Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
1.
Int J Mol Sci ; 24(12)2023 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-37373195

RESUMO

The arachidonic acid lipoxygenase 15B (ALOX15B) orthologs of men and mice form different reaction products when arachidonic acid is used as the substrate. Tyr603Asp+His604Val double mutation in mouse arachidonic acid lipoxygenase 15b humanized the product pattern and an inverse mutagenesis strategy murinized the specificity of the human enzyme. As the mechanistic basis for these functional differences, an inverse substrate binding at the active site of the enzymes has been suggested, but experimental proof for this hypothesis is still pending. Here we expressed wildtype mouse and human arachidonic acid lipoxygenase 15B orthologs as well as their humanized and murinized double mutants as recombinant proteins and analyzed the product patterns of these enzymes with different polyenoic fatty acids. In addition, in silico substrate docking studies and molecular dynamics simulation were performed to explore the mechanistic basis for the distinct reaction specificities of the different enzyme variants. Wildtype human arachidonic acid lipoxygenase 15B converted arachidonic acid and eicosapentaenoic acid to their 15-hydroperoxy derivatives but the Asp602Tyr+Val603His exchange murinized the product pattern. The inverse mutagenesis strategy in mouse arachidonic acid lipoxygenase 15b (Tyr603Asp+His604Val exchange) humanized the product pattern with these substrates, but the situation was different with docosahexaenoic acid. Here, Tyr603Asp+His604Val substitution in mouse arachidonic acid lipoxygenase 15b also humanized the specificity but the inverse mutagenesis (Asp602Tyr+Val603His) did not murinize the human enzyme. With linoleic acid Tyr603Asp+His604Val substitution in mouse arachidonic acid lipoxygenase 15b humanized the product pattern but the inverse mutagenesis in human arachidonic acid lipoxygenase 15B induced racemic product formation. Amino acid exchanges at critical positions of human and mouse arachidonic acid lipoxygenase 15B orthologs humanized/murinized the product pattern with C20 fatty acids, but this was not the case with fatty acid substrates of different chain lengths. Asp602Tyr+Val603His exchange murinized the product pattern of human arachidonic acid lipoxygenase 15B with arachidonic acid, eicosapentaenoic acid, and docosahexaenoic acid. An inverse mutagenesis strategy on mouse arachidonic acid lipoxygenase 15b (Tyr603Asp+His604Val exchange) did humanize the reaction products with arachidonic acid and eicosapentaenoic acid, but not with docosahexaenoic acid.


Assuntos
Araquidonato Lipoxigenases , Ácido Eicosapentaenoico , Humanos , Animais , Camundongos , Araquidonato Lipoxigenases/metabolismo , Ácido Eicosapentaenoico/metabolismo , Ácidos Docosa-Hexaenoicos , Ácido Araquidônico/metabolismo , Ácidos Graxos , Especificidade por Substrato , Araquidonato 15-Lipoxigenase/metabolismo
2.
Cells ; 11(16)2022 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-36010555

RESUMO

Endothelial cells regulate vascular homeostasis through the secretion of various paracrine molecules, including bioactive lipids, but little is known regarding the enzymes responsible for generating these lipids under either physiological or pathophysiological conditions. Arachidonate lipoxygenase (ALOX) expression was therefore investigated in confluent and nonconfluent EA.h926 endothelial cells, which represent the normal quiescent and proliferative states, respectively. mRNAs for ALOX15, ALOX15B, and ALOXE3 were detected in EA.hy926 cells, with the highest levels present in confluent cells compared to nonconfluent cells. In contrast, ALOX5, ALOX12, and ALOX12B mRNAs were not detected. At the protein level, only ALOX15B and ALOXE3 were detected but only in confluent cells. ALOXE3 was also observed in confluent human umbilical artery endothelial cells (HUAEC), indicating that its expression, although previously unreported, may be a general feature of endothelial cells. Exposure to laminar flow further increased ALOXE3 levels in EA.hy926 cells and HUAECs. The evidence obtained in this study indicates that proliferative status and shear stress are both important factors that mediate endothelial ALOX gene expression. The presence of ALOX15B and ALOXE3 exclusively in quiescent human endothelial cells suggests their activity likely contributes to the maintenance of a healthy endothelium.


Assuntos
Araquidonato Lipoxigenases , Células Endoteliais , Araquidonato Lipoxigenases/metabolismo , Linhagem Celular , Células Endoteliais/metabolismo , Endotélio , Humanos , Lipídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
Eur J Nutr ; 61(8): 4059-4075, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35804267

RESUMO

PURPOSE: Ferroptosis is a form of regulated cell death that has the potential to be targeted as a cancer therapeutic strategy. But cancer cells have a wide range of sensitivities to ferroptosis, which limits its therapeutic potential. Accumulation of lipid peroxides determines the occurrence of ferroptosis. However, the type of lipid involved in peroxidation and the mechanism of lipid peroxide accumulation are less studied. METHODS: The effects of fatty acids (10 µM) with different carbon chain length and unsaturation on ferroptosis were evaluated by MTT and LDH release assay in cell lines derived from prostate cancer (PC3, 22RV1, DU145 and LNCaP), colorectal cancer (HT-29), cervical cancer (HeLa) and liver cancer (HepG2). Inhibitors of apoptosis, necroptosis, autophagy and ferroptosis were used to determine the type of cell death. Then the regulation of reactive oxygen species (ROS) and lipid peroxidation by docosahexaenoic acid (DHA) was measured by HPLC-MS and flow cytometry. The avtive form of DHA was determined by siRNA mediated gene silencing. The role of lipoxygenases was checked by inhibitors and gene silencing. Finally, the effect of DHA on ferroptosis-mediated tumor killing was verified in xenografts. RESULTS: The sensitivity of ferroptosis was positively correlated with the unsaturation of exogenously added fatty acid. DHA (22:6 n-3) sensitized cancer cells to ferroptosis-inducing reagents (FINs) at the highest level in vitro and in vivo. In this process, DHA increased ROS accumulation, lipid peroxidation and protein oxidation independent of its membrane receptor, GPR120. Inhibition of long chain fatty acid-CoA ligases and lysophosphatidylcholine acyltransferases didn't affect the role of DHA. DHA-involved ferroptosis can be induced in both arachidonate lipoxygenase 5 (ALOX5) negative and positive cells. Down regulation of ALOX5 inhibited ferroptosis, while overexpression of ALOX5 promoted ferroptosis. CONCLUSION: DHA can effectively promote ferroptosis-mediated tumor killing by increasing intracellular lipid peroxidation. Both ALOX5 dependent and independent pathways are involved in DHA-FIN induced ferroptosis. And during this process, free DHA plays an important role.


Assuntos
Ácidos Docosa-Hexaenoicos , Neoplasias , Masculino , Humanos , Ácidos Docosa-Hexaenoicos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Peróxidos Lipídicos , Lipoxigenase/metabolismo , Lipoxigenase/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Lisofosfatidilcolinas/farmacologia , Linhagem Celular Tumoral , Morte Celular , Peroxidação de Lipídeos , Lipoxigenases/metabolismo , Araquidonato Lipoxigenases/metabolismo , Araquidonato Lipoxigenases/farmacologia , Aciltransferases/metabolismo , Aciltransferases/farmacologia , Carbono , Coenzima A/metabolismo , Coenzima A/farmacologia
4.
Biotechnol Lett ; 44(9): 1027-1036, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35834094

RESUMO

PURPOSE: Peroxidation and reduction of 11S- and 13S-positions on C20 and C22 polyunsaturated fatty acids (PUFAs) by Escherichia coli expressing highly active arachidonate (ARA) 11S-lipoxygenase (11S-LOX) from Enhygromyxa salina with the reducing agent cysteine. RESULTS: The specific activity and catalytic efficiency of ARA 11S-LOX from E. salina were 4.1- and 91-fold higher than those of only reported ARA 11S-LOX from Myxococcus xanthus, respectively. The hydroxy fatty acids (HFAs) obtained by the biotransformation of ARA, eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and docosahexanoic acid (DHA) by Escherichia coli expressing 11S-LOX from E. salina in the presence of cysteine were identified as 11S-hydroxyeicosatetraenoic acid (11S-HETE), 11S-hydroxyeicosapentaenoic acid (11S-HEPE), 13S-hydroxydocosapentaenoic acid (13S-HDPA), and 13S-hydroxydocosahexaenoic acid (13S-HDHA), respectively. The recombinant cells converted 3 mM of ARA, EPA, DPA, and DHA into 2.9 mM of 11S-HETE, 2.4 mM 11S-HEPE, 1. 9 mM 13S-HDPA, and 2.2 mM 13S-HDHA in 60, 80, 120, and 120 min, corresponding to productivities of 72.5, 40.4, 18.5, and 22.4 µM min-1 and conversion yields of 96.7, 80.0, 62.3, and 74.6%, respectively. CONCLUSIONS: We report the highest concentrations, conversion yields, and productivities of 11S- and 13S-hydroxy fatty acids from C20- and C22-PUFAs achieved via E. coli expressing highly active E. salina 11S-LOX.


Assuntos
Escherichia coli , Lipoxigenase , Araquidonato Lipoxigenases/metabolismo , Biotransformação , Cisteína/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Ácidos Graxos , Ácidos Graxos Insaturados/metabolismo , Ácidos Hidroxieicosatetraenoicos , Lipoxigenase/metabolismo , Myxococcales
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(12): 158806, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32841762

RESUMO

Lipoxygenases (ALOX) are lipid peroxidizing enzymes that catalyze the biosynthesis of pro- and anti-inflammatory lipid mediators and have been implicated in (patho-)physiological processes. In humans, six functional ALOX isoforms exist and their arachidonic acid oxygenation products have been characterized. Products include leukotrienes and lipoxins which are involved in the regulation of inflammation and resolution. Oxygenation of n3-polyunsaturated fatty acids gives rise to specialized pro-resolving mediators, e.g. resolvins. However, the catalytic activity of different ALOX isoforms can lead to a multitude of potentially bioactive products. Here, we characterized the patterns of oxygenation products formed by human recombinant ALOX5, ALOX15, ALOX15B and ALOX12 from eicosapentaenoic acid (EPA) and its 18-hydroxy derivative 18-HEPE with particular emphasis on double and triple oxygenation products. ALOX15 and ALOX5 formed a complex mixture of various double oxygenation products from EPA, which include 5,15-diHEPE and various 8,15-diHEPE isomers. Their biosynthetic mechanisms were explored using heavy oxygen isotopes (H218O, 18O2 gas) and three catalytic activities contributed to product formation: i) fatty acid oxygenase activity, ii) leukotriene synthase activity, iii) lipohydroperoxidase activity. For ALOX15B and ALOX12 more specific product patterns were identified, which was also the case when these enzymes reacted in concert with ALOX5. Several double oxygenated compounds were formed from 18-HEPE by ALOX5, ALOX15B and ALOX12 including previously identified resolvins (RvE2, RvE3), while formation of triple oxygenation products, e.g. 5,17,18-triHEPE, required ALOX5. Taken together our data show that EPA can be converted by human ALOX isoforms to a large number of secondary oxygenation products, which might exhibit bioactivity.


Assuntos
Araquidonato Lipoxigenases/metabolismo , Ácido Eicosapentaenoico/metabolismo , Oxigênio/metabolismo , Ácido Eicosapentaenoico/análogos & derivados , Humanos , Hidroxilação , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/metabolismo
6.
Phys Chem Chem Phys ; 22(28): 16013-16022, 2020 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-32632422

RESUMO

Lipoxygenases are non-heme iron containing enzymes that catalyze oxygenation of poly-unsaturated fatty acids in different animal and plant species with extremely high regio- and stereospecificity. Nature employs 8-lipoxygenase to produce 8R-hydroperoxide from the oxygenation of arachidonic acid. A single-point L434F mutation of 8-lipoxygenase alters the regio- and stereospecificity of the final products, with a product ratio of 66 : 34 for 8R- and 12S-hydroperoxide, respectively. A molecular level explanation of this flipped regiospecificity is presented in this work on the basis of molecular dynamics simulations and transition network analysis of oxygen migration in the protein matrix. Phe434 is shown to exist in two conformations, the so-called open and closed conformations. In the closed conformation, the phenyl group of Phe434 shields the C8 site of the substrate, thereby preventing access of the oxygen molecule to this site, which leads to a quenching of the 8R-product. On the other hand, both closed and open conformations of Phe434 allow the oxygen molecule to approach the pro-S face of the C12 site of the substrate, which enhances the propensity of the 12S-hydroperoxide.


Assuntos
Araquidonato Lipoxigenases/genética , Animais , Araquidonato Lipoxigenases/química , Araquidonato Lipoxigenases/metabolismo , Cristalografia por Raios X , Simulação de Dinâmica Molecular , Mutação , Conformação Proteica
7.
J Phys Chem B ; 123(50): 10605-10621, 2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31775504

RESUMO

Lipoxygenases (lox's) are a group of non-heme iron containing enzymes that catalyze oxygenation of polyunsaturated fatty acids with precise regio- and stereoselectivities. The origin of regio- and stereospecific catalysis by 8-lox is explored in its wild-type (wt) form and in three mutants (Arg185Ala, Ala592Met, and Ala623His). The catalytic action of this enzyme progresses in two steps, namely, hydrogen abstraction from one double allylic carbon atom of substrate followed by oxygen insertion at the resulting prochiral carbon radical of the substrate. It is shown that the positional specificity of the hydrogen abstraction is a result of conformational dynamics of the bound substrate. While the C10 atom of the substrate is found to be the most probable site of hydrogen abstraction in the wt-lox, hydrogen abstraction from C13 is more favorable in the mutants. The present study discovers the presence of an interconnected network of a three-channel migration pathway operating in the protein matrix for efficient oxygen transport. Each migration channel is bestowed with a pocket at the peripheral region of protein as an oxygen access site, which transfers the oxygen to the active site through a well-connected migration path on a time scale of a few hundred picoseconds. By a careful geometric analysis of the oxygen pockets near the substrate binding cleft, the present study identifies the launching sites for oxygenation at the prochiral carbon centers C8, C11, C12, and C15 and the stereochemistry (R/S) of the corresponding products. It is found that the dominating 8R product in the wt-lox is due to the presence of the aromatic ring pair of Tyr181 and Phe173 acting as a gatekeeper for efficient delivery of oxygen at the pro-R face of C8. The change in the stereochemistry of the products in mutants is explained in terms of dynamic interactions between substrate and the surrounding residues.


Assuntos
Araquidonato Lipoxigenases/metabolismo , Biocatálise , Simulação de Dinâmica Molecular , Araquidonato Lipoxigenases/química , Araquidonato Lipoxigenases/genética , Mutação , Conformação Proteica , Estereoisomerismo , Especificidade por Substrato
8.
Biochem Biophys Res Commun ; 519(1): 81-85, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31477267

RESUMO

Lipoxygenases (LOXs) are dioxygenases that catalyze the oxygenation of polyunsaturated fatty acids to hydroperoxyl derivates. These products are precursors for different lipid mediators which are associated with pathogenesis of various diseases such as asthma, atherosclerosis and cancer. Several LOXs suffer from substrate inhibition, a potential regulatory mechanism, yet it is unclear what is the cause of this phenomenon. One such enzyme is the coral 11R-LOX which displays a significant decrease in turnover rate at arachidonic acid concentrations above 30 µM. In this report, site-directed mutagenesis and inhibition assays were employed to shed light on the mechanism of substrate inhibition in 11R-LOX. We found that introduction of a positive charge to the active site entrance with Gly188Arg substitution completely eliminates the slow-down at higher substrate concentrations. Inhibition of 11R-LOX by its catalysis product, 11(R)-hydroperoxyeicosatetraenoic acid, suggests an uncompetitive mechanism. We reason that substrate inhibition in 11R-LOX is due to additional fatty acid binding by the enzyme:substrate complex at an allosteric site situated in the very vicinity of the active site entrance.


Assuntos
Araquidonato Lipoxigenases/antagonistas & inibidores , Ácidos Araquidônicos/farmacologia , Arginina/genética , Inibidores Enzimáticos/farmacologia , Glicina/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Araquidonato Lipoxigenases/genética , Araquidonato Lipoxigenases/metabolismo , Humanos , Mutagênese Sítio-Dirigida , Alinhamento de Sequência , Especificidade por Substrato/efeitos dos fármacos
9.
Poult Sci ; 98(10): 4346-4358, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31287882

RESUMO

Cytochrome P-450 2C45 (CYP2C45) is the most highly expressed cytochrome P-450 isoform in chicken liver, and may play an important role in avian liver biology. However, information regarding the function of CYP2C45 in fatty liver is generally limited. The aim of this study was to investigate the role of CYP2C45 during the development of goose fatty liver. Our result indicated that the transcription of CYP2C45, together with PK and ALOX5, was increased in goose liver upon overfeeding for 19 D (P < 0.05). In goose primary hepatocytes, CYP2C45 RNA expression was also upgraded by the treatment with various chemicals like insulin, the fatty acids, and PPAR agonists (P < 0.05). We also found that both CYP2C45 overexpression and troglitazone treatment could increase the expression of pyruvate kinase (PK) and arachidonate 5-lipoxygenase (ALOX5), and furthermore, showed that the up-regulation of PK and ALOX5 induced by troglitazone could be suppressed by small interfering RNAs targeting CYP2C45 (P < 0.05). These findings suggest that fatty acids treatment and the overfeeding can induce the up-regulation of CYP2C45 expression possibly via PPARγ and that the induction of PK and ALOX5 in goose fatty liver is at least partially attributed to fatty acid-induced expression of CYP2C45. Thus, our data provides an insight into the mechanism by which glycolysis and arachidonic acid metabolism are modulated in goose fatty liver.


Assuntos
Araquidonato Lipoxigenases/genética , Proteínas Aviárias/genética , Ácidos Graxos/metabolismo , Fígado Gorduroso/veterinária , Gansos , Doenças das Aves Domésticas/genética , Piruvato Quinase/genética , Animais , Araquidonato Lipoxigenases/metabolismo , Proteínas Aviárias/metabolismo , Sequência de Bases , Sistema Enzimático do Citocromo P-450/genética , Fígado Gorduroso/genética , Masculino , PPAR gama/genética , Piruvato Quinase/metabolismo , Transdução de Sinais/genética
10.
Biotechnol Lett ; 41(4-5): 575-582, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30825045

RESUMO

OBJECTIVE: To quantitatively hydroxylate 8S- and 10S-positions on polyunsaturated fatty acids by recombinant Escherichia coli cells expressing mouse arachidonate 8S-lipoxygenase (8S-LOX). RESULTS: Hydroxylated products gained from the conversion of arachidonic acid (20:4Δ5Z,8Z,11Z,14Z, AA), eicosapentanoic acid (20:5Δ5Z,8Z,11Z,14Z,17Z, EPA), and (22:6Δ4Z,7Z,10Z,13Z,16Z,19Z, DHA) by recombinant E. coli cells containing 8S-LOX from mouse were identified as 8S-hydroxy-5,9,11,14(Z,E,Z,Z)-eicosatetranoic acid (8S-HETE), 8S-hydroxy-5,9,11,14,17(Z,E,Z,Z,Z)-eicosapentanoic acid (8S-HEPE), and 10S-hydroxy-4,8,12,14,16,19(Z,E,Z,Z,Z,Z)-docosahexaenoic acid (10S-HDoHE), respectively. Under the optimal hydroxylation conditions of pH 7.5, 30 °C, 5% (v/v) ethanol, 15 g cells l-1, and 5 mM substrate, AA, EPA, and DHA were hydroxylated into 4.37 mM 8S-HETE, 3.77 mM 8S-HEPE, and 3.13 mM 10S-HDoHE for 60, 90, and 60 min, with 87, 75, and 63% molar conversions, respectively. CONCLUSION: To the best of our knowledge, this is the first quantitatively biotechnological production of 8S-HETE, 8S-HEPE, and 10S-HDoHE.


Assuntos
Araquidonato Lipoxigenases/metabolismo , Escherichia coli/metabolismo , Ácidos Graxos Insaturados/metabolismo , Proteínas Recombinantes/metabolismo , Animais , Araquidonato Lipoxigenases/genética , Escherichia coli/genética , Concentração de Íons de Hidrogênio , Camundongos , Proteínas Recombinantes/genética , Temperatura
11.
J Lipid Res ; 59(11): 2153-2163, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30257932

RESUMO

Lipoxygenases (LOXs) catalyze the dioxygenation of PUFAs to produce regio- and stereospecific oxygenated fatty acids. The identification of regio- and stereospecific LOXs is important because their specific products are involved in different physiological activities in various organisms. Bacterial LOXs are found only in some proteobacteria and cyanobacteria, and they are not stable in vitro. Here, we used C20 and C22 PUFAs such as arachidonic acid (ARA), eicosapentaenoic acid, and docosahexaenoic acid to identify an 11S-specific LOX from the proteobacterium Myxococcus xanthus and explore its in vitro stability and activity. The activity and stability of M. xanthus ARA 11S-LOX as well as the production of 11S-hydroxyeicosatetraenoic acid from ARA were significantly increased by the addition of phosphatidylcholine, Ca2+, and coactosin-like protein (newly identified in the yeast Rhodosporidium toluroides) as stimulatory factors; in fact, LOX activity in the presence of all three factors increased approximately 3-fold. Our results indicate that these stimulatory factors can be used to increase the activity and stability of bacterial LOX and the production of bioactive hydroxy fatty acids, which can contribute to new academic research.


Assuntos
Araquidonato Lipoxigenases/metabolismo , Myxococcus xanthus/enzimologia , Araquidonato Lipoxigenases/genética , Cinética , Mutagênese Sítio-Dirigida , Fosfatidilcolinas/metabolismo , Filogenia
12.
Methods Enzymol ; 605: 33-49, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29909831

RESUMO

Methods are presented for the use of the coral 8R-lipoxygenase from the Caribbean sea whip coral Plexaura homomalla as a model enzyme for structural studies of animal lipoxygenases. The 8R-lipoxygenase is remarkably stable and can be stored at 4°C for 3 months with virtually no loss of activity. In addition, an engineered "pseudo wild-type" enzyme is soluble in the absence of detergents, which helps facilitate the preparation of enzyme:substrate complexes.


Assuntos
Antozoários/metabolismo , Araquidonato Lipoxigenases/isolamento & purificação , Ácido Araquidônico/metabolismo , Ensaios Enzimáticos/métodos , Domínios Proteicos/genética , Animais , Araquidonato Lipoxigenases/química , Araquidonato Lipoxigenases/genética , Araquidonato Lipoxigenases/metabolismo , Ácido Araquidônico/química , Sítios de Ligação/genética , Cristalografia por Raios X/métodos , Escherichia coli/metabolismo , Engenharia Metabólica/métodos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Especificidade por Substrato/genética
13.
Nat Commun ; 9(1): 128, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29317615

RESUMO

Hepoxilins (HXs) and trioxilins (TrXs) are involved in physiological processes such as inflammation, insulin secretion and pain perception in human. They are metabolites of polyunsaturated fatty acids (PUFAs), including arachidonic acid, eicosapentaenoic acid and docosahexaenoic acid, formed by 12-lipoxygenase (LOX) and epoxide hydrolase (EH) expressed by mammalian cells. Here, we identify ten types of HXs and TrXs, produced by the prokaryote Myxococcus xanthus, of which six types are new, namely, HXB5, HXD3, HXE3, TrXB5, TrXD3 and TrXE3. We succeed in the biotransformation of PUFAs into eight types of HXs (>35% conversion) and TrXs (>10% conversion) by expressing M. xanthus 12-LOX or 11-LOX with or without EH in Escherichia coli. We determine 11-hydroxy-eicosatetraenoic acid, HXB3, HXB4, HXD3, TrXB3 and TrXD3 as potential peroxisome proliferator-activated receptor-γ partial agonists. These findings may facilitate physiological studies and drug development based on lipid mediators.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácidos Graxos Insaturados/metabolismo , Myxococcus xanthus/enzimologia , Ácido 8,11,14-Eicosatrienoico/química , Ácido 8,11,14-Eicosatrienoico/metabolismo , Araquidonato 12-Lipoxigenase/genética , Araquidonato 12-Lipoxigenase/metabolismo , Araquidonato Lipoxigenases/genética , Araquidonato Lipoxigenases/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biotransformação , Epóxido Hidrolases/genética , Epóxido Hidrolases/metabolismo , Ácidos Graxos Insaturados/química , Redes e Vias Metabólicas/genética , Estrutura Molecular , Myxococcus xanthus/genética
14.
J Immunol Res ; 2016: 7509653, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27556049

RESUMO

Rheumatoid arthritis (RA) is a chronic inflammatory disease, leading to progressive destruction of joints and extra-articular tissues, including organs such as liver and spleen. The purpose of this study was to compare the effects of a potential immunomodulator, natural polyphenol N-feruloylserotonin (N-f-5HT), with methotrexate (MTX), the standard in RA therapy, in the chronic phase of adjuvant-induced arthritis (AA) in male Lewis rats. The experiment included healthy controls (CO), arthritic animals (AA), AA given N-f-5HT (AA-N-f-5HT), and AA given MTX (AA-MTX). N-f-5HT did not affect the body weight change and clinical parameters until the 14th experimental day. Its positive effect was rising during the 28-day experiment, indicating a delayed onset of N-f-5HT action. Administration of either N-f-5HT or MTX caused reduction of inflammation measured as the level of CRP in plasma and the activity of LOX in the liver. mRNA transcription of TNF-α and iNOS in the liver was significantly attenuated in both MTX and N-f-5HT treated groups of arthritic rats. Interestingly, in contrast to MTX, N-f-5HT significantly lowered the level of IL-1ß in plasma and IL-1ß mRNA expression in the liver and spleen of arthritic rats. This speaks for future investigations of N-f-5HT as an agent in the treatment of RA in combination therapy with MTX.


Assuntos
Artrite Experimental/genética , Artrite Experimental/patologia , Mediadores da Inflamação , Fígado/efeitos dos fármacos , Fígado/metabolismo , Metotrexato/farmacologia , Serotonina/análogos & derivados , Transcriptoma , Animais , Araquidonato Lipoxigenases/genética , Araquidonato Lipoxigenases/metabolismo , Artrite Experimental/tratamento farmacológico , Biomarcadores , Proteína C-Reativa , Citocinas/sangue , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Especificidade de Órgãos , Ratos , Serotonina/farmacologia , Índice de Gravidade de Doença , Fatores de Tempo
15.
Phytother Res ; 29(1): 108-13, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25278440

RESUMO

Acanthus mollis (Acanthaceae), Achillea ligustica, Artemisia arborescens and Inula viscosa (Asteraceae) are used in Southern Italy against psoriasis and other skin diseases that occur with an imbalanced production of eicosanoids. We here assessed their in vitro effects upon 5-, 12-, 15-LOX and COX-1 enzymes as well as NFκB activation in intact cells as their possible therapeutic targets. All methanol crude extracts inhibited both 5-LOX and COX-1 activities under 200 µg/mL, without significant effects on the 12-LOX pathway or any relevant in vitro free radical scavenging activity. NFκB activation was prevented by all extracts but A. mollis. Interestingly, A. ligustica, A. arborescens and A. mollis increased the biosynthesis of 15(S)-HETE, an anti-inflammatory eicosanoid. A. ligustica (IC50 =49.5 µg/mL) was superior to Silybum marianum (IC50 =147.8 µg/mL), which we used as antipsoriatic herbal medicine of reference. Its n-hexane, dichloromethane and ethyl acetate fractions had also inhibitory effects on the LTB4 biosynthesis (IC50 s=9.6, 20.3 and 68 µg/mL, respectively) evidencing that the apolar extracts of A. ligustica are promising active herbal ingredients for future phytotherapeutical products targeting psoriasis.


Assuntos
Anti-Inflamatórios/farmacologia , Fármacos Dermatológicos/farmacologia , Extratos Vegetais/farmacologia , Psoríase/tratamento farmacológico , Dermatopatias/tratamento farmacológico , Acanthaceae/química , Achillea/química , Animais , Araquidonato Lipoxigenases/metabolismo , Artemisia/química , Plaquetas/efeitos dos fármacos , Ciclo-Oxigenase 1/metabolismo , Células HeLa , Humanos , Inula/química , Itália , Leucócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Fitoterapia , Plantas Medicinais/química , Ratos
16.
Compr Physiol ; 6(1): 423-41, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26756638

RESUMO

Arachidonic acid metabolites have a myriad of biological actions including effects on the kidney to alter renal hemodynamics and tubular transport processes. Cyclooxygenase metabolites are products of an arachidonic acid enzymatic pathway that has been extensively studied in regards to renal function. Two lesser-known enzymatic pathways of arachidonic acid metabolism are the lipoxygenase (LO) and cytochrome P450 (CYP) pathways. The importance of LO and CYP metabolites to renal hemodynamics and tubular transport processes is now being recognized. LO and CYP metabolites have actions to alter renal blood flow and glomerular filtration rate. Proximal and distal tubular sodium transport and fluid and electrolyte homeostasis are also significantly influenced by renal CYP and LO levels. Metabolites of the LO and CYP pathways also have renal actions that influence renal inflammation, proliferation, and apoptotic processes at vascular and epithelial cells. These renal LO and CYP pathway actions occur through generation of specific metabolites and cell-signaling mechanisms. Even though the renal physiological importance and actions for LO and CYP metabolites are readily apparent, major gaps remain in our understanding of these lipid mediators to renal function. Future studies will be needed to fill these major gaps regarding LO and CYP metabolites on renal function.


Assuntos
Araquidonato Lipoxigenases/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Rim/metabolismo , Animais , Apoptose , Araquidonato Lipoxigenases/genética , Sistema Enzimático do Citocromo P-450/genética , Hemodinâmica , Humanos , Rim/fisiologia , Reabsorção Renal
17.
Fertil Steril ; 102(1): 282-290.e4, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24796762

RESUMO

OBJECTIVE: To test whether lipoxin A4 (LXA4) deficiency results in preeclampsia. DESIGN: Prospective experimental study. SETTING: Patient and animal research facilities. ANIMAL(S): Sprague-Dawley rats. INTERVENTION(S): We measured LXA4 and its biosynthetic enzymes, blocked the LXA4 signaling pathway, treated experimental rats with preeclampsia with LXA4, and detected inflammatory factors, FPR2/ALX, and 11ß-HSD2 to systematically test whether lack of LXA4 results in preeclampsia. MAIN OUTCOME MEASURE(S): We measured serum levels of LXA4 and inflammatory factors using enzyme-linked immunosorbent assay; detected LXA4 biosynthetic enzymes, inflammatory factors, FPR2/ALX, and 11ß-HSD2 mRNA expression using reverse transcriptase-polymerase chain reaction (RT-PCR) and real-time RT-PCR; and localized protein expression using immunohistochemistry. RESULT(S): FPR2/ALX and LXA4 and its biosynthetic enzymes were found to be decreased in women with preeclampsia. Replenishing LXA4 improved the symptoms of lipopolysaccharide-induced rats with preeclampsia, while blocking LXA4 signaling resulted in preeclampsia. LXA4 significantly reduced interleukin-6 (IL-6), tumor necrosis factor-α, and IFN-γ but increased IL-10, LXA4 up-regulated 11ß-HSD2. CONCLUSION(S): A deficiency of LXA4 may result in preeclampsia, which might be ascribed to a reduction in inflammation response, oxidative stress, and regulation of 11ß-HSD2.


Assuntos
Lipoxinas/deficiência , Pré-Eclâmpsia/metabolismo , Trofoblastos/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Animais , Araquidonato Lipoxigenases/metabolismo , Biomarcadores/sangue , Estudos de Casos e Controles , Linhagem Celular , Modelos Animais de Doenças , Feminino , Ácidos Heptanoicos/farmacologia , Humanos , Mediadores da Inflamação/sangue , Interferon gama/sangue , Interleucina-6/sangue , Lipopolissacarídeos , Lipoxinas/sangue , Lipoxinas/farmacologia , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/induzido quimicamente , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/tratamento farmacológico , Pré-Eclâmpsia/genética , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Lipoxinas/sangue , Transdução de Sinais , Fatores de Tempo , Trofoblastos/efeitos dos fármacos , Trofoblastos/enzimologia , Fator de Necrose Tumoral alfa/sangue
18.
J Biol Inorg Chem ; 18(3): 343-55, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23361122

RESUMO

Lipoxygenases (LOXs) are ubiquitous in nature and catalyze a range of life-essential reactions within organisms. In particular they are critical to the formation of eicosanoids, which are critical for normal cell function. However, a number of important questions about the reactivity and mechanism of these enzymes still remain. Specifically, although the initial step in the mechanism of LOXs has been well studied, little is known of subsequent steps. Thus, with use of a quantum mechanical/molecular mechanical approach, the complete catalytic mechanism of (8R)-LOX was investigated. The results have provided a better understanding of the general chemistry of LOXs as a whole. In particular, from comparisons with soybean LOX-1, it appears that the initial proton-coupled electron transfer may be very similar among all LOXs. Furthermore, LOXs appear to undergo multistate reactivity where potential spin inversion of an electron may occur either in the attack of O(2) or in the regeneration of the active site. Lastly, it is shown that with the explicit modeling of the environment, the regeneration of the active center likely occurs via the rotation of the intermediate followed by an outer-sphere [Formula: see text] transfer as opposed to the formation of a "purple intermediate" complex.


Assuntos
Antozoários/enzimologia , Araquidonato Lipoxigenases/química , Araquidonato Lipoxigenases/metabolismo , Animais , Antozoários/química , Domínio Catalítico , Ativação Enzimática , Lipoxigenase/química , Lipoxigenase/metabolismo , Simulação de Acoplamento Molecular , Peróxidos/química , Peróxidos/metabolismo , Conformação Proteica , Teoria Quântica , Glycine max/enzimologia
19.
J Lipid Res ; 53(2): 292-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22158855

RESUMO

Conversion of fatty acid hydroperoxides to epoxyalcohols is a well known secondary reaction of lipoxygenases, described for S-specific lipoxygenases forming epoxyalcohols with a trans-epoxide configuration. Here we report on R-specific lipoxygenase synthesis of a cis-epoxyalcohol. Although arachidonic and dihomo-γ-linolenic acids are metabolized by extracts of the Caribbean coral Plexaura homomalla via 8R-lipoxygenase and allene oxide synthase activities, 20:3ω6 forms an additional prominent product, identified using UV, GC-MS, and NMR in comparison to synthetic standards as 8R,9S-cis-epoxy-10S-erythro-hydroxy-eicosa-11Z,14Z-dienoic acid. Both oxygens of (18)O-labeled 8R-hydroperoxide are retained in the product, indicating a hydroperoxide isomerase activity. Recombinant allene oxide synthase formed only allene epoxide from 8R-hydroperoxy-20:3ω6, whereas two different 8R-lipoxygenases selectively produced the epoxyalcohol.A biosynthetic scheme is proposed in which a partial rotation of the reacting intermediate is required to give the observed erythro epoxyalcohol product. This characteristic and the synthesis of cis-epoxy epoxyalcohol may be a feature of R-specific lipoxygenases.


Assuntos
Ácido 8,11,14-Eicosatrienoico/química , Ácido 8,11,14-Eicosatrienoico/metabolismo , Araquidonato Lipoxigenases/metabolismo , Lipoxigenase/metabolismo , Animais , Antozoários/enzimologia , Compostos de Epóxi/química , Compostos de Epóxi/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Oxirredutases Intramoleculares/metabolismo , Espectroscopia de Ressonância Magnética , Especificidade por Substrato
20.
Cancer Metastasis Rev ; 30(3-4): 277-94, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22002716

RESUMO

Cancer initiation and progression are multistep events that require cell proliferation, migration, extravasation to the blood or lymphatic vessels, arrest to the metastatic site, and ultimately secondary growth. Tumor cell functions at both primary or secondary sites are controlled by many different factors, including growth factors and their receptors, chemokines, nuclear receptors, cell-cell interactions, cell-matrix interactions, as well as oxygenated metabolites of arachidonic acid. The observation that cyclooxygenases and lipoxygenases and their arachidonic acid-derived eicosanoid products (prostanoids and HETEs) are expressed and produced by tumor cells, together with the finding that these enzymes can regulate cell growth, survival, migration, and invasion, has prompted investigators to analyze the roles of these enzymes in cancer progression. In this review, we focus on the contribution of cyclooxygenase- and lipoxygenase-derived eicosanoids to tumor cell function in vitro and in vivo and discuss hope and tribulations of targeting these enzymes for cancer prevention and treatment.


Assuntos
Araquidonato Lipoxigenases/metabolismo , Neoplasias/enzimologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Animais , Antineoplásicos/farmacologia , Araquidonato Lipoxigenases/antagonistas & inibidores , Transformação Celular Neoplásica/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Eicosanoides/biossíntese , Eicosanoides/metabolismo , Humanos , Ácidos Linoleicos/metabolismo , Neoplasias/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...