused the largest degree of structural relaxation. These data suggest that HisRS-linked CMT disease arises from open conformation-induced mechanisms distinct from loss of aminoacylation.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Enfermedad de Charcot-Marie-Tooth/genética , Histidina-ARNt Ligasa/genética , Secuencia de Aminoácidos , Aminoacil-ARNt Sintetasas/metabolismo , Aminoacilación/genética , Axones , Enfermedad de Charcot-Marie-Tooth/metabolismo , Mutación con Ganancia de Función/genética , Histidina-ARNt Ligasa/metabolismo , Humanos , Mutación , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Relación Estructura-Actividad , Tirosina-ARNt Ligasa/genética , Tirosina-ARNt Ligasa/metabolismo
5.
Crit Rev Immunol ; 38(4): 263-278, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30806243

RESUMEN

Among the inflammatory myopathies, anti-tRNA-synthetase syndrome (ASyS) is a severe autoimmune condition characterized by extramuscular involvement, affecting especially the lungs. ASyS specific serological markers are anti-tRNA-synthetase autoantibodies, among which anti-histidyl-tRNA-synthetase is the most common. In the past decades, ASyS has been distinguished by unique histological features attributed to a specific pathogenesis. Research has highlighted the role of environmental factors and infections as possible triggers. Tissue modifications of histidyl-tRNA-synthetase (HisRS) expression might be responsible for the recruitment and activation of both innate and adaptive immune cells. HisRS not only acts through antigenic properties, but also through many others, including chemoattraction, innate pathway activation, and cytokine-like functions. Favored by a certain genetic background, this whole activation of immunity results in widespread and specific tissue damage and finally leads to visible heterogeneous symptoms characterizing the disease state. Understanding the pathogenesis of ASyS is essential to improving patient care by identifying biomarkers and designing new therapeutic strategies accordingly. Therefore, this review details the recent hypotheses concerning the dynamic of ASyS pathogenesis with the aim of enlightening the development of new therapeutic axes in the future.


Asunto(s)
Miositis/inmunología , Miositis/patología , Animales , Histidina-ARNt Ligasa/biosíntesis , Histidina-ARNt Ligasa/inmunología , Histidina-ARNt Ligasa/metabolismo , Humanos , Miositis/genética
6.
Hum Mutat ; 39(3): 415-432, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29235198

RESUMEN

Histidyl-tRNA synthetase (HARS) ligates histidine to cognate tRNA molecules, which is required for protein translation. Mutations in HARS cause the dominant axonal peripheral neuropathy Charcot-Marie-Tooth disease type 2W (CMT2W); however, the precise molecular mechanism remains undefined. Here, we investigated three HARS missense mutations associated with CMT2W (p.Tyr330Cys, p.Ser356Asn, and p.Val155Gly). The three mutations localize to the HARS catalytic domain and failed to complement deletion of the yeast ortholog (HTS1). Enzyme kinetics, differential scanning fluorimetry (DSF), and analytical ultracentrifugation (AUC) were employed to assess the effect of these substitutions on primary aminoacylation function and overall dimeric structure. Notably, the p.Tyr330Cys, p.Ser356Asn, and p.Val155Gly HARS substitutions all led to reduced aminoacylation, providing a direct connection between CMT2W-linked HARS mutations and loss of canonical ARS function. While DSF assays revealed that only one of the variants (p.Val155Gly) was less thermally stable relative to wild-type, all three HARS mutants formed stable dimers, as measured by AUC. Our work represents the first biochemical analysis of CMT-associated HARS mutations and underscores how loss of the primary aminoacylation function can contribute to disease pathology.


Asunto(s)
Axones/patología , Histidina-ARNt Ligasa/metabolismo , Enfermedades del Sistema Nervioso Periférico/enzimología , Enfermedades del Sistema Nervioso Periférico/patología , Secuencia de Aminoácidos , Aminoacilación , Biocatálisis , Dominio Catalítico , Secuencia Conservada , Femenino , Prueba de Complementación Genética , Histidina-ARNt Ligasa/química , Histidina-ARNt Ligasa/genética , Histidina-ARNt Ligasa/aislamiento & purificación , Humanos , Cinética , Masculino , Mutación/genética , Linaje , Enfermedades del Sistema Nervioso Periférico/genética , Multimerización de Proteína , Especificidad por Sustrato
7.
SLAS Discov ; 23(1): 65-75, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28745975

RESUMEN

Pseudomonas aeruginosa histidyl-tRNA synthetase (HisRS) was selected as a target for antibiotic drug development. The HisRS protein was overexpressed in Escherichia coli and kinetically evaluated. The KM values for interaction of HisRS with its three substrates, histidine, ATP, and tRNAHis, were 37.6, 298.5, and 1.5 µM, while the turnover numbers were 8.32, 16.8, and 0.57 s-1, respectively. A robust screening assay was developed, and 800 natural products and 890 synthetic compounds were screened for inhibition of activity. Fifteen compounds with inhibitory activity were identified, and the minimum inhibitory concentration (MIC) was determined for each against a panel of nine pathogenic bacteria. Each compound exhibited broad-spectrum activity. Based on structural similarity and MIC results, four compounds, BT02C02, BT02D04, BT08E04, and BT09C11, were selected for additional analysis. These compounds inhibited the activity of HisRS with IC50 values of 4.4, 9.7, 14.1, and 11.3 µM, respectively. Time-kill studies indicated a bacteriostatic mode of inhibition for each compound. BT02D04 and BT08E04 were noncompetitive with both histidine and ATP, BT02C02 was competitive with histidine but noncompetitive with ATP, and BT09C11 was uncompetitive with histidine and noncompetitive with ATP. These compounds were not observed to be toxic to human cell cultures.


Asunto(s)
Antibacterianos/farmacología , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Histidina-ARNt Ligasa/antagonistas & inhibidores , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/enzimología , Secuencia de Aminoácidos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Expresión Génica , Histidina-ARNt Ligasa/genética , Histidina-ARNt Ligasa/metabolismo , Humanos , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Pseudomonas aeruginosa/genética
8.
PLoS One ; 12(9): e0185317, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28934368

RESUMEN

Histidyl tRNA Synthetase (HARS) is a member of the aminoacyl tRNA synthetase (ARS) family of enzymes. This family of 20 enzymes is responsible for attaching specific amino acids to their cognate tRNA molecules, a critical step in protein synthesis. However, recent work highlighting a growing number of associations between ARS genes and diverse human diseases raises the possibility of new and unexpected functions in this ancient enzyme family. For example, mutations in HARS have been linked to two different neurological disorders, Usher Syndrome Type IIIB and Charcot Marie Tooth peripheral neuropathy. These connections raise the possibility of previously undiscovered roles for HARS in metazoan development, with alterations in these functions leading to complex diseases. In an attempt to establish Danio rerio as a model for studying HARS functions in human disease, we characterized the Danio rerio hars gene and compared it to that of human HARS. Using a combination of bioinformatics, molecular biology, and cellular approaches, we found that while the human genome encodes separate genes for cytoplasmic and mitochondrial HARS protein, the Danio rerio genome encodes a single hars gene which undergoes alternative splicing to produce the respective cytoplasmic and mitochondrial versions of Hars. Nevertheless, while the HARS genes of humans and Danio differ significantly at the genomic level, we found that they are still highly conserved at the amino acid level, underscoring the potential utility of Danio rerio as a model organism for investigating HARS function and its link to human diseases in vivo.


Asunto(s)
Citoplasma/enzimología , Citoplasma/genética , Histidina-ARNt Ligasa/genética , Mitocondrias/enzimología , Pez Cebra/genética , Animales , Células COS , Chlorocebus aethiops , Secuencia Conservada , Regulación Enzimológica de la Expresión Génica , Histidina-ARNt Ligasa/química , Histidina-ARNt Ligasa/metabolismo , Humanos , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especificidad de la Especie
9.
Biochemistry ; 56(28): 3619-3631, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28632987

RESUMEN

Histidyl-tRNA synthetase (HARS) is a highly conserved translation factor that plays an essential role in protein synthesis. HARS has been implicated in the human syndromes Charcot-Marie-Tooth (CMT) Type 2W and Type IIIB Usher (USH3B). The USH3B mutation, which encodes a Y454S substitution in HARS, is inherited in an autosomal recessive fashion and associated with childhood deafness, blindness, and episodic hallucinations during acute illness. The biochemical basis of the pathophysiologies linked to USH3B is currently unknown. Here, we present a detailed functional comparison of wild-type (WT) and Y454S HARS enzymes. Kinetic parameters for enzymes and canonical substrates were determined using both steady state and rapid kinetics. Enzyme stability was examined using differential scanning fluorimetry. Finally, enzyme functionality in a primary cell culture was assessed. Our results demonstrate that the Y454S substitution leaves HARS amino acid activation, aminoacylation, and tRNAHis binding functions largely intact compared with those of WT HARS, and the mutant enzyme dimerizes like the wild type does. Interestingly, during our investigation, it was revealed that the kinetics of amino acid activation differs from that of the previously characterized bacterial HisRS. Despite the similar kinetics, differential scanning fluorimetry revealed that Y454S is less thermally stable than WT HARS, and cells from Y454S patients grown at elevated temperatures demonstrate diminished levels of protein synthesis compared to those of WT cells. The thermal sensitivity associated with the Y454S mutation represents a biochemical basis for understanding USH3B.


Asunto(s)
Histidina-ARNt Ligasa/genética , Histidina-ARNt Ligasa/metabolismo , Mutación Puntual , Síndromes de Usher/enzimología , Síndromes de Usher/genética , Secuencia de Aminoácidos , Aminoacilación , Células Cultivadas , Estabilidad de Enzimas , Células HEK293 , Histidina-ARNt Ligasa/química , Humanos , Cinética , Modelos Moleculares , Biosíntesis de Proteínas , ARN de Transferencia/metabolismo , Alineación de Secuencia , Temperatura , Síndromes de Usher/metabolismo
10.
Cell Mol Life Sci ; 74(14): 2663-2677, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28321488

RESUMEN

The discriminator base N73 is a key identity element of tRNAHis. In eukaryotes, N73 is an "A" in cytoplasmic tRNAHis and a "C" in mitochondrial tRNAHis. We present evidence herein that yeast histidyl-tRNA synthetase (HisRS) recognizes both A73 and C73, but somewhat prefers A73 even within the context of mitochondrial tRNAHis. In contrast, humans possess two distinct yet closely related HisRS homologues, with one encoding the cytoplasmic form (with an extra N-terminal WHEP domain) and the other encoding its mitochondrial counterpart (with an extra N-terminal mitochondrial targeting signal). Despite these two isoforms sharing high sequence similarities (81% identity), they strongly preferred different discriminator bases (A73 or C73). Moreover, only the mitochondrial form recognized the anticodon as a strong identity element. Most intriguingly, swapping the discriminator base between the cytoplasmic and mitochondrial tRNAHis isoacceptors conveniently switched their enzyme preferences. Similarly, swapping seven residues in the active site between the two isoforms readily switched their N73 preferences. This study suggests that the human HisRS genes, while descending from a common ancestor with dual function for both types of tRNAHis, have acquired highly specialized tRNA recognition properties through evolution.


Asunto(s)
Evolución Molecular , Histidina-ARNt Ligasa/metabolismo , ARN de Transferencia/metabolismo , Secuencia de Aminoácidos , Aminoacilación , Bacillus subtilis/enzimología , Escherichia coli/enzimología , Histidina-ARNt Ligasa/química , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Mitocondrias/metabolismo , Proteínas Mutantes/metabolismo , Filogenia , Saccharomyces cerevisiae/enzimología , Especificidad por Sustrato
11.
Biochim Biophys Acta Gen Subj ; 1861(11 Pt B): 3009-3015, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28288813

RESUMEN

BACKGROUND: Development of new aminoacyl-tRNA synthetase (aaRS)•tRNA pairs is central for incorporation of novel non-canonical amino acids (ncAAs) into proteins via genetic code expansion (GCE). The Escherichia coli and Caulobacter crescentus histidyl-tRNA synthetases (HisRS) evolved divergent mechanisms of tRNAHis recognition that prevent their cross-reactivity. Although the E. coli HisRS•tRNAHis pair is a good candidate for GCE, its use in C. crescentus is limited by the lack of established genetic selection methods and by the low transformation efficiency of C. crescentus. METHODS: E. coli was genetically engineered to use a C. crescentus HisRS•tRNAHis pair. Super-folder green fluorescent protein (sfGFP) and chloramphenicol acetyltransferase (CAT) were used as reporters for read-through assays. A library of 313 ncAAs coupled with the sfGFP reporter system was employed to investigate the specificity of E. coli HisRS in vivo. RESULTS: A genomically modified E. coli strain (named MEOV1) was created. MEVO1 requires an active C. crescentus HisRS•tRNAHis pair for growth, and displays a similar doubling time as the parental E. coli strain. sfGFP- and CAT-based assays showed that the E. coli HisRS•tRNAHis pair is orthogonal in MEOV1 cells. A mutation in the anticodon loop of E. coli tRNAHisCUA elevated its suppression efficiency by 2-fold. CONCLUSIONS: The C. crescentus HisRS•tRNAHis pair functionally complements an E. coli ΔhisS strain. The E. coli HisRS•tRNAHis is orthogonal in MEOV1 cells. E. coli tRNAHisCUA is an efficient amber suppressor in MEOV1. GENERAL SIGNIFICANCE: We developed a platform that allows protein engineering of E. coli HisRS that should facilitate GCE in E. coli. This article is part of a Special Issue entitled "Biochemistry of Synthetic Biology - Recent Developments" Guest Editor: Dr. Ilka Heinemann and Dr. Patrick O'Donoghue.


Asunto(s)
Escherichia coli/genética , Escherichia coli/metabolismo , Histidina-ARNt Ligasa/metabolismo , Ingeniería de Proteínas/métodos , ARN de Transferencia de Histidina/metabolismo , Clonación Molecular/métodos , Biblioteca de Genes , Ingeniería Genética/métodos , Histidina-ARNt Ligasa/genética , Mutagénesis Sitio-Dirigida , ARN de Transferencia de Histidina/genética
12.
Methods ; 113: 64-71, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27794454

RESUMEN

Differential scanning fluorimetry (DSF) is a fluorescence-based assay to evaluate protein stability by determining protein melting temperatures. Here, we describe the application of DSF to investigate aminoacyl-tRNA synthetase (AARS) stability and interaction with ligands. Employing three bacterial AARS enzymes as model systems, methods are presented here for the use of DSF to measure the apparent temperatures at which AARSs undergo melting transitions, and the effect of AARS substrates and inhibitors. One important observation is that the extent of temperature stability realized by an AARS in response to a particular bound ligand cannot be predicted a priori. The DSF method thus serves as a rapid and highly quantitative approach to measure AARS stability, and the ability of ligands to influence the temperature at which unfolding transitions occur.


Asunto(s)
Alanina-ARNt Ligasa/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Histidina-ARNt Ligasa/química , ARN de Transferencia Aminoácido-Específico/metabolismo , Treonina-ARNt Ligasa/química , Alanina-ARNt Ligasa/antagonistas & inhibidores , Alanina-ARNt Ligasa/genética , Alanina-ARNt Ligasa/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Benzopiranos/química , Inhibidores Enzimáticos/química , Estabilidad de Enzimas , Escherichia coli/genética , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Colorantes Fluorescentes/química , Fluorometría/métodos , Histidina-ARNt Ligasa/antagonistas & inhibidores , Histidina-ARNt Ligasa/genética , Histidina-ARNt Ligasa/metabolismo , Muramidasa/química , Muramidasa/metabolismo , Transición de Fase , Unión Proteica , Desplegamiento Proteico , ARN de Transferencia Aminoácido-Específico/genética , Especificidad por Sustrato , Treonina-ARNt Ligasa/antagonistas & inhibidores , Treonina-ARNt Ligasa/genética , Treonina-ARNt Ligasa/metabolismo , Aminoacilación de ARN de Transferencia
13.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 8): 1684-98, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26249349

RESUMEN

American trypanosomiasis, commonly known as Chagas disease, is a neglected tropical disease caused by the protozoan parasite Trypanosoma cruzi. The chronic form of the infection often causes debilitating morbidity and mortality. However, the current treatment for the disease is typically inadequate owing to drug toxicity and poor efficacy, necessitating a continual effort to discover and develop new antiparasitic therapeutic agents. The structure of T. cruzi histidyl-tRNA synthetase (HisRS), a validated drug target, has previously been reported. Based on this structure and those of human cytosolic HisRS, opportunities for the development of specific inhibitors were identified. Here, efforts are reported to identify small molecules that bind to T. cruzi HisRS through fragment-based crystallographic screening in order to arrive at chemical starting points for the development of specific inhibitors. T. cruzi HisRS was soaked into 68 different cocktails from the Medical Structural Genomics of Pathogenic Protozoa (MSGPP) fragment library and diffraction data were collected to identify bound fragments after soaking. A total of 15 fragments were identified, all bound to the same site on the protein, revealing a fragment-binding hotspot adjacent to the ATP-binding pocket. On the basis of the initial hits, the design of reactive fragments targeting the hotspot which would be simultaneously covalently linked to a cysteine residue present only in trypanosomatid HisRS was initiated. Inhibition of T. cruzi HisRS was observed with the resultant reactive fragments and the anticipated binding mode was confirmed crystallographically. These results form a platform for the development of future generations of selective inhibitors for trypanosomatid HisRS.


Asunto(s)
Inhibidores Enzimáticos/química , Histidina-ARNt Ligasa/antagonistas & inhibidores , Histidina-ARNt Ligasa/química , Bibliotecas de Moléculas Pequeñas/química , Trypanosoma cruzi/enzimología , Sitios de Unión , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/microbiología , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Histidina-ARNt Ligasa/metabolismo , Humanos , Modelos Moleculares , Bibliotecas de Moléculas Pequeñas/farmacología , Trypanosoma cruzi/química , Trypanosoma cruzi/efectos de los fármacos , Trypanosoma cruzi/metabolismo
14.
Nucleic Acids Res ; 43(5): 2980-90, 2015 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-25722375

RESUMEN

Aminoacyl-tRNA synthetases (aaRSs) play a crucial role in protein translation by linking tRNAs with cognate amino acids. Among all the tRNAs, only tRNA(His) bears a guanine base at position -1 (G-1), and it serves as a major recognition element for histidyl-tRNA synthetase (HisRS). Despite strong interests in the histidylation mechanism, the tRNA recognition and aminoacylation details are not fully understood. We herein present the 2.55 Å crystal structure of HisRS complexed with tRNA(His), which reveals that G-1 recognition is principally nonspecific interactions on this base and is made possible by an enlarged binding pocket consisting of conserved glycines. The anticodon triplet makes additional specific contacts with the enzyme but the rest of the loop is flexible. Based on the crystallographic and biochemical studies, we inferred that the uniqueness of histidylation system originates from the enlarged binding pocket (for the extra base G-1) on HisRS absent in other aaRSs, and this structural complementarity between the 5' extremity of tRNA and enzyme is probably a result of coevolution of both.


Asunto(s)
Guanina/química , Histidina-ARNt Ligasa/química , Conformación de Ácido Nucleico , Estructura Terciaria de Proteína , ARN de Transferencia de Histidina/química , Secuencia de Aminoácidos , Aminoacilación , Secuencia de Bases , Dominio Catalítico , Cristalografía por Rayos X , Guanina/metabolismo , Histidina-ARNt Ligasa/genética , Histidina-ARNt Ligasa/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , ARN de Transferencia de Histidina/genética , ARN de Transferencia de Histidina/metabolismo , Homología de Secuencia de Aminoácido , Thermus thermophilus/enzimología , Thermus thermophilus/genética
15.
Biochimie ; 106: 111-20, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25151410

RESUMEN

As part of a project aimed at obtaining selective inhibitors and drug-like compounds targeting tRNA synthetases from trypanosomatids, we have elucidated the crystal structure of human cytosolic histidyl-tRNA synthetase (Hs-cHisRS) in complex with histidine in order to be able to compare human and parasite enzymes. The resultant structure of Hs-cHisRS•His represents the substrate-bound state (H-state) of the enzyme. It provides an interesting opportunity to compare with ligand-free and imidazole-bound structures Hs-cHisRS published recently, both of which represent the ligand-free state (F-state) of the enzyme. The H-state Hs-cHisRS undergoes conformational changes in active site residues and several conserved motif of HisRS, compared to F-state structures. The histidine forms eight hydrogen bonds with HisRS of which six engage the amino and carboxylate groups of this amino acid. The availability of published imidazole-bound structure provides a unique opportunity to dissect the structural roles of individual chemical groups of histidine. The analysis revealed the importance of the amino and carboxylate groups, of the histidine in leading to these dramatic conformational changes of the H-state. Further, comparison with previously published trypanosomatid HisRS structures reveals a pocket in the F-state of the parasite enzyme that may provide opportunities for developing specific inhibitors of Trypanosoma brucei HisRS.


Asunto(s)
Histidina-ARNt Ligasa/química , Histidina/química , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Trypanosoma brucei brucei/enzimología , Secuencia de Aminoácidos , Sitios de Unión/genética , Biocatálisis/efectos de los fármacos , Dominio Catalítico , Cristalografía por Rayos X , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Histidina/metabolismo , Histidina-ARNt Ligasa/genética , Histidina-ARNt Ligasa/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Trypanosoma brucei brucei/genética
16.
J Biol Chem ; 289(28): 19269-75, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24898250

RESUMEN

Inflammatory and debilitating myositis and interstitial lung disease are commonly associated with autoantibodies (anti-Jo-1 antibodies) to cytoplasmic histidyl-tRNA synthetase (HisRS). Anti-Jo-1 antibodies from different disease-afflicted patients react mostly with spatially separated epitopes in the three-dimensional structure of human HisRS. We noted that two HisRS splice variants (SVs) include these spatially separated regions, but each SV lacks the HisRS catalytic domain. Despite the large deletions, the two SVs cross-react with a substantial population of anti-Jo-l antibodies from myositis patients. Moreover, expression of at least one of the SVs is up-regulated in dermatomyositis patients, and cell-based experiments show that both SVs and HisRS can be secreted. We suggest that, in patients with inflammatory myositis, anti-Jo-1 antibodies may have extracellular activity.


Asunto(s)
Empalme Alternativo , Autoanticuerpos/metabolismo , Epítopos/metabolismo , Histidina-ARNt Ligasa/metabolismo , Miositis/enzimología , Autoanticuerpos/inmunología , Línea Celular Tumoral , Epítopos/genética , Epítopos/inmunología , Histidina-ARNt Ligasa/genética , Histidina-ARNt Ligasa/inmunología , Humanos , Inflamación/enzimología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Miositis/genética , Miositis/inmunología , Miositis/patología , Estructura Terciaria de Proteína
17.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 27(9): 1128-33, 2013 Sep.
Artículo en Chino | MEDLINE | ID: mdl-24279029

RESUMEN

OBJECTIVE: To explore the effects of mechanical stimulation on the expression of autoantigens in myoblasts. METHODS: According to different processing methods, C2C12 cells were divided into the experimental group and control group; the experimental group was divided into 4 subgroups: 2-, 4-, and 6-day and 1-day stretch groups. In 2-, 4-, and 6-day stretch groups, mechanical loading was added on the C2C12 cells at a stretching frequency of 0.25 Hz and cellular deformation amplitude of 10%, 2 hours a day for 2, 4, and 6 days respectively by Flexercell 5000 strain unit, and at a stretching frequency of 1 Hz and cellular deformation amplitude of 15% for 1 hour in 1-day stretch group. In the control group, the cells were routinely cultured for 1, 2, 4, and 6 days (1-, 2-, 4-, and 6-day control). The cells were observed by inverted phase contrast microscope. The cell proliferation was detected by flow cytometry; the expressions of autoantigens were detected by Western blot method, including the Ku/the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs), U1-70 (A part of ATP-dependent DNA helicase II), histidyl tRNA synthetase (HRS), and Mi-2 (reconfigurable components deacetylase complexes of NuRD). RESULTS: The exfoliated cells were found in 1-day stretch group, but no exfoliated cell was seen in the control group for 1-day culture. The cells proliferated more obviously in 2-day stretch group than in the control group for 2-day culture; cell differentiation was found in 4-day stretch group, and cell fusion in 6-day stretch group, which were similar to those in the control group for 4- and 6-day culture. After single stretching, cell apoptosis was found in 1-day stretch group, showing no significant difference in the relative DNA proliferation index (DPI) when compared with DPI of control group for 1-day culture (t = 0.346, P = 0.747). After cyclic stretching, DPIs of 2- and 4- day stretch groups were significantly increased when compared with those of the control group for 2- and 4-day culture (P < 0.05), but no significant difference was found between control group for 6-day culture and 6-day stretch group (t = 1.191, P = 0.303). Compared with the control group for 2-day culture, the relative protein expression of autoantigens (DNA-Pkcs, Mi-2, HRS, and U1-70) in 2-day stretch group decreased significantly (P < 0.05), but no significant difference was found between control group for 4-day culture and 4-day stretch group (P > 0.05). The relative protein expressions of autoantigens in 4-day stretch group significantly increased when compared with those of 2-day stretch group (P < 0.05), but the relative protein expressions of autoantigens in the control group for 4-day culture significantly decreased when compared with those of the control group for 2-day culture (P < 0.05). CONCLUSION: Short-term mechanical stimulation can inhibit the expressions of autoantigens in myoblasts, but with the time prolonging, cell differentiation and fusion and adaptation to mechanical stimulation would result in diminished inhibitory effect.


Asunto(s)
Autoantígenos/metabolismo , Proliferación Celular , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/fisiología , Estrés Mecánico , Western Blotting , Diferenciación Celular , Células Cultivadas , Proteína Quinasa Activada por ADN/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica , Histidina-ARNt Ligasa/metabolismo , Humanos , Miositis/metabolismo , Miositis/patología , Resistencia a la Tracción
18.
Mater Sci Eng C Mater Biol Appl ; 33(8): 4867-70, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24094198

RESUMEN

An amino acid-sensing system with absorption spectrophotometric detection was developed. To ensure specific recognition of each amino acid, aminoacyl-tRNA synthetases were employed and the concentration of NADH produced by way of several enzymatic reactions was measured. Using this detection system, from 1.5 to 55 µM of histidine and from 15 to 95 µM of lysine could be measured selectively in HEPES-KOH buffer (pH 8.0).


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Histidina/análisis , Lisina/análisis , Espectrofotometría , Histidina-ARNt Ligasa/metabolismo , Concentración de Iones de Hidrógeno , Lisina-ARNt Ligasa/metabolismo , NAD/metabolismo
19.
Curr Rheumatol Rep ; 15(6): 335, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23591825

RESUMEN

Inflammatory myopathies are a group of acquired diseases, characterized by immunoflogistic processes primarily involving the skeletal muscle. According to recent classification criteria, four major diseases have been identified: polymyositis (PM), dermatomyositis (DM), sporadic inclusion body myositis (IBM), and necrotizing autoimmune myositis (NAM). Autoantibodies can be found in the sera of most patients with myositis. Myositis-specific autoantibodies (MSAs) are markers of very specific disease entities within the spectrum of myositis, and target proteins involved in key processes of protein synthesis. Myositis autoantigens comprise the well-defined aminoacyl-tRNA synthetases, the Mi-2 helicase/histone deacetylase protein complex, and the signal recognition particle (SRP) ribonucleoprotein, together with novel targets such as TIF1-γ, MDA5, NXP2, SAE, and HMGCR. Recent studies suggest that autoantigens drive a B cell antigen-specific immune response in muscles. Interestingly, an increased expression of Jo-1 and Mi-2 in regenerating fibers in muscle biopsies from PM and DM patients compared to normal was demonstrated. Myositis autoantigen up-regulation was observed in neoplastic tissues, thus representing a potential link between cancer and autoimmunity in myositis. Non-immunological mechanisms seem to participate to the pathogenesis of inflammatory myopathies; induction of endoplasmic reticulum stress response in response to abnormal muscle regeneration and inflammation has recently been reported in patients with myositis. This review article provides an update of new emerging insights about the clinical and pathophysiologic role of principal autoantibodies in myositis.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Autoanticuerpos/inmunología , Autoantígenos/metabolismo , Dermatomiositis/sangre , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Músculo Esquelético/metabolismo , Partícula de Reconocimiento de Señal/metabolismo , Aminoacil-ARNt Sintetasas/inmunología , Autoanticuerpos/metabolismo , Autoantígenos/inmunología , Biomarcadores/metabolismo , Dermatomiositis/inmunología , Dermatomiositis/fisiopatología , Histidina-ARNt Ligasa/inmunología , Histidina-ARNt Ligasa/metabolismo , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/inmunología , Fibras Musculares Esqueléticas/inmunología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Regeneración , Partícula de Reconocimiento de Señal/inmunología
20.
Nucleic Acids Res ; 41(3): 1885-94, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23241387

RESUMEN

The additional G(-1) nucleotide on tRNA(His) is a nearly universal feature that specifies tRNA(His) identity in all three domains of life. In eukaryotes, the G(-1) identity element is obtained by a post-transcriptional pathway, through the unusual 3'-5' polymerase activity of the highly conserved tRNA(His) guanylyltransferase (Thg1) enzyme, and no examples of eukaryotic histidyl-tRNAs that lack this essential element have been identified. Here we report that the eukaryote Acanthamoeba castellanii lacks the G(-1) identity element on its tRNA(His), consistent with the lack of a gene encoding a bona fide Thg1 ortholog in the A. castellanii genome. Moreover, the cytosolic histidyl-tRNA synthetase in A. castellanii exhibits an unusual tRNA substrate specificity, efficiently aminoacylating tRNA(His) regardless of the presence of G(-1). A. castellanii does contain two Thg1-related genes (encoding Thg1-like proteins, TLPs), but the biochemical properties we associate here with these proteins are consistent with a function for these TLPs in separate pathways unrelated to tRNA(His) metabolism, such as mitochondrial tRNA repair during 5'-editing.


Asunto(s)
Acanthamoeba castellanii/genética , ARN de Transferencia de Histidina/química , Acanthamoeba castellanii/enzimología , ARN Polimerasas Dirigidas por ADN/metabolismo , Histidina-ARNt Ligasa/metabolismo , Nucleotidiltransferasas/metabolismo , ARN/metabolismo , Edición de ARN , ARN Mitocondrial , ARN de Transferencia/metabolismo , ARN de Transferencia de Histidina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA