Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 347
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pharm Res ; 39(11): 2951-2963, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35799081

RESUMEN

Lipocalin-type prostaglandin D synthase was previously known as ß-trace protein (BTP), a low-molecular-weight glycoprotein that is heavily expressed in human cerebrospinal fluid. Nevertheless, it is also seen to be expressed in numerous other tissues including the kidney, liver, lung, heart, adipose, muscle, and pancreas. Functionally, L-PGDS behaves like a lipocalin type protein where it helps in binding and transportation of small lipophilic substances, such as steroids, retinoids, and other lipophilic ligands. Enzymatically, L-PGDS functions as a prostaglandin synthase where it helps in the production of PGD2 by catalyzing the isomerization of PGH2, a common precursor of the two series of prostaglandins. PGD2 regulates its physiological function through two individual receptors named DP1 and DP2. L-PGDS has been a central player in many diseases, its role in metabolism including diabetes, fatty liver disease, and obesity has gathered a large attention. In this review, we summarize the current state of knowledge about L-PGDS and it's signaling in adipose, hepatic, skeletal muscle, and pancreas tissues, which are core targets for metabolic studies. Modulation of L-PGDS signaling can be considered as a potential future therapeutic target for the treatment of insulin resistance as well as fatty liver disease.


Asunto(s)
Hepatopatías , Prostaglandina D2 , Humanos , Prostaglandina D2/metabolismo , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/química , Lipocalinas/metabolismo
2.
J Biol Chem ; 295(3): 850-867, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31811089

RESUMEN

Human macrophage migration-inhibitory factor (MIF) is an evolutionarily-conserved protein that has both extracellular immune-modulating and intracellular cell-regulatory functions. MIF plays a role in various diseases, including inflammatory diseases, atherosclerosis, autoimmunity, and cancer. It serves as an inflammatory cytokine and chemokine, but also exhibits enzymatic activity. Secreted MIF binds to cell-surface immune receptors such as CD74 and CXCR4. Plants possess MIF orthologs but lack the associated receptors, suggesting functional diversification across kingdoms. Here, we characterized three MIF orthologs (termed MIF/d-dopachrome tautomerase-like proteins or MDLs) of the model plant Arabidopsis thaliana Recombinant Arabidopsis MDLs (AtMDLs) share similar secondary structure characteristics with human MIF, yet only have minimal residual tautomerase activity using either p-hydroxyphenylpyruvate or dopachrome methyl ester as substrate. Site-specific mutagenesis suggests that this is due to a distinct amino acid difference at the catalytic cavity-defining residue Asn-98. Surprisingly, AtMDLs bind to the human MIF receptors CD74 and CXCR4. Moreover, they activate CXCR4-dependent signaling in a receptor-specific yeast reporter system and in CXCR4-expressing human HEK293 transfectants. Notably, plant MDLs exert dose-dependent chemotactic activity toward human monocytes and T cells. A small molecule MIF inhibitor and an allosteric CXCR4 inhibitor counteract this function, revealing its specificity. Our results indicate cross-kingdom conservation of the receptor signaling and leukocyte recruitment capacities of human MIF by its plant orthologs. This may point toward a previously unrecognized interplay between plant proteins and the human innate immune system.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Histocompatibilidad Clase II/genética , Inmunidad Innata/genética , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Receptores CXCR4/genética , Antígenos de Diferenciación de Linfocitos B/química , Arabidopsis/genética , Arabidopsis/inmunología , Quimiotaxis/genética , Quimiotaxis/inmunología , Secuencia Conservada/genética , Secuencia Conservada/inmunología , Citocinas/genética , Citocinas/inmunología , Células HEK293 , Antígenos de Histocompatibilidad Clase II/química , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/inmunología , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/inmunología , Monocitos/química , Monocitos/metabolismo , Unión Proteica/genética , Receptores CXCR4/química , Homología de Secuencia , Linfocitos T/química , Linfocitos T/metabolismo
3.
Biochem Biophys Res Commun ; 569: 66-71, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34237429

RESUMEN

Prostaglandin D2 (PGD2), an endogenous somnogen, is a unique PG that is secreted into the cerebrospinal fluid. PGD2 is a relatively fragile molecule and should be transported to receptors localized in the basal forebrain without degradation. However, it remains unclear how PGD2 is stably carried to such remote receptors. Here, we demonstrate that the PGD2-synthesizing enzyme, Lipocalin-type prostaglandin D synthase (L-PGDS), binds not only its substrate PGH2 but also its product PGD2 at two distinct binding sites for both ligands. This behaviour implys its PGD2 carrier function. Nevertheless, since the high affinity (Kd = âˆ¼0.6 µM) of PGD2 in the catalytic binding site is comparable to that of PGH2, it may act as a competitive inhibitor, while our binding assay exhibits only weak inhibition (Ki = 189 µM) of the catalytic reaction. To clarify this enigmatic behavior, we determined the solution structure of L-PGDS bound to one substrate analog by NMR and compared it with the two structures: one in the apo form and the other in substrate analogue complex with 1:2 stoichiometry. The structural comparisons showed clearly that open or closed forms of loops at the entrance of ligand binding cavity are regulated by substrate binding to two sites, and that the binding to a second non-catalytic binding site, which apparently substrate concentration dependent, induces opening of the cavity that releases the product. From these results, we propose that L-PGDS is a unique enzyme having a carrier function and a substrate-induced product-release mechanism.


Asunto(s)
Dominio Catalítico , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Prostaglandina D2/metabolismo , Prostaglandina H2/metabolismo , Animales , Sitios de Unión , Biocatálisis , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/genética , Cinética , Lipocalinas/química , Lipocalinas/genética , Espectroscopía de Resonancia Magnética , Ratones , Estructura Molecular , Mutación , Prostaglandina D2/química , Prostaglandina H2/química , Unión Proteica , Conformación Proteica , Especificidad por Sustrato
4.
Bioorg Med Chem Lett ; 49: 128284, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34311085

RESUMEN

Jasmonic acid (JA) is a plant hormone involved in the defense response against insects and fungi. JA is synthesized from α-linolenic acid (LA) by the octadecanoid pathway in plants. 12-oxo-Phytodienoic acid (OPDA) is one of the biosynthetic intermediates in this pathway. The reported stereo selective total synthesis of cis-(+)-OPDA is not very efficient due to the many steps involved in the reaction as well as the use of water sensitive reactions. Therefore, we developed an enzymatic method for the synthesis of OPDA using acetone powder of flax seed and allene oxide cyclase (PpAOC2) from Physcomitrella patens. From this method, natural cis-(+)-OPDA can be synthesized in the high yield of approximately 40%. In this study, we investigated the substrate specificity of the enzymatic synthesis of other OPDA analogs with successions to afford OPDA amino acid conjugates, dinor-OPDA (dn-OPDA), and OPDA monoglyceride, and it was suggested that the biosynthetic pathway of arabidopsides could occur via MGDG.


Asunto(s)
Ácidos Grasos Insaturados/síntesis química , Oxidorreductasas Intramoleculares/química , Proteínas de Plantas/química , Bryopsida/enzimología , Lino/enzimología , Semillas/enzimología , Estereoisomerismo
5.
Angew Chem Int Ed Engl ; 60(32): 17514-17521, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34018657

RESUMEN

Macrophage migration inhibitory factor (MIF) is involved in protein-protein interactions that play key roles in inflammation and cancer. Current strategies to develop small molecule modulators of MIF functions are mainly restricted to the MIF tautomerase active site. Here, we use this site to develop proteolysis targeting chimera (PROTAC) in order to eliminate MIF from its protein-protein interaction network. We report the first potent MIF-directed PROTAC, denoted MD13, which induced almost complete MIF degradation at low micromolar concentrations with a DC50 around 100 nM in A549 cells. MD13 suppresses the proliferation of A549 cells, which can be explained by deactivation of the MAPK pathway and subsequent induction of cell cycle arrest at the G2/M phase. MD13 also exhibits antiproliferative effect in a 3D tumor spheroid model. In conclusion, we describe the first MIF-directed PROTAC (MD13) as a research tool, which also demonstrates the potential of PROTACs in cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Benzoxazinas/farmacología , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ftalimidas/farmacología , Células A549 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos/síntesis química , Benzoxazinas/síntesis química , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Oxidorreductasas Intramoleculares/química , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/química , Ftalimidas/síntesis química , Proteolisis/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo
6.
J Biol Chem ; 294(45): 16865-16883, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31575663

RESUMEN

Accumulating evidence indicates that G protein-coupled receptors (GPCRs) interact with Rab GTPases during their intracellular trafficking. How GPCRs recruit and activate the Rabs is unclear. Here, we report that depletion of endogenous L-type prostaglandin D synthase (L-PGDS) in HeLa cells inhibited recycling of the prostaglandin D2 (PGD2) DP1 receptor (DP1) to the cell surface after agonist-induced internalization and that L-PGDS overexpression had the opposite effect. Depletion of endogenous Rab4 prevented l-PGDS-mediated recycling of DP1, and l-PGDS depletion inhibited Rab4-dependent recycling of DP1, indicating that both proteins are mutually involved in this pathway. DP1 stimulation promoted its interaction through its intracellular C terminus with Rab4, which was increased by l-PGDS. Confocal microscopy revealed that DP1 activation induces l-PGDS/Rab4 co-localization. l-PGDS/Rab4 and DP1/Rab4 co-immunoprecipitation levels were increased by DP1 agonist treatment. Pulldown assays with purified GST-l-PGDS and His6-Rab4 indicated that both proteins interact directly. l-PGDS interacted preferentially with the inactive, GDP-locked Rab4S22N variant rather than with WT Rab4 or with constitutively active Rab4Q67L proteins. Overexpression and depletion experiments disclosed that l-PGDS partakes in Rab4 activation following DP1 stimulation. Experiments with deletion mutants and synthetic peptides revealed that amino acids 85-92 in l-PGDS are involved in its interaction with Rab4 and in its effect on DP1 recycling. Of note, GTPγS loading and time-resolved FRET assays with purified proteins suggested that l-PGDS enhances GDP-GTP exchange on Rab4. Our results reveal how l-PGDS, which produces the agonist for DP1, regulates DP1 recycling by participating in Rab4 recruitment and activation.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Prostaglandina D2/metabolismo , Receptores de Prostaglandina/metabolismo , Proteínas de Unión al GTP rab4/metabolismo , Activación Enzimática , Células HeLa , Humanos , Oxidorreductasas Intramoleculares/química , Lipocalinas/química , Unión Proteica , Dominios Proteicos , Transporte de Proteínas
7.
J Biol Chem ; 294(49): 18522-18531, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31578280

RESUMEN

Cytokine macrophage migration inhibitory factor-2 (MIF-2 or D-dopachrome tautomerase) is a recently characterized second member of the MIF cytokine superfamily in mammalian genomes. MIF-2 shares pro-inflammatory and tumorigenic properties with the clinical target MIF (MIF-1), but the precise contribution of MIF-2 to immune physiology or pathology is unclear. Like MIF-1, MIF-2 has intrinsic keto-enol tautomerase activity and mediates biological functions by engaging the cognate, common MIF family receptor CD74. Evidence that the catalytic site of MIF family cytokines has a structural role in receptor binding has prompted exploration of tautomerase inhibitors as potential biological antagonists and therapeutic agents, although few catalytic inhibitors inhibit receptor activation. Here we describe the discovery and biochemical characterization of a selective small-molecule inhibitor of MIF-2. An in silico screen of 1.6 million compounds targeting the MIF-2 tautomerase site yielded several hits for potential catalytic inhibitors of MIF-2 and identified 4-(3-carboxyphenyl)-2,5-pyridinedicarboxylic acid (4-CPPC) as the most functionally potent compound. We found that 4-CPPC has an enzymatic IC50 of 27 µm and 17-fold selectivity for MIF-2 versus MIF-1. An in vitro binding assay for MIF-1/MIF-2 to the CD74 ectodomain (sCD74) indicated that 4-CPPC inhibits MIF-2-CD74 binding in a dose-dependent manner (0.01-10 µm) without influencing MIF-1-CD74 binding. Notably, 4-CPPC inhibited MIF-2-mediated activation of CD74 and reduced CD74-dependent signal transduction. These results open opportunities for development of more potent and pharmacologically auspicious MIF-2 inhibitors to investigate the distinct functions of this MIF family member in vivo.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Hormona Inhibidora de la Liberación de MSH/metabolismo , Humanos , Inflamación/enzimología , Inflamación/metabolismo , Oxidorreductasas Intramoleculares/química , Hormona Inhibidora de la Liberación de MSH/química , Neoplasias/enzimología , Neoplasias/metabolismo , Estructura Secundaria de Proteína , Transducción de Señal
8.
Arch Biochem Biophys ; 696: 108669, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33189651

RESUMEN

Histoplasma capsulatum is an ascomyceteous fungus and a human lung pathogen, which is present in river valleys of the Americas and other continents. H. capsulatum and two related human pathogens, Blasmomyces dermatitidis and Paracoccidioides brasiliensis, belongs to the Ajellomycetaceae family. The genomes of all three species code for three homologous and tentative enzymes of the linoleate diol synthase (LDS) family of fusion enzymes with dioxygenase (DOX) and cytochrome P450 domains. One group aligned closely with 8R-DOX-5,8-LDS of Aspergilli, which oxidizes linoleic acid to 5S,8R-dihydroxylinoleic acid; this group was not further investigated. The second group aligned with 10R-DOX-epoxy alcohol synthase (EAS) of plant pathogens. Expression of this enzyme from B. dermatitidis revealed only 10R-DOX activities, i.e., oxidation of linoleic acid to 10R-hydroperoxy-8E,12Z-octadecadienoic acid. The third group aligned in a separate entity. Expression of these enzymes of H. capsulatum and B. dermatitidis revealed no DOX activities, but both enzymes transformed 13S-hydroperoxy-9Z,11E-octadecadienoic acid efficiently to 12(13S)epoxy-11-hydroperoxy-9Z-octadecenoic acid. Other 13-hydroperoxides of linoleic and α-linolenic acids were transformed with less efficiency and the 9-hydroperoxides of linoleic acid were not transformed. In conclusion, a novel EAS has been found in H. capsulatum and B. dermititidis with 13S-hydroperoxy-9Z,11E-octadecadienoic acid as the likely physiological substrate.


Asunto(s)
Blastomyces/enzimología , Dioxigenasas/química , Proteínas Fúngicas/química , Histoplasma/enzimología , Oxidorreductasas Intramoleculares/química , Oxigenasas/química , Secuencia de Aminoácidos , Catálisis , Ácidos Grasos Insaturados/química , Filogenia , Proteínas Recombinantes/química
9.
Plant Cell Rep ; 39(12): 1785-1801, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33001313

RESUMEN

KEY MESSAGE: An allene oxide cyclase gene which is involved in defense against biotic and abiotic stresses was cloned and characterized in sugarcane. Allene oxide cyclase (AOC), a key enzyme in jasmonate acid (JA) biosynthesis, affects the stereoisomerism and biological activity of JA molecules, and plays an important role in plant stress resistance. In this study, four SsAOC alleles (SsAOC1-SsAOC4), which shared similar gene structure and were located on Chr1A, Chr1B, Chr1C, and Chr1D, respectively, were mined from sugarcane wild species Saccharum spontaneum, and a homologous gene ScAOC1 (GenBank Accession Number: MK674849) was cloned from sugarcane hybrid variety Yacheng05-179 inoculated with Sporisorium scitamineum for 48 h. ScAOC1 and SsAOC1-SsAOC4 were alkaline, unstable, hydrophilic, and non-secretory proteins, which possess the same set of conserved motifs and were clustered into one group in the phylogenetic analysis. ScAOC1 was expressed in all sugarcane tissues, but with different levels. After infection by S. scitamineum, the transcripts of ScAOC1 were increased significantly both in the smut-susceptible (ROC22) and resistant (Yacheng05-179) varieties, but its transcripts were more accumulated and lasted for a longer period in the smut-resistant variety than in the smut-susceptible one. ScAOC1 was down-regulated under MeJA and NaCl treatments, but up-regulated under SA, ABA, PEG, and cold stresses. Transiently overexpressing ScAOC1 gene into Nicotiana benthamiana leaves regulated the responses of N. benthamiana to two pathogens Ralstonia solanacearum and Fusarium solani var. coeruleum. Furthermore, prokaryotic expression analysis showed overexpression of ScAOC1 in Escherichia coli BL21 could enhance its tolerance to NaCl, mannitol, and cold stimuli. These results indicated that ScAOC1 may play an active role in response to biotic and abiotic stresses in sugarcane.


Asunto(s)
Oxidorreductasas Intramoleculares/genética , Proteínas de Plantas/genética , Saccharum/fisiología , Estrés Fisiológico/fisiología , Mapeo Cromosómico , Respuesta al Choque por Frío , Escherichia coli/genética , Evolución Molecular , Fusarium/patogenicidad , Regulación de la Expresión Génica de las Plantas , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/metabolismo , Manitol/farmacología , Familia de Multigenes , Reguladores del Crecimiento de las Plantas/farmacología , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Plantas Modificadas Genéticamente , Regiones Promotoras Genéticas , Ralstonia solanacearum/patogenicidad , Secuencias Reguladoras de Ácidos Nucleicos , Saccharum/efectos de los fármacos , Saccharum/genética , Cloruro de Sodio/farmacología , Nicotiana/genética , Nicotiana/microbiología
10.
J Nanobiotechnology ; 18(1): 157, 2020 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-33129330

RESUMEN

BACKGROUND: The chemotherapy drug doxorubicin (Dox) is widely used for treating a variety of cancers. However, its high cardiotoxicity hampered its clinical use. Exosomes derived from stem cells showed a therapeutic effect against Dox-induced cardiomyopathy (DIC). Previous studies reported that exosomes derived from mesenchymal stem cells (MSCs) pretreated with macrophage migration inhibitory factor (MIF) (exosomeMIF) showed a cardioprotective effect through modulating long noncoding RNAs/microRNAs (lncRNAs/miRs). This study aimed to investigate the role of exosomeMIF in the treatment of DIC. RESULTS: Exosomes were isolated from control MSCs (exosome) and MIF-pretreated MSCs (exosomeMIF). Regulatory lncRNAs activated by MIF pretreatment were explored using genomics approaches. Fluorescence-labeled exosomes were tracked in vitro by fluorescence imaging. In vivo and in vitro, miR-221-3p mimic transfection enforced miR-221-3p overexpression, and senescence-associated ß-galactosidase assay was applied to test cellular senescence. Exosomal delivering LncRNA-NEAT1 induced therapeutic effect in vivo was confirmed by echocardiography. It demonstrated that exosomesMIF recovered the cardiac function and exerted the anti-senescent effect through LncRNA-NEAT1 transfer against Dox. TargetScan and luciferase assay showed that miR-221-3p targeted the Sirt2 3'-untranslated region. Silencing LncRNA-NEAT1 in MSCs, miR-221-3p overexpression or Sirt2 silencing in cardiomyocytes decreased the exosomeMIF-induced anti-senescent effect against Dox. CONCLUSIONS: The results indicated exosomeMIF serving as a promising anti-senescent effector against Dox-induced cardiotoxicity through LncRNA-NEAT1 transfer, thus inhibiting miR-221-3p and leading to Sirt2 activation. The study proposed that exosomeMIF might have the potential to serve as a cardioprotective therapeutic agent during cancer chemotherapy.


Asunto(s)
Cardiotoxicidad/prevención & control , Doxorrubicina/efectos adversos , Exosomas/química , Oxidorreductasas Intramoleculares/química , Factores Inhibidores de la Migración de Macrófagos/química , Células Madre Mesenquimatosas/química , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Doxorrubicina/farmacología , Regulación de la Expresión Génica , Lesiones Cardíacas/inducido químicamente , Lesiones Cardíacas/genética , Lesiones Cardíacas/prevención & control , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Miocitos Cardíacos/efectos de los fármacos , Transducción de Señal , Sirtuina 2/metabolismo
11.
Arch Biochem Biophys ; 676: 108126, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31589830

RESUMEN

A catalase-related allene oxide synthase (cAOS) or a hydroperoxide lyase (cHPL) fused together with an 8R-lipoxygenase is involved in the stress signaling of corals via an arachidonic acid pathway. cAOS gives rise to α-ketol and cyclopentenone, while cHPL catalyzes the cleavage of 8R-hydroperoxyeicosatetraenoic acid (8R-HpETE) to C8-oxo acid and C12 aldehyde. In silico analysis of the substrate entry sites of highly identical coral cAOS and cHPL indicated that two positively charged residues of cAOS, K60 and K107, and the corresponding residues of cHPL, E60 and K107, may be involved in the anchoring of the carboxy group of polyunsaturated fatty acid (PUFA) hydroperoxides. A mutational analysis of cAOS and cHPL revealed that K60 or E60 and K107 were not necessary in the tethering of 8R-HpETE, however, the E60 of cHPL was essential in the productive binding of PUFA hydroperoxides. The substrate preferences of cAOS and cHPL were determined with hydroperoxy derivatives of C18, C20, C22 PUFAs, anandamide (AEA), 1-arachidonoyl glycerol (1-AG) and selected methylated substrates. Although cAOS and cHPL were able to metabolize different free PUFA substrates and arachidonoyl derivatives, only cHPL catalyzed the reaction with methylated PUFA hydroperoxides. The differences in the substrate binding and preferences between cAOS and cHPL can be explained by the distinct properties of their substrate entry sites. The current study demonstrated that homologous PUFA metabolizing enzymes may contribute to the versatile usage of the substrate pool.


Asunto(s)
Aldehído-Liasas/química , Aldehído-Liasas/metabolismo , Catalasa/química , Catalasa/metabolismo , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/metabolismo , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/metabolismo , Homología de Secuencia de Aminoácido , Animales , Antozoos/enzimología , Simulación por Computador , Ácidos Grasos Insaturados/química , Ácidos Grasos Insaturados/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica , Electricidad Estática , Especificidad por Sustrato
12.
Bioorg Med Chem ; 27(8): 1456-1478, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30858025

RESUMEN

With the goal of discovering more selective anti-inflammatory drugs, than COX inhibitors, to attenuate prostaglandin signaling, a fragment-based screen of hematopoietic prostaglandin D synthase was performed. The 76 crystallographic hits were sorted into similar groups, with the 3-cyano-quinoline 1a (FP IC50 = 220,000 nM, LE = 0.43) being a potent member of the 6,6-fused heterocyclic cluster. Employing SAR insights gained from structural comparisons of other H-PGDS fragment binding mode clusters, the initial hit 1a was converted into the 70-fold more potent quinoline 1d (IC50 = 3,100 nM, LE = 0.49). A systematic substitution of the amine moiety of 1d, utilizing structural information and array chemistry, with modifications to improve inhibitor stability, resulted in the identification of the 300-fold more active H-PGDS inhibitor tool compound 1bv (IC50 = 9.9 nM, LE = 0.42). This selective inhibitor exhibited good murine pharmacokinetics, dose-dependently attenuated PGD2 production in a mast cell degranulation assay and should be suitable to further explore H-PGDS biology.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Lipocalinas/antagonistas & inhibidores , Quinolinas/química , Quinolinas/farmacología , Animales , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacocinética , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/química , Lipocalinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Quinolinas/farmacocinética
13.
Proc Natl Acad Sci U S A ; 113(4): 972-7, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26755582

RESUMEN

Microsomal prostaglandin E2 synthase type 1 (mPGES-1) is responsible for the formation of the potent lipid mediator prostaglandin E2 under proinflammatory conditions, and this enzyme has received considerable attention as a drug target. Recently, a high-resolution crystal structure of human mPGES-1 was presented, with Ser-127 being proposed as the hydrogen-bond donor stabilizing thiolate anion formation within the cofactor, glutathione (GSH). We have combined site-directed mutagenesis and activity assays with a structural dynamics analysis to probe the functional roles of such putative catalytic residues. We found that Ser-127 is not required for activity, whereas an interaction between Arg-126 and Asp-49 is essential for catalysis. We postulate that both residues, in addition to a crystallographic water, serve critical roles within the enzymatic mechanism. After characterizing the size or charge conservative mutations Arg-126-Gln, Asp-49-Asn, and Arg-126-Lys, we inferred that a crystallographic water acts as a general base during GSH thiolate formation, stabilized by interaction with Arg-126, which is itself modulated by its respective interaction with Asp-49. We subsequently found hidden conformational ensembles within the crystal structure that correlate well with our biochemical data. The resulting contact signaling network connects Asp-49 to distal residues involved in GSH binding and is ligand dependent. Our work has broad implications for development of efficient mPGES-1 inhibitors, potential anti-inflammatory and anticancer agents.


Asunto(s)
Dipéptidos/química , Oxidorreductasas Intramoleculares/química , Microsomas/enzimología , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Glutatión/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Ligandos , Mutagénesis Sitio-Dirigida , Prostaglandina-E Sintasas , Conformación Proteica
14.
Int J Mol Sci ; 20(12)2019 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-31234561

RESUMEN

The channeling of metabolites is an essential step of metabolic regulation in all living organisms. Multifunctional enzymes with defined domains for metabolite compartmentalization are rare, but in many cases, larger assemblies forming multimeric protein complexes operate in defined metabolic shunts. In Arabidopsis thaliana, a multimeric complex was discovered that contains a 13-lipoxygenase and allene oxide synthase (AOS) as well as allene oxide cyclase. All three plant enzymes are localized in chloroplasts, contributing to the biosynthesis of jasmonic acid (JA). JA and its derivatives act as ubiquitous plant defense regulators in responses to both biotic and abiotic stresses. AOS belongs to the superfamily of cytochrome P450 enzymes and is named CYP74A. Another CYP450 in chloroplasts, hydroperoxide lyase (HPL, CYP74B), competes with AOS for the common substrate. The products of the HPL reaction are green leaf volatiles that are involved in the deterrence of insect pests. Both enzymes represent non-canonical CYP450 family members, as they do not depend on O2 and NADPH-dependent CYP450 reductase activities. AOS and HPL activities are crucial for plants to respond to different biotic foes. In this mini-review, we aim to summarize how plants make use of the LOX2-AOS-AOC2 complex in chloroplasts to boost JA biosynthesis over volatile production and how this situation may change in plant communities during mass ingestion by insect pests.


Asunto(s)
Aldehído-Liasas/metabolismo , Arabidopsis/fisiología , Sistema Enzimático del Citocromo P-450/metabolismo , Resistencia a la Enfermedad , Oxidorreductasas Intramoleculares/metabolismo , Aldehído-Liasas/química , Aldehído-Liasas/genética , Secuencia de Aminoácidos , Cloroplastos/metabolismo , Ciclopentanos/metabolismo , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/genética , Resistencia a la Enfermedad/genética , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/genética , Redes y Vías Metabólicas , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Oxilipinas/metabolismo , Desarrollo de la Planta/genética , Unión Proteica , Relación Estructura-Actividad
15.
Biochemistry ; 57(9): 1523-1532, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29412660

RESUMEN

Macrophage migration inhibitory factor (MIF) is a pro-inflammatory and tumor-promoting cytokine that occurs in two redox-dependent immunologically distinct conformational isoforms. The disease-related structural isoform of MIF (oxMIF) can be specifically and predominantly detected in the circulation of patients with inflammatory diseases and in tumor tissue, whereas the ubiquitously expressed isoform of MIF (redMIF) is abundantly expressed in healthy and diseased subjects. In this article, we report that cysteine 81 within MIF serves as a "switch cysteine" for the conversion of redMIF to oxMIF. Modulating cysteine 81 by thiol reactive agents leads to significant structural rearrangements of the protein, resulting in a decreased ß-sheet content and an increased random coil content, but maintaining the trimeric quaternary structure. This conformational change in the MIF molecule enables binding of oxMIF-specific antibodies BaxB01 and BaxM159, which showed beneficial activity in animal models of inflammation and cancer. Crystal structure analysis of the MIF-derived EPCALCS peptide, bound in its oxMIF-like conformation by the Fab fragment of BaxB01, revealed that this peptide adopts a curved conformation, making the central thiol protein oxidoreductase motif competent to undergo disulfide shuffling. We conclude that redMIF might reflect a latent zymogenic form of MIF, and formation of oxMIF leads to a physiologically relevant, i.e., enzymatically active, state.


Asunto(s)
Cisteína/química , Cisteína/metabolismo , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Especificidad de Anticuerpos , Dicroismo Circular , Cisteína/inmunología , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Disulfuro de Glutatión/química , Disulfuro de Glutatión/metabolismo , Humanos , Oxidorreductasas Intramoleculares/inmunología , Factores Inhibidores de la Migración de Macrófagos/inmunología , Modelos Moleculares , Oxidación-Reducción , Conformación Proteica , Relación Estructura-Actividad
16.
Biochemistry ; 57(26): 3599-3605, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29847104

RESUMEN

We report the first reversible and selective small molecule inhibitor of pro-inflammatory protein macrophage migration inhibitory factor-2 (also known as MIF-2 or d-DT). 4-(3-Carboxyphenyl)-2,5-pyridinedicarboxylic acid (4-CPPC) shows competitive binding with a 13-fold selectivity for human MIF-2 versus human MIF-1. The crystal structure of MIF-2 complexed with 4-CPPC reveals an induced fit mechanism that is not observed in the numerous MIF-1/inhibitor complexes. Crystallographic analysis demonstrates the structural source of 4-CPPC binding and selectivity for MIF-2. 4-CPPC can be employed to reveal previously unrecognized functions of MIF-1 in biological systems in which both MIF-1 and MIF-2 are expressed, to improve our knowledge of the MIF family of proteins, and to provide new mechanistic insights that can be utilized for the development of potent and selective pharmacological modulators of MIF-2.


Asunto(s)
Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Piridinas/química , Piridinas/farmacología , Cristalografía por Rayos X , Humanos , Oxidorreductasas Intramoleculares/química , Factores Inhibidores de la Migración de Macrófagos/química , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica/efectos de los fármacos
17.
J Neuroinflammation ; 15(1): 291, 2018 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-30340642

RESUMEN

BACKGROUND: Bacterial meningitis remains a big threat to the integrity of the central nervous system (CNS), despite the advancements in antimicrobial reagents. Escherichia coli is a bacterial pathogen that can disrupt the CNS function, especially in neonates. E. coli meningitis occurs after bacteria invade the brain microvascular endothelial cells (BMECs) that form a direct and essential barrier restricting the entry of circulating microbes and toxins to the brain. Previous studies have reported on several cellular proteins that function during meningitic E. coli infections; however, more comprehensive investigations to elucidate the potential targets involved in E. coli meningitis are essential to better understand this disease and discover new treatments for it. METHODS: The isobaric tags for relative and absolute quantification (iTRAQ) approach coupled with LC-MS/MS were applied to compare and characterize the different proteomic profiles of BMECs in response to meningitic or non-meningitic E. coli strains. KEGG and gene ontology annotations, ingenuity pathways analysis, and functional experiments were combined to identify the key host molecules involved in the meningitic E. coli-induced tight junction breakdown and neuroinflammatory responses. RESULTS: A total of 13 cellular proteins were found to be differentially expressed by meningitic E. coli strains PCN033 and RS218, including one that was also affected by HB101, a non-meningitic E. coli strain. Through bioinformatics analysis, we identified the macrophage migration inhibitory factor (MIF), granzyme A, NF-κB signaling, and mitogen-activated protein kinase (MAPK) pathways as being biologically involved in the meningitic E. coli-induced tight junction breakdown and neuroinflammation. Functionally, we showed that MIF facilitated meningitic E. coli-induced production of cytokines and chemokines and also helped to disrupt the blood-brain barrier by decreasing the expression of tight junction proteins like ZO-1, occludin. Moreover, we demonstrated the significant activation of NF-κB and MAPK signaling in BMECs in response to meningitic E. coli strains, which dominantly determined the generation of the proinflammatory cytokines including IL-6, IL-8, TNF-α, and IL-1ß. CONCLUSIONS: Our work identified 12 host cellular targets that are affected by meningitic E. coli strains and revealed MIF to be an important contributor to meningitic E. coli-induced cytokine production and tight junction disruption, and also the NF-κB and MAPK signaling pathways that are mainly involved in the infection-induced cytokines production. Characterization of these distinct proteins and pathways in BMECs will facilitate further elucidation of meningitis-causing mechanisms in humans and animals, thereby enabling the development of novel preventative and therapeutic strategies against infection with meningitic E. coli.


Asunto(s)
Encéfalo/citología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Regulación Bacteriana de la Expresión Génica/fisiología , Proteómica/métodos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Células Cultivadas , Biología Computacional , Citocinas/genética , Citocinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/farmacología , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/farmacología , Meningitis por Escherichia coli/metabolismo , Meningitis por Escherichia coli/patología , FN-kappa B/metabolismo , Transducción de Señal/fisiología
18.
Chemistry ; 24(1): 47-55, 2018 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-29052256

RESUMEN

Melanin is the main pigment responsible for the color of human skin, hair and eye. Its biosynthesis requires three melanogenic enzymes, tyrosinase (TYR), and the tyrosinase-related proteins TYRP1 and TYRP2. The difficulty of isolating pure and homogeneous proteins from endogenous sources has hampered their study, and resulted in many contradictory findings regarding their physiological functions. In this review, we summarize recent advances on the structure and function of TYR and TYRPs by virtue of the crystal structure of human TYRP1, which is the first available structure of a mammalian melanogenic enzyme. This structure, combined with tyrosinase structures from other lower eukaryotes and mutagenesis studies of key active site residues, sheds light on the mechanism of TYR and TYRPs. Furthermore, a TYRP1-based homology model of TYR provides a high-quality platform to map and analyze albinism-related mutations, as well as the design of specific antimelanogenic compounds. Finally, we provide perspectives for future structure/function studies of TYR and TYRPs.


Asunto(s)
Oxidorreductasas Intramoleculares/química , Melaninas/biosíntesis , Glicoproteínas de Membrana/química , Monofenol Monooxigenasa/química , Mutagénesis/genética , Mutación/genética , Oxidorreductasas/química , Animales , Dominio Catalítico , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Monofenol Monooxigenasa/genética , Monofenol Monooxigenasa/metabolismo , Mutagénesis/fisiología , Mutación/fisiología , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Pigmentación
19.
Protein Expr Purif ; 148: 46-53, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29601965

RESUMEN

The HLA class II histocompatibility antigen gamma chain, also known as HLA-DR antigen-associated invariant chain or CD74, has been shown to be involved in many biological processes amongst which antigen loading and transport of MHC class II molecules from the endoplasmic reticulum to the Golgi complex. It is also part of a receptor complex for macrophage migration inhibitory factor (MIF), and participates in inflammatory signaling. The inhibition of MIF-CD74 complex formation is regarded as a potentially attractive therapeutic target in inflammation, cancer and immune diseases. In order to be able to produce large quantities of the extracellular moiety of human CD74, which has been reported to be unstable and protease-sensitive, different constructs were made as fusions with two solubility enhancers: the well-known maltose-binding domain and Fh8, a small protein secreted by the parasite Fasciola hepatica. The fusion proteins could be purified with high yields from Escherichia coli and were demonstrated to be active in binding to MIF. Moreover, our results strongly suggest that the MIF binding site is located in the sequence between the transmembrane and the membrane-distal trimerisation domain of CD74, and comprises at least amino acids 113-125 of CD74.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/aislamiento & purificación , Antígenos de Histocompatibilidad Clase II/aislamiento & purificación , Oxidorreductasas Intramoleculares/aislamiento & purificación , Factores Inhibidores de la Migración de Macrófagos/aislamiento & purificación , Péptidos/química , Aminoácidos/genética , Animales , Antígenos de Diferenciación de Linfocitos B/química , Antígenos de Diferenciación de Linfocitos B/genética , Retículo Endoplásmico/genética , Escherichia coli/genética , Fasciola hepatica/química , Aparato de Golgi/genética , Antígenos HLA-DR/química , Antígenos HLA-DR/genética , Antígenos de Histocompatibilidad Clase II/química , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/genética , Péptidos/genética , Unión Proteica , Solubilidad
20.
Bioorg Med Chem Lett ; 28(4): 783-786, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29306572

RESUMEN

We synthesized a novel linker (1) with biotin, alkyne and amino groups for the identification of target proteins using a small molecule that contains an azide group (azide probe). The alkyne in the linker bound the azide probe via an azide-alkyne Huisgen cycloaddition. A protein cross-linker effectively bound the conjugate of the linker and an azide probe with a target protein. The covalently bound complex was detected by western blotting. Linker 1 was applied to a model system using an abscisic acid receptor, RCAR/PYR/PYL (PYL). Cross-linked complexes of linker 1, the azide probes and the target proteins were successfully visualized by western blotting. This method of target protein identification was more effective than a previously developed method that uses a second linker with biotin, alkyne, and benzophenone (linker 2) that acts to photo-crosslink target proteins. The system developed in this study is a method for identifying the target proteins of small bioactive molecules and is different from photo-affinity labelling.


Asunto(s)
Alquinos/química , Proteínas de Arabidopsis/química , Biotina/análogos & derivados , Biotina/química , Sondas Moleculares/química , Alquinos/síntesis química , Arabidopsis/química , Azidas/síntesis química , Azidas/química , Biotina/síntesis química , Western Blotting , Química Clic , Reactivos de Enlaces Cruzados/síntesis química , Reactivos de Enlaces Cruzados/química , Reacción de Cicloadición , Escherichia coli/química , Proteínas de Escherichia coli/química , Oxidorreductasas Intramoleculares/química , Luminiscencia , Lisina/análogos & derivados , Lisina/síntesis química , Lisina/química , Sondas Moleculares/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA