Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proteins ; 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38666764

RESUMEN

Proteases that recognize linear amino acid sequences with high specificity became indispensable tools of recombinant protein technology for the removal of various fusion tags. Due to its stringent sequence specificity, the catalytic domain of the nuclear inclusion cysteine protease of tobacco etch virus (TEV PR) is also a widely applied reagent for enzymatic removal of fusion tags. For this reason, efforts have been made to improve its stability and modify its specificity. For example, P1' autoproteolytic cleavage-resistant mutant (S219V) TEV PR was found not only to be nearly impervious to self-inactivation, but also exhibited greater stability and catalytic efficiency than the wild-type enzyme. An R203G substitution has been reported to further relax the P1' specificity of the enzyme, however, these results were obtained from crude intracellular assays. Until now, there has been no rigorous comparison of the P1' specificity of the S219V and S219V/R203G mutants in vitro, under carefully controlled conditions. Here, we compare the P1' amino acid preferences of these single and double TEV PR mutants. The in vitro analysis was performed by using recombinant protein substrates representing 20 P1' variants of the consensus TENLYFQ*SGT cleavage site, and synthetic oligopeptide substrates were also applied to study a limited set of the most preferred variants. In addition, the enzyme-substrate interactions were analyzed in silico. The results indicate highly similar P1' preferences for both enzymes, many side-chains can be accommodated by the S1' binding sites, but the kinetic assays revealed lower catalytic efficiency for the S219V/R203G than for the S219V mutant.

2.
J Struct Biol ; 214(4): 107918, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36343842

RESUMEN

C/EBPß is a key regulator of numerous cellular processes, but it can also contribute to tumorigenesis and viral diseases. It binds to specific DNA sequences (C/EBP sites) and interacts with other transcription factors to control expression of multiple eukaryotic genes in a tissue and cell-type dependent manner. A body of evidence has established that cell-type-specific regulatory information is contained in the local DNA sequence of the binding motif. In human epithelial cells, C/EBPß is an essential cofactor for TGFß signaling in the case of Smad2/3/4 and FoxO-dependent induction of the cell cycle inhibitor, p15INK4b. In the TGFß-responsive region 2 of the p15INK4b promoter, the Smad binding site is flanked by a C/EBP site, CTTAA•GAAAG, which differs from the canonical, palindromic ATTGC•GCAAT motif. The X-ray crystal structure of C/EBPß bound to the p15INK4b promoter fragment shows how GCGC-to-AAGA substitution generates changes in the intermolecular interactions in the protein-DNA interface that enhances C/EBPß binding specificity, limits possible epigenetic regulation of the promoter, and generates a DNA element with a unique pattern of methyl groups in the major groove. Significantly, CT/GA dinucleotides located at the 5'ends of the double stranded element maintain local narrowing of the DNA minor groove width that is necessary for DNA recognition. Our results suggest that C/EBPß would accept all forms of modified cytosine in the context of the CpT site. This contrasts with the effect on the consensus motif, where C/EBPß binding is modestly increased by cytosine methylation, but substantially decreased by hydroxymethylation.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT , Epigénesis Genética , Humanos , Proteína beta Potenciadora de Unión a CCAAT/genética , Ciclo Celular , Citosina , ADN/genética
3.
Int J Mol Sci ; 23(19)2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36232729

RESUMEN

ATP-dependent Lon proteases are key participants in the quality control system that supports the homeostasis of the cellular proteome. Based on their unique structural and biochemical properties, Lon proteases have been assigned in the MEROPS database to three subfamilies (A, B, and C). All Lons are single-chain, multidomain proteins containing an ATPase and protease domains, with different additional elements present in each subfamily. LonA and LonC proteases are soluble cytoplasmic enzymes, whereas LonBs are membrane-bound. Based on an analysis of the available sequences of Lon proteases, we identified a number of enzymes currently assigned to the LonB subfamily that, although presumably membrane-bound, include structural features more similar to their counterparts in the LonA subfamily. This observation was confirmed by the crystal structure of the proteolytic domain of the enzyme previously assigned as Bacillus subtilis LonB, combined with the modeled structure of its ATPase domain. Several structural features present in both domains differ from their counterparts in either LonA or LonB subfamilies. We thus postulate that this enzyme is the founding member of a newly identified LonBA subfamily, so far found only in the gene sequences of firmicutes.


Asunto(s)
Proteasa La , Proteasas ATP-Dependientes/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Péptido Hidrolasas/metabolismo , Proteasa La/genética , Proteasa La/metabolismo , Proteoma/metabolismo
4.
Nucleic Acids Res ; 47(19): 10134-10150, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31199869

RESUMEN

Tyrosyl DNA-phosphodiesterase I (TDP1) repairs type IB topoisomerase (TOP1) cleavage complexes generated by TOP1 inhibitors commonly used as anticancer agents. TDP1 also removes DNA 3' end blocking lesions generated by chain-terminating nucleosides and alkylating agents, and base oxidation both in the nuclear and mitochondrial genomes. Combination therapy with TDP1 inhibitors is proposed to synergize with topoisomerase targeting drugs to enhance selectivity against cancer cells exhibiting deficiencies in parallel DNA repair pathways. A crystallographic fragment screening campaign against the catalytic domain of TDP1 was conducted to identify new lead compounds. Crystal structures revealed two fragments that bind to the TDP1 active site and exhibit inhibitory activity against TDP1. These fragments occupy a similar position in the TDP1 active site as seen in prior crystal structures of TDP1 with bound vanadate, a transition state mimic. Using structural insights into fragment binding, several fragment derivatives have been prepared and evaluated in biochemical assays. These results demonstrate that fragment-based methods can be a highly feasible approach toward the discovery of small-molecule chemical scaffolds to target TDP1, and for the first time, we provide co-crystal structures of small molecule inhibitors bound to TDP1, which could serve for the rational development of medicinal TDP1 inhibitors.


Asunto(s)
Inhibidores Enzimáticos/química , Ligandos , Hidrolasas Diéster Fosfóricas/química , Conformación Proteica , Secuencia de Bases , Dominio Catalítico/genética , Cristalografía , Reparación del ADN/genética , Histidina/análogos & derivados , Histidina/química , Histidina/aislamiento & purificación , Humanos , Modelos Moleculares , Hidrolasas Diéster Fosfóricas/genética , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/química
5.
Int J Mol Sci ; 21(21)2020 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-33171880

RESUMEN

Protein splicing catalyzed by inteins utilizes many different combinations of amino-acid types at active sites. Inteins have been classified into three classes based on their characteristic sequences. We investigated the structural basis of the protein splicing mechanism of class 3 inteins by determining crystal structures of variants of a class 3 intein from Mycobacterium chimaera and molecular dynamics simulations, which suggested that the class 3 intein utilizes a different splicing mechanism from that of class 1 and 2 inteins. The class 3 intein uses a bond cleavage strategy reminiscent of proteases but share the same Hedgehog/INTein (HINT) fold of other intein classes. Engineering of class 3 inteins from a class 1 intein indicated that a class 3 intein would unlikely evolve directly from a class 1 or 2 intein. The HINT fold appears as structural and functional solution for trans-peptidyl and trans-esterification reactions commonly exploited by diverse mechanisms using different combinations of amino-acid types for the active-site residues.


Asunto(s)
Proteínas Hedgehog/fisiología , Inteínas/fisiología , Empalme de Proteína/fisiología , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Proteínas Hedgehog/genética , Inteínas/genética , Simulación de Dinámica Molecular , Mycobacterium/genética , Mycobacterium/metabolismo , Empalme de Proteína/genética , Empalme del ARN/fisiología
6.
Angew Chem Int Ed Engl ; 55(19): 5703-7, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27038327

RESUMEN

Conjugation of the small ubiquitin-like modifier (SUMO) to protein substrates is an important disease-associated posttranslational modification, although few inhibitors of this process are known. Herein, we report the discovery of an allosteric small-molecule binding site on Ubc9, the sole SUMO E2 enzyme. An X-ray crystallographic screen was used to identify two distinct small-molecule fragments that bind to Ubc9 at a site distal to its catalytic cysteine. These fragments and related compounds inhibit SUMO conjugation in biochemical assays with potencies of 1.9-5.8 mm. Mechanistic and biophysical analyses, coupled with molecular dynamics simulations, point toward ligand-induced rigidification of Ubc9 as a mechanism of inhibition.


Asunto(s)
Enzimas Ubiquitina-Conjugadoras/metabolismo , Regulación Alostérica , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Ligandos , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Especificidad por Sustrato , Sumoilación , Resonancia por Plasmón de Superficie , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/genética
7.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 5): 1102-11, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25945576

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) is a highly pathogenic virus that causes severe respiratory illness accompanied by multi-organ dysfunction, resulting in a case fatality rate of approximately 40%. As found in other coronaviruses, the majority of the positive-stranded RNA MERS-CoV genome is translated into two polyproteins, one created by a ribosomal frameshift, that are cleaved at three sites by a papain-like protease and at 11 sites by a 3C-like protease (3 CL(pro)). Since 3 CL(pro) is essential for viral replication, it is a leading candidate for therapeutic intervention. To accelerate the development of 3 CL(pro) inhibitors, three crystal structures of a catalytically inactive variant (C148A) of the MERS-CoV 3 CL(pro) enzyme were determined. The aim was to co-crystallize the inactive enzyme with a peptide substrate. Fortuitously, however, in two of the structures the C-terminus of one protomer is bound in the active site of a neighboring molecule, providing a snapshot of an enzyme-product complex. In the third structure, two of the three protomers in the asymmetric unit form a homodimer similar to that of SARS-CoV 3 CL(pro); however, the third protomer adopts a radically different conformation that is likely to correspond to a crystallographic monomer, indicative of substantial structural plasticity in the enzyme. The results presented here provide a foundation for the structure-based design of small-molecule inhibitors of the MERS-CoV 3 CL(pro) enzyme.


Asunto(s)
Antivirales/farmacología , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Inhibidores Enzimáticos/farmacología , Coronavirus del Síndrome Respiratorio de Oriente Medio/enzimología , Secuencia de Aminoácidos , Dominio Catalítico , Proteasas 3C de Coronavirus , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
8.
J Mol Biol ; 435(6): 168012, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36792007

RESUMEN

The Venezuelan equine encephalitis virus (VEEV) belongs to the Togaviridae family and is pathogenic to both humans and equines. The VEEV non-structural protein 2 (nsP2) is a cysteine protease (nsP2pro) that processes the polyprotein and thus it is a drug target for inhibitor discovery. The atomic structure of the VEEV nsP2 catalytic domain was previously characterized by both X-ray crystallography and computational studies. A modified nsP2pro harboring a N475A mutation in the N terminus was observed to exhibit an unexpected conformation: the N-terminal residues bind to the active site, mimicking binding of a substrate. The large conformational change of the N terminus was assumed to be induced by the N475A mutation, as N475 has an important role in stabilization of the N terminus and the active site. This conformation was first observed in the N475A mutant, but we also found it while determining a crystal structure of the catalytically active nsP2pro containing the wild-type N475 active site residue and K741A/K767A surface entropy reduction mutations. This suggests that the N475A mutation is not a prerequisite for self-inhibition. Here, we describe a high resolution (1.46 Å) crystal structure of a truncated nsP2pro (residues 463-785, K741A/K767A) and analyze the structure further by molecular dynamics to study the active and self-inhibited conformations of nsP2pro and its N475A mutant. A comparison of the different conformations of the N-terminal residues sheds a light on the interactions that play an important role in the stabilization of the enzyme.


Asunto(s)
Dominio Catalítico , Proteasas de Cisteína , Virus de la Encefalitis Equina Venezolana , Animales , Humanos , Cristalografía por Rayos X , Proteasas de Cisteína/química , Proteasas de Cisteína/genética , Virus de la Encefalitis Equina Venezolana/enzimología , Caballos , Simulación de Dinámica Molecular
9.
RSC Chem Biol ; 4(5): 334-343, 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37181631

RESUMEN

Tyrosyl-DNA phosphodiesterase 1 (TDP1) is a member of the phospholipase D family that can downregulate the anticancer effects of the type I topoisomerase (TOP1) inhibitors by hydrolyzing the 3'-phosphodiester bond between DNA and the TOP1 residue Y723 in the critical stalled intermediate that is the foundation of TOP1 inhibitor mechanism of action. Thus, TDP1 antagonists are attractive as potential enhancers of TOP1 inhibitors. However, the open and extended nature of the TOP1-DNA substrate-binding region has made the development of TDP1 inhibitors extremely challenging. In this study, starting from our recently identified small molecule microarray (SMM)-derived TDP1-inhibitory imidazopyridine motif, we employed a click-based oxime protocol to extend the parent platform into the DNA and TOP1 peptide substrate-binding channels. We applied one-pot Groebke-Blackburn-Bienayme multicomponent reactions (GBBRs) to prepare the needed aminooxy-containing substrates. By reacting these precursors with approximately 250 aldehydes in microtiter format, we screened a library of nearly 500 oximes for their TDP1 inhibitory potencies using an in vitro florescence-based catalytic assay. Select hits were structurally explored as their triazole- and ether-based isosteres. We obtained crystal structures of two of the resulting inhibitors bound to the TDP1 catalytic domain. The structures reveal that the inhibitors form hydrogen bonds with the catalytic His-Lys-Asn triads ("HKN" motifs: H263, K265, N283 and H493, K495, N516), while simultaneously extending into both the substrate DNA and TOP1 peptide-binding grooves. This work provides a structural model for developing multivalent TDP1 inhibitors capable of binding in a tridentate fashion with a central component situated within the catalytic pocket and extensions that project into both the DNA and TOP1 peptide substrate-binding regions.

10.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 3): 201-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22349221

RESUMEN

The Yersinia pestis YscD protein is an essential component of the type III secretion system. YscD consists of an N-terminal cytoplasmic domain (residues 1-121), a transmembrane linker (122-142) and a large periplasmic domain (143-419). Both the cytoplasmic and the periplasmic domains are required for the assembly of the type III secretion system. Here, the structure of the YscD cytoplasmic domain solved by SAD phasing is presented. Although the three-dimensional structure is similar to those of forkhead-associated (FHA) domains, comparison with the structures of canonical FHA domains revealed that the cytoplasmic domain of YscD lacks the conserved residues that are required for binding phosphothreonine and is therefore unlikely to function as a true FHA domain.


Asunto(s)
Proteínas Bacterianas/química , Proteínas de la Membrana/química , Yersinia pestis/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia , Análisis de Secuencia de Proteína , Homología Estructural de Proteína , Relación Estructura-Actividad , Yersinia pestis/metabolismo
11.
Front Chem ; 10: 910953, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36051621

RESUMEN

Tyrosyl-DNA phosphodiesterase 1 (TDP1) repairs stalled type I topoisomerase (TOP1)-DNA complexes by hydrolyzing the phosphodiester bond between the TOP1 Y723 residue and the 3'-phosphate of its DNA substrate. Although TDP1 antagonists could potentially reduce the dose of TOP1 inhibitors needed to achieve effective anticancer effects, the development of validated TDP1 inhibitors has proven to be challenging. This may, in part, be due to the open and extended nature of the TOP1 substrate binding region. We have previously reported imidazopyrazines and imidazopyridines that can inhibit TDP1 catalytic function in vitro. We solved the TDP1 crystal structures with bound inhibitors of this class and found that the dicarboxylic acid functionality within the N-(3,4-dicarboxyphenyl)-2-diphenylimidazo [1,2-a]pyridin-3-amine platform overlaps with aspects of phosphoryl substrate recognition. Yet phosphonic acids could potentially better-replicate cognate TOP1-DNA substrate binding interactions than carboxylic acids. As reported herein, we designed phosphonic acid-containing variants of our previously reported carboxylic acid-containing imidazopyrazine and imidazopyridine inhibitors and effected their synthesis using one-pot Groebke-Blackburn-Bienayme multicomponent reactions. We obtained crystal structures of TDP1 complexed with a subset of inhibitors. We discuss binding interactions of these inhibitors within the context of phosphate-containing substrate and carboxylic acid-based inhibitors. These compounds represent a new structural class of small molecule ligands that mimic aspects of the 3'-processed substrate that results from TDP1 catalysis.

12.
J Struct Biol ; 176(3): 292-301, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21963792

RESUMEN

Chk2 (checkpoint kinase 2) is a serine/threonine kinase that participates in a series of signaling networks responsible for maintaining genomic integrity and responding to DNA damage. The development of selective Chk2 inhibitors has recently attracted much interest as a means of sensitizing cancer cells to current DNA-damaging agents used in the treatment of cancer. Additionally, selective Chk2 inhibitors may reduce p53-mediated apoptosis in normal tissues, thereby helping to mitigate adverse side effects from chemotherapy and radiation. Thus far, relatively few selective inhibitors of Chk2 have been described and none have yet progressed into clinical trials. Here, we report crystal structures of the catalytic domain of Chk2 in complex with a novel series of potent and selective small molecule inhibitors. These compounds exhibit nanomolar potencies and are selective for Chk2 over Chk1. The structures reported here elucidate the binding modes of these inhibitors to Chk2 and provide information that can be exploited for the structure-assisted design of novel chemotherapeutics.


Asunto(s)
Antineoplásicos/química , Dominio Catalítico , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Sitios de Unión , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Cristalografía por Rayos X , Humanos , Estructura Molecular , Terapia Molecular Dirigida , Unión Proteica , Proteínas Quinasas/química
13.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 5): 471-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21543850

RESUMEN

There are over 100 genes in the human genome that encode protein tyrosine phosphatases (PTPs) and approximately 60 of these are classified as dual-specificity phosphatases (DUSPs). Although many dual-specificity phosphatases are still not well characterized, novel functions have been discovered for some of them that have led to new insights into a variety of biological processes and the molecular basis for certain diseases. Indeed, as the functions of DUSPs continue to be elucidated, a growing number of them are emerging as potential therapeutic targets for diseases such as cancer, diabetes and inflammatory disorders. Here, the overexpression, purification and structure determination of DUSP27 at 2.38 Šresolution are presented.


Asunto(s)
Fosfoproteínas Fosfatasas/química , Dominio Catalítico , Clonación Molecular , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/aislamiento & purificación , Homología Estructural de Proteína , Regulación hacia Arriba
14.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 7): 639-45, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21697602

RESUMEN

Isothiazolidinone (IZD) heterocycles can act as effective components of protein tyrosine phosphatase (PTP) inhibitors by simultaneously replicating the binding interactions of both a phosphoryl group and a highly conserved water molecule, as exemplified by the structures of several PTP1B-inhibitor complexes. In the first unambiguous demonstration of IZD interactions with a PTP other than PTP1B, it is shown by X-ray crystallography that the IZD motif binds within the catalytic site of the Yersinia pestis PTP YopH by similarly displacing a highly conserved water molecule. It is also shown that IZD-based bidentate ligands can inhibit YopH in a nonpromiscuous fashion at low micromolar concentrations. Hence, the IZD moiety may represent a useful starting point for the development of YopH inhibitors.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Materiales Biomiméticos/química , Inhibidores de Proteínas Quinasas/química , Proteínas Tirosina Fosfatasas/química , Tiazoles/química , Yersinia pestis/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Materiales Biomiméticos/metabolismo , Cristalografía por Rayos X , Modelos Moleculares , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Relación Estructura-Actividad , Tiazoles/antagonistas & inhibidores , Tiazoles/metabolismo , Yersinia pestis/metabolismo
15.
PLoS One ; 16(3): e0248385, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33784308

RESUMEN

N-hydroxylating flavin-dependent monooxygenases (FMOs) are involved in the biosynthesis of hydroxamate siderophores, playing a key role in microbial virulence. Herein, we report the first structural and kinetic characterization of a novel alkyl diamine N-hydroxylase DesB from Streptomyces sviceus (SsDesB). This enzyme catalyzes the first committed step in the biosynthesis of desferrioxamine B, a clinical drug used to treat iron overload disorders. X-ray crystal structures of the SsDesB holoenzyme with FAD and the ternary complex with bound NADP+ were solved at 2.86 Å and 2.37 Å resolution, respectively, providing a structural view of the active site environment. SsDesB crystallized as a tetramer and the structure of the individual protomers closely resembles the structures of homologous N-hydroxylating FMOs from Erwinia amylovora (DfoA), Pseudomonas aeruginosa (PvdA), and Aspergillus fumigatus (SidA). Using NADPH oxidation, oxygen consumption, and product formation assays, kinetic parameters were determined for various substrates with SsDesB. SsDesB exhibited typical saturation kinetics with substrate inhibition at high concentrations of NAD(P)H as well as cadaverine. The apparent kcat values for NADPH in steady-state NADPH oxidation and oxygen consumption assays were 0.28 ± 0.01 s-1 and 0.24 ± 0.01 s-1, respectively. However, in product formation assays used to measure the rate of N-hydroxylation, the apparent kcat for NADPH (0.034 ± 0.008 s-1) was almost 10-fold lower under saturating FAD and cadaverine concentrations, reflecting an uncoupled reaction, and the apparent NADPH KM was 33 ± 24 µM. Under saturating FAD and NADPH concentrations, the apparent kcat and KM for cadaverine in Csaky assays were 0.048 ± 0.004 s-1 and 19 ± 9 µM, respectively. SsDesB also N-hydroxylated putrescine, spermidine, and L-lysine substrates but not alkyl (di)amines that were branched or had fewer than four methylene units in an alkyl chain. These data demonstrate that SsDesB has wider substrate scope compared to other well-studied ornithine and lysine N-hydroxylases, making it an amenable biocatalyst for the production of desferrioxamine B, derivatives, and other N-substituted products.


Asunto(s)
Proteínas Bacterianas/metabolismo , Cadaverina/metabolismo , Deferoxamina/metabolismo , Oxigenasas de Función Mixta/biosíntesis , Sideróforos/biosíntesis , Streptomyces/enzimología , Biocatálisis , Dominio Catalítico , Dinitrocresoles/metabolismo , Flavina-Adenina Dinucleótido/metabolismo , Flavinas/metabolismo , Holoenzimas/metabolismo , Hidroxilación , Cinética , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/metabolismo , NADP/metabolismo , Ornitina/metabolismo , Oxidación-Reducción
16.
Chem Sci ; 12(11): 3876-3884, 2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-34163656

RESUMEN

Tyrosyl-DNA phosphodiesterase 1 (TDP1) is a member of the phospholipase D family of enzymes, which catalyzes the removal of both 3'- and 5'-DNA phosphodiester adducts. Importantly, it is capable of reducing the anticancer effects of type I topoisomerase (TOP1) inhibitors by repairing the stalled covalent complexes of TOP1 with DNA. It achieves this by promoting the hydrolysis of the phosphodiester bond between the Y723 residue of human TOP1 and the 3'-phosphate of its DNA substrate. Blocking TDP1 function is an attractive means of enhancing the efficacy of TOP1 inhibitors and overcoming drug resistance. Previously, we reported the use of an X-ray crystallographic screen of more than 600 fragments to identify small molecule variations on phthalic acid and hydroxyquinoline motifs that bind within the TDP1 catalytic pocket. Yet, the majority of these compounds showed limited (millimolar) TDP1 inhibitory potencies. We now report examining a 21 000-member library of drug-like Small Molecules in Microarray (SMM) format for their ability to bind Alexa Fluor 647 (AF647)-labeled TDP1. The screen identified structurally similar N,2-diphenylimidazo[1,2-a]pyrazin-3-amines as TDP1 binders and catalytic inhibitors. We then explored the core heterocycle skeleton using one-pot Groebke-Blackburn-Bienayme multicomponent reactions and arrived at analogs having higher inhibitory potencies. Solving TDP1 co-crystal structures of a subset of compounds showed their binding at the TDP1 catalytic site, while mimicking substrate interactions. Although our original fragment screen differed significantly from the current microarray protocol, both methods identified ligand-protein interactions containing highly similar elements. Importantly inhibitors identified through the SMM approach show competitive inhibition against TDP1 and access the catalytic phosphate-binding pocket, while simultaneously providing extensions into both the substrate DNA and peptide-binding channels. As such, they represent a platform for further elaboration of trivalent ligands, that could serve as a new genre of potent TDP1 inhibitors.

17.
Bioorg Med Chem Lett ; 20(9): 2813-6, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20350805

RESUMEN

A bivalent tethered approach toward YopH inhibitor development is presented that joins aldehydes with mixtures of bis-aminooxy-containing linkers using oxime coupling. The methodology is characterized by its facility and ease of use and its ability to rapidly identify low micromolar affinity inhibitors. The generality of the approach may potentially make it amenable to the development of bivalent inhibitors directed against other phosphatases.


Asunto(s)
Aldehídos/química , Proteínas de la Membrana Bacteriana Externa/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Oximas/química , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Yersinia pestis/enzimología , Aldehídos/síntesis química , Aldehídos/farmacología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Proteínas Tirosina Fosfatasas/metabolismo , Bibliotecas de Moléculas Pequeñas
18.
Bioorg Med Chem ; 18(12): 4535-41, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20472436

RESUMEN

The current paper presents the synthesis and evaluation of a series of maleimides that were designed to inhibit the Cdc25 phosphatase by alkylation of catalytically essential cysteine residues. Although in HepB3 cell culture assays the analogues did exhibit antiproliferative IC(50) values ranging from sub-micromolar to greater than 100 microM, inhibition of Cdc25 through cysteine alkylation could not be demonstrated. It was also found that analysis using fluorescence activated cell sorting (FACS) following treatment with the most potent analogue (1t) did not provide data consistent with inhibition at one specific point in the cell cycle, as would be expected if Cdc25A were inhibited. Further studies with a subset of analogues resulted in a correlation of antiproliferative potencies with activation of the unfolded protein response (UPR). The UPR is a regulatory pathway that temporarily suspends protein production when misfolding of proteins occurs within the endoplastic reticulum (ER). In addition, ER chaperones that promote proper refolding become up-regulated. If cellular damage cannot be resolved by these mechanisms, then the UPR can initiate apoptosis. The current study indicates that these maleimide analogues lead to UPR activation, which is predictive of the selective antiproliferative activity of the series.


Asunto(s)
Antineoplásicos Alquilantes/química , Maleimidas/química , Respuesta de Proteína Desplegada , Antineoplásicos Alquilantes/síntesis química , Antineoplásicos Alquilantes/farmacología , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Humanos , Maleimidas/síntesis química , Maleimidas/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Fosfatasas cdc25/metabolismo
19.
Appl Microbiol Biotechnol ; 88(1): 117-24, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20585771

RESUMEN

The objective of this study was to use protein engineering techniques to enhance the catalytic activity of glycerol dehydrogenase (GlyDH) on racemic 1, 3-butanediol (1, 3-BDO) for the bioproduction of the important pharmaceutical intermediate 4-hydroxy-2-butanone. Three GlyDH genes (gldA) from Escherichia coli K-12, Salmonella enterica, and Klebsiella pneumoniae MGH78578 were shuffled to generate a random mutagenesis library. The nitroblue tetrazolium/phenazine methosulfate high throughput screening protocol was used to select four chimeric enzymes with up to a 2.6-fold improved activity towards 1, 3-BDO. A rational design method was also employed to further improve the enzyme activity after DNA shuffling. Based on the homology model of GlyDH (Escherichia coli), Asp121 was predicted to influence 1, 3-BDO binding and replaced with Ala by site-directed mutagenesis. Combination of the mutations from both DNA shuffling and rational design produced the best mutant with a V (max) value of 126.6 U/mg, a 26-fold activity increase compared with that of the wild type GlyDH from E. coli.


Asunto(s)
Butileno Glicoles/metabolismo , Escherichia coli K12/enzimología , Hidroxibutiratos/metabolismo , Klebsiella pneumoniae/enzimología , Ingeniería de Proteínas , Salmonella enterica/enzimología , Deshidrogenasas del Alcohol de Azúcar/metabolismo , Barajamiento de ADN , ADN Bacteriano/genética , Escherichia coli K12/genética , Klebsiella pneumoniae/genética , Metosulfato de Metilfenazonio/metabolismo , Nitroazul de Tetrazolio/metabolismo , Salmonella enterica/genética , Deshidrogenasas del Alcohol de Azúcar/genética
20.
Biochemistry ; 48(4): 720-8, 2009 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-19133805

RESUMEN

Flavin C4a-OO(H) and C4a-OH adducts are critical intermediates proposed in many flavoenzyme reaction mechanisms, but they are rarely detected even by rapid transient kinetics methods. We observe a trapped flavin C4a-OH or C4a-OO(H) adduct by single-crystal spectroscopic methods and in the 1.86 A resolution X-ray crystal structure of choline oxidase. The microspectrophotometry results show that the adduct forms rapidly in situ at 100 K upon exposure to X-rays. Density functional theory calculations establish the electronic structures for the flavin C4a-OH and C4a-OO(H) adducts and estimate the stabilization energy of several active site hydrogen bonds deduced from the crystal structure. We propose that the enzyme-bound FAD is reduced in the X-ray beam. The aerobic crystals then form either a C4a-OH or C4a-OO(H) adduct, but an insufficient proton inventory prevents their decay at cryogenic temperatures.


Asunto(s)
Oxidorreductasas de Alcohol/química , Biología Computacional , Flavina-Adenina Dinucleótido/química , Flavoproteínas/química , Oxígeno/química , Oxidorreductasas de Alcohol/análisis , Oxidorreductasas de Alcohol/metabolismo , Arthrobacter/enzimología , Proteínas Bacterianas/análisis , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión/fisiología , Biología Computacional/métodos , Cristalografía por Rayos X/métodos , Flavina-Adenina Dinucleótido/análisis , Flavina-Adenina Dinucleótido/metabolismo , Flavinas/análisis , Flavinas/química , Flavinas/metabolismo , Flavoproteínas/análisis , Flavoproteínas/metabolismo , Oxígeno/análisis , Oxígeno/metabolismo , Estructura Secundaria de Proteína/fisiología , Espectrometría por Rayos X/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA