Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochemistry ; 59(35): 3247-3257, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32786412

RESUMEN

Trehalose-6-phosphate phosphatase (T6PP) catalyzes the dephosphorylation of trehalose 6-phosphate (T6P) to the disaccharide trehalose. The enzyme is not present in mammals but is essential to the viability of multiple lower organisms as trehalose is a critical metabolite, and T6P accumulation is toxic. Hence, T6PP is a target for therapeutics of human pathologies caused by bacteria, fungi, and parasitic nematodes. Here, we report the X-ray crystal structures of Salmonella typhimurium T6PP (StT6PP) in its apo form and in complex with the cofactor Mg2+ and the substrate analogue trehalose 6-sulfate (T6S), the product trehalose, or the competitive inhibitor 4-n-octylphenyl α-d-glucopyranoside 6-sulfate (OGS). OGS replaces the substrate phosphoryl group with a sulfate group and the glucosyl ring distal to the sulfate group with an octylphenyl moiety. The structures of these substrate-analogue and product complexes with T6PP show that specificity is conferred via hydrogen bonds to the glucosyl group proximal to the phosphoryl moiety through Glu123, Lys125, and Glu167, conserved in T6PPs from multiple species. The structure of the first-generation inhibitor OGS shows that it retains the substrate-binding interactions observed for the sulfate group and the proximal glucosyl ring. The OGS octylphenyl moiety binds in a unique manner, indicating that this subsite can tolerate various chemotypes. Together, these findings show that these conserved interactions at the proximal glucosyl ring binding site could provide the basis for the development of broad-spectrum therapeutics, whereas variable interactions at the divergent distal subsite could present an opportunity for the design of potent organism-specific therapeutics.


Asunto(s)
Monoéster Fosfórico Hidrolasas/metabolismo , Salmonella typhimurium/enzimología , Fosfatos de Azúcar/metabolismo , Trehalosa/análogos & derivados , Sitios de Unión , Cristalografía por Rayos X , Ligandos , Modelos Moleculares , Monoéster Fosfórico Hidrolasas/química , Unión Proteica , Pliegue de Proteína , Estructura Cuaternaria de Proteína , Especificidad por Sustrato , Fosfatos de Azúcar/química , Trehalosa/química , Trehalosa/metabolismo
2.
Biochemistry ; 57(30): 4504-4517, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-29952545

RESUMEN

α-Phosphoglucomutase (αPGM), in its phosphorylated state, catalyzes the interconversion of α-d-glucose 1-phosphate and α-d-glucose 6-phosphate. The αPGM of Lactococcus lactis is a type C2B member of the haloalkanoic acid dehalogenase (HAD) enzyme family and is comprised of a Rossmann-fold catalytic domain and inserted α/ß-fold cap domain. The active site is formed at the domain-domain interface. Herein, we report the results from a kinetic-based study of L. lactis αPGM catalysis, which demonstrate enzyme activation by autocatalyzed phosphorylation of Asp8 with αG1P, the intermediacy of αG1,6bisP in the phospho Ll-αPGM-catalyzed conversion of αG1P to G6P, and the reorientation of the αG1,6bisP intermediate via dissociation to solvent and rebinding. In order to provide insight into the structural determinants of L. lactis αPGM substrate recognition and catalysis, metal cofactor and substrate specificities were determined as were the contributions made by active-site residues toward catalytic efficiency. Lastly, the structure and catalytic mechanism of L. lactis αPGM are compared with those of HAD family phosphomutases L. lactis ß-phosphoglucomutase and eukayotic α-phosphomannomutase to provide insight into the evolution of phosphohexomutases from HAD family phosphatases.


Asunto(s)
Lactococcus lactis/enzimología , Fosfoglucomutasa/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Activación Enzimática , Glucosa-6-Fosfato/metabolismo , Glucofosfatos/metabolismo , Cinética , Lactococcus lactis/química , Lactococcus lactis/metabolismo , Modelos Moleculares , Fosfoglucomutasa/química , Fosforilación , Conformación Proteica , Especificidad por Sustrato
3.
Biochemistry ; 57(25): 3480-3492, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29695157

RESUMEN

The human phosphomannomutases PMM1 and PMM2 catalyze the interconversion of hexose 6-phosphates and hexose 1-phosphates. The two isoforms share 66% sequence identity and have kinetic properties similar to those of mutases in vitro but differ in their functional roles in vivo. Though the physiological role of PMM2 is catalysis of the mutase reaction that provides the mannose 1-phosphate (Man-1-P) essential for protein glycosylation, PMM1 is thought to provide a phosphohydrolase activity in the presence of inosine monophosphate (IMP), converting glucose 1,6-bisphosphate (Glu-1,6-P2) to glucose 6-phosphate (Glu-6-P), rescuing glycolysis during brain ischemia. To uncover the structural basis of how IMP binding converts PMM1 from a mutase to a phosphatase, the 1.93 Å resolution structure of PMM1 complexed with IMP was determined. The structure reveals IMP bound at the substrate recruitment site, thus inhibiting the mutase activity while simultaneously activating a phosphatase activity (IMP Kact = 1.5 µM) resulting from the hydrolysis of the phospho-enzyme. The bound structure and site-directed mutagenesis confirm that the long-range electrostatic interactions provided by Arg180 and Arg183 conserved in PMM1 are the major contributors to IMP binding, and their oblation removes phosphatase but not mutase activity. These residues are not present in the PMM2 isoform, which consequently lacks significant phosphatase activity in the presence of IMP. T2 relaxation nuclear magnetic resonance and small angle X-ray scattering together support the hypothesis that binding of IMP to PMM1 favors an enzyme conformation that is catalytically competent for water attack at the phosphoaspartyl intermediate. Such a mechanism may be generalizable to other enzymes that act through covalent intermediates.


Asunto(s)
Inosina Monofosfato/metabolismo , Fosfotransferasas (Fosfomutasas)/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Isquemia Encefálica/metabolismo , Cristalografía por Rayos X , Glucólisis , Humanos , Modelos Moleculares , Fosfotransferasas (Fosfomutasas)/química , Unión Proteica , Conformación Proteica , Alineación de Secuencia , Especificidad por Sustrato
4.
Chembiochem ; 18(19): 1935-1943, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28741300

RESUMEN

Thioesterase activity accounts for the majority of the activities in the hotdog-fold superfamily. The structures and mechanisms of catalysis for many hotdog enzymes have been elucidated by X-ray crystallography and kinetics to probe the specific substrate usage and cellular functions. However, structures of hotdog thioesterases in complexes with substrate analogues reported to date utilize ligands that either represent truncations of the substrate or include additional atoms to prevent hydrolysis. Here we present the synthesis of an isosteric and isoelectronic substrate analogue-benzoyl-OdCoA-and the X-ray crystal structure of a complex of the analogue with Pseudomonas aeruginosa hotdog thioesterase PA1618 (at 1.72 Šresolution). The complex is compared with that of the "imperfect" substrate analogue phenacyl-CoA, refined to a resolution of 1.62 Å. Kinetic and structural results are consistent with Glu64 as the catalytic residue and with the involvement of Gln49 in stabilization of the transition state. Structural comparison of the two ligand-bound structures revealed a crucial ordered water molecule coordinated in the active site of the benzoyl-OdCoA structure but not present in the phenacyl-CoA-bound structure. This suggests a general base mechanism of catalysis in which Glu64 activates the coordinated water nucleophile. Together, our findings reveal the importance of a closely similar substrate analogue to determine the true substrate binding and catalytic mechanism.


Asunto(s)
Ésteres/metabolismo , Oxígeno/metabolismo , Tioléster Hidrolasas/metabolismo , Biocatálisis , Cristalografía por Rayos X , Ésteres/química , Modelos Moleculares , Estructura Molecular , Oxígeno/química , Pseudomonas aeruginosa/enzimología , Tioléster Hidrolasas/química , Tioléster Hidrolasas/genética
5.
Eur J Med Chem ; 128: 274-286, 2017 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-28192710

RESUMEN

In some organisms, environmental stress triggers trehalose biosynthesis that is catalyzed collectively by trehalose 6-phosphate synthase, and trehalose 6-phosphate phosphatase (T6PP). T6PP catalyzes the hydrolysis of trehalose 6-phosphate (T6P) to trehalose and inorganic phosphate and is a promising target for the development of antibacterial, antifungal and antihelminthic therapeutics. Herein, we report the design, synthesis and evaluation of a library of aryl d-glucopyranoside 6-sulfates to serve as prototypes for small molecule T6PP inhibitors. Steady-state kinetic techniques were used to measure inhibition constants (Ki) of a panel of structurally diverse T6PP orthologs derived from the pathogens Brugia malayi, Ascaris suum, Mycobacterium tuberculosis, Shigella boydii and Salmonella typhimurium. The binding affinities of the most active inhibitor of these T6PP orthologs, 4-n-octylphenyl α-d-glucopyranoside 6-sulfate (9a), were found to be in the low micromolar range. The Ki of 9a with the B. malayi T6PP ortholog is 5.3 ± 0.6 µM, 70-fold smaller than the substrate Michaelis constant. The binding specificity of 9a was demonstrated using several representative sugar phosphate phosphatases from the HAD enzyme superfamily, the T6PP protein fold family of origin. Lastly, correlations drawn between T6PP active site structure, inhibitor structure and inhibitor binding affinity suggest that the aryl d-glucopyranoside 6-sulfate prototypes will find future applications as a platform for development of tailored second-generation T6PP inhibitors.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Glucosiltransferasas/antagonistas & inhibidores , Monosacáridos/farmacología , Fosfatos de Azúcar/metabolismo , Trehalosa/análogos & derivados , Animales , Brugia Malayi/enzimología , Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Mycobacterium tuberculosis/enzimología , Salmonella typhimurium/enzimología , Shigella boydii/enzimología , Trehalosa/metabolismo
6.
Curr Opin Struct Biol ; 41: 172-179, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27526404

RESUMEN

A single genome encodes a large number of phosphoryl hydrolases for the purposes of phosphate recycling, primary and secondary metabolism, signal transduction and regulation, and protection from xenobiotics. Phosphate monoester hydrolysis faces a high kinetic barrier, yet there are multiple solutions to the problem both in terms of catalytic mechanisms and three-dimensional structure of the hydrolases. Recent structural and mechanistic findings highlight the trigonal-bipyramidal nature of the transition state for enzyme promoted phosphate monoester hydrolysis and the evolution and role of inserted loops/domains in governing substrate specificity and promiscuity. Important questions remain as to how electrostatics modulate water networks and critical proton-transfer events. How substrate targeting and catalysis is achieved by the independently evolved catalytic platforms is compared and contrasted in this article.


Asunto(s)
Biocatálisis , Fosfatos/metabolismo , Humanos , Protones , Especificidad por Sustrato
7.
Proc Natl Acad Sci U S A ; 112(16): E1974-83, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25848029

RESUMEN

Large-scale activity profiling of enzyme superfamilies provides information about cellular functions as well as the intrinsic binding capabilities of conserved folds. Herein, the functional space of the ubiquitous haloalkanoate dehalogenase superfamily (HADSF) was revealed by screening a customized substrate library against >200 enzymes from representative prokaryotic species, enabling inferred annotation of ∼35% of the HADSF. An extremely high level of substrate ambiguity was revealed, with the majority of HADSF enzymes using more than five substrates. Substrate profiling allowed assignment of function to previously unannotated enzymes with known structure, uncovered potential new pathways, and identified iso-functional orthologs from evolutionarily distant taxonomic groups. Intriguingly, the HADSF subfamily having the least structural elaboration of the Rossmann fold catalytic domain was the most specific, consistent with the concept that domain insertions drive the evolution of new functions and that the broad specificity observed in HADSF may be a relic of this process.


Asunto(s)
Familia de Multigenes , Monoéster Fosfórico Hidrolasas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Cinética , Reproducibilidad de los Resultados , Especificidad por Sustrato
8.
J Biol Chem ; 289(44): 30229-30236, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25210039

RESUMEN

Catalytic promiscuity and substrate ambiguity are keys to evolvability, which in turn is pivotal to the successful acquisition of novel biological functions. Action on multiple substrates (substrate ambiguity) can be harnessed for performance of functions in the cell that supersede catalysis of a single metabolite. These functions include proofreading, scavenging of nutrients, removal of antimetabolites, balancing of metabolite pools, and establishing system redundancy. In this review, we present examples of enzymes that perform these cellular roles by leveraging substrate ambiguity and then present the structural features that support both specificity and ambiguity. We focus on the phosphatases of the haloalkanoate dehalogenase superfamily and the thioesterases of the hotdog fold superfamily.


Asunto(s)
Enzimas/química , Evolución Molecular , Animales , Biocatálisis , Dominio Catalítico , Enzimas/genética , Humanos , Modelos Moleculares , Especificidad por Sustrato
9.
PLoS Pathog ; 10(7): e1004245, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24992307

RESUMEN

Parasitic nematodes are responsible for devastating illnesses that plague many of the world's poorest populations indigenous to the tropical areas of developing nations. Among these diseases is lymphatic filariasis, a major cause of permanent and long-term disability. Proteins essential to nematodes that do not have mammalian counterparts represent targets for therapeutic inhibitor discovery. One promising target is trehalose-6-phosphate phosphatase (T6PP) from Brugia malayi. In the model nematode Caenorhabditis elegans, T6PP is essential for survival due to the toxic effect(s) of the accumulation of trehalose 6-phosphate. T6PP has also been shown to be essential in Mycobacterium tuberculosis. We determined the X-ray crystal structure of T6PP from B. malayi. The protein structure revealed a stabilizing N-terminal MIT-like domain and a catalytic C-terminal C2B-type HAD phosphatase fold. Structure-guided mutagenesis, combined with kinetic analyses using a designed competitive inhibitor, trehalose 6-sulfate, identified five residues important for binding and catalysis. This structure-function analysis along with computational mapping provided the basis for the proposed model of the T6PP-trehalose 6-phosphate complex. The model indicates a substrate-binding mode wherein shape complementarity and van der Waals interactions drive recognition. The mode of binding is in sharp contrast to the homolog sucrose-6-phosphate phosphatase where extensive hydrogen-bond interactions are made to the substrate. Together these results suggest that high-affinity inhibitors will be bi-dentate, taking advantage of substrate-like binding to the phosphoryl-binding pocket while simultaneously utilizing non-native binding to the trehalose pocket. The conservation of the key residues that enforce the shape of the substrate pocket in T6PP enzymes suggest that development of broad-range anthelmintic and antibacterial therapeutics employing this platform may be possible.


Asunto(s)
Antihelmínticos , Brugia Malayi/enzimología , Diseño de Fármacos , Proteínas del Helminto/química , Modelos Moleculares , Monoéster Fosfórico Hidrolasas/química , Animales , Filariasis/tratamiento farmacológico , Filariasis/enzimología , Estructura Terciaria de Proteína
10.
Biochemistry ; 53(29): 4775-87, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-24992697

RESUMEN

The work described in this paper, and its companion paper (Wu, R., Latham, J. A., Chen, D., Farelli, J., Zhao, H., Matthews, K. Allen, K. N., and Dunaway-Mariano, D. (2014) Structure and Catalysis in the Escherichia coli Hotdog-fold Thioesterase Paralogs YdiI and YbdB. Biochemistry, DOI: 10.1021/bi500334v), focuses on the evolution of a pair of paralogous hotdog-fold superfamily thioesterases of E. coli, YbdB and YdiI, which share a high level of sequence identity but perform different biological functions (viz., proofreader of 2,3-dihydroxybenzoyl-holoEntB in the enterobactin biosynthetic pathway and catalyst of the 1,4-dihydoxynapthoyl-CoA hydrolysis step in the menaquinone biosynthetic pathway, respectively). In vitro substrate activity screening of a library of thioester metabolites showed that YbdB displays high activity with benzoyl-holoEntB and benzoyl-CoA substrates, marginal activity with acyl-CoA thioesters, and no activity with 1,4-dihydoxynapthoyl-CoA. YdiI, on the other hand, showed a high level of activity with its physiological substrate, significant activity toward a wide range of acyl-CoA thioesters, and minimal activity toward benzoyl-holoEntB. These results were interpreted as evidence for substrate promiscuity that facilitates YbdB and YdiI evolvability, and divergence in substrate preference, which correlates with their assumed biological function. YdiI support of the menaquinone biosynthetic pathway was confirmed by demonstrating reduced anaerobic growth of the E. coli ydiI-knockout mutant (vs wild-type E. coli) on glucose in the presence of the electron acceptor fumarate. Bioinformatic analysis revealed that a small biological range exists for YbdB orthologs (i.e., limited to Enterobacteriales) relative to that of YdiI orthologs. The divergence in YbdB and YdiI substrate specificity detailed in this paper set the stage for their structural analyses reported in the companion paper.


Asunto(s)
Proteínas de Escherichia coli/química , Tioléster Hidrolasas/química , Biocatálisis , Biología Computacional , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Técnicas de Inactivación de Genes , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Mutagénesis Sitio-Dirigida , Filogenia , Conformación Proteica , Pliegue de Proteína , Especificidad por Sustrato , Tioléster Hidrolasas/genética , Tioléster Hidrolasas/metabolismo , Vitamina K 2/metabolismo
11.
Biochemistry ; 53(29): 4788-805, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25010423

RESUMEN

Herein, the structural determinants for substrate recognition and catalysis in two hotdog-fold thioesterase paralogs, YbdB and YdiI from Escherichia coli, are identified and analyzed to provide insight into the evolution of biological function in the hotdog-fold enzyme superfamily. The X-ray crystal structures of YbdB and YdiI, in complex with inert substrate analogs, determined in this study revealed the locations of the respective thioester substrate binding sites and the identity of the residues positioned for substrate binding and catalysis. The importance of each of these residues was assessed through amino acid replacements followed by steady-state kinetic analyses of the corresponding site-directed mutants. Transient kinetic and solvent (18)O-labeling studies were then carried out to provide insight into the role of Glu63 posited to function as the nucleophile or general base in catalysis. Finally, the structure-function-mechanism profiles of the two paralogs, along with that of a more distant homolog, were compared to identify conserved elements of substrate recognition and catalysis, which define the core traits of the hotdog-fold thioesterase family, as well as structural features that are unique to each thioesterase. Founded on the insight gained from this analysis, we conclude that the promiscuity revealed by in vitro substrate activity determinations, and posited to facilitate the evolution of new biological function, is the product of intrinsic plasticity in substrate binding as well as in the catalytic mechanism.


Asunto(s)
Proteínas de Escherichia coli/química , Tioléster Hidrolasas/química , Acilcoenzima A/química , Sustitución de Aminoácidos , Sitios de Unión , Biocatálisis , Cristalografía por Rayos X , Proteínas de Escherichia coli/genética , Hidrólisis , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad , Especificidad por Sustrato , Tioléster Hidrolasas/genética
12.
Proteins ; 82(9): 1896-906, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24577717

RESUMEN

In multi-domain proteins, the domains typically run end-to-end, that is, one domain follows the C-terminus of another domain. However, approximately 10% of multi-domain proteins are formed by insertion of one domain sequence into that of another domain. Detecting such insertions within protein sequences is a fundamental challenge in structural biology. The haloacid dehalogenase superfamily (HADSF) serves as a challenging model system wherein a variable cap domain (∼5-200 residues in length) accessorizes the ubiquitous Rossmann-fold core domain, with variations in insertion site and topology corresponding to different classes of cap types. Herein, we describe a comprehensive computational strategy, CapPredictor, for determining large, variable domain insertions in protein sequences. Using a novel sequence-alignment algorithm in conjunction with a structure-guided sequence profile from 154 core-domain-only structures, more than 40,000 HADSF member sequences were assigned cap types. The resulting data set afforded insight into HADSF evolution. Notably, a similar distribution of cap-type classes across different phyla was observed, indicating that all cap types existed in the last universal common ancestor. In addition, comparative analyses of the predicted cap-type and functional assignments showed that different cap types carry out similar chemistries. Thus, while cap domains play a role in substrate recognition and chemical reactivity, cap-type does not strictly define functional class. Through this example, we have shown that CapPredictor is an effective new tool for the study of form and function in protein families where domain insertion occurs.


Asunto(s)
Dominio Catalítico/genética , Hidrolasas/genética , Modelos Moleculares , Algoritmos , Secuencia de Aminoácidos , Alineación de Secuencia , Relación Estructura-Actividad
13.
Biochemistry ; 52(51): 9092-6, 2013 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-24308306

RESUMEN

Professor W. Wallace Cleland, the architect of modern steady-state enzyme kinetics, died on March 6, 2013, from injuries sustained in a fall outside of his home. He will be most remembered for giving the enzyme community Ping-Pong kinetics and the invention of dithiothreitol (DTT). He pioneered the utilization of heavy atom isotope effects for the elucidation of the chemical mechanisms of enzyme-catalyzed reactions. His favorite research journal was Biochemistry, in which he published more than 135 papers beginning in 1964 with the disclosure of DTT.


Asunto(s)
Biocatálisis , Bioquímica/historia , Enzimas/metabolismo , Cinética , Modelos Biológicos , Biocatálisis/efectos de los fármacos , Medición de Intercambio de Deuterio/historia , Ditiotreitol/química , Ditiotreitol/farmacología , Inhibidores Enzimáticos/farmacología , Enzimas/química , Historia del Siglo XX , Historia del Siglo XXI , Malato-Deshidrogenasa (NADP+)/antagonistas & inhibidores , Malato-Deshidrogenasa (NADP+)/química , Malato-Deshidrogenasa (NADP+)/metabolismo , Oxidación-Reducción/efectos de los fármacos , Filatelia/historia , Proteínas/química , Proteínas/metabolismo , Sustancias Reductoras/química , Sustancias Reductoras/farmacología , Espectrofotometría/historia , Reactivos de Sulfhidrilo/química , Reactivos de Sulfhidrilo/farmacología , Estados Unidos , Wisconsin
14.
J Org Chem ; 78(18): 9431-43, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-23992466

RESUMEN

To gain information about how alkoxy substitution in arene rings of ß-O-4 structural units within lignin governs the efficiencies/rates of radical cation C1-C2 bond cleavage reactions, single electron transfer (SET) photochemical and lignin peroxidase-catalyzed oxidation reactions of dimeric/tetrameric model compounds have been explored. The results show that the radical cations derived from less alkoxy-substituted dimeric ß-O-4 models undergo more rapid C1-C2 bond cleavage than those of more alkoxy-substituted analogues. These findings gained support from the results of DFT calculations, which demonstrate that C1-C2 bond dissociation energies of ß-O-4 radical cations decrease as the degree of alkoxy substitution decreases. In SET reactions of tetrameric compounds consisting of two ß-O-4 units, containing different degrees of alkoxy substitution, regioselective radical cation C-C bond cleavage was observed to occur in one case at the C1-C2 bond in the less alkoxy-substituted ß-O-4 moiety. However, regioselective C1-C2 cleavage in the more alkoxy-substituted ß-O-4 moiety was observed in another case, suggesting that other factors might participate in controlling this process. These observations show that lignins containing greater proportions of less rather than more alkoxylated rings as part of ß-O-4 units would be more efficiently cleaved by SET mechanisms.


Asunto(s)
Alcoholes/metabolismo , Lignina/metabolismo , Peroxidasas/metabolismo , Alcoholes/química , Biocatálisis , Transporte de Electrón , Lignina/química , Modelos Moleculares , Estructura Molecular , Oxidación-Reducción , Peroxidasas/química , Procesos Fotoquímicos
15.
Proc Natl Acad Sci U S A ; 110(36): E3381-7, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959887

RESUMEN

Although the universe of protein structures is vast, these innumerable structures can be categorized into a finite number of folds. New functions commonly evolve by elaboration of existing scaffolds, for example, via domain insertions. Thus, understanding structural diversity of a protein fold evolving via domain insertions is a fundamental challenge. The haloalkanoic dehalogenase superfamily serves as an excellent model system wherein a variable cap domain accessorizes the ubiquitous Rossmann-fold core domain. Here, we determine the impact of the cap-domain insertion on the sequence and structure divergence of the core domain. Through quantitative analysis on a unique dataset of 154 core-domain-only and cap-domain-only structures, basic principles of their evolution have been uncovered. The relationship between sequence and structure divergence of the core domain is shown to be monotonic and independent of the corresponding type of domain insert, reflecting the robustness of the Rossmann fold to mutation. However, core domains with the same cap type share greater similarity at the sequence and structure levels, suggesting interplay between the cap and core domains. Notably, results reveal that the variance in structure maps to α-helices flanking the central ß-sheet and not to the domain-domain interface. Collectively, these results hint at intramolecular coevolution where the fold diverges differentially in the context of an accessory domain, a feature that might also apply to other multidomain superfamilies.


Asunto(s)
Hidrolasas/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Evolución Molecular , Variación Genética , Hidrolasas/clasificación , Hidrolasas/genética , Modelos Moleculares , Mutagénesis Insercional , Filogenia , Análisis de Componente Principal , Pliegue de Proteína
16.
Artículo en Inglés | MEDLINE | ID: mdl-23989152

RESUMEN

The enzyme 2-keto-3-deoxy-9-O-phosphonononic acid phosphatase (KDN9P phosphatase) functions in the pathway for the production of 2-keto-3-deoxy-D-glycero-D-galacto-nononic acid, a sialic acid that is important for the survival of commensal bacteria in the human intestine. The enzyme is a member of the haloalkanoate dehalogenase superfamily and represents a good model for the active-site protonation state of family members. Crystals of approximate dimensions 1.5 × 1.0 × 1.0 mm were obtained in space group P2(1)2(1)2, with unit-cell parameters a = 83.1, b = 108.9, c = 75.7 Å. A complete neutron data set was collected from a medium-sized H/D-exchanged crystal at BIODIFF at the Heinz Maier-Leibnitz Zentrum (MLZ), Garching, Germany in 18 d. Initial refinement to 2.3 Šresolution using only neutron data showed significant density for catalytically important residues.


Asunto(s)
Proteínas Bacterianas/química , Magnesio/química , Monoéster Fosfórico Hidrolasas/química , Protones , Ácidos Siálicos/química , Proteínas Bacterianas/genética , Sitios de Unión , Dominio Catalítico , Cationes Bivalentes , Cristalografía , Medición de Intercambio de Deuterio , Escherichia coli/genética , Expresión Génica , Ligandos , Modelos Moleculares , Difracción de Neutrones , Monoéster Fosfórico Hidrolasas/genética , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Dispersión del Ángulo Pequeño , Especificidad por Sustrato
17.
Biochemistry ; 52(32): 5372-86, 2013 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-23848398

RESUMEN

The haloacid dehalogenase enzyme superfamily (HADSF) is largely composed of phosphatases that have been particularly successful at adaptating to novel biological functions relative to members of other phosphatase families. Herein, we examine the structural basis for the divergence of function in two bacterial homologues: 2-keto-3-deoxy-d-manno-octulosonate 8-phosphate phosphohydrolase (KDO8P phosphatase, KDO8PP) and 2-keto-3-deoxy-9-O-phosphonononic acid phosphohydrolase (KDN9P phosphatase, KDN9PP). KDO8PP and KDN9PP catalyze the final step in KDO and KDN synthesis, respectively, prior to transfer to CMP to form the activated sugar nucleotide. KDO8PP and KDN9PP orthologs derived from an evolutionarily diverse collection of bacterial species were subjected to steady-state kinetic analysis to determine their specificities toward catalyzed KDO8P and KDN9P hydrolysis. Although each enzyme was more active with its biological substrate, the degree of selectivity (as defined by the ratio of kcat/Km for KDO8P vs KDN9P) varied significantly. High-resolution X-ray structure determination of Haemophilus influenzae KDO8PP bound to KDO/VO3(-) and Bacteriodes thetaiotaomicron KDN9PP bound to KDN/VO3(-) revealed the substrate-binding residues. The structures of the KDO8PP and KDN9PP orthologs were also determined to reveal the differences in their active-site structures that underlie the variation in substrate preference. Bioinformatic analysis was carried out to define the sequence divergence among KDN9PP and KDO8PP orthologs. The KDN9PP orthologs were found to exist as single-domain proteins or fused with the pathway nucleotidyl transferases; the fusion of KDO8PP with the transferase is rare. The KDO8PP and KDN9PP orthologs share a stringently conserved Arg residue that forms a salt bridge with the substrate carboxylate group. The split of the KDN9PP lineage from the KDO8PP orthologs is easily tracked by the acquisition of a Glu/Lys pair that supports KDN9P binding. Moreover, independently evolved lineages of KDO8PP orthologs exist, and are separated by diffuse active-site sequence boundaries. We infer a high tolerance of the KDO8PP catalytic platform to amino acid replacements that in turn influence substrate specificity changes and thereby facilitate the divergence in biological function.


Asunto(s)
Proteínas Bacterianas/química , Hidrolasas/química , Lipopolisacáridos/biosíntesis , Ácido N-Acetilneuramínico/biosíntesis , Monoéster Fosfórico Hidrolasas/química , Proteínas Bacterianas/metabolismo , Bacteroidaceae/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Haemophilus influenzae/metabolismo , Hidrolasas/metabolismo , Cinética , Monoéster Fosfórico Hidrolasas/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato , Azúcares Ácidos/metabolismo
18.
FEBS Lett ; 587(17): 2851-9, 2013 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-23851007

RESUMEN

The function of a Bacteroidetes menaquinone biosynthetic pathway fusion protein comprised of an N-terminal haloacid dehalogenase (HAD) family domain and a C-terminal hotdog-fold family domain is described. Whereas the thioesterase domain efficiently catalyzes 1,4-dihydroxynapthoyl-CoA hydrolysis, an intermediate step in the menaquinone pathway, the HAD domain is devoid of catalytic activity. In some Bacteroidetes a homologous, catalytically active 1,4-dihydroxynapthoyl-CoA thioesterase replaces the fusion protein. Following the gene fusion event, sequence divergence resulted in a HAD domain that functions solely as the oligomerization domain of an otherwise inactive thioesterase domain.


Asunto(s)
Proteínas Bacterianas/química , Bacteroidetes/enzimología , Monoéster Fosfórico Hidrolasas/química , Proteínas Recombinantes de Fusión/química , Tioléster Hidrolasas/química , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Vías Biosintéticas , Dominio Catalítico , Evolución Molecular , Hidrolasas/química , Hidrolasas/genética , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Monoéster Fosfórico Hidrolasas/genética , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/genética , Tioléster Hidrolasas/genética , Vitamina K 2/metabolismo
19.
J Org Chem ; 78(5): 1910-22, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23094589

RESUMEN

Pyruvate phosphate dikinase (PPDK) catalyzes the phosphorylation reaction of pyruvate that forms phosphoenolpyruvate (PEP) via two partial reactions: PPDK + ATP + P(i) → PPDK-P + AMP + PP(i) and PPDK-P + pyruvate → PEP + PPDK. Based on its role in the metabolism of microbial human pathogens, PPDK is a potential drug target. A screen of substances that bind to the PPDK ATP-grasp domain active site revealed that flavone analogues are potent inhibitors of the Clostridium symbiosum PPDK. In silico modeling studies suggested that placement of a 3­6 carbon-tethered ammonium substituent at the 3'- or 4'-positions of 5,7-dihydroxyflavones would result in favorable electrostatic interactions with the PPDK Mg-ATP binding site. As a result, polymethylene-tethered amine derivatives of 5,7-dihydroxyflavones were prepared. Steady-state kinetic analysis of these substances demonstrates that the 4'-aminohexyl-5,7-dyhydroxyflavone 10 is a potent competitive PPDK inhibitor (K(i) = 1.6 ± 0.1 µM). Single turnover experiments were conducted using 4'-aminopropyl-5,7-dihydroxyflavone 7 to show that this flavone specifically targets the ATP binding site and inhibits catalysis of only the PPDK + ATP + P(i) → PPDK-P + AMP PP(i) partial reaction. Finally, the 4'-aminopbutyl-5,7-dihydroxyflavone 8 displays selectivity for inhibition of PPDK versus other enzymes that utilize ATP and NAD.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Flavonas/síntesis química , Piruvato Ortofosfato Diquinasa/antagonistas & inhibidores , Piruvato Ortofosfato Diquinasa/química , Sitios de Unión , Catálisis , Inhibidores Enzimáticos/química , Flavonas/química , Espectroscopía de Resonancia Magnética , Estructura Molecular , Fosforilación , Piruvato Ortofosfato Diquinasa/metabolismo
20.
Biochemistry ; 51(35): 6990-9, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22897136

RESUMEN

The mammalian brown fat inducible thioesterase variant 2 (BFIT2), also known as ACOT11, is a multimodular protein containing two consecutive hotdog-fold domains and a C-terminal steroidogenic acute regulatory protein-related lipid transfer domain (StarD14). In this study, we demonstrate that the N-terminal region of human BFIT2 (hBFIT2) constitutes a mitochondrial location signal sequence, which undergoes mitochondrion-dependent posttranslational cleavage. The mature hBFIT2 is shown to be located in the mitochondrial matrix, whereas the paralog "cytoplasmic acetyl-CoA hydrolase" (CACH, also known as ACOT12) was found in the cytoplasm. In vitro activity analysis of full-length hBFIT2 isolated from stably transfected HEK293 cells demonstrates selective thioesterase activity directed toward long chain fatty acyl-CoA thioesters, thus distinguishing the catalytic function of BFIT2 from that of CACH. The results from a protein-lipid overlay test indicate that the hBFIT2 StarD14 domain binds phosphatidylinositol 4-phosphate.


Asunto(s)
Mitocondrias/metabolismo , Palmitoil-CoA Hidrolasa/análisis , Palmitoil-CoA Hidrolasa/metabolismo , Tioléster Hidrolasas/análisis , Tioléster Hidrolasas/metabolismo , Acilcoenzima A/metabolismo , Secuencia de Aminoácidos , Células HEK293 , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA