Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Methods Enzymol ; 704: 27-38, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39300651

RESUMEN

Rieske non-heme iron oxygenases are ubiquitously expressed in prokaryotes. These enzymes catalyze a wide variety of reactions, including cis-dihydroxylation, mono-hydroxylation, sulfoxidation, and demethylation. They contain a Rieske-type [2Fe-2S] cluster and an active site with a mono-nuclear iron bound to a 2-His carboxylate triad. Naphthalene 1,2 dioxygenase, a representative of this family, catalyzes the conversion of naphthalene to (+)-cis-(1R,2S)-dihydroxy-1,2-dihydronaphthalene. This transformation requires naphthalene, two electrons, and an oxygen molecule. The first structure of the terminal oxygenase component of a Rieske non-heme iron oxygenase to be determined was naphthalene 1,2 dioxygenase (NDO-O). In this article, we describe in detail the methods used to recombinantly express and purify NDO-O in rich and minimal salts media, the crystallization of NDO-O for structure determination by X-ray crystallography, the challenges faced, and the methods used for the preparation of enzyme ligand complexes. The methods used here resulted in the determination of several NDO-O complexes with aromatic substrates, nitric oxide, oxygen molecule, and products, leading to an initial understanding of the mechanism of enzyme catalysis and the molecular determinants of the regio- and stereo-specificity of this class of enzymes.


Asunto(s)
Dioxigenasas , Dioxigenasas/química , Dioxigenasas/metabolismo , Dioxigenasas/genética , Cristalografía por Rayos X/métodos , Naftalenos/química , Naftalenos/metabolismo , Oxigenasas/química , Oxigenasas/metabolismo , Dominio Catalítico , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Cristalización/métodos , Modelos Moleculares , Complejos Multienzimáticos
2.
Cell Chem Biol ; 31(3): 514-522.e4, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38460516

RESUMEN

It is a challenge for the traditional affinity methods to capture transient interactions of enzyme-post-translational modification (PTM) substrates in vivo. Herein we presented a strategy termed proximity labeling-based orthogonal trap approach (ProLORT), relying upon APEX2-catalysed proximity labeling and an orthogonal trap pipeline as well as quantitative proteomics to directly investigate the transient interactome of enzyme-PTM substrates in living cells. As a proof of concept, ProLORT allows for robust evaluation of a known HDAC8 substrate, histone H3K9ac. By leveraging this approach, we identified numerous of putative acetylated proteins targeted by HDAC8, and further confirmed CTTN as a bona fide substrate in vivo. Next, we demonstrated that HDAC8 facilitates cell motility via deacetylation of CTTN at lysine 144 that attenuates its interaction with F-actin, expanding the underlying regulatory mechanisms of HDAC8. We developed a general strategy to profile the transient enzyme-substrate interactions mediated by PTMs, providing a powerful tool for identifying the spatiotemporal PTM-network regulated by enzymes in living cells.


Asunto(s)
Cortactina , Histona Desacetilasas , Histona Desacetilasas/metabolismo , Acetilación , Cortactina/metabolismo , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Movimiento Celular
3.
BioTech (Basel) ; 13(1)2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38390907

RESUMEN

Glycosyltransferases (GTs) are pivotal enzymes in the biosynthesis of various biological molecules. This study focuses on the scale-up, expression, and purification of a plant flavonol-specific 3-O glucosyltransferase (Cp3GT), a key enzyme from Citrus paradisi, for structural analysis and modeling. The challenges associated with recombinant protein production in Pichia pastoris, such as proteolytic degradation, were addressed through the optimization of culture conditions and purification processes. The purification strategy employed affinity, anion exchange, and size exclusion chromatography, leading to greater than 95% homogeneity for Cp3GT. In silico modeling, using D-I-TASSER and COFACTOR integrated with the AlphaFold2 pipeline, provided insights into the structural dynamics of Cp3GT and its ligand binding sites, offering predictions for enzyme-substrate interactions. These models were compared to experimentally derived structures, enhancing understanding of the enzyme's functional mechanisms. The findings present a comprehensive approach to produce a highly purified Cp3GT which is suitable for crystallographic studies and to shed light on the structural basis of flavonol specificity in plant GTs. The significant implications of these results for synthetic biology and enzyme engineering in pharmaceutical applications are also considered.

4.
Biochem Soc Trans ; 51(3): 1225-1233, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37140261

RESUMEN

Sister chromatid segregation is the final irreversible step of mitosis. It is initiated by a complex regulatory system that ultimately triggers the timely activation of a conserved cysteine protease named separase. Separase cleaves the cohesin protein ring that links the sister chromatids and thus facilitates their separation and segregation to the opposite poles of the dividing cell. Due to the irreversible nature of this process, separase activity is tightly controlled in all eukaryotic cells. In this mini-review, we summarize the latest structural and functional findings on the regulation of separase, with an emphasis on the regulation of the human enzyme by two inhibitors, the universal inhibitor securin and the vertebrate-specific inhibitor CDK1-cyclin B. We discuss the two fundamentally different inhibitory mechanisms by which these inhibitors block separase activity by occluding substrate binding. We also describe conserved mechanisms that facilitate substrate recognition and point out open research questions that will guide studies of this fascinating enzyme for years to come.


Asunto(s)
Proteínas de Ciclo Celular , Mitosis , Humanos , Separasa/química , Separasa/genética , Separasa/metabolismo , Proteínas de Ciclo Celular/metabolismo , Endopeptidasas/genética
5.
Comput Biol Chem ; 95: 107582, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34571426

RESUMEN

Aminoacyl-tRNA synthetases are an indispensable component of ribosomal protein translational machinery and Plasmodium Tyrosyl-tRNA synthetase (PfTyrRS) is a validated drug target. This manuscript illustrates the dynamic conformational landscape of PfTyrRS in the context of substrate binding. Molecular dynamics simulations of PfTyrRS in the presence and absence of ligand show conformational heterogeneity for both the protein and the bound ligand. Diverse conformations for the evolutionarily conserved ATP binding motif (KMSKS) have been observed in both apo- and holo PfTyrRS. Further, the presented attributes of the tyrosyl-adenylate conformational sub-states in situ along with their implications on the strength of intermolecular interactions would be a pertinent benchmark for molecular design studies. In addition, an analysis of the ligand hydration pattern foregrounds the structurally conserved water-mediated inter-molecular interactions. The quantitative assessment of the conformational landscape, based on the fluctuations of the distance between the ligand binding pockets, of apo-PfTyrRS and holo-PfTyrRS highlights the nature of diversity in conformational sampling for the two cases. Evidently, the holo-PfTyrRS adopts a rather compact conformation compared to the apo-PfTyrRS. An intriguing asymmetry in the dynamics of the two monomers is contextualized with the functional asymmetry of the symmetrically dimeric PfTyrRS. Importantly, the network of non-bonded contacts in the apo- and holo- simulated ensembles has been analyzed. The graph-theoretic analysis-based novel insights concerning the nature of information flow as a function of ligation state would prove valuable in understanding PfTyrRS functions. The results presented here contend that understanding allostery in PfTyrRS is essential to astutely design structure-based inhibitors.


Asunto(s)
Plasmodium/enzimología , Polimorfismo Genético/genética , Tirosina-ARNt Ligasa/química , Tirosina-ARNt Ligasa/genética , Biología Computacional , Conformación Proteica , Especificidad por Sustrato , Tirosina-ARNt Ligasa/metabolismo
6.
Biochem Mol Biol Educ ; 49(6): 904-916, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34418262

RESUMEN

Biochemistry curricula present a particular challenge to undergraduate students with abstract concepts which can lead to misconceptions that impede learning. In particular, these students have difficulty understanding enzyme structure and function concepts. Targeted learning activities and three-dimensional (3D) physical models are proposed to help students challenge these misconceptions and increase conceptual understanding. Here we assessed such pedagogical tools using the Enzyme-Substrate Interactions Concept Inventory (ESICI) to measure (mis)conceptual changes from Pre- to Post- time points in a single semester undergraduate biochemistry course. A Control group of students engaged with the active learning activities without the 3D physical models and students in the Intervention group utilized these activities with the 3D physical models. At the Post- time point both groups had higher, yet similar ESICI scores of the same magnitude as the highest scoring group from the national sample. Concomitantly, many misconception markers decreased compared to the national sample, although some of these differed between the Control and Intervention groups. Based on this assessment, both pedagogical approaches successfully increased conceptual understanding and targeted many of the misconceptions measured by the ESICI, however, several misconceptions persisted. Surprisingly, the students who used the 3D physical models did not demonstrate a further decrease in the misconception markers. Additionally, psychometric evaluation of the ESICI with our sample recommends the revision of several questions to improve the validity of this assessment. We also offer suggestions to improve instruction and pedagogical tools with further avenues for research on learning.


Asunto(s)
Bioquímica , Estudiantes , Bioquímica/educación , Curriculum , Humanos , Aprendizaje Basado en Problemas
7.
Biochem J ; 478(14): 2811-2823, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34190988

RESUMEN

The human protein kinase ULK3 regulates the timing of membrane abscission, thus being involved in exosome budding and cytokinesis. Herein, we present the first high-resolution structures of the ULK3 kinase domain. Its unique features are explored against the background of other ULK kinases. An inhibitor fingerprint indicates that ULK3 is highly druggable and capable of adopting a wide range of conformations. In accordance with this, we describe a conformational switch between the active and an inactive ULK3 conformation, controlled by the properties of the attached small-molecule binder. Finally, we discuss a potential substrate-recognition mechanism of the full-length ULK3 protein.


Asunto(s)
Dominio Catalítico , Conformación Proteica , Dominios Proteicos , Proteínas Serina-Treonina Quinasas/química , Compuestos de Anilina/metabolismo , Compuestos de Anilina/farmacología , Benzamidas/metabolismo , Benzamidas/farmacología , Biocatálisis/efectos de los fármacos , Humanos , Modelos Moleculares , Nitrilos/metabolismo , Nitrilos/farmacología , Proteínas Oncogénicas/química , Proteínas Oncogénicas/metabolismo , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacología , Quinolinas/metabolismo , Quinolinas/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
8.
Biochem J ; 478(4): 943-959, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33565573

RESUMEN

Members of the glycoside hydrolase family 4 (GH4) employ an unusual glycosidic bond cleavage mechanism utilizing NAD(H) and a divalent metal ion, under reducing conditions. These enzymes act upon a diverse range of glycosides, and unlike most other GH families, homologs here are known to accommodate both α- and ß-anomeric specificities within the same active site. Here, we report the catalytic properties and the crystal structures of TmAgu4B, an α-d-glucuronidase from the hyperthermophile Thermotoga maritima. The structures in three different states include the apo form, the NADH bound holo form, and the ternary complex with NADH and the reaction product d-glucuronic acid, at 2.15, 1.97 and 1.85 Šresolutions, respectively. These structures reveal the step-wise route of conformational changes required in the active site to achieve the catalytically competent state, and illustrate the direct role of residues that determine the reaction mechanism. Furthermore, a structural transition of a helical region in the active site to a turn geometry resulting in the rearrangement of a unique arginine residue governs the exclusive glucopyranosiduronic acid recognition in TmAgu4B. Mutational studies show that modifications of the glycone binding site geometry lead to catalytic failure and indicate overlapping roles of specific residues in catalysis and substrate recognition. The data highlight hitherto unreported molecular features and associated active site dynamics that determine the structure-function relationships within the unique GH4 family.


Asunto(s)
Proteínas Bacterianas/química , Apoenzimas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Ditiotreitol/metabolismo , Ácido Glucurónico/química , Ácido Glucurónico/metabolismo , Glicósido Hidrolasas/metabolismo , Holoenzimas/química , Cinética , Manganeso/metabolismo , Modelos Moleculares , Familia de Multigenes , Mutagénesis Sitio-Dirigida , NAD/metabolismo , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato , Thermotoga maritima/enzimología , Thermotoga maritima/genética
9.
Sci Total Environ ; 758: 143997, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33333309

RESUMEN

Cytochrome P450 1A1 (CYP1A1) has served as a known metabolic enzyme that mediates the carcinogenesis of polycyclic aromatic hydrocarbons (PAHs). However, the structural mechanism involved in the metabolic capacity remains unclear. In this study, thirty-three calculated properties representing the physicochemical and electronic properties of PAH and PAH-CYP1A1 interactions were utilized to identify the key structural properties that affect metabolic processes, including binding ability, metabolic clearance, and mutagenicity, using a quantitative structure-activity relationship (QSAR) strategy combined with docking methods, QM/MM calculations and ab initio calculations. van der Waals interactions (glide vdw) appeared to be important for PAH binding to CYP1A1 and were mainly affected by the molecular weight and hydrophobic structures of PAHs. Interaction features between PAHs and heme, including the distance between iron and carbons of PAHs (Fe_Cmin) and heme vdw, coordinately influence the metabolic clearance of PAHs. Furthermore, the electronic properties (ESP neg variance) appeared to be critical for the mutagenicity of PAHs by CYP1A1 through influencing epoxide metabolite formation. The QSAR models with these key properties provide a new perspective on the structural mechanism of PAH metabolism and provide a useful in silico tool for screening, classifying and predicting PAHs for their metabolism-related toxicities and risk assessment in the environment.


Asunto(s)
Citocromo P-450 CYP1A1 , Hidrocarburos Policíclicos Aromáticos , Citocromo P-450 CYP1A1/metabolismo , Cinética , Hidrocarburos Policíclicos Aromáticos/toxicidad , Relación Estructura-Actividad Cuantitativa
10.
Biochem Soc Trans ; 49(1): 131-144, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33367567

RESUMEN

Enzyme catalysis is omnipresent in the cell. The mechanisms by which highly evolved protein folds enable rapid and specific chemical transformation of substrates belong to the marvels of structural biology. Targeting of enzymes with inhibitors has immediate application in drug discovery, from chemotherapeutics over antibiotics to antivirals. NMR spectroscopy combines multiple assets for the investigation of enzyme function. The non-invasive technique can probe enzyme structure and dynamics and map interactions with substrates, cofactors and inhibitors at the atomic level. With experiments performed at close to native conditions, catalytic transformations can be monitored in real time, giving access to kinetic parameters. The power of NMR in the solid state, in contrast with solution, lies in the absence of fundamental size limitations, which is crucial for enzymes that are either membrane-embedded or assemble into large soluble complexes exceeding hundreds of kilodaltons in molecular weight. Here we review recent progress in solid-state NMR methodology, which has taken big leaps in the past years due to steady improvements in hardware design, notably magic angle spinning, and connect it to parallel biochemical advances that enable isotope labelling of increasingly complex enzymes. We first discuss general concepts and requirements of the method and then highlight the state-of-the-art in sample preparation, structure determination, dynamics and interaction studies. We focus on examples where solid-state NMR has been instrumental in elucidating enzyme mechanism, alone or in integrative studies.


Asunto(s)
Enzimas , Espectroscopía de Resonancia Magnética/métodos , Complejos Multiproteicos/química , Animales , Activadores de Enzimas/química , Activadores de Enzimas/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Enzimas/química , Enzimas/metabolismo , Humanos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/análisis , Complejos Multiproteicos/metabolismo , Especificidad por Sustrato
11.
PeerJ ; 8: e9586, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32765969

RESUMEN

Recycling biomass is indispensable these days not only because fossil energy sources are gradually depleted, but also because pollution of the environment, caused by the increasing use of energy, must be reduced. This article intends to overview the results of plant biomass processing methods that are currently in use. Our aim was also to review published methods that are not currently in use. It is intended to explore the possibilities of new methods and enzymes to be used in biomass recycling. The results of this overview are perplexing in almost every area. Advances have been made in the pre-treatment of biomass and in the diversity and applications of the enzymes utilized. Based on molecular modeling, very little progress has been made in the modification of existing enzymes for altered function and adaptation for the environmental conditions during the processing of biomass. There are hardly any publications in which molecular modeling techniques are used to improve enzyme function and to adapt enzymes to various environmental conditions. Our view is that using modern computational, biochemical, and biotechnological methods would enable the purposeful design of enzymes that are more efficient and suitable for biomass processing.

12.
Biochem J ; 476(7): 1109-1119, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30898847

RESUMEN

The biologically important carnitine biosynthesis pathway in humans proceeds via four enzymatic steps. The first step in carnitine biosynthesis is catalyzed by trimethyllysine hydroxylase (TMLH), a non-heme Fe(II) and 2-oxoglutarate (2OG)-dependent oxygenase, which catalyzes the stereospecific hydroxylation of (2S)-Nε-trimethyllysine to (2S,3S)-3-hydroxy-Nε-trimethyllysine. Here, we report biocatalytic studies on human TMLH and its 19 variants introduced through site-directed mutagenesis. Amino acid substitutions at the sites involved in binding of the Fe(II) cofactor, 2OG cosubstrate and (2S)-Nε-trimethyllysine substrate provide a basic insight into the binding requirements that determine an efficient TMLH-catalyzed conversion of (2S)-Nε-trimethyllysine to (2S,3S)-3-hydroxy-Nε-trimethyllysine. This work demonstrates the importance of the recognition sites that contribute to the enzymatic activity of TMLH: the Fe(II)-binding H242-D244-H389 residues, R391-R398 involved in 2OG binding and several residues (D231, N334 and the aromatic cage comprised of W221, Y217 and Y234) associated with binding of (2S)-Nε-trimethyllysine.


Asunto(s)
Oxigenasas de Función Mixta/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Biocatálisis , Carnitina/biosíntesis , Dominio Catalítico/genética , Humanos , Cinética , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , gamma-Butirobetaína Dioxigenasa/química , gamma-Butirobetaína Dioxigenasa/genética , gamma-Butirobetaína Dioxigenasa/metabolismo
13.
Biochem J ; 476(4): 719-732, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30718305

RESUMEN

d-aminoacyl-tRNA-deacylase (DTD) prevents the incorporation of d-amino acids into proteins during translation by hydrolyzing the ester bond between mistakenly attached amino acids and tRNAs. Despite extensive study of this proofreading enzyme, the precise catalytic mechanism remains unknown. Here, a combination of biochemical and computational investigations has enabled the discovery of a new substrate-assisted mechanism of d-Tyr-tRNATyr hydrolysis by Thermus thermophilus DTD. Several functional elements of the substrate, misacylated tRNA, participate in the catalysis. During the hydrolytic reaction, the 2'-OH group of the А76 residue of d-Tyr-tRNATyr forms a hydrogen bond with a carbonyl group of the tyrosine residue, stabilizing the transition-state intermediate. Two water molecules participate in this reaction, attacking and assisting ones, resulting in a significant decrease in the activation energy of the rate-limiting step. The amino group of the d-Tyr aminoacyl moiety is unprotonated and serves as a general base, abstracting the proton from the assisting water molecule and forming a more nucleophilic ester-attacking species. Quantum chemical methodology was used to investigate the mechanism of hydrolysis. The DFT-calculated deacylation reaction is in full agreement with the experimental data. The Gibbs activation energies for the first and second steps were 10.52 and 1.05 kcal/mol, respectively, highlighting that the first step of the hydrolysis process is the rate-limiting step. Several amino acid residues of the enzyme participate in the coordination of the substrate and water molecules. Thus, the present work provides new insights into the proofreading details of misacylated tRNAs and can be extended to other systems important for translation fidelity.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Biosíntesis de Proteínas/fisiología , ARN Bacteriano , Aminoacil-ARN de Transferencia , Thermus thermophilus , Proteínas Bacterianas/química , Catálisis , Hidrólisis , ARN Bacteriano/química , ARN Bacteriano/metabolismo , Aminoacil-ARN de Transferencia/química , Aminoacil-ARN de Transferencia/metabolismo , Thermus thermophilus/química , Thermus thermophilus/metabolismo
14.
Biosci Rep ; 38(6)2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30401732

RESUMEN

Despite the enormous number of therapeutic advances in medicine, nowadays many diseases are still incurable, mainly due to the lack of knowledge of the pathological biochemical pathways triggering those diseases. For this reason, it is compulsory for the scientific community to investigate and unveil the biomolecular mechanisms responsible for the development of those diseases, such as Alzheimer's disease and diabetes, which are widespread all over the world. In this scenario, it is of paramount importance to develop new analytical techniques and experimental procedures that are capable to make the above-mentioned investigations feasible. These new methods should allow easy performable analysis carried out in a label-free environment, in order to give reliable answers to specific biochemical questions. A recent paper published on Bioscience Reports by Ivancic et al. (https://doi.org/10.1042/BSR20181416) proposes a new analytical technique capable to reveal some mechanistic insights into the regulation of insulin-degrading enzyme (IDE), a protein involved in the above-mentioned diseases. IDE is a multifaceted enzyme having different and not well-defined roles in the cell, but it is primarily a proteolytic enzyme capable to degrade several different amyloidogenic substrates involved in different diseases. Moreover, many molecules are responsible for IDE activity modulation so that understanding how IDE activity is regulated represents a very challenging analytical task. The new analytical approach proposed by Ivancic et al. reports on the possibility to study IDE activity in an unbiased and label-free manner, representing a valid alternative assay for the investigation of any proteases degradative activity.


Asunto(s)
Enfermedad de Alzheimer , Insulisina , Dicroismo Circular , Humanos , Insulina , Cinética
15.
Biochem J ; 475(4): 787-802, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29367265

RESUMEN

Polyamines are linear polycationic compounds that play a crucial role in the growth and development of higher plants. One triamine (spermidine, SPD) and two tetraamine isomers (spermine, SPM, and thermospermine, TSPM) are obtained by the transfer of the aminopropyl group from decarboxylated S-adenosylmethionine to putrescine and SPD. These reactions are catalyzed by the specialized aminopropyltransferases. In that respect, plants are unique eukaryotes that have independently evolved two enzymes, thermospermine synthase (TSPS), encoded by the gene ACAULIS5, and spermine synthase, which produce TSPM and SPM, respectively. In this work, we structurally characterize the ACAULIS5 gene product, TSPS, from the model legume plant Medicago truncatula (Mt). Six crystal structures of MtTSPS - one without ligands and five in complexes with either reaction substrate (SPD), reaction product (TSPM), or one of three cofactor analogs (5'-methylthioadenosine, S-adenosylthiopropylamine, and adenosine) - give detailed insights into the biosynthesis of TSPM. Combined with small-angle X-ray scattering data, the crystal structures show that MtTSPS is a symmetric homotetramer with an interdomain eight-stranded ß-barrel. Such an assembly and the presence of a hinge-like feature between N-terminal and C-terminal domains give the protein additional flexibility which potentially improves loading substrates and discarding products after the catalytic event. We also discuss the sequence and structural features around the active site of the plant aminopropyltransferases that distinguish them from each other and determine their characteristic substrate discrimination.


Asunto(s)
Medicago truncatula/enzimología , Conformación Proteica , Espermidina Sintasa/química , Espermina Sintasa/química , Dominio Catalítico , Cristalografía por Rayos X , Espermidina Sintasa/genética , Espermina/análogos & derivados , Espermina/química , Espermina/metabolismo , Espermina Sintasa/genética , Especificidad por Sustrato
16.
Neurochem Int ; 113: 112-119, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29224965

RESUMEN

Compelling evidence suggests a crucial role of amyloid beta peptides (Aß(1-40/42)) in the etiology of Alzheimer's disease (AD). The N-terminal truncation of Aß(1-40/42) and their modification, e.g. by glutaminyl cyclase (QC), is expected to enhance the amyloid toxicity. In this work, the MALDI-TOF mass spectrometry application proved N-terminal cleavage of Aß(1-40/42) by purified dipeptidyl peptidase IV (DPPIV) in vitro observed earlier. The subsequent transformation of resulted Aß(3-40/42) to pE-Aß(3-40/42) in QC catalyzed glutamate cyclization was manifested. Hence, consecutive conversion of Aß(1-40/42) by DPPIV and QC can be assumed as a potential mechanism of formation of non-degrading pyroglutamated pE-Aß(3-40/42), which might accumulate and contribute to AD progression. The in vitro acceleration of Aß(1-40) aggregation in the simultaneous presence of DPPIV and QC was shown also.


Asunto(s)
Aminoaciltransferasas/metabolismo , Péptidos beta-Amiloides/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Fragmentos de Péptidos/metabolismo , Ácido Pirrolidona Carboxílico/farmacología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/análisis , Péptidos beta-Amiloides/genética , Animales , Bovinos , Humanos , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/genética , Agregado de Proteínas/efectos de los fármacos , Agregado de Proteínas/fisiología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos
17.
Biochem Soc Trans ; 45(5): 1159-1171, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28900015

RESUMEN

Pif1 family helicases have multiple roles in the maintenance of nuclear and mitochondrial DNA in eukaryotes. Saccharomyces cerevisiae Pif1 is involved in replication through barriers to replication, such as G-quadruplexes and protein blocks, and reduces genetic instability at these sites. Another Pif1 family helicase in S. cerevisiae, Rrm3, assists in fork progression through replication fork barriers at the rDNA locus and tRNA genes. ScPif1 (Saccharomyces cerevisiae Pif1) also negatively regulates telomerase, facilitates Okazaki fragment processing, and acts with polymerase δ in break-induced repair. Recent crystal structures of bacterial Pif1 helicases and the helicase domain of human PIF1 combined with several biochemical and biological studies on the activities of Pif1 helicases have increased our understanding of the function of these proteins. This review article focuses on these structures and the mechanism(s) proposed for Pif1's various activities on DNA.


Asunto(s)
ADN Helicasas/química , ADN Helicasas/metabolismo , ADN/metabolismo , Bacterias/química , Bacterias/enzimología , Cristalografía por Rayos X , ADN/química , Replicación del ADN , G-Cuádruplex , Humanos , Modelos Moleculares , Estructura Cuaternaria de Proteína , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo
18.
Biotechnol Bioeng ; 114(3): 503-515, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27617791

RESUMEN

The enzymatic hydrolysis of cellulose is a thermodynamically challenging catalytic process that is influenced by both substrate-related and enzyme-related factors. In this study, a proteolysis approach was applied to recover and clean the partially converted cellulose at the different stages of enzymatic hydrolysis to monitor the hydrolysis rate as a function of substrate reactivity/accessibility and investigate surface characteristics of the partially converted cellulose. Enzyme-substrate interactions between individual key cellulase components from wild-type Trichoderma reesei and partially converted cellulose were followed and correlated to the enzyme adsorption capacity and dynamic sugar release. Results suggest that cellobiohydrolase CBH1 (Cel7A) and endoglucanases EG2 (Cel5A) adsorption capacities decreased as cellulose was progressively hydrolyzed, likely due to the "depletion" of binding sites. Furthermore, the degree of synergism between CBH1 and EG2 varied depending on the enzyme loading and the substrates. The results provide a better understanding of the relationship between dynamic change of substrate features and the functionality of various cellulase components during enzymatic hydrolysis. Biotechnol. Bioeng. 2017;114: 503-515. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Celulasa/metabolismo , Celulosa/metabolismo , Adsorción , Celulasa/química , Celulosa/análisis , Celulosa/química , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Hidrólisis , Unión Proteica , Trichoderma/enzimología
19.
Biosens Bioelectron ; 84: 7-14, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27140308

RESUMEN

In the fields of clinical diagnostics and point-of-care diagnosis as well as food and environmental monitoring there is a high demand for reliable high-throughput, rapid and highly sensitive assays for a simultaneous detection of several analytes in complex and low-volume samples. Sensor platforms based on solution-processable electrolyte-gated carbon nanotube field-effect transistors (CNT-FETs) are a simple and cost-effective alternative for conventional assays. In this work we demonstrate a selective as well as direct detection of the products of an enzyme-substrate interaction, here the for metabolic processes important urea-urease system, with sensors based on spray-coated CNT-FETs. The selective and direct detection is achieved by immobilizing the enzyme urease via certain surface functionalization techniques on the sensor surface and further modifying the active interfaces with polymeric ion-selective membranes as well as pH-sensitive layers. Thereby, we can avoid the generally applied approach for a field-effect based detection of enzyme reactions via detecting changes in the pH value due to an on-going enzymatic reaction and directly detect selectively the products of the enzymatic conversion. Thus, we can realize a buffering-capacity independent monitoring of changes in the substrate concentration.


Asunto(s)
Técnicas Biosensibles/instrumentación , Pruebas de Enzimas/instrumentación , Enzimas Inmovilizadas/metabolismo , Nanotubos de Carbono/química , Transistores Electrónicos , Urea/metabolismo , Ureasa/metabolismo , Enzimas Inmovilizadas/química , Diseño de Equipo , Humanos , Urea/análisis , Ureasa/química
20.
Bioresour Technol ; 164: 143-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24852646

RESUMEN

Enzymatic hydrolysis of biomass undergoes a significant decrease in rate, which is often attributed to activity loss of enzyme during the incubation. Activity loss due to both interaction with substrate (for example inactivation of adsorbed enzyme) and all combined environmental mechanisms in a substrate free buffer solution were compared in this study. Enzyme-substrate interactions contributed more towards the overall activity loss than did the combined environmental sources as evidenced from three independent metrics. (1) Relative extents of inactivation were higher for enzyme-substrate interactions than for environmental mechanisms. (2) Apparent half-lives (1.37-11.01 h) following interaction with substrate were relatively small compared to environmental inactivation, which was 21.5h. (3) The inactivation rate constant for enzyme-substrate interactions (0.56 h(-1)) was 46 times higher than that of environmental inactivation (0.0123 h(-1)). These results suggest enzyme-substrate interaction is the main cause of cellulase activity loss and contributes significantly to the slow rate of hydrolysis.


Asunto(s)
Celulasa/metabolismo , Activación Enzimática , Semivida , Cinética , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA