Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 674
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 596(7873): 603-607, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34381213

RESUMEN

Single-particle cryogenic electron microscopy (cryo-EM) has become a standard technique for determining protein structures at atomic resolution1-3. However, cryo-EM studies of protein-free RNA are in their early days. The Tetrahymena thermophila group I self-splicing intron was the first ribozyme to be discovered and has been a prominent model system for the study of RNA catalysis and structure-function relationships4, but its full structure remains unknown. Here we report cryo-EM structures of the full-length Tetrahymena ribozyme in substrate-free and bound states at a resolution of 3.1 Å. Newly resolved peripheral regions form two coaxially stacked helices; these are interconnected by two kissing loop pseudoknots that wrap around the catalytic core and include two previously unforeseen (to our knowledge) tertiary interactions. The global architecture is nearly identical in both states; only the internal guide sequence and guanosine binding site undergo a large conformational change and a localized shift, respectively, upon binding of RNA substrates. These results provide a long-sought structural view of a paradigmatic RNA enzyme and signal a new era for the cryo-EM-based study of structure-function relationships in ribozymes.


Asunto(s)
Microscopía por Crioelectrón , Conformación de Ácido Nucleico , ARN Catalítico/química , ARN Catalítico/ultraestructura , Tetrahymena thermophila , Apoenzimas/química , Apoenzimas/ultraestructura , Holoenzimas/química , Holoenzimas/ultraestructura , Modelos Moleculares , Tetrahymena thermophila/enzimología , Tetrahymena thermophila/genética
2.
Nature ; 579(7800): 615-619, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32214249

RESUMEN

Arenaviruses can cause severe haemorrhagic fever and neurological diseases in humans and other animals, exemplified by Lassa mammarenavirus, Machupo mammarenavirus and lymphocytic choriomeningitis virus, posing great threats to public health1-4. These viruses encode a large multi-domain RNA-dependent RNA polymerase for transcription and replication of the viral genome5. Viral polymerases are one of the leading antiviral therapeutic targets. However, the structure of arenavirus polymerase is not yet known. Here we report the near-atomic resolution structures of Lassa and Machupo virus polymerases in both apo and promoter-bound forms. These structures display a similar overall architecture to influenza virus and bunyavirus polymerases but possess unique local features, including an arenavirus-specific insertion domain that regulates the polymerase activity. Notably, the ordered active site of arenavirus polymerase is inherently switched on, without the requirement for allosteric activation by 5'-viral RNA, which is a necessity for both influenza virus and bunyavirus polymerases6,7. Moreover, dimerization could facilitate the polymerase activity. These findings advance our understanding of the mechanism of arenavirus replication and provide an important basis for developing antiviral therapeutics.


Asunto(s)
Arenavirus del Nuevo Mundo/enzimología , Microscopía por Crioelectrón , Virus Lassa/enzimología , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/ultraestructura , Replicación Viral , Apoenzimas/química , Apoenzimas/metabolismo , Apoenzimas/ultraestructura , Arenavirus del Nuevo Mundo/ultraestructura , Dominio Catalítico , Virus Lassa/ultraestructura , Virus de la Coriomeningitis Linfocítica/enzimología , Virus de la Coriomeningitis Linfocítica/ultraestructura , Modelos Moleculares , Regiones Promotoras Genéticas/genética , ARN Polimerasa Dependiente del ARN/metabolismo
3.
Nature ; 575(7783): 540-544, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31723264

RESUMEN

Transposons have had a pivotal role in genome evolution1 and are believed to be the evolutionary progenitors of the RAG1-RAG2 recombinase2, an essential component of the adaptive immune system in jawed vertebrates3. Here we report one crystal structure and five cryo-electron microscopy structures of Transib4,5, a RAG1-like transposase from Helicoverpa zea, that capture the entire transposition process from the apo enzyme to the terminal strand transfer complex with transposon ends covalently joined to target DNA, at resolutions of 3.0-4.6 Å. These structures reveal a butterfly-shaped complex that undergoes two cycles of marked conformational changes in which the 'wings' of the transposase unfurl to bind substrate DNA, close to execute cleavage, open to release the flanking DNA and close again to capture and attack target DNA. Transib possesses unique structural elements that compensate for the absence of a RAG2 partner, including a loop that interacts with the transposition target site and an accordion-like C-terminal tail that elongates and contracts to help to control the opening and closing of the enzyme and assembly of the active site. Our findings reveal the detailed reaction pathway of a eukaryotic cut-and-paste transposase and illuminate some of the earliest steps in the evolution of the RAG recombinase.


Asunto(s)
Biocatálisis , Proteínas de Homeodominio , Mariposas Nocturnas/enzimología , Transposasas/química , Transposasas/metabolismo , Secuencia de Aminoácidos , Animales , Apoenzimas/química , Apoenzimas/metabolismo , Apoenzimas/ultraestructura , Secuencia de Bases , Microscopía por Crioelectrón , Cristalografía por Rayos X , ADN/química , ADN/genética , ADN/metabolismo , División del ADN , Proteínas de Unión al ADN , Evolución Molecular , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/ultraestructura , Modelos Moleculares , Mariposas Nocturnas/ultraestructura , Dominios Proteicos , Transposasas/ultraestructura
4.
J Biol Chem ; 299(9): 105152, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37567475

RESUMEN

The ESKAPE bacteria are the six highly virulent and antibiotic-resistant pathogens that require the most urgent attention for the development of novel antibiotics. Detailed knowledge of target proteins specific to bacteria is essential to develop novel treatment options. The methylerythritol-phosphate (MEP) pathway, which is absent in humans, represents a potentially valuable target for the development of novel antibiotics. Within the MEP pathway, the enzyme 1-deoxy-D-xylulose-5-phosphate synthase (DXPS) catalyzes a crucial, rate-limiting first step and a branch point in the biosynthesis of the vitamins B1 and B6. We report the high-resolution crystal structures of DXPS from the important ESKAPE pathogens Pseudomonas aeruginosa and Klebsiella pneumoniae in both the co-factor-bound and the apo forms. We demonstrate that the absence of the cofactor thiamine diphosphate results in conformational changes that lead to disordered loops close to the active site that might be important for the design of potent DXPS inhibitors. Collectively, our results provide important structural details that aid in the assessment of DXPS as a potential target in the ongoing efforts to combat antibiotic resistance.


Asunto(s)
Coenzimas , Klebsiella pneumoniae , Pseudomonas aeruginosa , Transferasas , Humanos , Antibacterianos/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/enzimología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/enzimología , Transferasas/química , Transferasas/metabolismo , Conformación Proteica , Coenzimas/metabolismo , Vitamina B 6/biosíntesis , Tiamina/biosíntesis , Apoenzimas/química , Apoenzimas/metabolismo , Tiamina Pirofosfato/metabolismo , Dominio Catalítico , Farmacorresistencia Bacteriana
5.
Nature ; 550(7677): 481-486, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29045389

RESUMEN

Ubiquitination controls the stability of most cellular proteins, and its deregulation contributes to human diseases including cancer. Deubiquitinases remove ubiquitin from proteins, and their inhibition can induce the degradation of selected proteins, potentially including otherwise 'undruggable' targets. For example, the inhibition of ubiquitin-specific protease 7 (USP7) results in the degradation of the oncogenic E3 ligase MDM2, and leads to re-activation of the tumour suppressor p53 in various cancers. Here we report that two compounds, FT671 and FT827, inhibit USP7 with high affinity and specificity in vitro and within human cells. Co-crystal structures reveal that both compounds target a dynamic pocket near the catalytic centre of the auto-inhibited apo form of USP7, which differs from other USP deubiquitinases. Consistent with USP7 target engagement in cells, FT671 destabilizes USP7 substrates including MDM2, increases levels of p53, and results in the transcription of p53 target genes, induction of the tumour suppressor p21, and inhibition of tumour growth in mice.


Asunto(s)
Piperidinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Peptidasa Específica de Ubiquitina 7/antagonistas & inhibidores , Animales , Apoenzimas/antagonistas & inhibidores , Apoenzimas/química , Apoenzimas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Femenino , Humanos , Ratones , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Piperidinas/síntesis química , Proteínas Proto-Oncogénicas c-mdm2/química , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Pirazoles/síntesis química , Pirimidinas/síntesis química , Especificidad por Sustrato , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Peptidasa Específica de Ubiquitina 7/química , Peptidasa Específica de Ubiquitina 7/metabolismo , Ubiquitinación/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Nucleic Acids Res ; 49(17): 9607-9624, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-33880546

RESUMEN

Tpt1, an essential component of the fungal and plant tRNA splicing machinery, catalyzes transfer of an internal RNA 2'-PO4 to NAD+ yielding RNA 2'-OH and ADP-ribose-1',2'-cyclic phosphate products. Here, we report NMR structures of the Tpt1 ortholog from the bacterium Runella slithyformis (RslTpt1), as apoenzyme and bound to NAD+. RslTpt1 consists of N- and C-terminal lobes with substantial inter-lobe dynamics in the free and NAD+-bound states. ITC measurements of RslTpt1 binding to NAD+ (KD ∼31 µM), ADP-ribose (∼96 µM) and ADP (∼123 µM) indicate that substrate affinity is determined primarily by the ADP moiety; no binding of NMN or nicotinamide is observed by ITC. NAD+-induced chemical shift perturbations (CSPs) localize exclusively to the RslTpt1 C-lobe. NADP+, which contains an adenylate 2'-PO4 (mimicking the substrate RNA 2'-PO4), binds with lower affinity (KD ∼1 mM) and elicits only N-lobe CSPs. The RslTpt1·NAD+ binary complex reveals C-lobe contacts to adenosine ribose hydroxyls (His99, Thr101), the adenine nucleobase (Asn105, Asp112, Gly113, Met117) and the nicotinamide riboside (Ser125, Gln126, Asn163, Val165), several of which are essential for RslTpt1 activity in vivo. Proximity of the NAD+ ß-phosphate to ribose-C1″ suggests that it may stabilize an oxocarbenium transition-state during the first step of the Tpt1-catalyzed reaction.


Asunto(s)
Proteínas Bacterianas/química , Cytophagaceae/enzimología , NAD/química , Fosfotransferasas/química , Apoenzimas/química , Proteínas Bacterianas/genética , Sitios de Unión , Ligandos , Modelos Moleculares , Mutagénesis , Resonancia Magnética Nuclear Biomolecular , Nucleótidos/química , Fosfotransferasas/genética , Unión Proteica , Conformación Proteica , ARN/metabolismo
7.
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
8.
Proteins ; 89(9): 1216-1225, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33983654

RESUMEN

The main protease Mpro , 3CLpro is an important target from coronaviruses. In spite of having 96% sequence identity among Mpros from SARS-CoV-1 and SARS-CoV-2; the inhibitors used to block the activity of SARS-CoV-1 Mpro so far, were found to have differential inhibitory effect on Mpro of SARS-CoV-2. The possible reason could be due to the difference of few amino acids among the peptidases. Since, overall 3-D crystallographic structure of Mpro from SARS-CoV-1 and SARS-CoV-2 is quite similar and mapping a subtle structural variation is seemingly impossible. Hence, we have attempted to study a structural comparison of SARS-CoV-1 and SARS-CoV-2 Mpro in apo and inhibitor bound states using protein structure network (PSN) based approach at contacts level. The comparative PSNs analysis of apo Mpros from SARS-CoV-1 and SARS-CoV-2 uncovers small but significant local changes occurring near the active site region and distributed throughout the structure. Additionally, we have shown how inhibitor binding perturbs the PSG and the communication pathways in Mpros . Moreover, we have also investigated the network connectivity on the quaternary structure of Mpro and identified critical residue pairs for complex formation using three centrality measurement parameters along with the modularity analysis. Taken together, these results on the comparative PSN provide an insight into conformational changes that may be used as an additional guidance towards specific drug development.


Asunto(s)
Proteasas 3C de Coronavirus/química , SARS-CoV-2/enzimología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Apoenzimas/antagonistas & inhibidores , Apoenzimas/química , Apoenzimas/metabolismo , Sitios de Unión , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/metabolismo , Holoenzimas/química , Holoenzimas/metabolismo , Modelos Moleculares , Inhibidores de Proteasas/farmacología , Multimerización de Proteína/efectos de los fármacos , Estructura Cuaternaria de Proteína/efectos de los fármacos
9.
Biochem Biophys Res Commun ; 553: 85-91, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33765558

RESUMEN

Glucose-6-phosphate dehydrogenase is the first enzyme in the pentose phosphate pathway. The reaction catalyzed by the enzyme is considered to be the main source of reducing power for nicotinamide adenine dinucleotide phosphate (NADPH) and is a precursor of 5-carbon sugar used by cells. To uncover the structural features of the enzyme, we determined the crystal structures of glucose-6-phosphate dehydrogenase from Kluyveromyces lactis (KlG6PD) in both the apo form and a binary complex with its substrate glucose-6-phosphate. KlG6PD contains a Rossman-like domain for cofactor NADPH binding; it also presents a typical antiparallel ß sheet at the C-terminal domain with relatively the same pattern as those of other homologous structures. Moreover, our structural and biochemical analyses revealed that Lys153 contributes significantly to substrate G6P recognition. This study may provide insights into the structural variation and catalytic features of the G6PD enzyme.


Asunto(s)
Glucosafosfato Deshidrogenasa/química , Glucosafosfato Deshidrogenasa/metabolismo , Kluyveromyces/enzimología , Secuencia de Aminoácidos , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Glucosafosfato Deshidrogenasa/genética , Cinética , Modelos Moleculares , Mutagénesis , Relación Estructura-Actividad , Especificidad por Sustrato
10.
Nature ; 527(7576): 114-7, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26503046

RESUMEN

Negative-sense RNA viruses, such as influenza, encode large, multidomain RNA-dependent RNA polymerases that can both transcribe and replicate the viral RNA genome. In influenza virus, the polymerase (FluPol) is composed of three polypeptides: PB1, PB2 and PA/P3. PB1 houses the polymerase active site, whereas PB2 and PA/P3 contain, respectively, cap-binding and endonuclease domains required for transcription initiation by cap-snatching. Replication occurs through de novo initiation and involves a complementary RNA intermediate. Currently available structures of the influenza A and B virus polymerases include promoter RNA (the 5' and 3' termini of viral genome segments), showing FluPol in transcription pre-initiation states. Here we report the structure of apo-FluPol from an influenza C virus, solved by X-ray crystallography to 3.9 Å, revealing a new 'closed' conformation. The apo-FluPol forms a compact particle with PB1 at its centre, capped on one face by PB2 and clamped between the two globular domains of P3. Notably, this structure is radically different from those of promoter-bound FluPols. The endonuclease domain of P3 and the domains within the carboxy-terminal two-thirds of PB2 are completely rearranged. The cap-binding site is occluded by PB2, resulting in a conformation that is incompatible with transcription initiation. Thus, our structure captures FluPol in a closed, transcription pre-activation state. This reveals the conformation of newly made apo-FluPol in an infected cell, but may also apply to FluPol in the context of a non-transcribing ribonucleoprotein complex. Comparison of the apo-FluPol structure with those of promoter-bound FluPols allows us to propose a mechanism for FluPol activation. Our study demonstrates the remarkable flexibility of influenza virus RNA polymerase, and aids our understanding of the mechanisms controlling transcription and genome replication.


Asunto(s)
Gammainfluenzavirus/enzimología , ARN Polimerasa Dependiente del ARN/química , Apoenzimas/química , Apoenzimas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Endonucleasas/química , Endonucleasas/metabolismo , Activación Enzimática , Modelos Moleculares , Iniciación de la Cadena Peptídica Traduccional , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Caperuzas de ARN/metabolismo , ARN Viral/biosíntesis , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleoproteínas/química
11.
Mol Cell ; 47(1): 76-86, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22793692

RESUMEN

NAD(P)H:quinone-oxidoreductase-1 (NQO1) is a cytosolic enzyme that catalyzes the reduction of various quinones using flavin adenine dinucleotide (FAD) as a cofactor. NQO1 has been also shown to rescue proteins containing intrinsically unstructured domains, such as p53 and p73, from degradation by the 20S proteasome through an unknown mechanism. Here, we studied the nature of interaction between NQO1 and the 20S proteasome. Our study revealed a double negative feedback loop between NQO1 and the 20S proteasome, whereby NQO1 prevents the proteolytic activity of the 20S proteasome and the 20S proteasome degrades the apo form of NQO1. Furthermore, we demonstrate, both in vivo and in vitro, that NQO1 levels are highly dependent on FAD concentration. These observations suggest a link between 20S proteolysis and the metabolic cellular state. More generally, the results may represent a regulatory mechanism by which associated cofactors dictate the stability of proteins, thus coordinating protein levels with the metabolic status.


Asunto(s)
Retroalimentación Fisiológica , Flavina-Adenina Dinucleótido/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Western Blotting , Línea Celular Tumoral , Estabilidad de Enzimas , Flavina-Adenina Dinucleótido/química , Células HEK293 , Células HeLa , Humanos , Espectrometría de Masas , Modelos Biológicos , Modelos Moleculares , NAD(P)H Deshidrogenasa (Quinona)/química , NAD(P)H Deshidrogenasa (Quinona)/genética , Complejo de la Endopetidasa Proteasomal/química , Unión Proteica , Pliegue de Proteína , Proteolisis , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Temperatura
12.
Nucleic Acids Res ; 46(14): 7309-7322, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-29917149

RESUMEN

DNA polymerase ß (pol ß) plays a central role in the DNA base excision repair pathway and also serves as an important model polymerase. Dynamic characterization of pol ß from methyl-TROSY 13C-1H multiple quantum CPMG relaxation dispersion experiments of Ile and Met sidechains and previous backbone relaxation dispersion measurements, reveals transitions in µs-ms dynamics in response to highly variable substrates. Recognition of a 1-nt-gapped DNA substrate is accompanied by significant backbone and sidechain motion in the lyase domain and the DNA binding subdomain of the polymerase domain, that may help to facilitate binding of the apoenzyme to the segments of the DNA upstream and downstream from the gap. Backbone µs-ms motion largely disappears after formation of the pol ß-DNA complex, giving rise to an increase in uncoupled µs-ms sidechain motion throughout the enzyme. Formation of an abortive ternary complex using a non-hydrolyzable dNTP results in sidechain motions that fit to a single exchange process localized to the catalytic subdomain, suggesting that this motion may play a role in catalysis.


Asunto(s)
ADN Polimerasa beta/química , Reparación del ADN , ADN/química , Conformación Proteica , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Biocatálisis , Cristalografía por Rayos X , ADN/genética , ADN/metabolismo , ADN Polimerasa beta/genética , ADN Polimerasa beta/metabolismo , Cinética , Modelos Moleculares , Movimiento (Física) , Resonancia Magnética Nuclear Biomolecular , Conformación de Ácido Nucleico , Unión Proteica , Especificidad por Sustrato , Factores de Tiempo
13.
Biophys J ; 116(10): 1823-1835, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31003762

RESUMEN

A critical step in injury-induced initiation of blood coagulation is the formation of the complex between the trypsin-like protease coagulation factor VIIa (FVIIa) and its cofactor tissue factor (TF), which converts FVIIa from an intrinsically poor enzyme to an active protease capable of activating zymogens of downstream coagulation proteases. Unlike its constitutively active ancestor trypsin, FVIIa is allosterically activated (by TF). Here, ensemble refinement of crystallographic structures, which uses multiple copies of the entire structure as a means of representing structural flexibility, is applied to explore the impacts of inhibitor binding to trypsin and FVIIa, as well as cofactor binding to FVIIa. To assess the conformational flexibility and its role in allosteric pathways in these proteases, main-chain hydrogen bond networks are analyzed by calculating the hydrogen-bond propensity. Mapping pairwise propensity differences between relevant structures shows that binding of the inhibitor benzamidine to trypsin has a minor influence on the protease flexibility. For FVIIa, in contrast, the protease domain is "locked" into the catalytically competent trypsin-like configuration upon benzamidine binding as indicated by the stabilization of key structural features: the nonprime binding cleft and the oxyanion hole are stabilized, and the effect propagates from the active site region to the calcium-binding site and to the vicinity of the disulphide bridge connecting with the light chain. TF binding to FVIIa furthermore results in stabilization of the 170 loop, which in turn propagates an allosteric signal from the TF-binding region to the active site. Analyses of disulphide bridge energy and flexibility reflect the striking stability difference between the unregulated enzyme and the allosterically activated form after inhibitor or cofactor binding. The ensemble refinement analyses show directly, for the first time to our knowledge, whole-domain structural footprints of TF-induced allosteric networks present in x-ray crystallographic structures of FVIIa, which previously only have been hypothesized or indirectly inferred.


Asunto(s)
Factor VIIa/química , Factor VIIa/metabolismo , Regulación Alostérica , Apoenzimas/química , Apoenzimas/metabolismo , Benzamidinas/farmacología , Cristalografía por Rayos X , Disulfuros/química , Activación Enzimática/efectos de los fármacos , Modelos Moleculares , Dominios Proteicos , Pliegue de Proteína , Tripsina/química , Tripsina/metabolismo , Tripsinógeno/metabolismo
14.
Nucleic Acids Res ; 45(11): 6684-6697, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28531330

RESUMEN

5-methylcytosine (m5C) modifications of RNA are ubiquitous in nature and play important roles in many biological processes such as protein translational regulation, RNA processing and stress response. Aberrant expressions of RNA:m5C methyltransferases are closely associated with various human diseases including cancers. However, no structural information for RNA-bound RNA:m5C methyltransferase was available until now, hindering elucidation of the catalytic mechanism behind RNA:m5C methylation. Here, we have solved the structures of NSun6, a human tRNA:m5C methyltransferase, in the apo form and in complex with a full-length tRNA substrate. These structures show a non-canonical conformation of the bound tRNA, rendering the base moiety of the target cytosine accessible to the enzyme for methylation. Further biochemical assays reveal the critical, but distinct, roles of two conserved cysteine residues for the RNA:m5C methylation. Collectively, for the first time, we have solved the complex structure of a RNA:m5C methyltransferase and addressed the catalytic mechanism of the RNA:m5C methyltransferase family, which may allow for structure-based drug design toward RNA:m5C methyltransferase-related diseases.


Asunto(s)
ARNt Metiltransferasas/química , Secuencia de Aminoácidos , Apoenzimas/química , Biocatálisis , Dominio Catalítico , Secuencia Conservada , Cristalografía por Rayos X , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , ARN de Transferencia/química , Especificidad por Sustrato
15.
Nucleic Acids Res ; 45(6): 3547-3558, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28039325

RESUMEN

Although all Type II restriction endonucleases catalyze phosphodiester bond hydrolysis within or close to their DNA target sites, they form different oligomeric assemblies ranging from monomers, dimers, tetramers to higher order oligomers to generate a double strand break in DNA. Type IIP restriction endonuclease AgeI recognizes a palindromic sequence 5΄-A/CCGGT-3΄ and cuts it ('/' denotes the cleavage site) producing staggered DNA ends. Here, we present crystal structures of AgeI in apo and DNA-bound forms. The structure of AgeI is similar to the restriction enzymes that share in their target sites a conserved CCGG tetranucleotide and a cleavage pattern. Structure analysis and biochemical data indicate, that AgeI is a monomer in the apo-form both in the crystal and in solution, however, it binds and cleaves the palindromic target site as a dimer. DNA cleavage mechanism of AgeI is novel among Type IIP restriction endonucleases.


Asunto(s)
División del ADN , Desoxirribonucleasas de Localización Especificada Tipo II/química , Apoenzimas/química , Emparejamiento Base , Dominio Catalítico , ADN/química , ADN/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Modelos Moleculares , Unión Proteica , Multimerización de Proteína
16.
Int J Mol Sci ; 20(5)2019 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-30836629

RESUMEN

Human triokinase/flavin mononucleotide (FMN) cyclase (hTKFC) catalyzes the adenosine triphosphate (ATP)-dependent phosphorylation of D-glyceraldehyde and dihydroxyacetone (DHA), and the cyclizing splitting of flavin adenine dinucleotide (FAD). hTKFC structural models are dimers of identical subunits, each with two domains, K and L, with an L2-K1-K2-L1 arrangement. Two active sites lie between L2-K1 and K2-L1, where triose binds K and ATP binds L, although the resulting ATP-to-triose distance is too large (≈14 Å) for phosphoryl transfer. A 75-ns trajectory of molecular dynamics shows considerable, but transient, ATP-to-DHA approximations in the L2-K1 site (4.83 Å or 4.16 Å). To confirm the trend towards site closure, and its relationship to kinase activity, apo-hTKFC, hTKFC:2DHA:2ATP and hTKFC:2FAD models were submitted to normal mode analysis. The trajectory of hTKFC:2DHA:2ATP was extended up to 160 ns, and 120-ns trajectories of apo-hTKFC and hTKFC:2FAD were simulated. The three systems were comparatively analyzed for equal lengths (120 ns) following the principles of essential dynamics, and by estimating site closure by distance measurements. The full trajectory of hTKFC:2DHA:2ATP was searched for in-line orientations and short distances of DHA hydroxymethyl oxygens to ATP γ-phosphorus. Full site closure was reached only in hTKFC:2DHA:2ATP, where conformations compatible with an associative phosphoryl transfer occurred in L2-K1 for significant trajectory time fractions.


Asunto(s)
Apoenzimas/genética , Simulación de Dinámica Molecular , Liasas de Fósforo-Oxígeno/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Adenosina Trifosfato/química , Apoenzimas/química , Sitios de Unión , Catálisis , Dominio Catalítico/genética , Dihidroxiacetona/química , Mononucleótido de Flavina/química , Mononucleótido de Flavina/genética , Flavina-Adenina Dinucleótido/química , Gliceraldehído/química , Humanos , Liasas de Fósforo-Oxígeno/genética , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Especificidad por Sustrato
17.
J Biol Chem ; 292(44): 18290-18302, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-28935670

RESUMEN

Strain SYK-6 of the bacterium Sphingobium sp. catabolizes lignin-derived biphenyl via a meta-cleavage pathway. In this pathway, LigY is proposed to catalyze the hydrolysis of the meta-cleavage product (MCP) 4,11-dicarboxy-8-hydroxy-9-methoxy-2-hydroxy-6-oxo-6-phenyl-hexa-2,4-dienoate. Here, we validated this reaction by identifying 5-carboxyvanillate and 4-carboxy-2-hydroxypenta-2,4-dienoate as the products and determined the kcat and kcat/Km values as 9.3 ± 0.6 s-1 and 2.5 ± 0.2 × 107 m-1 s-1, respectively. Sequence analyses and a 1.9 Å resolution crystal structure established that LigY belongs to the amidohydrolase superfamily, unlike previously characterized MCP hydrolases, which are serine-dependent enzymes of the α/ß-hydrolase superfamily. The active-site architecture of LigY resembled that of α-amino-ß-carboxymuconic-ϵ-semialdehyde decarboxylase, a class III amidohydrolase, with a single zinc ion coordinated by His-6, His-8, His-179, and Glu-282. Interestingly, we found that LigY lacks the acidic residue proposed to activate water for hydrolysis in other class III amidohydrolases. Moreover, substitution of His-223, a conserved residue proposed to activate water in other amidohydrolases, reduced the kcat to a much lesser extent than what has been reported for other amidohydrolases, suggesting that His-223 has a different role in LigY. Substitution of Arg-72, Tyr-190, Arg-234, or Glu-282 reduced LigY activity over 100-fold. On the basis of these results, we propose a catalytic mechanism involving substrate tautomerization, substrate-assisted activation of water for hydrolysis, and formation of a gem-diol intermediate. This last step diverges from what occurs in serine-dependent MCP hydrolases. This study provides insight into C-C-hydrolyzing enzymes and expands the known range of reactions catalyzed by the amidohydrolase superfamily.


Asunto(s)
Amidohidrolasas/metabolismo , Proteínas Bacterianas/metabolismo , Hidrolasas/metabolismo , Modelos Moleculares , Sphingomonadaceae/enzimología , Zinc/metabolismo , Amidohidrolasas/química , Amidohidrolasas/clasificación , Amidohidrolasas/genética , Sustitución de Aminoácidos , Apoenzimas/química , Apoenzimas/clasificación , Apoenzimas/genética , Apoenzimas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/genética , Sitios de Unión , Biocatálisis , Caproatos/metabolismo , Cristalografía por Rayos X , Glutaratos/metabolismo , Hidrolasas/química , Hidrolasas/clasificación , Hidrolasas/genética , Hidrólisis , Ligandos , Mutagénesis Sitio-Dirigida , Mutación , Parabenos/metabolismo , Ácidos Ftálicos/metabolismo , Filogenia , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/clasificación , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Homología Estructural de Proteína , Especificidad por Sustrato , Ácido Vanílico/análogos & derivados , Ácido Vanílico/metabolismo
18.
J Biol Chem ; 292(24): 10002-10013, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28455448

RESUMEN

Nonribosomal peptide synthesis involves the interplay between covalent protein modifications, conformational fluctuations, catalysis, and transient protein-protein interactions. Delineating the mechanisms involved in orchestrating these various processes will deepen our understanding of domain-domain communication in nonribosomal peptide synthetases (NRPSs) and lay the groundwork for the rational reengineering of NRPSs by swapping domains handling different substrates to generate novel natural products. Although many structural and biochemical studies of NRPSs exist, few studies have focused on the energetics and dynamics governing the interactions in these systems. Here, we present detailed binding studies of an adenylation domain and its partner carrier protein in apo-, holo-, and substrate-loaded forms. Results from fluorescence anisotropy, isothermal titration calorimetry, and NMR titrations indicated that covalent modifications to a carrier protein modulate domain communication, suggesting that chemical modifications to carrier proteins during NRPS synthesis may impart directionality to sequential NRPS domain interactions. Comparison of the structure and dynamics of an apo-aryl carrier protein with those of its modified forms revealed structural fluctuations induced by post-translational modifications and mediated by modulations of protein dynamics. The results provide a comprehensive molecular description of a carrier protein throughout its life cycle and demonstrate how a network of dynamic residues can propagate the molecular impact of chemical modifications throughout a protein and influence its affinity toward partner domains.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Coenzima A Ligasas/metabolismo , Modelos Moleculares , Péptido Sintasas/metabolismo , Modificación Traduccional de las Proteínas , Procesamiento Proteico-Postraduccional , Yersinia pestis/metabolismo , Sustitución de Aminoácidos , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Apoproteínas/química , Apoproteínas/genética , Apoproteínas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Calorimetría , Isótopos de Carbono , Proteínas Portadoras/química , Proteínas Portadoras/genética , Coenzima A Ligasas/química , Coenzima A Ligasas/genética , Polarización de Fluorescencia , Holoenzimas/química , Holoenzimas/genética , Holoenzimas/metabolismo , Cinética , Mutación , Isótopos de Nitrógeno , Resonancia Magnética Nuclear Biomolecular , Péptido Sintasas/química , Péptido Sintasas/genética , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Volumetría , Yersinia pestis/enzimología
19.
J Biol Chem ; 292(29): 12025-12040, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28533431

RESUMEN

Metallochaperones are a diverse family of trafficking molecules that provide metal ions to protein targets for use as cofactors. The copper chaperone for superoxide dismutase (Ccs1) activates immature copper-zinc superoxide dismutase (Sod1) by delivering copper and facilitating the oxidation of the Sod1 intramolecular disulfide bond. Here, we present structural, spectroscopic, and cell-based data supporting a novel copper-induced mechanism for Sod1 activation. Ccs1 binding exposes an electropositive cavity and proposed "entry site" for copper ion delivery on immature Sod1. Copper-mediated sulfenylation leads to a sulfenic acid intermediate that eventually resolves to form the Sod1 disulfide bond with concomitant release of copper into the Sod1 active site. Sod1 is the predominant disulfide bond-requiring enzyme in the cytoplasm, and this copper-induced mechanism of disulfide bond formation obviates the need for a thiol/disulfide oxidoreductase in that compartment.


Asunto(s)
Cobre/metabolismo , Cistina/metabolismo , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas de Saccharomyces cerevisiae/metabolismo , Superóxido Dismutasa/metabolismo , Sustitución de Aminoácidos , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Cisteína/metabolismo , Activación Enzimática , Estabilidad de Enzimas , Humanos , Ligandos , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Mutagénesis Sitio-Dirigida , Mutación , Oxidación-Reducción , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Superóxido Dismutasa/química , Superóxido Dismutasa/genética
20.
J Biol Chem ; 292(32): 13323-13332, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28607149

RESUMEN

Polycomb-group proteins control many fundamental biological processes, such as anatomical development in mammals and vernalization in plants. Polycomb repressive complex 2 (PRC2) is responsible for methylation of histone H3 lysine 27 (H3K27), and trimethylated H3K27 (H3K27me3) is implicated in epigenetic gene silencing. Recent genomic, biochemical, and structural data indicate that PRC2 is broadly conserved from yeast to human in many aspects. Here, we determined the crystal structure of an apo-PRC2 from the fungus Chaetomium thermophilum captured in a bona fide autoinhibited state, which represents a novel conformation of PRC2 associated with enzyme regulation in light of the basal and stimulated states that we reported previously. We found that binding by the cofactor S-adenosylmethionine mitigates this autoinhibited structural state. Using steady-state enzyme kinetics, we also demonstrated that disrupting the autoinhibition results in a vastly activated enzyme complex. Autoinhibition provides a novel structural platform that may enable control of PRC2 activity in response to diverse transcriptional states and chromatin contexts and set a ground state to allow PRC2 activation by other cellular mechanisms as well.


Asunto(s)
Chaetomium/enzimología , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteínas Fúngicas/metabolismo , Histonas/metabolismo , Modelos Moleculares , Procesamiento Proteico-Postraduccional , S-Adenosilmetionina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Coenzimas/química , Coenzimas/metabolismo , Secuencia Conservada , Proteína Potenciadora del Homólogo Zeste 2/química , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Histonas/química , Lisina/metabolismo , Metilación , Mutación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , S-Adenosilmetionina/química , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA