Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Inflamm Res ; 72(8): 1649-1664, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37498393

RESUMEN

BACKGROUND, OBJECTIVES AND DESIGN: Arachidonic acid 15-lipoxygenase (ALOX15) has been implicated in the pathogenesis of inflammatory diseases but since pro- and anti-inflammatory roles have been suggested, the precise function of this enzyme is still a matter of discussion. To contribute to this discussion, we created transgenic mice, which express human ALOX15 under the control of the activating protein 2 promoter (aP2-ALOX15 mice) and compared the sensitivity of these gain-of-function animals in two independent mouse inflammation models with Alox15-deficient mice (loss-of-function animals) and wildtype control animals. MATERIALS AND METHODS: Transgenic aP2-ALOX15 mice were tested in comparison with Alox15 knockout mice (Alox15-/-) and corresponding wildtype control animals (C57BL/6J) in the complete Freund's adjuvant induced hind-paw edema model and in the dextran sulfate sodium induced colitis (DSS-colitis) model. In the paw edema model, the degree of paw swelling and the sensitivity of the inflamed hind-paw for mechanic (von Frey test) and thermal (Hargreaves test) stimulation were quantified as clinical readout parameters. In the dextran sodium sulfate induced colitis model the loss of body weight, the colon lengths and the disease activity index were determined. RESULTS: In the hind-paw edema model, systemic inactivation of the endogenous Alox15 gene intensified the inflammatory symptoms, whereas overexpression of human ALOX15 reduced the degree of hind-paw inflammation. These data suggest anti-inflammatory roles for endogenous and transgenic ALOX15 in this particular inflammation model. As mechanistic reason for the protective effect downregulation of the pro-inflammatory ALOX5 pathways was suggested. However, in the dextran sodium sulfate colitis model, in which systemic inactivation of the Alox15 gene protected female mice from DSS-induced colitis, transgenic overexpression of human ALOX15 did hardly impact the intensity of the inflammatory symptoms. CONCLUSION: The biological role of ALOX15 in the pathogenesis of inflammation is variable and depends on the kind of the animal inflammation model.


Asunto(s)
Araquidonato 15-Lipooxigenasa , Colitis , Humanos , Ratones , Femenino , Animales , Ratones Transgénicos , Adyuvante de Freund , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/uso terapéutico , Dextranos/efectos adversos , Dextranos/metabolismo , Ratones Endogámicos C57BL , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/tratamiento farmacológico , Colitis/metabolismo , Colon/metabolismo , Antiinflamatorios/farmacología , Ratones Noqueados , Edema/inducido químicamente , Edema/genética , Edema/metabolismo , Sulfato de Dextran/efectos adversos , Sulfato de Dextran/metabolismo , Modelos Animales de Enfermedad
2.
Cell Mol Biol Lett ; 28(1): 97, 2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38030974

RESUMEN

Arachidonic acid 15-lipoxygenases (ALOX15) play a role in mammalian erythropoiesis but they have also been implicated in inflammatory processes. Seven intact Alox genes have been detected in the mouse reference genome and the mouse Alox15 gene is structurally similar to the orthologous genes of other mammals. However, mouse and human ALOX15 orthologs have different functional characteristics. Human ALOX15 converts C20 polyenoic fatty acids like arachidonic acid mainly to the n-6 hydroperoxide. In contrast, the n-9 hydroperoxide is the major oxygenation product formed by mouse Alox15. Previous experiments indicated that Leu353Phe exchange in recombinant mouse Alox15 humanized the catalytic properties of the enzyme. To investigate whether this functional humanization might also work in vivo and to characterize the functional consequences of mouse Alox15 humanization we generated Alox15 knock-in mice (Alox15-KI), in which the Alox15 gene was modified in such a way that the animals express the arachidonic acid 15-lipoxygenating Leu353Phe mutant instead of the arachidonic acid 12-lipoxygenating wildtype enzyme. These mice develop normally, they are fully fertile but display modified plasma oxylipidomes. In young individuals, the basic hematological parameters were not different when Alox15-KI mice and outbred wildtype controls were compared. However, when growing older male Alox15-KI mice develop signs of dysfunctional erythropoiesis such as reduced hematocrit, lower erythrocyte counts and attenuated hemoglobin concentration. These differences were paralleled by an improved ex vivo osmotic resistance of the peripheral red blood cells. Interestingly, such differences were not observed in female individuals suggesting gender specific effects. In summary, these data indicated that functional humanization of mouse Alox15 induces defective erythropoiesis in aged male individuals.


Asunto(s)
Araquidonato 15-Lipooxigenasa , Peróxido de Hidrógeno , Animales , Femenino , Humanos , Masculino , Ratones , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/genética , Ácido Araquidónico , Mamíferos
3.
Int J Mol Sci ; 24(18)2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37762455

RESUMEN

The human genome involves six functional arachidonic acid lipoxygenase (ALOX) genes and the corresponding enzymes (ALOX15, ALOX15B, ALOX12, ALOX12B, ALOXE3, ALOX5) have been implicated in cell differentiation and in the pathogenesis of inflammatory, hyperproliferative, metabolic, and neurological disorders. In other vertebrates, ALOX-isoforms have also been identified, but they occur less frequently. Since bony fish represent the most abundant subclass of vertebrates, we recently expressed and characterized putative ALOX15 orthologs of three different bony fish species (Nothobranchius furzeri, Pundamilia nyererei, Scleropages formosus). To explore whether these enzymes represent functional equivalents of mammalian ALOX15 orthologs, we here compared a number of structural and functional characteristics of these ALOX-isoforms with those of mammalian enzymes. We found that in contrast to mammalian ALOX15 orthologs, which exhibit a broad substrate specificity, a membrane oxygenase activity, and a special type of dual reaction specificity, the putative bony fish ALOX15 orthologs strongly prefer C20 fatty acids, lack any membrane oxygenase activity and exhibit a different type of dual reaction specificity with arachidonic acid. Moreover, mutagenesis studies indicated that the Triad Concept, which explains the reaction specificity of all mammalian ALOX15 orthologs, is not applicable for the putative bony fish enzymes. The observed functional differences between putative bony fish ALOX15 orthologs and corresponding mammalian enzymes suggest a targeted optimization of the catalytic properties of ALOX15 orthologs during vertebrate development.

4.
Int J Mol Sci ; 24(5)2023 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-36902243

RESUMEN

Arachidonic acid lipoxygenases (ALOX) have been implicated in the pathogenesis of inflammatory, hyperproliferative, neurodegenerative, and metabolic diseases, but the physiological function of ALOX15 still remains a matter of discussion. To contribute to this discussion, we created transgenic mice (aP2-ALOX15 mice) expressing human ALOX15 under the control of the aP2 (adipocyte fatty acid binding protein 2) promoter, which directs expression of the transgene to mesenchymal cells. Fluorescence in situ hybridization and whole-genome sequencing indicated transgene insertion into the E1-2 region of chromosome 2. The transgene was highly expressed in adipocytes, bone marrow cells, and peritoneal macrophages, and ex vivo activity assays proved the catalytic activity of the transgenic enzyme. LC-MS/MS-based plasma oxylipidome analyses of the aP2-ALOX15 mice suggested in vivo activity of the transgenic enzyme. The aP2-ALOX15 mice were viable, could reproduce normally, and did not show major phenotypic alterations when compared with wildtype control animals. However, they exhibited gender-specific differences with wildtype controls when their body-weight kinetics were evaluated during adolescence and early adulthood. The aP2-ALOX15 mice characterized here can now be used for gain-of-function studies evaluating the biological role of ALOX15 in adipose tissue and hematopoietic cells.


Asunto(s)
Araquidonato 15-Lipooxigenasa , Expresión Génica , Espectrometría de Masas en Tándem , Adulto , Animales , Humanos , Ratones , Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Cromatografía Liquida , Hibridación Fluorescente in Situ , Ratones Transgénicos
5.
Int J Mol Sci ; 24(13)2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37446212

RESUMEN

Mammalian arachidonic acid lipoxygenases (ALOXs) have been implicated in the pathogenesis of inflammatory diseases, and its pro- and anti-inflammatory effects have been reported for different ALOX-isoforms. Human ALOX15B oxygenates arachidonic acid to its 15-hydroperoxy derivative, whereas the corresponding 8-hydroperoxide is formed by mouse Alox15b (Alox8). This functional difference impacts the biosynthetic capacity of the two enzymes for creating pro- and anti-inflammatory eicosanoids. To explore the functional consequences of the humanization of the reaction specificity of mouse Alox15b in vivo, we tested Alox15b knock-in mice that express the arachidonic acid 15-lipoxygenating Tyr603Asp and His604Val double mutant of Alox15b, instead of the arachidonic acid 8-lipoxygenating wildtype enzyme, in two different animal inflammation models. In the dextran sodium sulfate-induced colitis model, female Alox15b-KI mice lost significantly more bodyweight during the acute phase of inflammation and recovered less rapidly during the resolution phase. Although we observed significant differences in the colonic levels of selected pro- and anti-inflammatory eicosanoids during the time-course of inflammation, there were no differences between the two genotypes at any time-point of the disease. In Freund's complete adjuvant-induced paw edema model, Alox15b-KI mice were less susceptible than outbred wildtype controls, though we did not observe significant differences in pain perception (Hargreaves-test, von Frey-test) when the two genotypes were compared. our data indicate that humanization of the reaction specificity of mouse Alox15b (Alox8) sensitizes mice for dextran sodium sulfate-induced experimental colitis, but partly protects the animals in the complete Freund's adjuvant-induced paw edema model.


Asunto(s)
Colitis , Dextranos , Humanos , Ratones , Femenino , Animales , Ácido Araquidónico , Inflamación/genética , Mamíferos , Antiinflamatorios , Edema/inducido químicamente , Edema/genética , Modelos Animales de Enfermedad
6.
Int J Mol Sci ; 23(24)2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36555666

RESUMEN

Eicosanoids and related compounds are pleiotropic lipid mediators, which are biosynthesized in mammals via three distinct metabolic pathways (cyclooxygenase pathway, lipoxygenase pathway, epoxygenase pathway). These mediators have been implicated in the pathogenesis of inflammatory diseases and drugs interfering with eicosanoid signaling are currently available as antiphlogistics. Eicosanoid biosynthesis has well been explored in mammals including men, but much less detailed information is currently available on eicosanoid biosynthesis in other vertebrates including bony fish. There are a few reports in the literature describing the expression of arachidonic acid lipoxygenases (ALOX isoforms) in several bony fish species but except for two zebrafish ALOX-isoforms (zfALOX1 and zfALOX2) bony fish eicosanoid biosynthesizing enzymes have not been characterized. To fill this gap and to explore the possible roles of ALOX15 orthologs in bony fish inflammation we cloned and expressed putative ALOX15 orthologs from three different bony fish species (N. furzeri, P. nyererei, S. formosus) as recombinant N-terminal his-tag fusion proteins and characterized the corresponding enzymes with respect to their catalytic properties (temperature-dependence, activation energy, pH-dependence, substrate affinity and substrate specificity with different polyenoic fatty acids). Furthermore, we identified the chemical structure of the dominant oxygenation products formed by the recombinant enzymes from different free fatty acids and from more complex lipid substrates. Taken together, our data indicate that functional ALOX isoforms occur in bony fish but that their catalytic properties are different from those of mammalian enzymes. The possible roles of these ALOX-isoforms in bony fish inflammation are discussed.


Asunto(s)
Lipooxigenasa , Pez Cebra , Animales , Lipooxigenasa/genética , Lipooxigenasa/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Mamíferos/metabolismo , Eicosanoides/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Inflamación/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo
7.
Proc Natl Acad Sci U S A ; 113(30): E4266-75, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27412860

RESUMEN

ALOX15 (12/15-lipoxygenase) orthologs have been implicated in maturational degradation of intracellular organelles and in the biosynthesis of antiinflammatory and proresolving eicosanoids. Here we hypothesized that lower mammals (mice, rats, pigs) express 12-lipoxygenating ALOX15 orthologs. In contrast, 15-lipoxygenating isoforms are found in higher primates (orangutans, men), and these results suggest an evolution of ALOX15 specificity. To test this hypothesis we first cloned and characterized ALOX15 orthologs of selected Catarrhini representing different stages of late primate evolution and found that higher primates (men, chimpanzees) express 15-lipoxygenating orthologs. In contrast, lower primates (baboons, rhesus monkeys) express 12-lipoxygenating enzymes. Gibbons, which are flanked in evolution by rhesus monkeys (12-lipoxygenating ALOX15) and orangutans (15-lipoxygenating ALOX15), express an ALOX15 ortholog with pronounced dual specificity. To explore the driving force for this evolutionary alterations, we quantified the lipoxin synthase activity of 12-lipoxygenating (rhesus monkey, mouse, rat, pig, humIle418Ala) and 15-lipoxygenating (man, chimpanzee, orangutan, rabbit, ratLeu353Phe) ALOX15 variants and found that, when normalized to their arachidonic acid oxygenase activities, the lipoxin synthase activities of 15-lipoxygenating ALOX15 variants were more than fivefold higher (P < 0.01) [corrected]. Comparative molecular dynamics simulations and quantum mechanics/molecular mechanics calculations indicated that, for the 15-lipoxygenating rabbit ALOX15, the energy barrier for C13-hydrogen abstraction (15-lipoxygenation) was 17 kJ/mol lower than for arachidonic acid 12-lipoxygenation. In contrast, for the 12-lipoxygenating Ile418Ala mutant, the energy barrier for 15-lipoxygenation was 10 kJ/mol higher than for 12-lipoxygenation. Taken together, our data suggest an evolution of ALOX15 specificity, which is aimed at optimizing the biosynthetic capacity for antiinflammatory and proresolving lipoxins.


Asunto(s)
Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Evolución Molecular , Lipoxinas/biosíntesis , Animales , Antiinflamatorios/química , Antiinflamatorios/metabolismo , Araquidonato 15-Lipooxigenasa/química , Dominio Catalítico , Humanos , Lipoxinas/química , Ratones , Mutación , Primates , Conejos , Ratas , Especificidad de la Especie , Especificidad por Sustrato , Porcinos
8.
Skin Pharmacol Physiol ; 32(4): 192-200, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31096247

RESUMEN

BACKGROUND: Atopic diseases constitute a major health challenge for industrialized countries, and elevated levels of interleukin 4 (IL-4) frequently characterize these disorders. Previous in vitroanalyses have indicated that IL-4 strongly upregulates the expression of IL-4-sensitive genes in human monocytes. OBJECTIVE: To explore whether similar expression alterations may contribute to the pathomechanisms of atopic diseases in vivo we carried out a small-scale case-control clinical study (n = 43), in which we quantified the plasma levels of IgE and IL-4 as well as the expression of selected IL-4-sensitive genes in blood leukocytes. METHODS: 34 allergic patients suffering from allergic rhinitis (n = 11), atopic eczema (n = 11) and allergic asthma (n = 12) as well as 9 healthy control individuals were recruited. IgE and IL-4 plasma levels were determined by ELISA, and the expression of selected IL-4-sensitive gene products in blood leukocytes was quantified by qRT-PCR. In addition, the fatty acid oxygenase activity of isolated monocytes was measured by RP-HPLC analysis of the arachidonic acid oxygenation products (ex vivo activity assays). RESULTS: We found that plasma levels of IgE and IL-4 were significantly elevated in atopic patients but the degree of elevation was not sufficient to upregulate the expression of the selected IL-4-sensitive genes in circulating leukocytes. Moreover, the arachidonic acid oxygenase activity of blood monocytes was not significantly altered in atopic patients. CONCLUSION: Our data suggest that the IL-4 plasma levels of atopic patients are not high enough to impact the expression of IL-4-sensitive genes.


Asunto(s)
Hipersensibilidad Inmediata/sangre , Hipersensibilidad Inmediata/genética , Inmunoglobulina E/biosíntesis , Interleucina-4/biosíntesis , Leucocitos/fisiología , Adulto , Asma/sangre , Asma/genética , Estudios de Casos y Controles , Dermatitis Atópica/sangre , Dermatitis Atópica/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oxigenasas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Rinitis Alérgica/sangre , Rinitis Alérgica/genética , Regulación hacia Arriba
9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(2): 152-164, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29146531

RESUMEN

Pseudomonas aeruginosa is a gram-negative pathogen, which causes life-threatening infections in immunocompromized patients. These bacteria express a secreted lipoxygenase (PA-LOX), which oxygenates free arachidonic acid to 15S-hydro(pero)xyeicosatetraenoic acid. It binds phospholipids at its active site and physically interacts with lipid vesicles. When incubated with red blood cells membrane lipids are oxidized and hemolysis is induced but the structures of the oxygenated membrane lipids have not been determined. Using a lipidomic approach, we analyzed the formation of oxidized phospholipids generated during the in vitro incubation of recombinant PA-LOX with human erythrocytes and cultured human lung epithelial cells. Precursor scanning of lipid extracts prepared from these cells followed by multiple reaction monitoring and MS/MS analysis revealed a complex mixture of oxidation products. For human red blood cells this mixture comprised forty different phosphatidylethanolamine and phosphatidylcholine species carrying oxidized fatty acid residues, such as hydroxy-octadecadienoic acids, hydroxy- and keto-eicosatetraenoic acid, hydroxy-docosahexaenoic acid as well as oxygenated derivatives of less frequently occurring polyenoic fatty acids. Similar oxygenation products were also detected when cultured lung epithelial cells were employed but here the amounts of oxygenated lipids were smaller and under identical experimental conditions we did not detect major signs of cell lysis. However, live imaging indicated an impaired capacity for trypan blue exclusion and an augmented mitosis rate. Taken together these data indicate that PA-LOX can oxidize the membrane lipids of eukaryotic cells and that the functional consequences of this reaction strongly depend on the cell type.


Asunto(s)
Proteínas Bacterianas/metabolismo , Membrana Eritrocítica/metabolismo , Lipooxigenasa/metabolismo , Fosfolípidos/metabolismo , Pseudomonas aeruginosa/enzimología , Células A549 , Proteínas Bacterianas/química , Membrana Eritrocítica/química , Membrana Eritrocítica/patología , Humanos , Lipooxigenasa/química , Oxidación-Reducción , Fosfolípidos/química
10.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(8): 866-880, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29702245

RESUMEN

Lipoxygenases (ALOXs) are involved in the regulation of cellular redox homeostasis. They also have been implicated in the biosynthesis of pro- and anti-inflammatory lipid mediators and play a role in the pathogenesis of inflammatory diseases, which constitute a major health challenge owing to increasing incidence and prevalence in all industrialized countries around the world. To explore the pathophysiological role of Alox15 (leukocyte-type 12-LOX) in mouse experimental colitis we tested the impact of systemic inactivation of the Alox15 gene on the extent of dextrane sulfate sodium (DSS) colitis. We found that in wildtype mice expression of the Alox15 gene was augmented during DSS-colitis while expression of other Alox genes (Alox5, Alox15b) was hardly altered. Systemic Alox15 (leukocyte-type 12-LOX) deficiency induced less severe colitis symptoms and suppressed in vivo formation of 12-hydroxyeicosatetraenoic acid (12-HETE), the major Alox15 (leukocyte-type 12-LOX) product in mice. These alterations were paralleled by reduced expression of pro-inflammatory gene products, by sustained expression of the zonula occludens protein 1 (ZO-1) and by a less impaired intestinal epithelial barrier function. These results are consistent with in vitro incubations of colon epithelial cells, in which addition of 12S-HETE compromised enantioselectively transepithelial electric resistance. Consistent with these data transgenic overexpression of human ALOX15 intensified the inflammatory symptoms. In summary, our results indicate that systemic Alox15 (leukocyte-type 12-LOX) deficiency protects mice from DSS-colitis. Since exogenous 12-HETE compromises the expression of the tight junction protein ZO-1 the protective effect has been related to a less pronounced impairment of the intestinal epithelial barrier function.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Colitis/patología , Colon/patología , Mucosa Intestinal/patología , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/biosíntesis , Animales , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/genética , Colitis/inducido químicamente , Colitis/genética , Colon/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Permeabilidad , Factores Sexuales , Proteína de la Zonula Occludens-1/metabolismo
11.
Chemistry ; 24(4): 962-973, 2018 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-29154477

RESUMEN

The reaction specificity of lipoxygenases is of physiological relevance since the various oxygenation products exhibit different biological activities. Among mammalian ALOX15 orthologs there are arachidonic acid 12- and 15-lipoxygenating enzymes and recent studies suggested an evolutionary switch in that reaction specificity during late primate development. Previous reports showed that 12-lipoxygenating ALOX15 orthologs can be converted to 15-lipoxygenating enzymes by site-directed mutagenesis of some sequence determinants. Unfortunately, the molecular basis for those alterations are not well understood. Here, the arachidonic acid 12-lipoxygenating N-terminal truncation variant of pig ALOX15, for which a crystal structure is available, was used to explore the catalytic mechanism of the specificity switch induced by mutagenesis of Val418 and Val419 sequence determinants. We found that Val418Ile+Val419Met double mutant is dominantly 15-lipoxygenating. Docking and MD simulations, and quantum mechanics/molecular mechanics calculations indicated that the wildtype energy barrier for arachidonic acid 15-lipoxygenation is 3.4 kcal mol-1 higher than for 12-lipoxygenation. In contrast, for the Val418Ile+Val419Met double mutant the energy barrier for 12-lipoxygenation is 6.0 kcal mol-1 higher than for 15-lipoxygenation. Our data suggest that enzyme-substrate complex geometries determine the value of these energy barriers and, as a consequence, the reaction specificity of ALOX15 orthologs.

12.
Biochim Biophys Acta ; 1861(1): 1-11, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26456699

RESUMEN

The zebrafish (Danio rerio) is frequently employed as model organism to explore vertebrate embryogenesis but little is known about the lipoxygenase pathway in this lower vertebrate. When we searched the zebrafish genome for lipoxygenase genes we detected seven different genes localized on four different chromosomes. Four of these genes (ALOX2, ALOX3a, ALOX3b, ALOX3c) share a high degree of sequence conservation with the human ALOX5 gene, which encodes for the key enzyme of mammalian leukotriene biosynthesis. Expression profiles of the corresponding transcripts indicated that the ALOX2 gene is high level expressed during zebrafish embryogenesis whereas transcripts originating from the other three paralog genes could not be detected. To functionally compare human ALOX5 with the putative zebrafish ortholog (zebrafish ALOX2) we cloned and expressed the zebrafish enzyme in pro- and eukaryotic expression systems and characterized it as arachidonic acid 5S-lipoxygenating enzyme, which also exhibits leukotriene synthase activity. Mutagenesis studies of the triad determinants (Phe359Trp, Ala424Ile, Asn425Met) altered the reaction specificity from 5S- to dominant 15S-lipoxygenation. The finding that zebrafish expresses a functional ALOX5 together with the observation that most other human leukotriene relevant genes have an ortholog in the zebrafish genome suggests the biological relevance of leukotriene signaling in lower vertebrates.


Asunto(s)
Araquidonato 5-Lipooxigenasa/genética , Proteínas de Pez Cebra/genética , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Araquidonato 5-Lipooxigenasa/química , Humanos , Isoenzimas/química , Isoenzimas/genética , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Filogenia , Alineación de Secuencia , Pez Cebra/embriología , Proteínas de Pez Cebra/química
13.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(7): 666-675, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28400162

RESUMEN

Mammalian lipoxygenases (LOX) have been implicated in cell differentiation and in the pathogenesis of inflammatory, hyperproliferative and neurological diseases. Although the reaction specificity of mammalian LOX with n-6 fatty acids (linoleic acid, arachidonic acid) has been explored in detail little information is currently available on the product patterns formed from n-3 polyenoic fatty acids, which are of particular nutritional importance and serve as substrate for the biosynthesis of pro-resolving inflammatory mediators such as resolvins and maresins. Here we expressed the ALOX15 orthologs of eight different mammalian species as well as human ALOX12 and ALOX15B as recombinant his-tag fusion proteins and characterized their reaction specificity with the most abundantly occurring polyunsaturated fatty acids (PUFAs) including 5,8,11,14,17-eicosapentaenoic acid (EPA) and 4,7,10,13,16,19-docosahexaenoic acid (DHA). We found that the LOX isoforms tested accept these fatty acids as suitable substrates and oxygenate them with variable positional specificity to the corresponding n-6 and n-9 hydroperoxy derivatives. Surprisingly, human ALOX15 as well as the corresponding orthologs of chimpanzee and orangutan, which oxygenates arachidonic acid mainly to 15S-H(p)ETE, exhibit a pronounced dual reaction specificity with DHA forming similar amounts of 14- and 17-H(p)DHA. Moreover, ALOX15 orthologs prefer DHA and EPA over AA when equimolar concentrations of n-3 and n-6 PUFA were supplied simultaneously. Taken together, these data indicate that the reaction specificity of mammalian LOX isoforms is variable and strongly depends on the chemistry of fatty acid substrates. Most mammalian ALOX15 orthologs exhibit dual positional specificity with highly unsaturated n-3 polyunsaturated fatty acids.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Mamíferos/metabolismo , Animales , Ácido Eicosapentaenoico/metabolismo , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/metabolismo , Ácidos Grasos Insaturados/metabolismo , Humanos , Sensibilidad y Especificidad
14.
Chemphyschem ; 17(20): 3321-3332, 2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27459330

RESUMEN

Molecular dynamics simulations and quantum mechanics/molecular mechanics calculations were performed on the in silico Leu597Ala/Ile663Ala double mutant of rabbit ALOX15 (12/15 lipoxygenase). The computational results suggested that subtle steric hindrance by the conserved Leu597 and C-terminal Ile663 residues disturbed H10 abstractions in wildtype ALOX15 (which abstracts H13), but if these two bulky residues were mutated to smaller ones, H10 abstraction was no longer impeded and the regioselectivity of the initial H-abstraction step was changed. However, site-directed mutagenesis with HPLC analysis of the products of the whole oxidation process showed that the regioselectivity of the hydroperoxidation was not altered. This disagreement may be explained by the conformational reorganization of the system needed to rotate the -OO. group from an antarafacial to a suprafacial arrangement prior to back-hydrogen transfer. After H10 abstraction and O2 insertion, the evolution of the peroxy radical at C12 was sterically impeded, whereas peroxyl group rotation at C15 (after H13 abstraction) could easily evolve to a suprafacial arrangement, which thus led to the final product. For this reason, the global regiospecificity was not affected in the mutant. These findings exemplify that the regioselectivity of initial hydrogen abstraction and the regioselectivity of the final product do not necessarily coincide (in fact, they can be opposite) for the hydroperoxidation of arachidonic acid catalyzed by a lipoxygenase.


Asunto(s)
Araquidonato 12-Lipooxigenasa/genética , Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Ácido Araquidónico/metabolismo , Biocatálisis , Hidrógeno/metabolismo , Animales , Ácido Araquidónico/química , Hidrógeno/química , Simulación de Dinámica Molecular , Mutación , Oxidación-Reducción , Teoría Cuántica , Conejos , Estereoisomerismo
15.
Biochim Biophys Acta ; 1842(10): 1460-6, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25025884

RESUMEN

5-Lipoxygenase (ALOX5) plays a key role in the biosynthesis of pro-inflammatory leukotrienes whereas 15-lipoxygenases (ALOX15) have been implicated in the formation of pro-resolving eicosanoids (lipoxins, resolvins). Recently, it has been suggested that a phosphorylation mimicking mutant (Ser663Asp) of a stabilized variant of human ALOX5 exhibits dominant arachidonic acid 15-lipoxygenase activity (>95%). To test whether similar alterations in the reaction specificity can also be observed for ALOX5 orthologs of other species we expressed wildtype and phosphorylation mimicking mutants (Ser271Asp, Ser523Asp, Ser663Asp, Ser663Glu) of human, mouse and zebrafish ALOX5 in pro- and eukaryotic overexpression systems and characterized their reaction specificities. We found that neither of the phosphorylation mimicking mutants produced significant amounts of 15-hydroperoxyeicosatetraenoic acid and the 5-lipoxygenation/15-lipoxygenation ratio for all wildtype and mutant enzyme species was lower than 100:2. Taken together, this data suggest that phosphorylation of native ALOX5 orthologs of different vertebrates may not induce major alterations in the reaction specificity and thus may not inverse their biological activity.

16.
Arch Biochem Biophys ; 584: 116-24, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26361973

RESUMEN

Pseudomonas aeruginosa (PA) expresses a secreted lipoxygenase (LOX), which oxygenates free arachidonic acid predominantly to 15S-H(p)ETE. The enzyme is capable of binding phospholipids at its active site and physically interacts with model membranes. However, its membrane oxygenase activity has not been quantified. To address this question, we overexpressed PA-LOX as intracellular his-tag fusion protein in Escherichia coli, purified it to electrophoretic homogeneity and compared its biomembrane oxygenase activity with that of rabbit ALOX15. We found that both enzymes were capable of oxygenating mitochondrial membranes to specific oxygenation products and 13S-H(p)ODE and 15S-H(p)ETE esterified to phosphatidylcholine and phosphatidylethanolamine were identified as major oxygenation products. When normalized to similar linoleic acid oxygenase activity, the rabbit enzyme exhibited a much more effective mitochondrial membrane oxygenase activity. In contrast, during long-term incubations (24 h) with red blood cells PA-LOX induced significant (50%) hemolysis whereas rabbit ALOX15 was more or less ineffective. These data indicate the principle capability of PA-LOX of oxygenating membrane bound phospholipids which is likely to alter the barrier function of the biomembranes. Although the membrane oxygenase activity was lower than the fatty acid oxygenase activity of PA-LOX red blood cell membrane oxygenation might be of biological relevance for P. aeruginosa septicemia.


Asunto(s)
Proteínas Bacterianas/metabolismo , Eritrocitos/metabolismo , Hemólisis , Lipooxigenasa/metabolismo , Fosfolípidos/metabolismo , Pseudomonas aeruginosa/enzimología , Animales , Proteínas Bacterianas/genética , Humanos , Lipooxigenasa/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Conejos
17.
Arch Biochem Biophys ; 571: 50-7, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25731857

RESUMEN

Among lipoxygenases ALOX15 orthologs are somewhat peculiar because of their capability of oxygenating polyenoic fatty acids even if they are incorporated in complex lipid-protein assemblies. ALOX15 orthologs of different species have been characterized before, but little is known about the corresponding rat enzyme. Since rats are frequently employed as models in biomedical research we expressed rat Alox15 as recombinant protein in pro- and eukaryotic expression systems and characterized the enzyme with respect to its enzymatic properties. The enzyme oxygenated free arachidonic acid mainly to 12S-HpETE with 15S-HpETE only contributing 10% to the product mixture. Multiple directed mutagenesis studies indicated applicability of the triad concept with particular importance of Leu353 and Ile593 as specificity determinants. Ala404Gly exchange induced subtle alterations in enantioselectivity suggesting partial applicability of the Coffa/Brash concept. Wildtype rat Alox15 and its 15-lipoxygenating Leu353Phe mutant are capable of oxygenating ester lipids of biomembranes and high-density lipoproteins. For the wildtype enzyme 13S-HODE and 12S-HETE were identified as major oxygenation products but for the Leu353Phe mutant 13S-HODE and 15S-HETE prevailed. These data indicate for the first time that mutagenesis of triad determinants modifies the reaction specificity of ALOX15 orthologs with free fatty acids and complex ester lipids in a similar way.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , Ácidos Grasos/metabolismo , Fosfolípidos/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/química , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/metabolismo , Secuencia de Aminoácidos , Animales , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/genética , Bovinos , Línea Celular Tumoral , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Membranas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Mutación , Conejos , Ratas , Estereoisomerismo , Especificidad por Sustrato
18.
Arch Biochem Biophys ; 565: 17-24, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25447821

RESUMEN

Mammalian lipoxygenases (LOXs) have been implicated in cell differentiation and in the biosynthesis of pro- and anti-inflammatory lipid mediators. The initial draft sequence of the Homo neanderthalensis genome (coverage of 1.3-fold) suggested defective leukotriene signaling in this archaic human subspecies since expression of essential proteins appeared to be corrupted. Meanwhile high quality genomic sequence data became available for two extinct human subspecies (H. neanderthalensis, Homo denisovan) and completion of the human 1000 genome project provided a comprehensive database characterizing the genetic variability of the human genome. For this study we extracted the nucleotide sequences of selected eicosanoid relevant genes (ALOX5, ALOX15, ALOX12, ALOX15B, ALOX12B, ALOXE3, COX1, COX2, LTA4H, LTC4S, ALOX5AP, CYSLTR1, CYSLTR2, BLTR1, BLTR2) from the corresponding databases. Comparison of the deduced amino acid sequences in connection with site-directed mutagenesis studies and structural modeling suggested that the major enzymes and receptors of leukotriene signaling as well as the two cyclooxygenase isoforms were fully functional in these two extinct human subspecies.


Asunto(s)
Genoma Humano/fisiología , Leucotrienos/genética , Lipooxigenasas/genética , Hombre de Neandertal/genética , Transducción de Señal/genética , Animales , Bases de Datos Genéticas , Humanos , Leucotrienos/metabolismo , Lipooxigenasas/metabolismo , Hombre de Neandertal/metabolismo , Especificidad de la Especie
19.
J Lipid Res ; 54(5): 1397-409, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23475662

RESUMEN

Lipoxygenases (LOX) have been implicated in biosynthesis of pro- and anti-inflammatory mediators, and a previous report suggested compromised leukotriene signaling in H. neanderthalensis. To search for corresponding differences in leukotriene biosynthesis, we screened the Neandertal genome for LOX genes and found that, as modern humans, this archaic hominid contains six LOX genes (nALOX15, nALOX12, nALOX5, nALOX15B, nALOX12B, and nALOXE3) and one pseudogene. In the Neandertal genome, 60-75% of the amino acids of the different human LOX isoforms have been identified, and the degree of identity varies between 96 and 99%. Most functional amino acids (iron ligands, specificity determinants, calcium and ATP-binding sites, membrane-binding determinants, and phosphorylation sites) are well conserved in the Neandertal LOX isoforms, and expression of selected neandertalized human LOX mutants revealed no major functional defects. However, in nALOX12 and nALOXE3, two premature stop codons were found, leading to inactive enzyme species. These data suggest that ALOX15, ALOX5, ALOX15B, and ALOX12B should have been present as functional enzymes in H. neanderthalensis and that in contrast to lower nonhuman primates (M. mulatta) and other mammals (mice, rats), this ancient hominid expressed a 15-lipoxygenating ALOX15. Expression of ALOXE3 and ALOX12 was compromised, which might have caused problems in epidermal differentiation.


Asunto(s)
Aminoácidos/química , Lipooxigenasa/química , Lipooxigenasa/metabolismo , Relación Estructura-Actividad , Secuencia de Aminoácidos , Aminoácidos/genética , Animales , Codón sin Sentido/genética , Genoma , Humanos , Ligandos , Lipooxigenasa/genética , Ratones , Mutación , Hombre de Neandertal , Primates/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA