Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
2.
Am J Hum Genet ; 111(4): 778-790, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38531365

RESUMEN

Selenophosphate synthetase (SEPHS) plays an essential role in selenium metabolism. Two mammalian SEPHS paralogues, SEPHS1 and SEPHS2, share high sequence identity and structural homology with SEPHS. Here, we report nine individuals from eight families with developmental delay, growth and feeding problems, hypotonia, and dysmorphic features, all with heterozygous missense variants in SEPHS1. Eight of these individuals had a recurrent variant at amino acid position 371 of SEPHS1 (p.Arg371Trp, p.Arg371Gln, and p.Arg371Gly); seven of these variants were known to be de novo. Structural modeling and biochemical assays were used to understand the effect of these variants on SEPHS1 function. We found that a variant at residue Trp352 results in local structural changes of the C-terminal region of SEPHS1 that decrease the overall thermal stability of the enzyme. In contrast, variants of a solvent-exposed residue Arg371 do not impact enzyme stability and folding but could modulate direct protein-protein interactions of SEPSH1 with cellular factors in promoting cell proliferation and development. In neuronal SH-SY5Y cells, we assessed the impact of SEPHS1 variants on cell proliferation and ROS production and investigated the mRNA expression levels of genes encoding stress-related selenoproteins. Our findings provided evidence that the identified SEPHS1 variants enhance cell proliferation by modulating ROS homeostasis. Our study supports the hypothesis that SEPHS1 plays a critical role during human development and provides a basis for further investigation into the molecular mechanisms employed by SEPHS1. Furthermore, our data suggest that variants in SEPHS1 are associated with a neurodevelopmental disorder.


Asunto(s)
Discapacidad Intelectual , Anomalías Musculoesqueléticas , Trastornos del Neurodesarrollo , Animales , Niño , Humanos , Discapacidades del Desarrollo/genética , Exones , Discapacidad Intelectual/genética , Mamíferos/genética , Hipotonía Muscular/genética , Anomalías Musculoesqueléticas/genética , Neuroblastoma/genética , Trastornos del Neurodesarrollo/genética , Especies Reactivas de Oxígeno
3.
Nucleic Acids Res ; 51(8): 4012-4026, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36929010

RESUMEN

O-Phosphoseryl-tRNASec selenium transferase (SepSecS) catalyzes the terminal step of selenocysteine (Sec) synthesis in archaea and eukaryotes. How the Sec synthetic machinery recognizes and discriminates tRNASec from the tRNA pool is essential to the integrity of the selenoproteome. Previously, we suggested that SepSecS adopts a competent conformation that is pre-ordered for catalysis. Herein, using high-resolution X-ray crystallography, we visualized tRNA-dependent conformational changes in human SepSecS that may be a prerequisite for achieving catalytic competency. We show that tRNASec binding organizes the active sites of the catalytic protomer, while stabilizing the N- and C-termini of the non-catalytic protomer. Binding of large anions to the catalytic groove may further optimize the catalytic site for substrate binding and catalysis. Our biochemical and mutational analyses demonstrate that productive SepSecS•tRNASec complex formation is enthalpically driven and primarily governed by electrostatic interactions between the acceptor-, TΨC-, and variable arms of tRNASec and helices α1 and α14 of SepSecS. The detailed visualization of the tRNA-dependent activation of SepSecS provides a structural basis for a revised model of the terminal reaction of Sec formation in archaea and eukaryotes.


Asunto(s)
ARN de Transferencia , Selenocisteína , Humanos , Selenocisteína/química , Subunidades de Proteína , Cristalografía por Rayos X , Dominio Catalítico
4.
Science ; 376(6599): 1338-1343, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35709277

RESUMEN

The elongation of eukaryotic selenoproteins relies on a poorly understood process of interpreting in-frame UGA stop codons as selenocysteine (Sec). We used cryo-electron microscopy to visualize Sec UGA recoding in mammals. A complex between the noncoding Sec-insertion sequence (SECIS), SECIS-binding protein 2 (SBP2), and 40S ribosomal subunit enables Sec-specific elongation factor eEFSec to deliver Sec. eEFSec and SBP2 do not interact directly but rather deploy their carboxyl-terminal domains to engage with the opposite ends of the SECIS. By using its Lys-rich and carboxyl-terminal segments, the ribosomal protein eS31 simultaneously interacts with Sec-specific transfer RNA (tRNASec) and SBP2, which further stabilizes the assembly. eEFSec is indiscriminate toward l-serine and facilitates its misincorporation at Sec UGA codons. Our results support a fundamentally distinct mechanism of Sec UGA recoding in eukaryotes from that in bacteria.


Asunto(s)
Codón de Terminación , Extensión de la Cadena Peptídica de Translación , Proteínas de Unión al ARN , Ribosomas , Selenocisteína , Selenoproteínas , Codón de Terminación/genética , Microscopía por Crioelectrón , Humanos , Extensión de la Cadena Peptídica de Translación/genética , Conformación Proteica , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Ribosomas/química , Selenocisteína/química , Selenocisteína/genética , Selenocisteína/metabolismo , Selenoproteínas/biosíntesis , Selenoproteínas/genética
5.
Nat Struct Mol Biol ; 26(8): 679-685, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31285607

RESUMEN

The RNA-guided Cas9 endonuclease from Streptococcus pyogenes is a single-turnover enzyme that displays a stable product state after double-stranded-DNA cleavage. Here, we present cryo-EM structures of precatalytic, postcatalytic and product states of the active Cas9-sgRNA-DNA complex in the presence of Mg2+. In the precatalytic state, Cas9 adopts the 'checkpoint' conformation with the HNH nuclease domain positioned far away from the DNA. Transition to the postcatalytic state involves a dramatic ~34-Å swing of the HNH domain and disorder of the REC2 recognition domain. The postcatalytic state captures the cleaved substrate bound to the catalytically competent HNH active site. In the product state, the HNH domain is disordered, REC2 returns to the precatalytic conformation, and additional interactions of REC3 and RuvC with nucleic acids are formed. The coupled domain motions and interactions between the enzyme and the RNA-DNA hybrid provide new insights into the mechanism of genome editing by Cas9.


Asunto(s)
Proteína 9 Asociada a CRISPR/ultraestructura , Sistemas CRISPR-Cas , ADN/metabolismo , Proteína 9 Asociada a CRISPR/química , Proteína 9 Asociada a CRISPR/metabolismo , Microscopía por Crioelectrón , ADN/ultraestructura , Sustancias Macromoleculares/ultraestructura , Modelos Moleculares , Movimiento (Física) , Conformación Proteica , Dominios Proteicos , Edición de ARN , ARN Guía de Kinetoplastida/metabolismo , Streptococcus pyogenes/enzimología
6.
Biochim Biophys Acta Gen Subj ; 1862(11): 2463-2472, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29555379

RESUMEN

BACKGROUND: Selenium, an essential dietary micronutrient, is incorporated into proteins as the amino acid selenocysteine (Sec) in response to in-frame UGA codons. Complex machinery ensures accurate recoding of Sec codons in higher organisms. A specialized elongation factor eEFSec is central to the process. SCOPE OF REVIEW: Selenoprotein synthesis relies on selenocysteinyl-tRNASec (Sec-tRNASec), selenocysteine inserting sequence (SECIS) and other selenoprotein mRNA elements, an in-trans SECIS binding protein 2 (SBP2) protein factor, and eEFSec. The exact mechanisms of discrete steps of the Sec UGA recoding are not well understood. However, recent studies on mammalian model systems have revealed the first insights into these mechanisms. Herein, we summarize the current knowledge about the structure and role of mammalian eEFSec. MAJOR CONCLUSIONS: eEFSec folds into a chalice-like structure resembling that of the archaeal and bacterial orthologues SelB and the initiation protein factor IF2/eIF5B. The three N-terminal domains harbor major functional sites and adopt an EF-Tu-like fold. The C-terminal domain 4 binds to Sec-tRNASec and SBP2, senses distinct binding domains, and modulates the GTPase activity. Remarkably, GTP hydrolysis does not induce a canonical conformational change in eEFSec, but instead promotes a slight ratchet of domains 1 and 2 and a lever-like movement of domain 4, which may be critical for the release of Sec-tRNASec on the ribosome. GENERAL SIGNIFICANCE: Based on current findings, a non-canonical mechanism for elongation of selenoprotein synthesis at the Sec UGA codon is proposed. Although incomplete, our understanding of this fundamental biological process is significantly improved, and it is being harnessed for biomedical and synthetic biology initiatives. This article is part of a Special Issue entitled "Selenium research" in celebration of 200 years of selenium discovery, edited by Dr. Elias Arnér and Dr. Regina Brigelius-Flohe.

7.
RNA Biol ; 15(4-5): 623-634, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28534666

RESUMEN

Aminoacyl-tRNA synthetases (AaRSs) are ubiquitously expressed enzymes that ensure accurate translation of the genetic information into functional proteins. These enzymes also execute a variety of non-canonical functions that are significant for regulation of diverse cellular processes and that reside outside the realm of protein synthesis. Associations between faults in AaRS-mediated processes and human diseases have been long recognized. Most recent research findings strongly argue that 10 cytosolic and 14 mitochondrial AaRSs are implicated in some form of pathology of the human nervous system. The advent of modern whole-exome sequencing makes it all but certain that similar associations between the remaining 15 ARS genes and neurologic illnesses will be defined in future. It is not surprising that an intense scientific debate about the role of translational machinery, in general, and AaRSs, in particular, in the development and maintenance of the healthy human neural cell types and the brain is sparked. Herein, we summarize the current knowledge about causative links between mutations in human AaRSs and diseases of the nervous system and briefly discuss future directions.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Encefalopatías/genética , Enfermedades Cerebelosas/genética , Enfermedades de los Nervios Craneales/genética , Mutación , ARN de Transferencia/genética , Aminoacil-ARNt Sintetasas/metabolismo , Encefalopatías/enzimología , Encefalopatías/patología , Enfermedades Cerebelosas/enzimología , Enfermedades Cerebelosas/patología , Enfermedades de los Nervios Craneales/enzimología , Enfermedades de los Nervios Craneales/patología , Citosol/metabolismo , Expresión Génica , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Sistema Nervioso/enzimología , Sistema Nervioso/patología , ARN de Transferencia/metabolismo
8.
RNA ; 23(11): 1685-1699, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28808125

RESUMEN

Seryl-tRNA synthetase (SerRS) attaches L-serine to the cognate serine tRNA (tRNASer) and the noncognate selenocysteine tRNA (tRNASec). The latter activity initiates the anabolic cycle of selenocysteine (Sec), proper decoding of an in-frame Sec UGA codon, and synthesis of selenoproteins across all domains of life. While the accuracy of SerRS is important for overall proteome integrity, it is its substrate promiscuity that is vital for the integrity of the selenoproteome. This raises a question as to what elements in the two tRNA species, harboring different anticodon sequences and adopting distinct folds, facilitate aminoacylation by a common aminoacyl-tRNA synthetase. We sought to answer this question by analyzing the ability of human cytosolic SerRS to bind and act on tRNASer, tRNASec, and 10 mutant and chimeric constructs in which elements of tRNASer were transposed onto tRNASec We show that human SerRS only subtly prefers tRNASer to tRNASec, and that discrimination occurs at the level of the serylation reaction. Surprisingly, the tRNA mutants predicted to adopt either the 7/5 or 8/5 fold are poor SerRS substrates. In contrast, shortening of the acceptor arm of tRNASec by a single base pair yields an improved SerRS substrate that adopts an 8/4 fold. We suggest that an optimal tertiary arrangement of structural elements within tRNASec and tRNASer dictate their utility for serylation. We also speculate that the extended acceptor-TΨC arm of tRNASec evolved as a compromise for productive binding to SerRS while remaining the major recognition element for other enzymes involved in Sec and selenoprotein synthesis.


Asunto(s)
ARN de Transferencia Aminoácido-Específico/metabolismo , ARN de Transferencia de Serina/metabolismo , Serina-ARNt Ligasa/metabolismo , Secuencia de Bases , Sitios de Unión , Citosol/enzimología , Humanos , Cinética , Modelos Moleculares , Mutagénesis , Conformación de Ácido Nucleico , Pliegue del ARN , ARN de Transferencia Aminoácido-Específico/química , ARN de Transferencia Aminoácido-Específico/genética , ARN de Transferencia de Serina/química , ARN de Transferencia de Serina/genética , Especificidad por Sustrato
9.
Nat Commun ; 7: 12941, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27708257

RESUMEN

Selenocysteine is the only proteinogenic amino acid encoded by a recoded in-frame UGA codon that does not operate as the canonical opal stop codon. A specialized translation elongation factor, eEFSec in eukaryotes and SelB in prokaryotes, promotes selenocysteine incorporation into selenoproteins by a still poorly understood mechanism. Our structural and biochemical results reveal that four domains of human eEFSec fold into a chalice-like structure that has similar binding affinities for GDP, GTP and other guanine nucleotides. Surprisingly, unlike in eEF1A and EF-Tu, the guanine nucleotide exchange does not cause a major conformational change in domain 1 of eEFSec, but instead induces a swing of domain 4. We propose that eEFSec employs a non-canonical mechanism involving the distinct C-terminal domain 4 for the release of the selenocysteinyl-tRNA during decoding on the ribosome.


Asunto(s)
Factores de Elongación de Péptidos/química , Selenocisteína/química , Codón de Terminación , Cristalografía por Rayos X , Guanosina Difosfato/química , Guanosina Trifosfato/química , Humanos , Filogenia , Biosíntesis de Proteínas , Dominios Proteicos , Estructura Secundaria de Proteína , Aminoacil-ARN de Transferencia/química , Ribosomas/metabolismo , Selenoproteínas/genética
10.
Sci Rep ; 6: 32563, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27576344

RESUMEN

Selenocysteine synthase (SepSecS) catalyzes the terminal reaction of selenocysteine, and is vital for human selenoproteome integrity. Autosomal recessive inheritance of mutations in SepSecS-Ala239Thr, Thr325Ser, Tyr334Cys and Tyr429*-induced severe, early-onset, neurological disorders in distinct human populations. Although harboring different mutant alleles, patients presented remarkably similar phenotypes typified by cerebellar and cerebral atrophy, seizures, irritability, ataxia, and extreme spasticity. However, it has remained unclear how these genetic alterations affected the structure of SepSecS and subsequently elicited the development of a neurological pathology. Herein, our biophysical and structural characterization demonstrates that, with the exception of Tyr429*, pathogenic mutations decrease protein stability and trigger protein misfolding. We propose that the reduced stability and increased propensity towards misfolding are the main causes for the loss of SepSecS activity in afflicted patients, and that these factors contribute to disease progression. We also suggest that misfolding of enzymes regulating protein synthesis should be considered in the diagnosis and study of childhood neurological disorders.


Asunto(s)
Aminoacil-ARNt Sintetasas/química , Ataxia/genética , Espasticidad Muscular/genética , ARN de Transferencia/química , Convulsiones/genética , Selenocisteína/química , Edad de Inicio , Sustitución de Aminoácidos , Aminoacil-ARNt Sintetasas/genética , Aminoacil-ARNt Sintetasas/metabolismo , Ataxia/enzimología , Ataxia/patología , Atrofia , Sitios de Unión , Cerebelo/enzimología , Cerebelo/patología , Corteza Cerebral/enzimología , Corteza Cerebral/patología , Cristalografía por Rayos X , Humanos , Genio Irritable , Modelos Moleculares , Espasticidad Muscular/enzimología , Espasticidad Muscular/patología , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , ARN de Transferencia/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Convulsiones/enzimología , Convulsiones/patología , Selenocisteína/metabolismo , Especificidad por Sustrato
11.
J Biol Chem ; 291(46): 24036-24040, 2016 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-27645994

RESUMEN

The human genome contains 25 genes coding for selenocysteine-containing proteins (selenoproteins). These proteins are involved in a variety of functions, most notably redox homeostasis. Selenoprotein enzymes with known functions are designated according to these functions: TXNRD1, TXNRD2, and TXNRD3 (thioredoxin reductases), GPX1, GPX2, GPX3, GPX4, and GPX6 (glutathione peroxidases), DIO1, DIO2, and DIO3 (iodothyronine deiodinases), MSRB1 (methionine sulfoxide reductase B1), and SEPHS2 (selenophosphate synthetase 2). Selenoproteins without known functions have traditionally been denoted by SEL or SEP symbols. However, these symbols are sometimes ambiguous and conflict with the approved nomenclature for several other genes. Therefore, there is a need to implement a rational and coherent nomenclature system for selenoprotein-encoding genes. Our solution is to use the root symbol SELENO followed by a letter. This nomenclature applies to SELENOF (selenoprotein F, the 15-kDa selenoprotein, SEP15), SELENOH (selenoprotein H, SELH, C11orf31), SELENOI (selenoprotein I, SELI, EPT1), SELENOK (selenoprotein K, SELK), SELENOM (selenoprotein M, SELM), SELENON (selenoprotein N, SEPN1, SELN), SELENOO (selenoprotein O, SELO), SELENOP (selenoprotein P, SeP, SEPP1, SELP), SELENOS (selenoprotein S, SELS, SEPS1, VIMP), SELENOT (selenoprotein T, SELT), SELENOV (selenoprotein V, SELV), and SELENOW (selenoprotein W, SELW, SEPW1). This system, approved by the HUGO Gene Nomenclature Committee, also resolves conflicting, missing, and ambiguous designations for selenoprotein genes and is applicable to selenoproteins across vertebrates.


Asunto(s)
Selenoproteínas/clasificación , Selenoproteínas/genética , Humanos , Terminología como Asunto
12.
Nucleic Acids Res ; 44(7): 3420-31, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26869582

RESUMEN

Cytosolic glutaminyl-tRNA synthetase (GlnRS) is the singular enzyme responsible for translation of glutamine codons. Compound heterozygous mutations in GlnRS cause severe brain disorders by a poorly understood mechanism. Herein, we present crystal structures of the wild type and two pathological mutants of human GlnRS, which reveal, for the first time, the domain organization of the intact enzyme and the structure of the functionally important N-terminal domain (NTD). Pathological mutations mapping in the NTD alter the domain structure, and decrease catalytic activity and stability of GlnRS, whereas missense mutations in the catalytic domain induce misfolding of the enzyme. Our results suggest that the reduced catalytic efficiency and a propensity of GlnRS mutants to misfold trigger the disease development. This report broadens the spectrum of brain pathologies elicited by protein misfolding and provides a paradigm for understanding the role of mutations in aminoacyl-tRNA synthetases in neurological diseases.


Asunto(s)
Aminoacil-ARNt Sintetasas/química , Aminoacil-ARNt Sintetasas/genética , Mutación , Enfermedades del Sistema Nervioso/genética , Aminoacil-ARNt Sintetasas/ultraestructura , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Pliegue de Proteína , Estructura Terciaria de Proteína
13.
Nucleic Acids Res ; 44(3): 1428-39, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26704982

RESUMEN

In mitochondria of Saccharomyces cerevisiae, a single aminoacyl-tRNA synthetase (aaRS), MST1, aminoacylates two isoacceptor tRNAs, tRNA1(Thr) and tRNA2(Thr), that harbor anticodon loops of different size and sequence. As a result of this promiscuity, reassignment of the CUN codon box from leucine to threonine is facilitated. However, the mechanism by which a single aaRS binds distinct anticodon loops with high specificity is not well understood. Herein, we present the crystal structure of MST1 in complex with the canonical tRNA2(Thr) and non-hydrolyzable analog of threonyl adenylate. Our structure reveals that the dimeric arrangement of MST1 is essential for binding the 5'-phosphate, the second base pair of the acceptor stem, the first two base pairs of the anticodon stem and the first nucleotide of the variable arm. Further, in contrast to the bacterial ortholog that 'reads' the entire anticodon sequence, MST1 recognizes bases in the second and third position and the nucleotide upstream of the anticodon sequence. We speculate that a flexible loop linking strands ß4 and ß5 may be allosteric regulator that establishes cross-subunit communication between the aminoacylation and tRNA-binding sites. We also propose that structural features of the anticodon-binding domain in MST1 permit binding of the enlarged anticodon loop of tRNA1(Thr).


Asunto(s)
Proteínas de Escherichia coli/metabolismo , ARN de Transferencia de Treonina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Treonina-ARNt Ligasa/metabolismo , Anticodón/química , Anticodón/genética , Anticodón/metabolismo , Secuencia de Bases , Sitios de Unión/genética , Cristalografía por Rayos X , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Cinética , Mitocondrias/genética , Mitocondrias/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Unión Proteica , Estructura Terciaria de Proteína , ARN de Hongos/química , ARN de Hongos/genética , ARN de Hongos/metabolismo , ARN de Transferencia de Treonina/química , ARN de Transferencia de Treonina/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Treonina-ARNt Ligasa/química , Treonina-ARNt Ligasa/genética
14.
Structure ; 23(12): 2256-2266, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26655472

RESUMEN

The role of internal dynamics in enzyme function is highly debated. Specifically, how small changes in structure far away from the reaction site alter protein dynamics and overall enzyme mechanisms is of wide interest in protein engineering. Using RNase A as a model, we demonstrate that elimination of a single methyl group located >10 Å away from the reaction site significantly alters conformational integrity and binding properties of the enzyme. This A109G mutation does not perturb structure or thermodynamic stability, both in the apo and ligand-bound states. However, significant enhancement in conformational dynamics was observed for the bound variant, as probed over nano- to millisecond timescales, resulting in major ligand repositioning. These results illustrate the large effects caused by small changes in structure on long-range conformational dynamics and ligand specificities within proteins, further supporting the importance of preserving wild-type dynamics in enzyme systems that rely on flexibility for function.


Asunto(s)
Dominio Catalítico , Simulación de Dinámica Molecular , Ribonucleasa Pancreática/química , Adenosina Monofosfato/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Datos de Secuencia Molecular , Mutación , Unión Proteica , Ribonucleasa Pancreática/genética , Ribonucleasa Pancreática/metabolismo , Uridina Monofosfato/metabolismo
15.
Neurology ; 85(4): 306-15, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26115735

RESUMEN

OBJECTIVE: We aimed to decipher the molecular genetic basis of disease in a cohort of children with a uniform clinical presentation of neonatal irritability, spastic or dystonic quadriplegia, virtually absent psychomotor development, axonal neuropathy, and elevated blood/CSF lactate. METHODS: We performed whole-exome sequencing of blood DNA from the index patients. Detected compound heterozygous mutations were confirmed by Sanger sequencing. Structural predictions and a bacterial activity assay were performed to evaluate the functional consequences of the mutations. Mass spectrometry, Western blotting, and protein oxidation detection were used to analyze the effects of selenoprotein deficiency. RESULTS: Neuropathology indicated laminar necrosis and severe loss of myelin, with neuron loss and astrogliosis. In 3 families, we identified a missense (p.Thr325Ser) and a nonsense (p.Tyr429*) mutation in SEPSECS, encoding the O-phosphoseryl-tRNA:selenocysteinyl-tRNA synthase, which was previously associated with progressive cerebellocerebral atrophy. We show that the mutations do not completely abolish the activity of SEPSECS, but lead to decreased selenoprotein levels, with demonstrated increase in oxidative protein damage in the patient brain. CONCLUSIONS: These results extend the phenotypes caused by defective selenocysteine biosynthesis, and suggest SEPSECS as a candidate gene for progressive encephalopathies with lactate elevation.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Encefalopatías Metabólicas Innatas/genética , Encefalopatías Metabólicas Innatas/metabolismo , Ácido Láctico/sangre , Ácido Láctico/líquido cefalorraquídeo , Selenoproteínas/deficiencia , Adolescente , Encéfalo/metabolismo , Encéfalo/patología , Encefalopatías Metabólicas Innatas/sangre , Encefalopatías Metabólicas Innatas/líquido cefalorraquídeo , Niño , Preescolar , Femenino , Humanos , Masculino , Mutación , Estrés Oxidativo/genética , Selenoproteínas/biosíntesis
16.
J Biol Chem ; 289(42): 28783-94, 2014 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-25190812

RESUMEN

Selenocysteine (Sec), the 21(st) amino acid, is synthesized from a serine precursor in a series of reactions that require selenocysteine tRNA (tRNA(Sec)). In archaea and eukaryotes, O-phosphoseryl-tRNA(Sec):selenocysteinyl-tRNA(Sec) synthase (SepSecS) catalyzes the terminal synthetic reaction during which the phosphoseryl intermediate is converted into the selenocysteinyl moiety while being attached to tRNA(Sec). We have previously shown that only the SepSecS tetramer is capable of binding to and recognizing the distinct fold of tRNA(Sec). Because only two of the four tRNA-binding sites were occupied in the crystal form, a question was raised regarding whether the observed arrangement and architecture faithfully recapitulated the physiologically relevant ribonucleoprotein complex important for selenoprotein formation. Herein, we determined the stoichiometry of the human terminal synthetic complex of selenocysteine by using small angle x-ray scattering, multi-angle light scattering, and analytical ultracentrifugation. In addition, we provided the first estimate of the ratio between SepSecS and tRNA(Sec) in vivo. We show that SepSecS preferentially binds one or two tRNA(Sec) molecules at a time and that the enzyme is present in large molar excess over the substrate tRNA in vivo. Moreover, we show that in a complex between SepSecS and two tRNAs, one enzyme homodimer plays a role of the noncatalytic unit that positions CCA ends of two tRNA(Sec) molecules into the active site grooves of the other, catalytic, homodimer. Finally, our results demonstrate that the previously determined crystal structure represents the physiologically and catalytically relevant complex and suggest that allosteric regulation of SepSecS might play an important role in regulation of selenocysteine and selenoprotein synthesis.


Asunto(s)
Aminoacil-ARNt Sintetasas/química , ARN de Transferencia/química , Selenocisteína/química , Sitio Alostérico , Dominio Catalítico , Difusión , Escherichia coli/metabolismo , Humanos , Luz , Mapeo de Interacción de Proteínas , Multimerización de Proteína , ARN/química , Dispersión de Radiación , Triptófano/química , Ultracentrifugación , Difracción de Rayos X
17.
Food Chem ; 164: 36-43, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24996302

RESUMEN

Polyphenols, the potent plant secondary metabolites, have beneficial effects on human health, but the mechanism(s) by which these effects are exerted is not well understood. Here, we present the detailed analysis of the interactions between the major green tea catechin, epigallo-catechin 3-gallate (EGCG), and the major dietary protein and allergen, ovalbumin (OVA). We show that EGCG binds to the pocket that partly overlaps with the previously identified IgE-binding region in OVA, and that this interaction induces structural changes in the allergen. Moreover, our ex vivo studies reveal that OVA binds IgE and stimulates degranulation of basophils, and that its uptake by monocytes proceeds at a slower rate in the presence of EGCG. This study provides further evidence in support of the proposed mechanism by which EGCG interactions with the food allergens contribute to its diverse biological activities and may impair antigen uptake by antigen-presenting cells.


Asunto(s)
Catequina/análogos & derivados , Clara de Huevo/química , Ovalbúmina/química , Alérgenos/química , Células Presentadoras de Antígenos/metabolismo , Basófilos/metabolismo , Catequina/química , Dicroismo Circular , Electroforesis en Gel de Poliacrilamida , Hipersensibilidad a los Alimentos , Humanos , Inmunoglobulina E/química , Monocitos/metabolismo , Polifenoles , Unión Proteica , Conformación Proteica , Espectrometría de Fluorescencia , Té/química
18.
Chemistry ; 19(47): 15872-8, 2013 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-24127424

RESUMEN

The twenty first amino acid, selenocysteine (Sec), is the only amino acid that is synthesized on its cognate transfer RNA (tRNA(Sec)) in all domains of life. The multistep pathway involves O-phosphoseryl-tRNA:selenocysteinyl-tRNA synthase (SepSecS), an enzyme that catalyzes the terminal chemical reaction during which the phosphoseryl-tRNA(Sec) intermediate is converted into selenocysteinyl-tRNA(Sec). The SepSecS architecture and the mode of tRNA(Sec) recognition have been recently determined at atomic resolution. The crystal structure provided valuable insights that gave rise to mechanistic proposals that could not be validated because of the lack of appropriate molecular probes. To further improve our understanding of the mechanism of the biosynthesis of selenocysteine in general and the mechanism of SepSecS in particular, stable tRNA(Sec) substrates carrying aminoacyl moieties that mimic particular reaction intermediates are needed. Here, we report on the accurate synthesis of methylated, phosphorylated, and phosphonated serinyl-derived tRNA(Sec) mimics that contain a hydrolysis-resistant ribose 3'-amide linkage instead of the natural ester bond. The procedures introduced allow for efficient site-specific methylation and/or phosphorylation directly on the solid support utilized in the automated RNA synthesis. For the preparation of (S)-2-amino-4-phosphonobutyric acid-oligoribonucleotide conjugates, a separate solid support was generated. Furthermore, we developed a three-strand enzymatic ligation protocol to obtain the corresponding full-length tRNA(Sec) derivatives. Finally, we developed an electrophoretic mobility shift assay (EMSA) for rapid, qualitative characterization of the SepSecS-tRNA interactions. The novel tRNA(Sec) mimics are promising candidates for further elucidation of the biosynthesis of selenocysteine by X-ray crystallography and other biochemical approaches, and could be attractive for similar studies on other tRNA-dependent enzymes.


Asunto(s)
Materiales Biocompatibles/síntesis química , Ácidos Fosforosos/química , ARN de Transferencia Aminoácido-Específico/química , Aminobutiratos/química , Secuencia de Bases , Materiales Biocompatibles/química , Metilación , Conformación de Ácido Nucleico , Fosforilación , ARN de Transferencia Aminoácido-Específico/síntesis química , Técnicas de Síntesis en Fase Sólida
19.
J Biol Chem ; 287(34): 28518-25, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22773845

RESUMEN

Accurate translation of mRNA into protein is a fundamental biological process critical for maintaining normal cellular functions. To ensure translational fidelity, aminoacyl-tRNA synthetases (aaRSs) employ pre-transfer and post-transfer editing activities to hydrolyze misactivated and mischarged amino acids, respectively. Whereas post-transfer editing, which requires either a specialized domain in aaRS or a trans-protein factor, is well described, the mechanism of pre-transfer editing is less understood. Here, we show that yeast mitochondrial threonyl-tRNA synthetase (MST1), which lacks an editing domain, utilizes pre-transfer editing to discriminate against serine. MST1 misactivates serine and edits seryl adenylate (Ser-AMP) in a tRNA-independent manner. MST1 hydrolyzes 80% of misactivated Ser-AMP at a rate 4-fold higher than that for the cognate threonyl adenylate (Thr-AMP) while releasing 20% of Ser-AMP into the solution. To understand the mechanism of pre-transfer editing, we solved the crystal structure of MST1 complexed with an analog of Ser-AMP. The binding of the Ser-AMP analog to MST1 induces conformational changes in the aminoacylation active site, and it positions a potential hydrolytic water molecule more favorably for nucleophilic attack. In addition, inhibition results reveal that the Ser-AMP analog binds the active site 100-fold less tightly than the Thr-AMP analog. In conclusion, we propose that the plasticity of the aminoacylation site in MST1 allows binding of Ser-AMP and the appropriate positioning of the hydrolytic water molecule.


Asunto(s)
Mitocondrias/enzimología , Proteínas Mitocondriales/química , Edición de ARN , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Treonina-ARNt Ligasa/química , Cristalografía por Rayos X , Mitocondrias/genética , Proteínas Mitocondriales/genética , Unión Proteica , Estructura Terciaria de Proteína , ARN de Hongos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Treonina-ARNt Ligasa/genética
20.
Proc Natl Acad Sci U S A ; 109(9): 3281-6, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22343532

RESUMEN

Aminoacyl-tRNA synthetases (aaRSs) ensure faithful translation of mRNA into protein by coupling an amino acid to a set of tRNAs with conserved anticodon sequences. Here, we show that in mitochondria of Saccharomyces cerevisiae, a single aaRS (MST1) recognizes and aminoacylates two natural tRNAs that contain anticodon loops of different size and sequence. Besides a regular tRNA(2Thr) with a threonine (Thr) anticodon, MST1 also recognizes an unusual tRNA(1Thr), which contains an enlarged anticodon loop and an anticodon triplet that reassigns the CUN codons from leucine to threonine. Our data show that MST1 recognizes the anticodon loop in both tRNAs, but employs distinct recognition mechanisms. The size but not the sequence of the anticodon loop is critical for tRNA(1Thr) recognition, whereas the anticodon sequence is essential for aminoacylation of tRNA(2Thr). The crystal structure of MST1 reveals that, while lacking the N-terminal editing domain, the enzyme closely resembles the bacterial threonyl-tRNA synthetase (ThrRS). A detailed structural comparison with Escherichia coli ThrRS, which is unable to aminoacylate tRNA(1Thr), reveals differences in the anticodon-binding domain that probably allow recognition of the distinct anticodon loops. Finally, our mutational and modeling analyses identify the structural elements in MST1 (e.g., helix α11) that define tRNA selectivity. Thus, MTS1 exemplifies that a single aaRS can recognize completely divergent anticodon loops of natural isoacceptor tRNAs and that in doing so it facilitates the reassignment of the genetic code in yeast mitochondria.


Asunto(s)
Aminoacil-ARN de Transferencia/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Treonina-ARNt Ligasa/metabolismo , Aeropyrum/enzimología , Secuencia de Aminoácidos , Anticodón/genética , Dominio Catalítico , Codón/genética , Cristalografía por Rayos X , Escherichia coli/enzimología , Evolución Molecular , Leucina , Mitocondrias/enzimología , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Estructura Terciaria de Proteína , Edición de ARN , Aminoacil-ARN de Transferencia/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Alineación de Secuencia , Especificidad de la Especie , Staphylococcus aureus/enzimología , Especificidad por Sustrato , Treonina , Treonina-ARNt Ligasa/química , Treonina-ARNt Ligasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA