Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Pregnancy Hypertens ; 26: 75-78, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34563982

ABSTRACT

BACKGROUND: LDA triggers biosynthesis of endogenous anti-inflammatory molecules, aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4), which may counteract inflammatory process of preeclampsia (PE), and play role in LDA's mechanism of action in PE prevention in high-risk patients. OBJECTIVE: Investigate the effects of daily LDA on levels of 15-epi-LXA4 in pregnancies at high-risk for developing PE. MATERIALS AND METHODS: Secondary analysis of multi-centered randomized controlled trial investigating effects of daily LDA (60 mg) in high-risk pregnancies. Maternal samples were drawn at three points: before LDA initiation (13-26 weeks' gestation), 24-28 weeks' gestation (at least two weeks after LDA) and 34-36 weeks' gestation. 15-epi-LXA4 levels were measured by ELISA. RESULTS: Analysis included 82 patients: 63 receiving daily LDA and 29 receiving daily placebo starting between 13 and 25 weeks gestation. Prior to randomization, baseline 15-epi-LXA4 levels were similar between both groups (75.9 pg/mL [IQR; 63.8-114.0] vs 136.2 pg/mL [52.4-476.2]; p = 0.10). Patients receiving daily LDA were noted to have significantly increased levels of 15-epi-LXA4 after LDA administration (136.2 pg/mL [IQR; 52.4-476.2] vs 1758.2 pg/mL [905.4-6638.5]; p < 0.001). They also had higher 15-epi-LXA4 levels compared to those receiving placebo at 24-28 weeks' (50.3 [38.1-94.2] vs 1758.2 [905.4-6638.5]; p < 0.001 and 34-38 weeks' gestation (57.9 [41.9-76.7] vs 2310.3 pg/mL [656.9-10609.4]; p < 0.001). After LDA administration in the second trimester, patients who developed PE had decrease in 15-epi-LXA4 levels compared to those without PE (942 pg/mL [348.3-1810.3] vs 1758.2 pg/mL [905.4-6638.5]; p = 0.129). CONCLUSION: Daily LDA administration increases 15-epi-LXA4 levels in high-risk pregnancies for PE. In LDA group, pregnancies complicated by PE have lower levels of 15-epi-LXA4 compared to pregnancies without PE.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Pre-Eclampsia/prevention & control , Adult , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Aspirin/administration & dosage , Female , Humans , Lipoxins/biosynthesis , Lipoxins/blood , Pregnancy , Pregnancy, High-Risk
2.
PLoS One ; 15(12): e0242543, 2020.
Article in English | MEDLINE | ID: mdl-33326419

ABSTRACT

Clinical studies using a range of omega-3 supplements have yielded conflicting results on their efficacy to control inflammation. Omega-3 fatty acids are substrate for the formation of potent immune-protective mediators, termed as specialized pro-resolving mediators (SPM). Herein, we investigated whether observed differences in the potencies of distinct omega-3 supplements were linked with their ability to upregulate SPM formation. Using lipid mediator profiling we found that four commercially available supplements conferred a unique SPM signature profile to human macrophages, with the overall increases in SPM concentrations being different between the four supplements. These increases in SPM concentrations were linked with an upregulation of macrophage phagocytosis and a decreased uptake of oxidized low-density lipoproteins. Pharmacological inhibition of two key SPM biosynthetic enzymes 5-Lipoxygenase or 15-Lipoxygenase reversed the macrophage-directed actions of each of the omega-3 supplements. Furthermore, administration of the two supplements that most potently upregulated macrophage SPM formation and reprogrammed their responses in vitro, to APOE-/- mice fed a western diet, increased plasma SPM concentrations and reduced vascular inflammation. Together these findings support the utility of SPM as potential prognostic markers in determining the utility of a given supplement to regulate macrophage responses and inflammation.


Subject(s)
Atherosclerosis/prevention & control , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Leukotrienes/biosynthesis , Lipoxins/biosynthesis , Macrophages/drug effects , Prostaglandins/biosynthesis , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/immunology , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/immunology , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/immunology , Atherosclerosis/etiology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Diet, Western/adverse effects , Fatty Acids, Omega-3/metabolism , Female , Gene Expression , Humans , Leukotrienes/immunology , Lipoproteins, LDL/antagonists & inhibitors , Lipoproteins, LDL/pharmacology , Lipoxins/immunology , Lipoxygenase Inhibitors/pharmacology , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Knockout, ApoE , Phagocytosis/drug effects , Primary Cell Culture , Principal Component Analysis , Prostaglandins/immunology
3.
J Phys Chem B ; 124(50): 11406-11418, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33274949

ABSTRACT

Chronic inflammation is now widely recognized to play important roles in many commonly occurring diseases, including COVID-19. The resolution response to this chronic inflammation is an active process governed by specialized pro-resolving mediators (SPMs) like the lipid mediators known as lipoxins. The biosynthesis of lipoxins is catalyzed by several lipoxygenases (LOXs) from arachidonic acid. However, the molecular details of the mechanisms involved are not well known yet. In this paper, we have combined molecular dynamics (MD) simulations and quantum mechanics/molecular mechanics (QM/MM) calculations to analyze how reticulocyte 15-LOX-1 catalyzes the production of lipoxins from 5(S),15(S)-diHpETE. Our results indicate that the dehydration mechanism from 5(S),15(S)-diHpETE, via the formation of an epoxide, presents huge energy barriers even though it was one of the two a priori synthetic proposals. This result is compatible with the fact that no epoxide has been directly detected as an intermediate in the catalytic formation of lipoxins from 5(S),15(S)-diHpETE. Conversely, the oxygenation of 5(S),15(S)-diHpETE at C14 is feasible because there is an open channel connecting the protein surface with this carbon atom, and the energy barrier for oxygen addition through this channel is small. The analysis of the following steps of this mechanism, leading to the corresponding hydroperoxide at the 15-LOX-1 active site, indicates that the oxygenation mechanism will lead to the formation of lipoxinB4 after the final action of a reductase. In contrast, our calculations are in agreement with experiments that lipoxinA4 cannot derive from 5(S),15(S)-diHpETE by either of the two proposed mechanisms and that 5(S),15(S)-diHETE is not an intermediate of lipoxin biosynthesis catalyzed by 15-LOX-1.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Leukotrienes/biosynthesis , Lipid Peroxides/biosynthesis , Lipoxins/biosynthesis , Reticulocytes/enzymology , Biosynthetic Pathways , COVID-19/complications , Catalysis , Humans , Inflammation/etiology , Inflammation/metabolism , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Oxygen/chemistry , Quantum Theory
4.
Elife ; 92020 03 02.
Article in English | MEDLINE | ID: mdl-32118582

ABSTRACT

The eicosanoid lipoxin A4 (LXA4) has emerging roles in lymphocyte-driven diseases. We identified reduced LXA4 levels in posterior segment uveitis patients and investigated the role of LXA4 in the pathogenesis of experimental autoimmune uveitis (EAU). Immunization for EAU with a retinal self-antigen caused selective downregulation of LXA4 in lymph nodes draining the site of immunization, while at the same time amplifying LXA4 in the inflamed target tissue. T cell effector function, migration and glycolytic responses were amplified in LXA4-deficient mice, which correlated with more severe pathology, whereas LXA4 treatment attenuated disease. In vivo deletion or supplementation of LXA4 identified modulation of CC-chemokine receptor 7 (CCR7) and sphingosine 1- phosphate receptor-1 (S1PR1) expression and glucose metabolism in CD4+ T cells as potential mechanisms for LXA4 regulation of T cell effector function and trafficking. Our results demonstrate the intrinsic lymph node LXA4 pathway as a significant checkpoint in the development and severity of adaptive immunity.


Subject(s)
Autoimmunity/physiology , Eye/immunology , Lipoxins/physiology , Lymph Nodes/physiology , Retina/immunology , Animals , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Homeostasis , Humans , Lipoxins/biosynthesis , Lipoxins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR7/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Uveitis/immunology
5.
Am J Reprod Immunol ; 83(2): e13207, 2020 02.
Article in English | MEDLINE | ID: mdl-31696583

ABSTRACT

PROBLEM: Oxidative stress and inflammation are key events leading to pre-eclampsia, involved in several maternal deaths. Low doses of acetylsalicylic acid (ASA) are used in the prevention and treatment of pre-eclampsia. The synthesis of aspirin-triggered lipoxin (ATL) by cyclooxygenase-2 acetylation is an alternative mechanism of ASA, which could explain the effectiveness of ASA treatments. The aim of this study was to evaluate the role of ASA, salicylates, and ATL in the modulation of the oxidative and inflammatory responses induced by plasma from women with pre-eclampsia. METHOD OF STUDY: Plasma from 14 women with pre-eclampsia and 17 normotensive pregnant women was probed for inducing oxidative and inflammatory responses on endothelial cells and U937 promonocytes. The role of ATL, ASA, and salicylic acid (SA) on these events was evaluated. RESULTS: Plasma from women with pre-eclampsia induced TBARS and nitrotyrosine production on endothelial and U937 cells. Pre-treatment with both ATL and ASA decreased the TBARS production, while ATL decreased the nitrotyrosine. Pre-eclamptic plasma augmented the translocation of NF-kB on U937 cells, which decreased by a high dose of ASA and SA. Finally, the pre-eclamptic plasma increased the adhesion of leukocytes-PMN and monocytes-to endothelium, and we were able to determine a state of resolution of inflammation, since ATL decreased the PMN adhesion, and conversely, it increased the monocytes adhesion to endothelium. CONCLUSION: Together, these results suggest that ATL could explain the beneficial actions of ASA and support further research on mechanisms, real efficacy, and rational use of ASA in pre-eclampsia.


Subject(s)
Aspirin/therapeutic use , Lipoxins/blood , Oxidative Stress/drug effects , Pre-Eclampsia/blood , Salicylic Acid/blood , Acetylation , Adolescent , Adult , Aspirin/blood , Aspirin/pharmacology , Cell Adhesion/drug effects , Cyclooxygenase 2/blood , Female , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/blood , Lipoxins/biosynthesis , Lipoxins/pharmacology , NF-kappa B/metabolism , Neutrophils/drug effects , Pre-Eclampsia/drug therapy , Pre-Eclampsia/prevention & control , Pregnancy , Protein Processing, Post-Translational/drug effects , Salicylic Acid/pharmacology , Thiobarbituric Acid Reactive Substances/analysis , Tyrosine/analogs & derivatives , Tyrosine/biosynthesis , U937 Cells , Young Adult
6.
Biochemistry ; 57(48): 6726-6734, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30407793

ABSTRACT

The reaction of 5 S,15 S-dihydroperoxyeicosatetraenoic acid (5,15-diHpETE) with human 5-lipoxygenase (LOX), human platelet 12-LOX, and human reticulocyte 15-LOX-1 was investigated to determine the reactivity and relative rates of producing lipoxins (LXs). 5-LOX does not react with 5,15-diHpETE, although it can produce LXA4 when 15-HpETE is the substrate. In contrast, both 12-LOX and 15-LOX-1 react with 5,15-diHpETE, forming specifically LXB4. For 12-LOX and 5,15-diHpETE, the kinetic parameters are kcat = 0.17 s-1 and kcat/ KM = 0.011 µM-1 s-1 [106- and 1600-fold lower than those for 12-LOX oxygenation of arachidonic acid (AA), respectively]. On the other hand, for 15-LOX-1 the equivalent parameters are kcat = 4.6 s-1 and kcat/ KM = 0.21 µM-1 s-1 (3-fold higher and similar to those for 12-HpETE formation by 15-LOX-1 from AA, respectively). This contrasts with the complete lack of reaction of 15-LOX-2 with 5,15-diHpETE [Green, A. R., et al. (2016) Biochemistry 55, 2832-2840]. Our data indicate that 12-LOX is markedly inferior to 15-LOX-1 in catalyzing the production of LXB4 from 5,15-diHpETE. Platelet aggregation was inhibited by the addition of 5,15-diHpETE, with an IC50 of 1.3 µM; however, LXB4 did not significantly inhibit collagen-mediated platelet activation up to 10 µM. In summary, LXB4 is the primary product of 12-LOX and 15-LOX-1 catalysis, if 5,15-diHpETE is the substrate, with 15-LOX-1 being 20-fold more efficient than 12-LOX. LXA4 is the primary product with 5-LOX but only if 15-HpETE is the substrate. Approximately equal proportions of LXA4 and LXB4 are produced by 12-LOX but only if LTA4 is the substrate, as described previously [Sheppard, K. A., et al. (1992) Biochim. Biophys. Acta 1133, 223-234].


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Arachidonate 5-Lipoxygenase/metabolism , Leukotrienes/metabolism , Lipid Peroxides/metabolism , Lipoxins/biosynthesis , Biocatalysis , Biosynthetic Pathways , Blood Platelets/metabolism , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Kinetics , Leukocytes/metabolism , Reticulocytes/metabolism , Substrate Specificity
7.
Biomed Res Int ; 2017: 2014583, 2017.
Article in English | MEDLINE | ID: mdl-29181388

ABSTRACT

Recruitment of neutrophil granulocytes to sites of infectious tissue damage is an early event in innate immune responses. Following chemotactic signals neutrophils establish a first line of defense in a swarm-like manner. Intracellular pathogens such as Leishmania major can, however, evade neutrophil-mediated killing and survive inside neutrophils. To achieve this the parasites evolved potent evasion mechanisms. Since neutrophils are a major source of inflammation regulating lipid mediators, we hypothesized that intracellular infection modifies the release of pro- and anti-inflammatory lipid mediators like leukotriene B4 (LTB4) and lipoxin A4 (LXA4), respectively. In the present study, we demonstrated in vitro that L. major-infected primary human neutrophils release an increased amount of LTB4, whereas LXA4 liberation is reduced during the first hours of infection. To investigate whether lipid mediator modulation is a common feature in intracellular infections, we tested the impact of an infection with Anaplasma phagocytophilum. Similarly to L. major, neutrophil infection with A. phagocytophilum led to an enhanced release of LTB4 and decreased LXA4 production. Together, our findings indicate that intracellular infections modulate the lipid mediator profile of neutrophils. This effect is likely to contribute to the survival of the pathogens in neutrophils and to the outcome of the infections.


Subject(s)
Leishmania major , Leishmaniasis, Cutaneous/metabolism , Leukotriene B4/biosynthesis , Lipoxins/biosynthesis , Neutrophils/metabolism , Neutrophils/parasitology , Humans
8.
Biofactors ; 43(2): 251-271, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27862450

ABSTRACT

OBJECTIVE: We studied whether polyunsaturated fatty acids (PUFAs) can protect rat insulinoma (RIN5F) cells against alloxan-induced apoptosis in vitro and type 1 diabetes mellitus (type 1 DM) in vivo and if so, mechanism of this beneficial action. MATERIAL AND METHODS: In vitro study was conducted using RIN5F cells while in vivo study was performed in Wistar rats. The effect of PUFAs, cyclo-oxygenase and lipoxygenase inhibitors, various eicosanoids and PUFAs metabolites: lipoxin A4 (LXA4), resolvin D2 and protectin against alloxan-induced cytotoxicity to RIN5F cells and type 1 DM was studied. Expression of PDX1, P65 NF-kB and IKB in RIN5F cells and Nrf2, GLUT2, COX2, iNOS protein levels in the pancreatic tissue and plasma glucose, insulin and tumor necrosis factor-α and antioxidants, lipid peroxides and nitric oxide were measured. RESULTS: Of all, arachidonic acid (AA) was found to be the most effective against alloxan-induced cytotoxicity to RIN5F cells and preventing type 1 DM. Both cyclo-oxygenase and lipoxygenase inhibitors did not block the beneficial actions of AA in vitro and in vivo. Alloxan inhibited LXA4 production by RIN5F cells and in alloxan-induced type 1 DM Wistar rats. AA-treatment restored LXA4 levels to normal both in vitro and in vivo. LXA4 protected RIN5F cells against alloxan-induced cytotoxicity and prevented type 1 DM and restored expression of Nrf2, Glut2, COX2, and iNOS genes and abnormal antioxidants to near normal. DISCUSSION: AA seems to bring about its beneficial actions against alloxan-induced cytotoxicity and type 1 DM by enhancing the production of LXA4. © 2016 BioFactors, 43(2):251-271, 2017.


Subject(s)
Arachidonic Acid/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Lipoxins/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Gene Expression Regulation, Neoplastic , Humans , Insulinoma , Lipoxins/biosynthesis , Neoplasm Proteins/biosynthesis , Rats
9.
Biochim Biophys Acta ; 1861(11): 1681-1692, 2016 11.
Article in English | MEDLINE | ID: mdl-27500637

ABSTRACT

Pseudomonas aeruginosa expresses a secreted LOX-isoform (PA-LOX, LoxA) capable of oxidizing polyenoic fatty acids to hydroperoxy derivatives. Here we report high-level expression of this enzyme in E. coli and its structural and functional characterization. Recombinant PA-LOX oxygenates polyenoic fatty acids including eicosapentaenoic acid and docosahexaenoic acid to the corresponding (n-6)S-hydroperoxy derivatives. This reaction involves abstraction of the proS-hydrogen from the n-8 bisallylic methylene. PA-LOX lacks major leukotriene synthase activity but converts 5S-HETE and 5S,6R/S-DiHETE to anti-inflammatory and pro-resolving lipoxins. It also exhibits phospholipid oxygenase activity as indicated by the formation of a specific pattern of oxygenation products from different phospholipid subspecies. Multiple mutagenesis studies revealed that PA-LOX does not follow classical concepts explaining the reaction specificity of mammalian LOXs. The crystal structure of PA-LOX was solved with resolutions of up to 1.48Å and its polypeptide chain is folded as single domain. The substrate-binding pocket consists of two fatty acid binding subcavities and lobby. Subcavity-1 contains the catalytic non-heme iron. A phosphatidylethanolamine molecule occupies the substrate-binding pocket and its sn1 fatty acid is located close to the catalytic non-heme iron. His377, His382, His555, Asn559 and the C-terminal Ile685 function as direct iron ligands and a water molecule (hydroxyl) completes the octahedral ligand sphere. Although the biological relevance of PA-LOX is still unknown its functional characteristics (lipoxin synthase activity) implicate this enzyme in a bacterial evasion strategy aimed at downregulating the hosts' immune system.


Subject(s)
Lipoxygenase/chemistry , Lipoxygenase/metabolism , Pseudomonas aeruginosa/enzymology , Animals , Arachidonic Acid/chemistry , Arachidonic Acid/metabolism , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Enzyme Stability , Fatty Acids/metabolism , Kinetics , Leukotrienes/metabolism , Ligands , Linoleic Acid/chemistry , Linoleic Acid/metabolism , Lipoxins/biosynthesis , Models, Molecular , Mutant Proteins/metabolism , Oxidation-Reduction , Rabbits , Recombinant Proteins/metabolism , Stereoisomerism , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity , Temperature
10.
Proc Natl Acad Sci U S A ; 113(30): E4266-75, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27412860

ABSTRACT

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.


Subject(s)
Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Evolution, Molecular , Lipoxins/biosynthesis , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Arachidonate 15-Lipoxygenase/chemistry , Catalytic Domain , Humans , Lipoxins/chemistry , Mice , Mutation , Primates , Rabbits , Rats , Species Specificity , Substrate Specificity , Swine
11.
Mediators Inflamm ; 2015: 852574, 2015.
Article in English | MEDLINE | ID: mdl-26635449

ABSTRACT

Cysteinyl leukotrienes (CysLTs) and lipoxins (LXs) are lipid mediators that control inflammation, with the former inducing and the latter inhibiting this process. Because the role played by these mediators in paracoccidioidomycosis was not investigated, we aimed to characterize the role of CysLT in the pulmonary infection developed by resistant (A/J) and susceptible (B10.A) mice. 48 h after infection, elevated levels of pulmonary LTC4 and LXA4 were produced by both mouse strains, but higher levels were found in the lungs of susceptible mice. Blocking the CysLTs receptor by MTL reduced fungal loads in B10.A, but not in A/J mice. In susceptible mice, MLT treatment led to reduced influx of PMN leukocytes, increased recruitment of monocytes, predominant synthesis of anti-inflammatory cytokines, and augmented expression of 5- and 15-lipoxygenase mRNA, suggesting a prevalent LXA4 activity. In agreement, MTL-treated macrophages showed reduced fungal burdens associated with decreased ingestion of fungal cells. Furthermore, the addition of exogenous LX reduced, and the specific blockade of the LX receptor increased the fungal loads of B10.A macrophages. This study showed for the first time that inhibition of CysLTs signaling results in less severe pulmonary paracoccidioidomycosis that occurs in parallel with elevated LX activity and reduced infection of macrophages.


Subject(s)
Lipoxins/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Paracoccidioides/pathogenicity , Paracoccidioidomycosis/etiology , Acetates/pharmacology , Animals , Arachidonate 5-Lipoxygenase/deficiency , Arachidonate 5-Lipoxygenase/genetics , Cyclopropanes , Dinoprostone/biosynthesis , Inflammation Mediators/metabolism , Leukotriene Antagonists/pharmacology , Leukotriene C4/biosynthesis , Lipoxins/biosynthesis , Lipoxins/immunology , Macrophages, Alveolar/drug effects , Male , Mice , Mice, 129 Strain , Mice, Inbred A , Mice, Knockout , Paracoccidioidomycosis/drug therapy , Paracoccidioidomycosis/immunology , Quinolines/pharmacology , Receptors, Leukotriene/metabolism , Receptors, Pattern Recognition/metabolism , Sulfides
12.
J Immunol ; 195(7): 3086-99, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26324767

ABSTRACT

Immune-driven dry eye disease primarily affects women; the cause for this sex-specific prevalence is unknown. Polymorphonuclear neutrophils (PMN) have distinct phenotypes that drive inflammation but also regulate lymphocytes and are the rate-limiting cell for generating anti-inflammatory lipoxin A4 (LXA4). Estrogen regulates the LXA4 circuit to induce delayed female-specific wound healing in the cornea. However, the role of PMNs in dry eye disease remains unexplored. We discovered an LXA4-producing tissue PMN population in the corneal limbus, lacrimal glands, and cervical lymph nodes of healthy male and female mice. These tissue PMNs, unlike inflammatory PMNs, expressed a highly amplified LXA4 circuit and were sex-specifically regulated during immune-driven dry eye disease. Desiccating stress in females, unlike in males, triggered a remarkable decrease in lymph node PMN and LXA4 formation that remained depressed during dry eye disease. Depressed lymph node PMN and LXA4 in females correlated with an increase in effector T cells (Th1 and Th17), a decrease in regulatory T cells (Treg), and increased dry eye pathogenesis. Ab depletion of tissue PMN abrogated LXA4 formation in lymph nodes, as well as caused a marked increase in Th1 and Th17 cells and a decrease in Tregs. To establish an immune-regulatory role for PMN-derived LXA4 in dry eye, females were treated with LXA4. LXA4 treatment markedly inhibited Th1 and Th17 and amplified Treg in draining lymph nodes, while reducing dry eye pathogenesis. These results identify female-specific regulation of LXA4-producing tissue PMN as a potential key factor in aberrant effector T cell activation and initiation of immune-driven dry eye disease.


Subject(s)
Autoimmune Diseases/immunology , Dry Eye Syndromes/immunology , Lipoxins/pharmacology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Down-Regulation , Estrogens/metabolism , Female , Lacrimal Apparatus/metabolism , Limbus Corneae/metabolism , Limbus Corneae/physiology , Lipoxins/biosynthesis , Lymph Nodes/metabolism , Lymphocyte Activation/immunology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Sex Factors , Wound Healing/physiology
13.
FASEB J ; 29(12): 5029-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26289316

ABSTRACT

Resolution of acute inflammation is an active process coordinated by proresolving lipid mediators (SPMs) such as lipoxins (LXs) and resolvins (Rvs), which are formed by the concerted action of 2 lipoxygenases (LOs). Because the exact molecular mechanisms of SPM biosynthesis are not completely understood, we aimed to investigate LX and D-type Rv formation in human leukocytes and HEK293T cells overexpressing leukotriene (LT) pathway enzymes. Activity assays in precursor (15-hydroxyeicosatetraenoic acids, 17-HDoHE)-treated granulocytes [polymorphonuclear leukocytes (PMNLs)] showed a strict dependence of LXA4/RvD1 biosynthesis on cell integrity, and incubation with recombinant human 5-LO did not lead to LX or Rv formation. Pharmacologic inhibition of 5-LO activating protein (FLAP) by MK-886 inhibited LXA4/RvD1 biosynthesis in precursor-treated PMNLs (drug concentration causing 50% inhibition ∼ 0.3/0.2 µM), as did knockdown of the enzyme in MM6 cells, and precursor-treated HEK293T overexpressing 5-LO produced high amounts of LXA4 only in the presence of FLAP. In addition, inhibition of cytosolic phospholipase A2α (cPLA2α) interfered with LXA4/RvD1 formation from exogenous precursors in PMNLs. Furthermore, inhibition of the LT synthases LTA4 hydrolase and LTC4 synthase in PMNL/platelet coincubations augmented LXA4 levels. These findings show that several enzymes known to be involved in the biosynthesis of proinflammatory LTs, such as FLAP and cPLA2α, also contribute to LX and Rv formation.


Subject(s)
5-Lipoxygenase-Activating Proteins/metabolism , Docosahexaenoic Acids/biosynthesis , Lipoxins/biosynthesis , Arachidonate 5-Lipoxygenase/metabolism , Cell Line, Tumor , Cytosol/enzymology , Group IV Phospholipases A2/metabolism , HEK293 Cells , Humans , Indoles/pharmacology , Macrophages/enzymology , Macrophages/metabolism , Neutrophils/drug effects , Recombinant Proteins/metabolism
14.
Biomed Res Int ; 2015: 781087, 2015.
Article in English | MEDLINE | ID: mdl-25866809

ABSTRACT

Lipoxin A4 has been described as a major signal for the resolution of inflammation and is abnormally produced in the lungs of patients with cystic fibrosis (CF). In CF, the loss of chloride transport caused by the mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel gene results in dehydration, mucus plugging, and reduction of the airway surface liquid layer (ASL) height which favour chronic lung infection and neutrophil based inflammation leading to progressive lung destruction and early death of people with CF. This review highlights the unique ability of LXA4 to restore airway surface hydration, to stimulate airway epithelial repair, and to antagonise the proinflammatory program of the CF airway, circumventing some of the most difficult aspects of CF pathophysiology. The report points out novel aspects of the cellular mechanism involved in the physiological response to LXA4, including release of ATP from airway epithelial cell via pannexin channel and subsequent activation of and P2Y11 purinoreceptor. Therefore, inadequate endogenous LXA4 biosynthesis reported in CF exacerbates the ion transport abnormality and defective mucociliary clearance, in addition to impairing the resolution of inflammation, thus amplifying the vicious circle of airway dehydration, chronic infection, and inflammation.


Subject(s)
Cystic Fibrosis/genetics , Inflammation/genetics , Lipoxins/biosynthesis , Lung/metabolism , Adenosine Triphosphate/metabolism , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelium/metabolism , Epithelium/pathology , Humans , Inflammation/pathology , Lipoxins/metabolism , Lung/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction/genetics
15.
PLoS One ; 10(4): e0123256, 2015.
Article in English | MEDLINE | ID: mdl-25886460

ABSTRACT

BACKGROUND: Colorectal cancer is common. Polyunsaturated fatty acids (PUFAs) exert growth-inhibitory and pro-apoptotic effects on colon cancer cells. Metabolites of PUFAs such as prostaglandins (PGs), leukotrienes (LTs) and lipoxins (LXs) play a significant role in colon cancer. METHODS: Human colon cancer LoVo and RKO cells were cultured with different concentration of PUFAs and 5-fluorouracil (5-FU) in vitro. Cell morphological changes, fatty acid composition, formation of PGE2, LTB4 and LXA4 and expression of COX-2, ALOX5, PGD synthase (PGDS), microsomal prostaglandin E synthase (mPGES) were assessed in LoVo and RKO cells when supplemented with PUFAs and 5-FU. RESULTS: PUFAs and 5-FU inhibited growth of LoVo and RKO cells to the same extent at the doses used and produced significant alterations in their shape. As expected, higher concentrations of supplemented PUFAs were noted in the cells compared to control. LA, GLA, AA, ALA and EPA supplementation to LoVo cells suppressed production of PGE2, LTB4,and ALOX5, mPGES expression, but enhanced that of LXA4; whereas DHA enhanced PGE2 and LXA4 synthesis but decreased LTB4 formation and COX-2, ALOX5, mPGES expression. In contrast, 5-FU enhanced formation of PGE2, LTB4 and mPGES expression, but suppressed LXA4 synthesis and COX-2 expression. PGE2, LTB4 synthesis and ALOX5 expression was suppressed by LA, GLA, ALA and DHA; whereas AA, EPA and 5-FU enhanced PGE2 but paradoxically AA decreased and EPA and 5-FU enhanced LTB4 synthesis in RKO cells. All the PUFAs tested enhanced, while 5-FU decreased LXA4 formation in RKO cells; whereas GLA, AA, and 5-FU augmented while LA, ALA, EPA and DHA enhanced COX-2 expression in RKO cells. CONCLUSIONS: Tumoricidal action of PUFAs on colorectal LoVo and RKO cancer cells in vitro was associated with increased formation of LXA4, decreased synthesis of PGE2 and LTB4 and suppressed expression of COX-2, ALOX5, mPGES, whereas 5-FU produced contrasting actions on these indices.


Subject(s)
Colorectal Neoplasms/metabolism , Fatty Acids, Unsaturated/pharmacology , Fatty Acids/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Antimetabolites, Antineoplastic/pharmacology , Arachidonate 5-Lipoxygenase/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Fluorouracil/pharmacology , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Leukotriene B4/biosynthesis , Lipocalins/genetics , Lipocalins/metabolism , Lipoxins/biosynthesis , Prostaglandin-E Synthases
16.
Infect Immun ; 83(3): 1210-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25583526

ABSTRACT

Brucella abortus is a Gram-negative bacterium that infects humans and cattle, causing a chronic inflammatory disease known as brucellosis. A Th1-mediated immune response plays a critical role in host control of this pathogen. Recent findings indicate contrasting roles for lipid mediators in host responses against infections. 5-Lipoxygenase (5-LO) is an enzyme required for the production of the lipid mediators leukotrienes and lipoxins. To determine the involvement of 5-LO in host responses to B. abortus infection, we intraperitoneally infected wild-type and 5-LO-deficient mice and evaluated the progression of infection and concomitant expression of immune mediators. Here, we demonstrate that B. abortus induced the upregulation of 5-LO mRNA in wild-type mice. Moreover, this pathogen upregulated the production of the lipid mediators leukotriene B4 and lipoxin A4 in a 5-LO-dependent manner. 5-LO-deficient mice displayed lower bacterial burdens in the spleen and liver and less severe liver pathology, demonstrating an enhanced resistance to infection. Host resistance paralleled an increased expression of the proinflammatory mediators interleukin-12 (IL-12), gamma interferon (IFN-γ), and inducible nitric oxide synthase (iNOS) during the course of infection. Moreover, we demonstrated that 5-LO downregulated the expression of IL-12 in macrophages during B. abortus infection. Our results suggest that 5-LO has a major involvement in B. abortus infection, by functioning as a negative regulator of the protective Th1 immune responses against this pathogen.


Subject(s)
Arachidonate 5-Lipoxygenase/immunology , Brucella abortus/immunology , Brucellosis/enzymology , Brucellosis/immunology , Th1 Cells/immunology , Animals , Arachidonate 5-Lipoxygenase/deficiency , Arachidonate 5-Lipoxygenase/genetics , Bacterial Load , Brucellosis/microbiology , Brucellosis/pathology , Disease Progression , Gene Expression Regulation , Host-Pathogen Interactions , Immunity, Innate , Injections, Intraperitoneal , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Leukotriene B4/biosynthesis , Lipoxins/biosynthesis , Liver/immunology , Liver/microbiology , Liver/pathology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Spleen/immunology , Spleen/microbiology , Spleen/pathology , Th1 Cells/microbiology , Th1 Cells/pathology
17.
Antimicrob Agents Chemother ; 58(8): 4298-307, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24820086

ABSTRACT

The accumulation of neutrophils and proinflammatory mediators, such as leukotriene B4 (LTB4), is a classic marker of inflammatory disease. The clearance of apoptotic neutrophils, inhibition of proinflammatory signaling, and production of proresolving lipids (including lipoxins, such as lipoxin A4 [LXA4]) are imperative for resolving inflammation. Tulathromycin (TUL), a macrolide used to treat bovine respiratory disease, confers immunomodulatory benefits via mechanisms that remain unclear. We recently reported the anti-inflammatory properties of TUL in bovine phagocytes in vitro and in Mannheimia haemolytica-challenged calves. The findings demonstrated that this system offers a powerful model for investigating novel mechanisms of pharmacological immunomodulation. In the present study, we examined the effects of TUL in a nonbacterial model of pulmonary inflammation in vivo and characterized its effects on lipid signaling. In bronchoalveolar lavage (BAL) fluid samples from calves challenged with zymosan particles (50 mg), treatment with TUL (2.5 mg/kg of body weight) significantly reduced pulmonary levels of LTB4 and prostaglandin E2 (PGE2). In calcium ionophore (A23187)-stimulated bovine neutrophils, TUL inhibited phospholipase D (PLD), cytosolic phospholipase A2 (PLA2) activity, and the release of LTB4. In contrast, TUL promoted the secretion of LXA4 in resting and A23187-stimulated neutrophils, while levels of its precursor, 15(S)-hydroxyeicosatetraenoic acid [15(S)-HETE], were significantly lower. These findings indicate that TUL directly modulates lipid signaling by inhibiting the production of proinflammatory eicosanoids and promoting the production of proresolving lipoxins.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dinoprostone/antagonists & inhibitors , Disaccharides/pharmacology , Heterocyclic Compounds/pharmacology , Leukotriene B4/antagonists & inhibitors , Lipoxins/agonists , Phospholipases A2/metabolism , Pneumonia/drug therapy , Animals , Bronchoalveolar Lavage Fluid/chemistry , Calcimycin/pharmacology , Cattle , Dinoprostone/biosynthesis , Hydroxyeicosatetraenoic Acids/metabolism , Leukotriene B4/biosynthesis , Lipoxins/biosynthesis , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Particulate Matter , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Primary Cell Culture , Zymosan
18.
Oncol Rep ; 31(6): 2785-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24737143

ABSTRACT

Aspirin has been proposed in recent years as a candidate for chemoprevention of adenocarcinoma in patients with Barrett's esophagus. The aim of the present study was to evaluate the effect of acetylsalicylic acid (ASA) in an experimental model of esophageal adenocarcinoma. An animal model of gastroenteroesophageal reflux was established using Wistar rats undergoing esophagojejunostomy with gastric preservation. Following surgery, rats were divided into three groups: i) control (vehicle); ii) ASA 50 mg/kg/day; and iii) ASA 5 mg/kg/day. Four months after surgery, the surviving animals were sacrificed and the rat esophagi were assessed for histological and biochemical [prostaglandin E2 (PGE2) and lipoxin A4 (LXA4 ) levels] analysis. As in the control rats, those receiving aspirin treatment showed no decrease in inflammation grade, extent of ulcerated esophageal mucosa, length of intestinal metaplasia in continuity with anastomosis, presence of intestinal metaplasia beyond anastomosis, severity of dysplasia or incidence of adenocarcinoma. In contrast, aspirin-treated rats showed decreased esophageal tissue levels of PGE2 and increased LXA4, significantly in the high-dose aspirin group (p=0.008 and p=0.01, respectively). In this rat model of gastroesophageal reflux, the administration of aspirin modified esophageal tissue levels of PGE2 and LXA4, but was not effective in preventing the development of esophageal adenocarcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Aspirin/administration & dosage , Barrett Esophagus/drug therapy , Esophageal Neoplasms/drug therapy , Gastroesophageal Reflux/drug therapy , Neoplasms, Experimental/drug therapy , Adenocarcinoma/pathology , Animals , Barrett Esophagus/pathology , Dinoprostone/biosynthesis , Disease Models, Animal , Esophageal Neoplasms/pathology , Gastroesophageal Reflux/pathology , Gastroesophageal Reflux/surgery , Gene Expression Regulation, Neoplastic , Humans , Lipoxins/biosynthesis , Neoplasms, Experimental/pathology , Neoplasms, Experimental/surgery , Rats
19.
Biol Reprod ; 90(4): 74, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24571985

ABSTRACT

Inflammation dysregulation in placenta is implicated in the pathogenesis of numerous pregnancy complications. Glucocorticoids (GCs), universally considered anti-inflammatory, can also exert proinflammatory actions under some conditions, whereas whether and how GCs promote placental inflammation have not been intensively investigated. In this paper we report the opposing regulation of rat placental inflammation by synthetic GC dexamethasone (Dex). When Dex was subcutaneously injected 1 h after we administered an intraperitoneal lipopolysaccharide (LPS) challenge, neutrophil infiltration and proinflammatory Il1b, Il6, and Tnfa expression in rat placenta were significantly reduced. In contrast, Dex pretreatment for 24 h potentiated rat placental proinflammatory response to LPS and delayed inflammation resolution, which involved MAPKs and NF-kappaB activation. Mechanically, Dex pretreatment promoted 5-lipoxygenase (ALOX5) activation and increased leukotriene B4 production, whereas it inhibited the anti-inflammatory and proresolving lipid mediator lipoxin A4 (LXA4) biosynthesis in rat placenta via downregulating ALOX15 and ALOX15B expression. Moreover, LXA4 supplementation dampened Dex-potentiated placental inflammation and suppressed Dex-mediated ALOX5 activation in vivo and in vitro. Taken together, these findings suggest that GCs exposure could promote placental inflammation initiation and delay resolution via disrupting LXA4 biosynthesis.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Inflammation/immunology , Lipoxins/immunology , Placenta/drug effects , Placenta/immunology , Animals , Arachidonate 5-Lipoxygenase/immunology , Arachidonate 5-Lipoxygenase/metabolism , Arachidonic Acid/immunology , Cell Line , Dexamethasone/immunology , Dinoprostone/immunology , Dinoprostone/metabolism , Female , Glucocorticoids/immunology , Humans , Inflammation/metabolism , Leukotriene B4/immunology , Leukotriene B4/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lipoxins/antagonists & inhibitors , Lipoxins/biosynthesis , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , NF-kappa B/immunology , NF-kappa B/metabolism , Placenta/cytology , Pregnancy , Rats , Rats, Sprague-Dawley
20.
J Leukoc Biol ; 95(4): 587-98, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24338629

ABSTRACT

PPARγ-achieved neuroprotection in experimental stroke has been explained by the inhibition of inflammatory genes, an action in which 5-LO, Alox5, is involved. In addition, PPARγ is known to promote the expression of CD36, a scavenger receptor that binds lipoproteins and mediates bacterial recognition and also phagocytosis. As phagocytic clearance of neutrophils is a requisite for resolution of the inflammatory response, PPARγ-induced CD36 expression might help to limit inflammatory tissue injury in stroke, an effect in which 5-LO might also be involved. Homogenates, sections, and cellular suspensions were prepared from brains of WT and Alox5(-/-) mice exposed to distal pMCAO. BMMs were obtained from Lys-M Cre(+) PPARγ(f/f) and Lys-M Cre(-) PPARγ(f/f) mice. Stereological counting of double-immunofluorescence-labeled brain sections and FACS analysis of cell suspensions was performed. In vivo and in vitro phagocytosis of neutrophils by microglia/macrophages was analyzed. PPARγ activation with RSG induced CD36 expression in resident microglia. This process was mediated by the 5-LO gene, which is induced in neurons by PPARγ activation and at least by one of its products--LXA4--which induced CD36 independently of PPARγ. Moreover, CD36 expression helped resolution of inflammation through phagocytosis, concomitantly to neuroprotection. Based on these findings, in addition to a direct modulation by PPARγ, we propose in brain a paracrine model by which products generated by neuronal 5-LO, such as LXA4, increase the microglial expression of CD36 and promote tissue repair in pathologies with an inflammatory component, such as stroke.


Subject(s)
Arachidonate 5-Lipoxygenase/physiology , CD36 Antigens/physiology , Hypoglycemic Agents/pharmacology , Inflammation/immunology , Neutrophils/immunology , PPAR gamma/physiology , Thiazolidinediones/pharmacology , Animals , Brain Ischemia/immunology , CD36 Antigens/analysis , Cells, Cultured , Lipoxins/biosynthesis , Mice , Mice, Inbred C57BL , PPAR gamma/agonists , Phagocytosis , Rats , Rosiglitazone , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...