Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Immunol ; 206(2): 329-334, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33277388

ABSTRACT

The COVID-19 pandemic has affected more than 20 million people worldwide, with mortality exceeding 800,000 patients. Risk factors associated with severe disease and mortality include advanced age, hypertension, diabetes, and obesity. Each of these risk factors pathologically disrupts the lipidome, including immunomodulatory eicosanoid and docosanoid lipid mediators (LMs). We hypothesized that dysregulation of LMs may be a defining feature of the severity of COVID-19. By examining LMs and polyunsaturated fatty acid precursor lipids in serum from hospitalized COVID-19 patients, we demonstrate that moderate and severe disease are separated by specific differences in abundance of immune-regulatory and proinflammatory LMs. This difference in LM balance corresponded with decreased LM products of ALOX12 and COX2 and an increase LMs products of ALOX5 and cytochrome p450. Given the important immune-regulatory role of LMs, these data provide mechanistic insight into an immuno-lipidomic imbalance in severe COVID-19.


Subject(s)
COVID-19 , Eicosanoids , Lipidomics , SARS-CoV-2 , Adult , Aged , Aged, 80 and over , Arachidonate 12-Lipoxygenase/immunology , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 5-Lipoxygenase/immunology , Arachidonate 5-Lipoxygenase/metabolism , Biomarkers/blood , COVID-19/blood , COVID-19/immunology , Cyclooxygenase 2/immunology , Cyclooxygenase 2/metabolism , Eicosanoids/blood , Eicosanoids/immunology , Female , Humans , Male , Middle Aged , SARS-CoV-2/immunology , SARS-CoV-2/metabolism
2.
Immunity ; 36(5): 834-46, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22503541

ABSTRACT

Noninflammatory clearance of apoptotic cells (ACs) is crucial to maintain self-tolerance. Here, we have reported a role for the enzyme 12/15-lipoxygenase (12/15-LO) as a central factor governing the sorting of ACs into differentially activated monocyte subpopulations. During inflammation, uptake of ACs was confined to a population of 12/15-LO-expressing, alternatively activated resident macrophages (resMΦ), which blocked uptake of ACs into freshly recruited inflammatory Ly6C(hi) monocytes in a 12/15-LO-dependent manner. ResMΦ exposed 12/15-LO-derived oxidation products of phosphatidylethanolamine (oxPE) on their plasma membranes and thereby generated a sink for distinct soluble receptors for ACs such as milk fat globule-EGF factor 8, which were essential for the uptake of ACs into inflammatory monocytes. Loss of 12/15-LO activity, in turn, resulted in an aberrant phagocytosis of ACs by inflammatory monocytes, subsequent antigen presentation of AC-derived antigens, and a lupus-like autoimmune disease. Our data reveal an unexpected key role for enzymatic lipid oxidation during the maintenance of self-tolerance.


Subject(s)
Apoptosis/immunology , Arachidonate 12-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/immunology , Self Tolerance/immunology , Animals , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Female , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism/immunology , Lipids/immunology , Macrophage Activation/immunology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Oxidation-Reduction
3.
Allergy ; 73(2): 350-360, 2018 02.
Article in English | MEDLINE | ID: mdl-28857185

ABSTRACT

BACKGROUND: As asthma progresses, the levels of IL-33 in serum are markedly increased and contribute to asthmatic development and exacerbation. Mast cells, one of the principal effector cells in the pathogenesis of asthma, express high levels of the IL-33 receptor ST2 and have been shown to be activated by IL-33. Thus, IL-33 stimulates mast cells to produce Th2-type cytokines such as IL-13, thus contributing to asthmatic development. However, the signaling mechanism for IL-33-induced synthesis of Th2 cytokines, particularly IL-13, has not been fully elucidated in mast cells. METHODS: The role of 5- or 12-LO in the IL-33-induced synthesis of IL-13 was investigated using knockdown or pharmacological inhibitors in bone marrow-derived mast cells (BMMCs) and animal model. RESULTS: Blockade of 5- or 12-LO significantly suppressed IL-33-induced synthesis of IL-13 in BMMCs. The subsequent action of 5- and 12-LO metabolites through their specific receptor, BLT2, was also critical for IL-33-induced synthesis of IL-13. We also demonstrated that the MyD88-p38 kinase cascade lies upstream of 5-/12-LO and that NF-κB lies downstream of 5-/12-LO to mediate the IL-33-induced synthesis of IL-13 in mast cells. Consistent with these findings, we observed that in an IL-33-administered asthmatic airway inflammation model, IL-13 levels were markedly increased in bronchoalveolar lavage fluid, but its levels were markedly suppressed by treatment with inhibitors of 5-LO, 12-LO or BLT2, further suggesting roles of 5-/12-LO in IL-33-induced IL-13 production. CONCLUSION: Our results suggest that "MyD88-5-/12-LO-BLT2-NF-κB" cascade significantly contributes to the IL-33-induced synthesis of IL-13 in mast cells, thus potentially contributing to asthmatic development and exacerbation.


Subject(s)
Arachidonate 12-Lipoxygenase/immunology , Arachidonate 5-Lipoxygenase/immunology , Asthma/immunology , Interleukin-13/immunology , Interleukin-33/immunology , Mast Cells/immunology , Animals , Asthma/blood , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Immunoblotting , Interleukin-13/metabolism , Interleukin-33/blood , Mast Cells/metabolism , Mice , Polymerase Chain Reaction
4.
Article in English | MEDLINE | ID: mdl-29763661

ABSTRACT

The profile of activation of lipid mediator (LM) pathways in asthmatic airway inflammation remains unclear. This experimental study quantified metabolite levels of ω3-, ω6- and ω9-derived polyunsaturated fatty acids in bronchoalveolar lavage fluid (BALF) after 4-weeks of repeated house dust mite (HDM) exposure in a murine (C57BL/6) asthma model. The challenge induced airway hyperresponsiveness, pulmonary eosinophil infiltration, but with low and unchanged mast cell numbers. Of the 112 screened LMs, 26 were increased between 2 to >25-fold in BALF with HDM treatment (p < 0.05, false discovery rate = 5%). While cysteinyl-leukotrienes were the most abundant metabolites at baseline, their levels did not increase after HDM treatment, whereas elevation of PGD2, LTB4 and multiple 12/15-lipoxygenase products, such as 5,15-DiHETE, 15-HEDE and 15-HEPE were observed. We conclude that this model has identified a global lipoxygenase activation signature, not linked to mast cells, but with aspects that mimic chronic allergic airway inflammation in asthma.


Subject(s)
Arachidonate 12-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/immunology , Arachidonate 5-Lipoxygenase/immunology , Asthma/immunology , Inflammation Mediators/immunology , Prostaglandins/immunology , Pyroglyphidae/immunology , Animals , Asthma/pathology , Bronchoalveolar Lavage , Disease Models, Animal , Mice , Mice, Transgenic
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(4): 371-381, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27480217

ABSTRACT

12/15-Lipoxygenase (12/15-LOX) mediates the enzymatic oxidation of polyunsaturated fatty acids, thereby contributing to the generation of various bioactive lipid mediators. Although 12/15-LOX has been implicated in the pathogenesis of multiple chronic inflammatory diseases, its physiologic functions seem to include potent immune modulatory properties that physiologically contribute to the resolution of inflammation and the clearance of inflammation-associated tissue damage. This review aims to give a comprehensive overview about our current knowledge on the role of this enzyme during the regulation of inflammation and immunity. This article is part of a Special Issue entitled: Lipid modification and lipid peroxidation products in innate immunity and inflammation edited by Christoph J. Binder.


Subject(s)
Arachidonate 12-Lipoxygenase/immunology , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/metabolism , Immunity/immunology , Inflammation/metabolism , Animals , Humans , Inflammation/immunology , Lipid Peroxidation/immunology , Lipid Peroxidation/physiology
6.
Infect Immun ; 84(3): 658-65, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26667839

ABSTRACT

Localized aggressive periodontitis (LAP) is a distinct form of early-onset periodontitis linked to periodontal infection with uncontrolled inflammation and leukocyte-mediated tissue destruction. The resolution of inflammation is an active process orchestrated by specialized proresolving lipid mediators (SPMs). Since the level of the Maresin pathway marker 14-hydroxy-docosahexaenoic acid (14-HDHA) was lower in activated peripheral blood from LAP patients, we investigated the Maresin 1 (MaR1) biosynthetic pathway in these subjects and its role in regulating phagocyte functions. Macrophages from LAP patients had a lower level of expression of 12-lipoxygenase (∼30%) and reduced MaR1 (LAP versus healthy controls [HC], 87.8 ± 50 pg/10(6) cells versus 239.1 ± 32 pg/10(6) cells). Phagocytosis by LAP macrophages was reduced ∼40% compared to that of HC, and killing of periodontal pathogens, including Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans, were similarly reduced. LAP neutrophils also displayed slower kinetics (∼30%) and decreased maximal phagocytosis (∼20% lower) with these pathogens than those of HC. The administration of MaR1 at 1 nM enhanced phagocytosis (31 to 65% increase), intracellular antimicrobial reactive oxygen species production (26 to 71% increase), bacterial killing of these periodontal pathogens (22 to 38% reduction of bacterial titers), and restored impairment of LAP phagocytes. Together, these results suggest that therapeutics targeting the Maresin pathway have clinical utility in treating LAP and other oral diseases associated with infection, inflammation, and altered phagocyte functions.


Subject(s)
Aggressive Periodontitis/immunology , Docosahexaenoic Acids/immunology , Leukocytes/immunology , Aggregatibacter actinomycetemcomitans/physiology , Aggressive Periodontitis/genetics , Aggressive Periodontitis/microbiology , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/immunology , Case-Control Studies , Cells, Cultured , Docosahexaenoic Acids/biosynthesis , Female , Humans , Macrophages/immunology , Macrophages/microbiology , Male , Phagocytosis , Porphyromonas gingivalis/physiology
7.
J Immunol ; 191(10): 5115-23, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24089193

ABSTRACT

Acute pulmonary infection by Streptococcus pneumoniae is characterized by high bacterial numbers in the lung, a robust alveolar influx of polymorphonuclear cells (PMNs), and a risk of systemic spread of the bacterium. We investigated host mediators of S. pneumoniae-induced PMN migration and the role of inflammation in septicemia following pneumococcal lung infection. Hepoxilin A3 (HXA3) is a PMN chemoattractant and a metabolite of the 12-lipoxygenase (12-LOX) pathway. We observed that S. pneumoniae infection induced the production of 12-LOX in cultured pulmonary epithelium and in the lungs of infected mice. Inhibition of the 12-LOX pathway prevented pathogen-induced PMN transepithelial migration in vitro and dramatically reduced lung inflammation upon high-dose pulmonary challenge with S. pneumoniae in vivo, thus implicating HXA3 in pneumococcus-induced pulmonary inflammation. PMN basolateral-to-apical transmigration in vitro significantly increased apical-to-basolateral transepithelial migration of bacteria. Mice suppressed in the expression of 12-LOX exhibited little or no bacteremia and survived an otherwise lethal pulmonary challenge. Our data suggest that pneumococcal pulmonary inflammation is required for high-level bacteremia and systemic infection, partly by disrupting lung epithelium through 12-LOX-dependent HXA3 production and subsequent PMN transepithelial migration.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Arachidonate 12-Lipoxygenase/metabolism , Neutrophils/immunology , Pneumococcal Infections/immunology , Transendothelial and Transepithelial Migration , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Arachidonate 12-Lipoxygenase/immunology , Bacillus subtilis , Bacteremia , Cell Line, Tumor , Cell Movement/immunology , Chemotactic Factors/metabolism , Humans , Inflammation/immunology , Lung/metabolism , Lung/microbiology , Lung Diseases/microbiology , Lung Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumococcal Infections/pathology , Streptococcus pneumoniae/pathogenicity
8.
J Immunol ; 189(10): 4960-9, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23045615

ABSTRACT

A feature shared by many inflammatory lung diseases is excessive neutrophilic infiltration. Neutrophil homing to airspaces involve multiple factors produced by several distinct cell types. Hepoxilin A(3) is a neutrophil chemoattractant produced by pathogen-infected epithelial cells that is hypothesized to facilitate neutrophil breach of mucosal barriers. Using a Transwell model of lung epithelial barriers infected with Pseudomonas aeruginosa, we explored the role of hepoxilin A(3) in neutrophil transepithelial migration. Pharmacological inhibitors of the enzymatic pathways necessary to generate hepoxilin A(3), including phospholipase A(2) and 12-lipoxygenase, potently interfere with P. aeruginosa-induced neutrophil transepithelial migration. Both transformed and primary human lung epithelial cells infected with P. aeruginosa generate hepoxilin A(3) precursor arachidonic acid. All four known lipoxygenase enzymes capable of synthesizing hepoxilin A(3) are expressed in lung epithelial cell lines, primary small airway epithelial cells, and human bronchial epithelial cells. Lung epithelial cells produce increased hepoxilin A(3) and lipid-derived neutrophil chemotactic activity in response to P. aeruginosa infection. Lipid-derived chemotactic activity is soluble epoxide hydrolase sensitive, consistent with hepoxilin A(3) serving a chemotactic role. Stable inhibitory structural analogs of hepoxilin A(3) are capable of impeding P. aeruginosa-induced neutrophil transepithelial migration. Finally, intranasal infection of mice with P. aeruginosa promotes enhanced cellular infiltrate into the airspace, as well as increased concentration of the 12-lipoxygenase metabolites hepoxilin A(3) and 12-hydroxyeicosa-5Z,8Z,10E,14Z-tetraenoic acid. Data generated from multiple models in this study provide further evidence that hepoxilin A(3) is produced in response to lung pathogenic bacteria and functions to drive neutrophils across epithelial barriers.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Arachidonate 12-Lipoxygenase/immunology , Blood-Air Barrier/immunology , Neutrophils/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Transendothelial and Transepithelial Migration/immunology , 8,11,14-Eicosatrienoic Acid/immunology , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Arachidonate 12-Lipoxygenase/metabolism , Blood-Air Barrier/metabolism , Blood-Air Barrier/microbiology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Humans , Male , Mice , Neutrophils/metabolism , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/metabolism , Pseudomonas Infections/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology
9.
Front Immunol ; 12: 687192, 2021.
Article in English | MEDLINE | ID: mdl-34093589

ABSTRACT

Dysregulated fatty acid metabolism is clinically associated with eosinophilic allergic diseases, including severe asthma and chronic rhinosinusitis. This study aimed to demonstrate the role of 12/15-lipoxygenase (12/15-LOX) in interleukin (IL)-33-induced eosinophilic airway inflammation; to this end, we used 12/15-LOX-deficient mice, which displayed augmented IL-33-induced lung inflammation, characterized by an increased number of infiltrated eosinophils and group 2 innate lymphoid cells (ILC2s) in the airway. Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based lipidomics revealed that the levels of a series of 12/15-LOX-derived metabolites were significantly decreased, and application of 14(S)-hydroxy docosahexaenoic acid (HDoHE), a major 12/15-LOX-derived product, suppressed IL-33-mediated eosinophilic inflammation in 12/15-LOX-deficient mice. Using bioactive lipid screening, we found that 14(S)-HDoHE and 10(S),17(S)-diHDoHE markedly attenuated ILC2 proliferation and cytokine production at micromolar concentration in vitro. In addition, maresin 1 (MaR1) and resolvin D1 (RvD1), 12/15-LOX-derived specialized proresolving mediators (SPMs), inhibited cytokine production of ILC2s at nanomolar concentration. These findings demonstrate the protective role of endogenous 12/15-LOX-derived lipid mediators in controlling ILC2-mediated eosinophilic airway inflammation and related diseases. Thus, 12/15-LOX-derived lipid mediators may represent a potential therapeutic strategy for ameliorating airway inflammation-associated conditions.


Subject(s)
Arachidonate 12-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/immunology , Docosahexaenoic Acids/pharmacology , Immunity, Innate/immunology , Interleukin-33/metabolism , Lymphocytes/immunology , Pneumonia/immunology , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Chromatography, Liquid , Interleukin-33/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/metabolism , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/metabolism , Tandem Mass Spectrometry
10.
J Immunol ; 181(11): 8044-52, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017997

ABSTRACT

Neutrophil transmigration across mucosal surfaces contributes to dysfunction of epithelial barrier properties, a characteristic underlying many mucosal inflammatory diseases. Thus, insight into the directional movement of neutrophils across epithelial barriers will provide important information relating to the mechanisms of such inflammatory disorders. The eicosanoid hepoxilin A(3), an endogenous product of 12-lipoxygenase activity, is secreted from the apical surface of the epithelial barrier and establishes a chemotactic gradient to guide neutrophils from the submucosa across epithelia to the luminal site of an inflammatory stimulus, the final step in neutrophil recruitment. Currently, little is known regarding how hepoxilin A(3) is secreted from the intestinal epithelium during an inflammatory insult. In this study, we reveal that hepoxilin A(3) is a substrate for the apical efflux ATP-binding protein transporter multidrug resistance-associated protein 2 (MRP2). Moreover, using multiple in vitro and in vivo models, we show that induction of intestinal inflammation profoundly up-regulates apical expression of MRP2, and that interfering with hepoxilin A(3) synthesis and/or inhibition of MRP2 function results in a marked reduction in inflammation and severity of disease. Lastly, examination of inflamed intestinal epithelia in human biopsies revealed up-regulation of MRP2. Thus, blocking hepoxilin A(3) synthesis and/or inhibiting MRP2 may lead to the development of new therapeutic strategies for the treatment of epithelial-associated inflammatory conditions.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Intestinal Diseases/immunology , Intestinal Mucosa/immunology , Multidrug Resistance-Associated Proteins/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , 8,11,14-Eicosatrienoic Acid/immunology , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Arachidonate 12-Lipoxygenase/immunology , Arachidonate 12-Lipoxygenase/metabolism , Gene Expression Regulation/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/biosynthesis , Neutrophils/metabolism , Neutrophils/pathology
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(11): 1669-1680, 2019 11.
Article in English | MEDLINE | ID: mdl-31349026

ABSTRACT

Recent studies suggest a potential role of bioactive lipids in acute kidney injury induced by lipopolysaccharide (LPS). The current study was designed to determine the profiling activities of various polyunsaturated fatty acid (PUFA) metabolizing enzymes, including lipoxygenases (LO), cyclooxygenase, and cytochrome P450 in the plasma of LPS-injected mice using LC-MS. Heat map analysis revealed that out of 126 bioactive lipids screened, only the 12/15-LO metabolite, 12-HETE, had a significant (2.24 ±â€¯0.4) fold increase relative to control (P = 0.0001) after Bonferroni Correction (BCF α = 0.003). We then determined the role of the 12/15-LO in LPS-induced acute kidney injury using genetic and pharmacological approaches. Treatment of LPS injected mice with the 12/15-LO inhibitor, baicalein, significantly reduced levels of renal injury and inflammation markers including urinary thiobarbituric acid reactive substance (TBARs), urinary monocyte chemoattractant protein-1 (MCP-1), renal interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). Similarly, knocking-out of 12/15-LO reduced levels of renal inflammation and injury markers elicited by LPS injection. Next, we tested whether exogenous supplementation with docosahexaenoic acid (DHA) as a substrate would divert the role of 12/15-LO from being pro-inflammatory to anti-inflammatory via increased production of the anti-inflammatory metabolite. DHA treatment restored the decreased in plasma level of resolvin D2 (RvD2) and reduced renal injury in LPS-injected mice whereas DHA treatment failed to provide any synergistic effects in reducing renal injury in LPS injected 12/15-LO knock-out mice. The ability of RvD2 to protect kidney against LPS-induced renal injury was further confirmed by exogenous RvD2 which significantly reduced the elevation in renal injury in LPS injected mice. These data suggest a double-edged sword role of 12/15-LO in LPS-induced acute renal inflammation and injury, depending on the type of substrate available for its activity.


Subject(s)
Acute Kidney Injury/immunology , Arachidonate 12-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/immunology , Inflammation/immunology , Lipopolysaccharides/immunology , Acute Kidney Injury/pathology , Animals , Inflammation/pathology , Male , Mice, Inbred C57BL
12.
Prog Lipid Res ; 45(4): 334-56, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16678271

ABSTRACT

12/15-Lipoxygenases (12/15-LOX) are members of the LOX family, which are expressed in mammals by monocytes and macrophages following induction by the T helper type 2 cytokines, interleukins-4 and -13. They oxygenate free polyenoic fatty acids but also ester lipids and even complex lipid-protein assemblies such as biomembranes and lipoproteins. The primary oxidation products are either reduced by glutathione peroxidases to corresponding hydroxy derivatives or metabolized into secondary oxidized lipids including leukotrienes, lipoxins and hepoxilins, which act as lipid mediators. Examination of knockout and transgenic animals revealed important roles for 12/15-LOX in inflammatory diseases, including atherosclerosis, cancer, osteoporosis, angiotension II-dependent hypertension and diabetes. In vitro studies suggested 12/15-LOX products as coactivators of peroxisomal proliferator activating-receptors (PPAR), regulators of cytokine generation, and modulators of gene expression related to inflammation resolution. Despite much work in this area, the biochemical mechanisms by which 12/15-LOX regulates physiological and pathological immune cell function are not fully understood. This review will summarize the biochemistry and tissue expression of 12/15-LOX and will describe the current knowledge regarding its immunobiology and regulation of inflammation.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Inflammation/enzymology , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/immunology , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/metabolism , Catalysis , Gene Expression Regulation, Enzymologic , Humans , Immunity, Cellular/physiology , Lipid Metabolism , Mice , Protein Processing, Post-Translational , Signal Transduction/immunology , Species Specificity , Vasculitis/enzymology
13.
Biochim Biophys Acta ; 1083(1): 89-93, 1991 Apr 24.
Article in English | MEDLINE | ID: mdl-1851637

ABSTRACT

Arachidonate 12-lipoxygenases of porcine and bovine leukocytes were different in substrate specificity and immunogenicity from the enzyme of bovine platelets (Arch. Biochem. Biophys. (1988) 266, 613). In order to extend the comparative studies on the two types of 12-lipoxygenase, we purified the enzyme from the cytosol of human platelets by immunoaffinity chromatography to a specific activity of about 0.3 mumol/min per mg protein at 37 degrees C. The purified enzyme was active with eicosapolyenoic acids and docosahexaenoic acid. Linoleic and linolenic acids were poor substrates in contrast to the high reactivity of the leukocyte enzymes with these octadecapolyenoic acids. The finding that the human platelet enzyme catalyzed 15-oxygenation of 5S-hydroxy-6,8,11,14-eicosatetraenoic acid, raised a question if lipoxins were produced by incubation of the enzyme with leukotriene A4. However, the leukotriene A4 was scarcely transformed to lipoxin isomers by 12-lipoxygenases of human and bovine platelets. In sharp contrast, the porcine and bovine leukocyte enzymes converted leukotriene A4 to various lipoxin isomers by the reaction rates of 3% and 2% of the arachidonate 12-oxygenation. Thus, 12-lipoxygenases of human and bovine platelets were catalytically distinct from the porcine and bovine leukocyte enzymes in terms of their reactivities not only with linoleic and linolenic acids, but also with leukotriene A4 as lipoxin precursor.


Subject(s)
Arachidonate 12-Lipoxygenase/blood , Blood Platelets/enzymology , Leukocytes/enzymology , Animals , Arachidonate 12-Lipoxygenase/immunology , Arachidonate 12-Lipoxygenase/isolation & purification , Cattle , Chromatography, Affinity , Humans , Isomerism , Kinetics , Leukotriene A4 , Leukotrienes/biosynthesis , Leukotrienes/blood , Substrate Specificity , Swine
14.
J Histochem Cytochem ; 48(10): 1411-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10990494

ABSTRACT

There are three isoforms of arachidonate 12-lipoxygenase in mammals: platelet, leukocyte, and epidermal types. We found in this study that the leukocyte-type enzyme was present in rat pineal gland, lung, spleen, aorta, adrenal gland, spinal cord, and pancreas, as assessed by RT-PCR. Immunohistochemical analysis showed that the enzyme was localized in macrophages in lung and spleen, alpha-cells of pancreatic islet, zona glomerulosa cells of adrenal cortex, and neuronal cells of spinal cord and superior cervical ganglion. The presence of the 12-lipoxygenase in pancreatic alpha-cells was confirmed by glucagon staining in a consecutive section. We overexpressed the leukocyte-type 12-lipoxygenase cDNA in a glucagon-secreting alphaTC clone 6 cell line that had been established from a transgenic mouse. Glucagon secretion was stimulated by approximately twofold in the 12-lipoxygenase-expressing cells compared to the mock-transfected and original cells. The results suggest that the 12-lipoxygenase of the leukocyte type augments glucagon secretion from pancreatic islets.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Glucagon/metabolism , Animals , Antibody Specificity , Arachidonate 12-Lipoxygenase/immunology , Blotting, Western , Cell Line , Immunohistochemistry , Islets of Langerhans/metabolism , Leukocytes/enzymology , Macrophages/metabolism , Mice , Mice, Transgenic , Organ Specificity , Pineal Gland/enzymology , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
15.
Sci Rep ; 3: 1540, 2013.
Article in English | MEDLINE | ID: mdl-23528921

ABSTRACT

The mechanisms underlying asthmatic airway epithelial injury are not clear. 12/15-lipoxygenase (an ortholog of human 15-LOX-1), which is induced by IL-13, is associated with mitochondrial degradation in reticulocytes at physiological conditions. In this study, we showed that 12/15-LOX expressed in nonepithelial cells caused epithelial injury in asthma pathogenesis. While 12/15-LOX overexpression or IL-13 administration to naïve mice showed airway epithelial injury, 12/15-LOX knockout/knockdown in allergic mice reduced airway epithelial injury. The constitutive expression of 15-LOX-1 in bronchial epithelia of normal human lungs further indicated that epithelial 15-LOX-1 may not cause epithelial injury. 12/15-LOX expression is increased in various inflammatory cells in allergic mice. Though non-epithelial cells such as macrophages or fibroblasts released 12/15-LOX metabolites upon IL-13 induction, bronchial epithelia didn't release. Further 12-S-HETE, arachidonic acid metabolite of 12/15-LOX leads to epithelial injury. These findings suggested 12/15-LOX expressed in non-epithelial cells such as macrophages and fibroblasts leads to bronchial epithelial injury.


Subject(s)
Arachidonate 12-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/immunology , Asthma/immunology , Fibroblasts/immunology , Macrophages/immunology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/blood , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/immunology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , 3T3 Cells , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Asthma/genetics , Asthma/metabolism , Blotting, Western , Cell Line , Cytochromes c/immunology , Cytochromes c/metabolism , Epithelium/drug effects , Epithelium/immunology , Epithelium/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Immunohistochemistry , Interleukin-13/administration & dosage , Interleukin-13/immunology , Interleukin-13/pharmacology , Lactones , Linoleic Acids/blood , Linoleic Acids/immunology , Linoleic Acids/metabolism , Lung/immunology , Lung/metabolism , Lung/ultrastructure , Macrophages/drug effects , Macrophages/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/immunology , Mitochondria/metabolism , Mitochondria/physiology , Sesquiterpenes, Eudesmane
17.
J Exp Med ; 206(7): 1565-74, 2009 Jul 06.
Article in English | MEDLINE | ID: mdl-19546247

ABSTRACT

To identify a novel target for the treatment of heart failure, we examined gene expression in the failing heart. Among the genes analyzed, Alox15 encoding the protein 12/15 lipoxygenase (LOX) was markedly up-regulated in heart failure. To determine whether increased expression of 12/15-LOX causes heart failure, we established transgenic mice that overexpressed 12/15-LOX in cardiomyocytes. Echocardiography showed that Alox15 transgenic mice developed systolic dysfunction. Cardiac fibrosis increased in Alox15 transgenic mice with advancing age and was associated with the infiltration of macrophages. Consistent with these observations, cardiac expression of monocyte chemoattractant protein 1 (MCP-1) was up-regulated in Alox15 transgenic mice compared with wild-type mice. Treatment with 12-hydroxy-eicosatetraenoic acid, a major metabolite of 12/15-LOX, increased MCP-1 expression in cardiac fibroblasts and endothelial cells but not in cardiomyocytes. Inhibition of MCP-1 reduced the infiltration of macrophages into the myocardium and prevented both systolic dysfunction and cardiac fibrosis in Alox15 transgenic mice. Likewise, disruption of 12/15-LOX significantly reduced cardiac MCP-1 expression and macrophage infiltration, thereby improving systolic dysfunction induced by chronic pressure overload. Our results suggest that cardiac 12/15-LOX is involved in the development of heart failure and that inhibition of 12/15-LOX could be a novel treatment for this condition.


Subject(s)
Arachidonate 12-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/immunology , Heart Failure , Inflammation , Myocardium , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression Profiling , Heart/physiology , Heart Failure/enzymology , Heart Failure/immunology , Humans , Inflammation/enzymology , Inflammation/immunology , Lipoxygenase Inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardium/enzymology , Myocardium/immunology , Rats , Rats, Inbred Dahl , Rats, Wistar , Sodium, Dietary
18.
J Biol Chem ; 282(28): 20151-63, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17519227

ABSTRACT

12/15-Lipoxygenase (LOX) mediates immune-regulatory activities not accounted for by its known free acid eicosanoids, suggesting that additional lipids may be generated by activated cells. To characterize novel LOX-derived lipids, a lipidomic approach was utilized. Ionophore-activated interleukin-4-treated human peripheral monocytes generated up to 10-fold more esterified 15-hydroxyeicosatetraenoic acid (15-HETE) than free in a phosphatidylinositol 3-kinase- and protein kinase C-sensitive manner. Precursor scanning electrospray ionization/tandem spectroscopy for m/z 319 (HETE, [M-H](-)) showed 4 ions at m/z 738, 764, 766, and 782 that were identified using tandem spectroscopy and MS3 as specific diacyl and plasmalogen 15-HETE phosphatidylethanolamines. Using H (18)(2)O water, the compounds were shown to form by direct oxidation of endogenous phosphatidylethanolamine (PE) by 15-LOX, with PE being the preferred phospholipid pool containing 15-HETE. Similarly, human platelets generated 4 analogous PE lipids that contained 12-HETE and increased significantly in response to ionophore, collagen, or convulxin. These products were retained in the cells, in contrast to free acids, which are primarily secreted. Precursor scanning of platelet extracts for the major platelet-derived prostanoid, thromboxane B2 (m/z 369.2), did not reveal PE esters, indicating that this modification is restricted to the LOX pathway. In summary, we show formation of PE-esterified HETEs in immune cells that may contribute to LOX signaling in inflammation.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Blood Platelets/enzymology , Monocytes/enzymology , Phosphatidylethanolamines/metabolism , Platelet Activation , Signal Transduction , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/immunology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Arachidonate 12-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/metabolism , Blood Platelets/immunology , Coculture Techniques , Collagen/pharmacology , Crotalid Venoms/pharmacology , Humans , Hydroxyeicosatetraenoic Acids/immunology , Hydroxyeicosatetraenoic Acids/metabolism , Interleukin-4/pharmacology , Ionophores/pharmacology , Lectins, C-Type , Monocytes/immunology , Oxidation-Reduction/drug effects , Phosphatidylethanolamines/immunology , Platelet Activation/drug effects , Platelet Activation/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Spectrometry, Mass, Electrospray Ionization
19.
J Immunol ; 176(1): 265-74, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365418

ABSTRACT

IL-12 drives type I immune responses and can mediate chronic inflammation that leads to host defense as well as disease. Recently, we discovered a novel role for 12/15-lipoxygenase (12/15-LO) in mediating IL-12p40 expression in atherosclerotic plaque and in isolated macrophages. We now demonstrate that 12/15-LO regulates IL-12 family cytokine production in a cell-type and stimulus-restricted fashion. LPS-stimulated elicited peritoneal macrophages derived from 12/15-LO-deficient (Alox15) mice produced reduced IL-12 and IL-23 levels, but comparable amounts of several other inflammatory mediators tested. Furthermore, LPS stimulation triggered an increase in wild-type macrophage 12/15-LO activity, whereas pharmacological inhibition of 12/15-LO activity suppressed LPS-induced IL-12 production in wild-type macrophages. 12/15-LO-deficient macrophages also produced reduced levels of IL-12 in response to TLR2 stimulation, but not in response to CpG (TLR9) or CD40/CD40L-mediated activation. In contrast to our previous finding of reduced IL-12 production in the setting of atherosclerosis, we found that comparable IL-12 levels were produced in Alox15 and wild-type mice during an acute response to LPS in vivo. This paradox may be explained by normal production of IL-12 by 12/15-LO-deficient neutrophils and dendritic cells, which are major sources of IL-12 during acute inflammation. Finally, we detected selectively decreased association of the transcription factors IFN consensus sequence binding protein and NF-kappaB with the IL-12p40 promoter in 12/15-LO-deficient macrophages. Taken together, these findings reveal a highly selective pathway to IL-12 production that may prove a useful target in chronic inflammation while sparing the acute response to infection.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Interleukin-12/biosynthesis , Macrophages, Peritoneal/immunology , Animals , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 12-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/immunology , Cells, Cultured , Cytokines/analysis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interferon Regulatory Factors , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12/immunology , Macrophage Activation/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , NF-kappa B/immunology , NF-kappa B/metabolism , Protein Subunits/biosynthesis , Protein Subunits/immunology , Reverse Transcriptase Polymerase Chain Reaction
20.
Article in English | MEDLINE | ID: mdl-1825573

ABSTRACT

Human tracheal epithelial cells contain an arachidonate 15-lipoxygenase, while the same cells from animals (including bovine, ovine, canine, porcine) cells express a 12-lipoxygenase. The epithelial 12-lipoxygenase is antigenically related to the leukocyte 12-lipoxygenase but is biochemically distinct from platelet and leukocyte forms of the enzyme, in that it is more efficient at metabolizing a wider array of fatty acid substrates. We have suggested that this lipoxygenase heterogeneity may provide a basis for different functional roles for the enzyme in different cell types. In addition, animal epithelial 12-lipoxygenase and human epithelial 15-lipoxygenase are antigenically related and have similar but distinct distributions in the lung. Our findings might suggest that the species diversity for epithelial lipoxygenases represents molecular divergence within a family of closely related genes with perhaps closely related functions.


Subject(s)
Arachidonate 12-Lipoxygenase/analysis , Arachidonate 15-Lipoxygenase/analysis , Epithelium/enzymology , Animals , Arachidonate 12-Lipoxygenase/immunology , Arachidonate 15-Lipoxygenase/immunology , Cattle , Dogs , Enzyme Induction , Humans , Sheep , Species Specificity , Swine , Trachea/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL