Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 129(4): 1684-1698, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30855276

ABSTRACT

Retinoic acid-related orphan receptor α (RORα) is considered a key regulator of polarization in liver macrophages that is closely related to nonalcoholic steatohepatitis (NASH) pathogenesis. However, hepatic microenvironments that support the function of RORα as a polarity regulator were largely unknown. Here, we identified maresin 1 (MaR1), a docosahexaenoic acid (DHA) metabolite with a function of specialized proresolving mediator, as an endogenous ligand of RORα. MaR1 enhanced the expression and transcriptional activity of RORα and thereby increased the M2 polarity of liver macrophages. Administration of MaR1 protected mice from high-fat diet-induced NASH in a RORα-dependent manner. Surprisingly, RORα increased the level of MaR1 through transcriptional induction of 12-lipoxygenase (12-LOX), a key enzyme in MaR1 biosynthesis. Furthermore, we demonstrated that modulation of 12-LOX activity enhanced the protective function of DHA against NASH. Together, these results suggest that the MaR1/RORα/12-LOX autoregulatory circuit could offer potential therapeutic strategies for curing NASH.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Docosahexaenoic Acids/pharmacology , Macrophages/metabolism , Non-alcoholic Fatty Liver Disease , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Animals , Arachidonate 12-Lipoxygenase/genetics , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/prevention & control , Macrophages/pathology , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics
2.
J Biol Chem ; 294(16): 6612-6620, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30792307

ABSTRACT

In type 1 diabetes, an autoimmune event increases oxidative stress in islet ß cells, giving rise to cellular dysfunction and apoptosis. Lipoxygenases are enzymes that catalyze the oxygenation of polyunsaturated fatty acids that can form lipid metabolites involved in several biological functions, including oxidative stress. 12-Lipoxygenase and 12/15-lipoxygenase are related but distinct enzymes that are expressed in pancreatic islets, but their relative contributions to oxidative stress in these regions are still being elucidated. In this study, we used mice with global genetic deletion of the genes encoding 12-lipoxygenase (arachidonate 12-lipoxygenase, 12S type [Alox12]) or 12/15-lipoxygenase (Alox15) to compare the influence of each gene deletion on ß cell function and survival in response to the ß cell toxin streptozotocin. Alox12-/- mice exhibited greater impairment in glucose tolerance following streptozotocin exposure than WT mice, whereas Alox15-/- mice were protected against dysglycemia. These changes were accompanied by evidence of islet oxidative stress in Alox12-/- mice and reduced oxidative stress in Alox15-/- mice, consistent with alterations in the expression of the antioxidant response enzymes in islets from these mice. Additionally, islets from Alox12-/- mice displayed a compensatory increase in Alox15 gene expression, and treatment of these mice with the 12/15-lipoxygenase inhibitor ML-351 rescued the dysglycemic phenotype. Collectively, these results indicate that Alox12 loss activates a compensatory increase in Alox15 that sensitizes mouse ß cells to oxidative stress.


Subject(s)
Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/biosynthesis , Gene Expression Regulation, Enzymologic , Insulin-Secreting Cells/enzymology , Oxidative Stress , Animals , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Gene Deletion , Isoxazoles/pharmacology , Mice , Mice, Knockout , Naphthalenes/pharmacology , Streptozocin/toxicity
3.
Dev Neurosci ; 35(2-3): 140-54, 2013.
Article in English | MEDLINE | ID: mdl-23838566

ABSTRACT

Oxidative stress involving premyelinating oligodendrocytes (OLs) is a major factor in the pathogenesis of preterm white matter injury. In animal and cell culture studies, activation of the lipid-oxidizing enzyme 12/15-lipoxygenase (12/15-LOX) plays a central role as an inflammatory mediator in the pathology of oxidative stress and OL cell death, as well as ischemia and neuronal death. The role of 12/15-LOX, however, is unclear in the developing human brain. The mechanism of 12/15-LOX involves the production of reactive oxygen species through the metabolism of arachidonic acid, as well as direct detrimental effects on organelle membranes. Here we tested the hypothesis that the density of 12/15-LOX-expressing cells is increased in periventricular leukomalacia (PVL). Using immunocytochemistry (ICC) in human paraffin-embedded tissue, 12/15-LOX expression was seen in macrophages of the focally necrotic lesions in the periventricular white matter, as well as in glial cells throughout the surrounding white matter with reactive gliosis. Interestingly, no significant 12/15-LOX expression was detected in neurons in the cerebral cortex overlying the damaged white matter. Using a scoring system from 0 to 3, we assessed the density of 12/15-LOX-expressing cells in diffusely gliotic white matter from 20 to 43 postconceptional (PC) weeks in 19 PVL cases (median = 36 PC weeks) and 10 control (non-PVL) cases (median = 34 PC weeks). The density of 12/15-LOX-positive cells was significantly increased in the diffuse component of PVL (score = 1.17 ± 0.15) compared to controls (score = 0.48 ± 0.21; p = 0.014). Using double-label ICC, 12/15-LOX was observed in PVL in OLs of the O4 and O1 premyelinating stages, as well as in mature OLs as determined with the mature OL marker adenomatous polyposis coli (APC). In addition, 12/15-LOX expression was present in a population of CD68-positive activated microglia. There was no 12/15-LOX expression in reactive astrocytes. Finally we observed terminal deoxynucleotide transferase dUTP nick end-labeling-positive cells within the white matter of PVL that expressed 12/15-LOX and/or within close proximity of 12/15-LOX-positive cells. Our data support a role for 12/15-LOX activation as an inflammatory mediator of injury in PVL, with a contribution of 12/15-LOX to PVL-induced damage to or cell death of OLs, including those at the O1 and O4 stages.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Leukomalacia, Periventricular/enzymology , Microglia/enzymology , Oligodendroglia/enzymology , Arachidonate 12-Lipoxygenase/analysis , Arachidonate 15-Lipoxygenase/analysis , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Infant, Newborn , Leukomalacia, Periventricular/pathology
4.
Exp Mol Pathol ; 94(1): 216-27, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23000425

ABSTRACT

Exposure to excessive quantities of bacterial-derived lipopolysaccharide (LPS) is associated with injury to the lung and the liver. Macrophages are thought to play a key role in the pathogenic response to LPS by releasing proinflammatory/cytotoxic mediators. Macrophage responses to LPS are mediated in large part by toll-like receptor 4 (TLR4). In the present studies we used C3H/HeJ mice, which possess a mutated nonfunctional TLR4, to examine its role in lung and liver macrophage responses to acute endotoxemia induced by LPS administration. Treatment of control C3H/HeOuJ mice with LPS (3 mg/ml, i.p.) was associated with a significant increase in the number of macrophages in both the lung and the liver. This was most prominent after 48 h, and was preceded by expression of proliferating cell nuclear antigen (PCNA), suggesting that macrophage proliferation contributes to the response. In liver, but not lung macrophages, LPS administration resulted in a rapid (within 3 h) increase in mRNA expression of Mn superoxide dismutase (SOD) and heme oxygenase-1 (HO-1), key enzymes in antioxidant defense. In contrast, HO-1 protein expression decreased 3 h after LPS administration in liver macrophages, while in lung macrophages it increased. mRNA expression of enzymes mediating the biosynthesis of eicosanoids, including cyclooxygenase-2 (COX-2) and microsomal prostaglandin E synthase-1 (mPGES-1), but not 12/15-lipoxygenase (LOX), was upregulated in liver macrophages 3-24 h after LPS, with no effect on lung macrophages. However, COX-2 protein expression increased in both cell types. Loss of functional TLR4 significantly blunted the effects of LPS. Thus, no major changes were observed after LPS administration in the number of lung and liver macrophages recovered from TLR4 mutant mice, or on expression of PCNA. Increases in HO-1, MnSOD, COX-2 and PGES-1 mRNA expression in liver macrophages were also reduced in these mice. Conversely, in lung macrophages, loss of functional TLR4 resulted in increased expression of COX-2 protein and 12/15-LOX mRNA. These results demonstrate distinct lung and liver macrophage responses to acute endotoxemia are mediated, in part, by functional TLR4.


Subject(s)
Endotoxemia/immunology , Kupffer Cells/immunology , Macrophages, Alveolar/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Animals , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Cyclooxygenase 2/biosynthesis , Enzyme Activation , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Intramolecular Oxidoreductases/biosynthesis , Kupffer Cells/metabolism , Lipopolysaccharides , Liver/immunology , Lung/immunology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Transgenic , Proliferating Cell Nuclear Antigen/biosynthesis , Prostaglandin-E Synthases , RNA, Messenger/genetics , RNA, Messenger/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Toll-Like Receptor 4/genetics , Up-Regulation
5.
Aging Cell ; 9(5): 851-67, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20698835

ABSTRACT

Because of recent insights into the pathogenesis of age-related bone loss, we investigated whether intermittent parathyroid hormone (PTH) administration antagonizes the molecular mechanisms of the adverse effects of aging on bone. Parathyroid hormone produced a greater increase in vertebral trabecular bone mineral density and bone volume as well as a greater expansion of the endocortical bone surface in the femur of 26- when compared to 6 -month-old female C57BL/6 mice. Moreover, PTH increased trabecular connectivity in vertebrae, and the toughness of both vertebrae and femora in old, but not young, mice. Parathyroid hormone also increased the rate of bone formation and reduced osteoblast apoptosis to a greater extent in the old mice. Most strikingly, PTH reduced reactive oxygen species, p66(Shc) phosphorylation, and expression of the lipoxygenase Alox15, and it increased glutathione and stimulated Wnt signaling in bone of old mice. Parathyroid hormone also antagonized the effects of oxidative stress on p66(Shc) phosphorylation, Forkhead Box O transcriptional activity, osteoblast apoptosis, and Wnt signaling in vitro. In contrast, administration of the antioxidants N-acetyl cysteine or pegylated catalase reduced osteoblast progenitors and attenuated proliferation and Wnt signaling. These results suggest that PTH has a greater bone anabolic efficacy in old age because in addition to its other positive actions on bone formation, it antagonizes the age-associated increase in oxidative stress and its adverse effects on the birth and survival of osteoblasts. On the other hand, ordinary antioxidants cannot restore bone mass in old age because they slow remodeling and attenuate osteoblastogenesis by interfering with Wnt signaling.


Subject(s)
Aging , Bone Density/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Oxidative Stress/drug effects , Parathyroid Hormone/administration & dosage , Parathyroid Hormone/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Cell Proliferation/drug effects , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Multienzyme Complexes/biosynthesis , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Shc Signaling Adaptor Proteins/metabolism , Signal Transduction/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 1 , Wnt Proteins/metabolism
6.
J Immunol ; 183(5): 3383-9, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19675173

ABSTRACT

Eicosanoids are essential mediators of the inflammatory response and contribute both to the initiation and the resolution of inflammation. Leukocyte-type 12/15-lipoxygenase (12/15-LO) represents a major enzyme involved in the generation of a subclass of eicosanoids, including the anti-inflammatory lipoxin A(4) (LXA(4)). Nevertheless, the impact of 12/15-LO on chronic inflammatory diseases such as arthritis has remained elusive. By using two experimental models of arthritis, the K/BxN serum-transfer and a TNF transgenic mouse model, we show that deletion of 12/15-LO leads to uncontrolled inflammation and tissue damage. Consistent with these findings, 12/15-LO-deficient mice showed enhanced inflammatory gene expression and decreased levels of LXA(4) within their inflamed synovia. In isolated macrophages, the addition of 12/15-LO-derived eicosanoids blocked both phosphorylation of p38MAPK and expression of a subset of proinflammatory genes. Conversely, 12/15-LO-deficient macrophages displayed significantly reduced levels of LXA(4), which correlated with increased activation of p38MAPK and an enhanced inflammatory gene expression after stimulation with TNF-alpha. Taken together, these results support an anti-inflammatory and tissue-protective role of 12/15-LO and its products during chronic inflammatory disorders such as arthritis.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Animals , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/deficiency , Arthritis, Experimental/immunology , Chronic Disease , Eicosanoids/antagonists & inhibitors , Eicosanoids/biosynthesis , Feedback, Physiological/immunology , Knee Joint/enzymology , Knee Joint/immunology , Knee Joint/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity/genetics , Organ Specificity/immunology
7.
J Immunol ; 181(9): 6514-24, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941242

ABSTRACT

The peritoneal macrophage (Mphi) is the site of greatest 12/15-lipoxygenase (12/15-LOX) expression in the mouse; however, its immunoregulatory role in this tissue has not been explored. Herein, we show that 12/15-LOX is expressed by 95% of resident peritoneal CD11b(high) cells, with the remaining 5% being 12/15-LOX(-). 12/15-LOX(+) cells are phenotypically defined by high F4/80, SR-A, and Siglec1 expression, and enhanced IL-10 and G-CSF generation. In contrast, 12/15-LOX(-) cells are a dendritic cell population. Resident peritoneal Mphi numbers were significantly increased in 12/15-LOX(-/-) mice, suggesting alterations in migratory trafficking or cell differentiation in vivo. In vitro, Mphi from 12/15-LOX(-/-) mice exhibit multiple abnormalities in the regulation of cytokine/growth factor production both basally and after stimulation with Staphylococcus epidermidis cell-free supernatant. Resident adherent cells from 12/15-LOX(-/-) mice generate more IL-1, IL-3, GM-CSF, and IL-17, but less CCL5/RANTES than do cells from wild-type mice, while Staphylococcus epidermidis cell-free supernatant-elicited 12/15-LOX(-/-) adherent cells release less IL-12p40, IL-12p70, and RANTES, but more GM-CSF. This indicates a selective effect of 12/15-LOX on peritoneal cell cytokine production. In acute sterile peritonitis, 12/15-LOX(+) cells and LOX products were cleared, then reappeared during the resolution phase. The peritoneal lavage of 12/15-LOX(-/-) mice showed elevated TGF-beta1, along with increased immigration of monocytes/Mphi, but decreases in several cytokines including RANTES/CCL5, MCP-1/CCL2, G-CSF, IL-12-p40, IL-17, and TNF-alpha. No changes in neutrophil or lymphocyte numbers were seen. In summary, endogenous 12/15-LOX defines the resident MPhi population and regulates both the recruitment of monocytes/Mphi and cytokine response to bacterial products in vivo.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Cytokines/biosynthesis , Inflammation Mediators/physiology , Peritonitis/enzymology , Peritonitis/microbiology , Staphylococcal Infections/enzymology , Staphylococcal Infections/pathology , Staphylococcus epidermidis/immunology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/biosynthesis , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/physiology , Acute Disease , Animals , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/deficiency , Cells, Cultured , Cytokines/metabolism , Cytokines/physiology , Hydroxyeicosatetraenoic Acids/biosynthesis , Hydroxyeicosatetraenoic Acids/physiology , Immunophenotyping , Inflammation Mediators/metabolism , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/microbiology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/immunology , Staphylococcal Infections/immunology , Staphylococcus epidermidis/metabolism
8.
Am J Physiol Gastrointest Liver Physiol ; 294(4): G948-62, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18258795

ABSTRACT

To identify novel genes associated with iron metabolism, we performed gene chip studies in two models of iron deficiency: iron-deprived rats and rats deficient in the principal intestinal iron transporter, divalent metal transporter 1 (i.e., Belgrade rats). Affymetrix rat genome gene chips were utilized (RAE230) with cRNA samples derived from duodenum and jejunum of experimental and control animals. Computational analysis and statistical data reduction identified 29 candidate genes, which were induced in both models of iron deficiency. Gene ontology analysis showed enrichment for genes related to lipid homeostasis, and one gene related to this physiological process, a leukocyte type, arachidonate 12-lipoxygenase (Alox15), was selected for further examination. TaqMan real-time PCR studies demonstrated strong induction of Alox15 throughout the small and large intestine, and in the liver of iron-deficient rats. Polyclonal antibodies were developed and utilized to demonstrate that proteins levels are significantly increased in the intestinal epithelium of iron-deprived rats. HPLC analysis revealed altered intestinal lipid metabolism indicative of Alox15 activity, which resulted in the production of biologically active lipid molecules (12-HETE, 13-HODE, and 13-HOTE). The overall effect is a perturbation of intestinal lipid homeostasis, which results in the production of lipids essentially absent in the intestine of control rats. We have thus provided mechanistic insight into the alteration in lipid metabolism that occurs during iron deficiency, in that induction of Alox15 mRNA expression may be the primary event. The resulting lipid mediators may be related to documented alterations in villus structure and cell proliferation rates in iron deficiency, or to structural alterations in membrane lipid composition.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Duodenum/metabolism , Iron Deficiencies , Iron Metabolism Disorders/metabolism , Jejunum/metabolism , Lipid Metabolism/genetics , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Algorithms , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Blotting, Western , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Chromatography, High Pressure Liquid , Cluster Analysis , Disease Models, Animal , Duodenum/enzymology , Duodenum/pathology , Enzyme Induction , Gene Expression Profiling/methods , Immunohistochemistry , Iron Metabolism Disorders/enzymology , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/pathology , Jejunum/enzymology , Jejunum/pathology , Linoleic Acids/metabolism , Liver/metabolism , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
9.
Dig Dis Sci ; 53(1): 181-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17522976

ABSTRACT

12-Lipoxygenase (12-LOX) is over-expressed in a variety of human tumors, but its exact effect on the tumorogenesis of gastric cancer remains largely obscure. To investigate the effect of 12-LOX and its inhibitor baicalein on proliferation and apoptosis of human gastric cancer, AGS cells were separately treated with 12-hydroxyeicosatetraenoic acid (12-HETE, a metabolite of 12-LOX) and baicalein. MTT assay revealed that the absorbance of the 12-HETE-treated group was significantly (P < 0.01) higher than that of control group and that the absorbance of baicalein-treated group was significantly (P < 0.01) less than that of the control group, and that 48 h after treatment the apoptosis index of the baicalein-treated group was significantly (P < 0.01) higher than that of the untreated group and was significantly (P < 0.01) lower in the 12-HETE-treated group. Western blotting analysis was used to investigate the mechanism of these effects. The results revealed that the concentration of phosphorylated ERK in cells treated with 100 nmol L(-1) 12-HETE was significantly (P < 0.05) higher than in the untreated group and that the concentration of phosphorylated ERK1/2 in cells treated with 40 micromol L(-1) baicalein was significantly (P < 0.05) lower than in the untreated group. The expression level of bcl-2 was up-regulated and down-regulated after separate treatment with 12-HETE and baicalein, respectively, and both of these effects could be blocked by PD98059. Protein kinase C (PKC) activity was increased by treatment with 12-HETE and reduced by treatment with baicalein (P < 0.05). The PKC inhibitor BIM (bisindolymaleimide-I) blocked the phosphorylation of ERK1/2 and activation of PKC induced by 12-LOX. When pretreated with BIM, the concentration of phospho-ERK1/2 or bcl-2 in the BIM + 12-HETE-treated group was significantly (P < 0.05) lower than in that treated with 12-HETE only, and the concentration in the BIM + baicalein-treated group was significantly (P < 0.05) higher than in that treated with baicalein only. On the basis of these data we conclude that, via its metabolite 12-HETE, 12-LOX abolishes proliferation of AGS cells and protect cells from apoptosis by activating the ERK1/2 pathway and, eventually, enhances expression of bcl-2. Because PKC is also involved in the activation of ERK1/2 induced by 12-LOX, 12-LOX inhibitors would be potentially powerful anticancer agents for prevention and cure of human gastric cancer.


Subject(s)
Apoptosis/physiology , Arachidonate 12-Lipoxygenase/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Signal Transduction/physiology , Stomach Neoplasms/enzymology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor , Blotting, Western , Cell Proliferation/drug effects , DNA Topoisomerases, Type II , Electrophoresis, Agar Gel , Enzyme Inhibitors/pharmacology , Flavanones/pharmacology , Humans , Lipoxygenase Inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Phosphorylation , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Tumor Cells, Cultured
10.
J Neurochem ; 103(1): 380-7, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17877641

ABSTRACT

12/15 Lipoxygenase (12/15LO) protein levels and activity are increased in pathologically affected regions of Alzheimer's disease (AD) brains, compared with controls. Its metabolic products are elevated in cerebrospinal fluid of patients with AD and individuals with mild cognitive impairment, suggesting that this enzyme may be involved early in AD pathogenesis. Herein, we investigate the effect of pharmacologic inhibition of 12/15LO on the amyloid beta precursor protein (APP) metabolism. To this end, we used CHO and N2A cells stably expressing human APP with the Swedish mutant, and two structurally distinct and selective 12/15LO inhibitors, PD146176 and CDC. Our results demonstrated that both drugs dose-dependently reduced Abeta formation without affecting total APP levels. Interestingly, in the same cells we observed a significant reduction in secreted (s)APPbeta and beta-secretase (BACE), but not sAPPalpha and ADAM10 protein levels. Together, these data show for the first time that this enzymatic pathway influences Abeta formation whereby modulating the BACE proteolytic cascade. We conclude that specific pharmacologic inhibition of 12/15LO could represent a novel therapeutic target for treating or preventing AD pathology in humans.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Blotting, Western , CHO Cells , Cell Line , Cricetinae , Cricetulus , Enzyme Inhibitors/pharmacology , Fluorenes/pharmacology , Humans , Lipoxygenase Inhibitors , Mutation
11.
Food Chem Toxicol ; 45(3): 508-14, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17027136

ABSTRACT

12(S)-Lipoxygenase (LOX) is regarded as a pro-tumorigenic enzyme and as a potential target for therapy and prevention of cancer so that the search for specific 12(S)-LOX inhibitors is part of drug development strategies. To facilitate the identification of specific 12(S)-LOX inhibitors we have created an assay cell line by introducing a12(S)-LOX expression vector into SW480 colorectal cancer cells. When arachidonic acid was supplied in the medium both transiently and stably overexpressing cells produced 12(S)-hydroxytetraenic acid (HETE) originating from the transfected gene at 4-5-fold the amount obtained from control transfectants. 12(S)-HETE production was 1913.7+/-17.2pg/ml and reached a steady state level 24h after addition of arachidonic acid. To demonstrate the models suitability of 12(S)-LOX overexpressing SW480 cells they were used to measure the inhibitory activity of the plant phenols baicalein, kaempferol, quercetin, nordihydroguaretic acid and resveratrol which are known for their chemopreventive as well as LOX-inhibitory activity in different tumour models. All 5 compounds inhibited 12(S)-HETE production at concentrations below those necessary for growth inhibition.


Subject(s)
Apoptosis/drug effects , Lipoxygenase Inhibitors , Lipoxygenase Inhibitors/pharmacology , Phytotherapy , Plants, Medicinal , Protective Agents/pharmacology , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 12-Lipoxygenase/genetics , Cell Line, Tumor , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , DNA Primers , Humans , Lipoxygenase Inhibitors/administration & dosage , Lipoxygenase Inhibitors/therapeutic use , Polymerase Chain Reaction , Protective Agents/administration & dosage , Protective Agents/therapeutic use , RNA/analysis
12.
J Biol Chem ; 281(27): 18601-9, 2006 Jul 07.
Article in English | MEDLINE | ID: mdl-16638750

ABSTRACT

12-Lipoxygenase utilizes arachidonic acid to synthesize 12(S)-hydroperoxyeicosatetraenoic acid, which is converted to the end product 12(S)-hydroxyeicosatetraenoic acid, an eicosanoid that promotes tumorigenesis and metastasis. Increased expression of 12-lipoxygenase has been documented in a number of carcinomas. When overexpressed in human prostate or breast cancer, 12-lipoxygenase promotes tumor angiogenesis and growth in vivo. The present study was undertaken to delineate the mechanisms by which 12-lipoxygenase enhances angiogenesis. Herein we report that nordihydroguaiaretic acid, a pan inhibitor of lipoxygenases and baicalein, a selective inhibitor of 12-lipoxygenase, reduced VEGF expression in human prostate cancer PC-3 cells. Overexpression of 12-lipoxygenase in PC-3 cells resulted in a 3-fold increase in VEGF protein level when compared with vector control cells. An increase in PI 3-kinase activity was found in 12-LOX-transfected PC-3 cells and inhibition of PI 3-kinase by LY294002 significantly reduced VEGF expression. Northern blot and real time PCR analyses revealed an elevated VEGF transcript level in PC-3 cells transfected with a 12-lipoxygenase expression construct. Using a VEGF promoter luciferase construct (-1176/+54), we found a 10-fold increase in VEGF promoter activity in 12-lipoxygenase-transfected PC-3 cells. The region located between -88 and -66 of the VEGF promoter was identified as 12-lipoxygenase responsive using VEGF promoter-based luciferase assays. Further analysis with mutant constructs indicated Sp1 as a transcription factor required for 12-lipoxygenase stimulation of VEGF. Neutralization of VEGF by a function-blocking antibody significantly decreased the ability of 12-lipoxygenase-transfected PC-3 cells to stimulate endothelial cell migration, suggesting VEGF as an important effector for 12-lipoxygenase-mediated stimulation of tumor angiogenesis.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/enzymology , Prostatic Neoplasms/blood supply , Vascular Endothelial Growth Factor A/biosynthesis , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Cell Line, Tumor , Cell Movement/genetics , Chromones/pharmacology , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Flavanones/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lipoxygenase Inhibitors/pharmacology , Male , Masoprocol/pharmacology , Morpholines/pharmacology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphoinositide-3 Kinase Inhibitors , Promoter Regions, Genetic , Prostatic Neoplasms/enzymology , Vascular Endothelial Growth Factor A/genetics
13.
Am J Physiol Renal Physiol ; 290(4): F864-72, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16219916

ABSTRACT

The arachidonate signaling pathways comprise prostanoids formed by cyclooxygenases, EETs, and HETEs formed by cytochrome P-450 (CYP) enzymes and HETEs and leukotrienes generated by lipoxygenases. Whereas the intrarenal localization of cyclooxygenases and of some CYP enzymes along the nephron has already been determined, the localization of lipoxygenases and leukotriene-forming enzymes together with leukotriene receptors in the kidney is less clear. This study therefore aimed to determine the expression of 5-, 12-, and 15-lipoxygenases as well as the leukotriene receptors along the rat nephron. The kidneys were dissected into cortex and outer and inner medulla, and the microdissected nephron segments were collected after a collagenase digestion. mRNA abundance was determined by RT-PCR and real-time PCR. 15-LOX mRNA showed a characteristic expression pattern along the distal nephron. 12-LOX mRNA was only found in the glomerulus. Similarly, 5-LOX mRNAs together with 5-LOX-activating protein mRNAs were expressed in the glomerulus and also in the vasa recta. The leukotriene A4 hydrolase was found in all nephron segments, whereas leukotriene C4 synthase mRNA could not be found in any nephron segment. The leukotriene receptor B4 and the cysteinyl leukotriene receptor type 1 were selectively expressed in the glomerulus, whereas cysteinyl receptor type 2 was not found in any nephron segment. Our data suggest that the glomerulus is a major source and target for 5- and 12-HETE and for leukotrienes. The collecting duct system, on the other hand, appears to be a major source of 15-HETE.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Arachidonate 5-Lipoxygenase/biosynthesis , Nephrons/physiology , Receptors, Leukotriene/biosynthesis , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/genetics , Gene Expression Profiling , Glomerular Filtration Rate , Male , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Receptors, Leukotriene/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Vasoconstriction
14.
Biochem Biophys Res Commun ; 338(1): 128-35, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16105647

ABSTRACT

Oxidation of low density lipoprotein (LDL) is a critical step for atherogenesis, and the role of the 12/15-lipoxygenase (12/15-LOX) as well as LDL receptor-related protein (LRP) expressed in macrophages in this process has been suggested. The oxygenation of cholesteryl linoleate in LDL by mouse macrophage-like J774A.1 cells overexpressing 12/15-LOX was inhibited by an anti-LRP antibody but not by an anti-LDL receptor antibody. When the cells were incubated with LDL double-labeled by [3H]cholesteryl linoleate and [125I]apoB, association with the cells of [3H]cholesteryl linoleate expressed as LDL protein equivalent exceeded that of [125I]apoB, indicating selective uptake of [3H]cholesteryl linoleate from LDL to these cells. An anti-LRP antibody inhibited the selective uptake of [3H]cholesteryl ester by 62% and 81% with the 12/15-LOX-expressing cells and macrophages, respectively. Furthermore, addition of LDL to the culture medium of the [3H]cholesteryl linoleate-labeled 12/15-LOX-expressing cells increased the release of [3H]cholesteryl linoleate to the medium in LDL concentration- and time-dependent manners. The transport of [3H]cholesteryl linoleate from the cells to LDL was also inhibited by an anti-LRP antibody by 75%. These results strongly suggest that LRP contributes to the LDL oxidation by 12/15-LOX in macrophages by selective uptake and efflux of cholesteryl ester in the LDL particle.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Cholesterol Esters/metabolism , Cholesterol, LDL/metabolism , Macrophages, Peritoneal/enzymology , Animals , CHO Cells , Cell Line , Cell Membrane/enzymology , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Macrophages, Peritoneal/metabolism , Mice , Oxygen/metabolism , Protein Transport/physiology
15.
Biochem Biophys Res Commun ; 338(1): 117-21, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16122700

ABSTRACT

The signal transduction of human 12(S)-lipoxygenase and the regulation of gene activation, induced by epidermal growth factor (EGF), are discussed in this review article. Treatment of human epidermoid carcinoma A431 cells with EGF induces the gene expression of human 12(S)-lipoxygenase, and two Sp1 binding sites residing at -158 to -150 bp and -123 to -114 bp are essential in the mediation of EGF induction of the 12(S)-lipoxygenase gene. EGF induces MAPK activation in cells, followed by the activation of AP1. Thus, the biosynthesis of c-Jun is enhanced, which subsequently interacts with Sp1. c-Jun on Sp1/c-Jun complex is then recruited to gene promoter through the binding of Sp1 to Sp1-binding sites on gene promoter. Subsequent transactivation of the promoter activation of the human 12(S)-lipoxygenase gene is induced. In addition to the functional role of Sp1 in gene regulation of 12(S)-lipoxygenase, recent studies have also demonstrated that Sp1 acting as an anchor protein to recruit transcription factor c-Jun is essential for growth factor and/or phorbol ester-induced expression of several genes.


Subject(s)
Arachidonate 12-Lipoxygenase/genetics , Sp1 Transcription Factor/chemistry , Sp1 Transcription Factor/physiology , Transcription, Genetic/physiology , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 12-Lipoxygenase/metabolism , Humans
16.
Am J Pathol ; 164(5): 1655-62, 2004 May.
Article in English | MEDLINE | ID: mdl-15111312

ABSTRACT

Alzheimer's disease (AD) is a chronic neurodegenerative disorder that impairs cognition and behavior. Although the initiating molecular events are not known, increasing evidence suggests that oxidative stress could play a functional role in its pathogenesis. Lipoxygenase (LOX) enzymes by oxidizing polyunsaturated fatty acids synthesize hydroperoxyacids, which are potent pro-oxidant mediators. Because circumstantial evidence suggests that 12/15-LOX is a major source of oxidative stress, we investigated the protein levels and activity of this enzyme in different brain regions of histopathologically confirmed AD and control cases. Using quantitative Western blot analysis we demonstrated that in affected frontal and temporal regions of AD brains the amount of 12/15-LOX was higher compared with controls, whereas no difference between the two groups was detected in the cerebellum. This observation was confirmed by immunohistochemical studies. Levels of 12/15-hydroxyeicosatetraenoic acids, metabolic products of 12/15-LOX, were also markedly elevated in AD brains compared to controls. This increase directly correlated with brain lipid peroxidation, and inversely with vitamin E levels. Finally, genetic deletion of this enzyme in vitro resulted in a reduction of the cellular oxidative stress response after incubation with H2O2 or amyloid beta. These data show that the 12/15-LOX metabolic pathway is increased and correlates with an oxidative imbalance in the AD brain, implying that this enzyme might contribute to the pathogenesis of this neurodegenerative disorder.


Subject(s)
Alzheimer Disease/enzymology , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Brain/metabolism , Oxidative Stress , Aged , Aged, 80 and over , Blotting, Western , Cerebellum/enzymology , Female , Frontal Lobe/enzymology , Humans , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Lipid Peroxidation , Male , Middle Aged , Oxidants/pharmacology , Oxygen/metabolism , Temporal Lobe/enzymology , Vitamin E/metabolism
17.
Int J Mol Med ; 13(1): 41-6, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14654968

ABSTRACT

The metabolism of arachidonic acid (AA) by either the cyclooxygenase (COX) or lipoxygenase (LOX) pathway generates eicosanoids, has been implicated in the pathogenesis of a variety of human diseases, including cancer, and may play important roles in tumor promotion, progression, and metastasis. The involvement of 12-LOX expression and function in tumor growth and metastasis has been reported in both murine and human tumor cell lines. The expression of 12-LOX in human renal cell carcinoma (RCC), normal kidney (NK) tissues and 3 kinds of RCC cell lines (Caki-1, A498, RC-1), and its effects on cell proliferation in 3 RCC cell lines were examined. Expression of 12-LOX protein was detected by immunohistochemistry and 12-LOX mRNA in RCC cell lines was detected by nested RT-PCR. Effects of 12-LOX inhibitor on RCC cell growth was examined by MTT assay, and to determine whether or not 12-LOX inhibitors induce apoptosis, we used Hoechst staining. While 12-LOX expression was detected slightly in NK tissues, a marked expression of 12-LOX was detected in RCC tissues. All human RCC cell lines expressed 12-LOX mRNA. The 12-LOX inhibitor baicalein caused marked inhibition of all three kinds of RCC cells in a concentration- and time-dependent manner. Cells treated with 12-LOX inhibitor showed chromatin condensation, cellular shrinkage, small membrane-bound bodies (apoptotic bodies), and cytoplasmic condensation. These results suggest 12-LOX may play a role in the progression of RCC in human tissue, and its inhibitors may become anticancer agents in human RCC.


Subject(s)
Apoptosis/drug effects , Arachidonate 12-Lipoxygenase/genetics , Carcinoma, Renal Cell/enzymology , Flavanones , Arachidonate 12-Lipoxygenase/biosynthesis , Carcinoma, Renal Cell/drug therapy , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Immunohistochemistry , Lipoxygenase Inhibitors , Reverse Transcriptase Polymerase Chain Reaction
18.
Prostaglandins Other Lipid Mediat ; 71(3-4): 189-204, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14518561

ABSTRACT

Platelet-type arachidonate 12-lipoxygenase (12-LOX) is highly expressed in many types of cancers and plays an important role in cancer pathophysiology. Arachidonic acid metabolism by 12-LOX results in the stable end product 12(S)-hydroxy eicosatetraenoic acid (12(S)-HETE), which is a signaling molecule with effects on cell proliferation, motility, invasiveness, angiogenesis, and inhibition of apoptosis. The myriad biological activities manifested by 12(S)-HETE appear to be mediated, at least in part, by the activation of NF-kappaB. Overexpression of the 12-LOX in PC-3 prostate cancer cells resulted in the constitutive activation of the transcription factor. The enzymatic product of arachidonic acid metabolism, 12(S)-HETE, mediates the activation of NF-kappaB by the 12-LOX. 12(S)-HETE treatment of PC-3 cells induced the degradation of IkappaB by the S6 proteasomal pathway and the activated NF-kappaB translocated to the nucleus causing kappaB-induced transcription. Specificity of the NF-kappaB activation by 12(S)-HETE was established by the use of a 12-LOX-specific inhibitor and 13(S)-HODE, a known 12(S)-HETE antagonist. Considering the known involvement of MAP kinase pathway in NF-kappaB activation and that of 12(S)-HETE in MAP kinase pathway, 12-LOX present in prostate cancer tissues may contribute to the constitutive activation of NF-kappaB in prostate cancer cells.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Antineoplastic Agents/pharmacology , Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 12-Lipoxygenase/genetics , Arachidonic Acid/metabolism , Blood Platelets/enzymology , Gene Expression Regulation, Neoplastic/drug effects , Humans , I-kappa B Proteins/biosynthesis , I-kappa B Proteins/metabolism , Leupeptins/pharmacology , Linoleic Acids/pharmacology , Lipoxygenase Inhibitors/pharmacology , Male , NF-kappa B/biosynthesis , NF-kappa B/genetics , Prostatic Neoplasms/genetics , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured/drug effects
19.
Prostaglandins Other Lipid Mediat ; 71(3-4): 277-85, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14518567

ABSTRACT

Human 12(S)-lipoxygenase is a platelet-type 12(S)-lipoxyenase. Its expression is detected in human erythroleukemia cells, human skin epidermal cells and human epidermoid carcinoma A431 cells. Treatment of A431 cells with EGF or PMA induces the gene expression of human 12(S)-lipoxygenase. The induction of gene expression is mediated through the cell signaling of MAPK activation, followed by the induction of c-Jun expression. The transcription factor Sp1 binding to the two Sp1 recognition motifs residing at -158 to 150 bp and -123 to 114 bp in the gene promoter is found to be essential for both EGF- and PMA-induced gene expression of human 12(S)-lipoxygenase. However, no change of Sp1 binding to GC-rich sequence was observed while no AP-1-binding site can be found in the responsive region of the promoter in EGF- and PMA-induced promoter activation of the human 12(S)-lipoxygenase gene. Since both of the transcription factors c-Jun and Sp1 are prerequisite for EGF and PMA response, interaction between c-Jun and Sp1 may account for the functional regulation of human 12(S)-lipoxygenase gene regulation. The direct and cooperative interaction between c-Jun and Sp1 induced by EGF or PMA activates the expression of the human 12(S)-lipoxygenase gene. Therefore, Sp1 may serve at least in part as a carrier to bring c-Jun to the promoter, thu's transactivating the transcriptional activity of the human 12(S)-lipoxygenase gene.


Subject(s)
Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/biosynthesis , Base Sequence , Epidermal Growth Factor/pharmacology , Gene Expression Regulation/drug effects , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Sequence Data , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/drug effects , Sp1 Transcription Factor/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transforming Growth Factor alpha/pharmacology , Tumor Cells, Cultured
20.
Cancer Res ; 63(14): 4258-67, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12874035

ABSTRACT

Arachidonic acid metabolism leads to the generation of biologically active metabolites that regulate cell growth and proliferation, as well as survival and apoptosis. We have demonstrated previously that platelet-type 12-lipoxygenase (LOX) regulates the growth and survival of a number of cancer cells. In this study, we show that overexpression of platelet-type 12-LOX in prostate cancer PC3 cells or epithelial cancer A431 cells significantly extended their survival and delayed apoptosis when cultured under serum-free conditions. These effects were shown to be a result of enhanced surface integrin expression, resulting in a more spread morphology of the cells in culture. PC3 cells transfected with 12-LOX displayed increased alpha(v)beta(3) and alpha(v)beta(5) integrin expression, whereas other integrins were unaltered. Transfected A431 cells did not express alpha(v)beta(3); however, alpha(v)beta(5) integrin expression was increased. Treatment of both transfected cell lines with monoclonal antibody to alpha(v)beta(5) (and in the case of PC3 cells, anti-alpha(v)beta(3)) resulted in significant apoptosis. In addition, treatment with 100 nM 12(S)-hydroxy-eicosatetraenoic acid, the end product of platelet-type 12-LOX, but not other hydroxy-eicosatetraenoic acids, enhanced the survival of wild-type PC3 and A431 cells and resulted in increased expression of alpha(v)beta(5). Furthermore, Baicalein or N-benzyl-N-hydroxy-5-phenylpentamide, specific 12-LOX inhibitors, significantly decreased alpha(v)beta(5)-mediated adhesion and survival in 12-LOX-overexpressing cells. The results show that 12-LOX regulates cell survival and apoptosis by affecting the expression and localization of the vitronectin receptors, alpha(v)beta(3) and alpha(v)beta(5), in two cancer cell lines.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Carcinoma, Squamous Cell/metabolism , Integrin alphaVbeta3/biosynthesis , Integrins/biosynthesis , Prostatic Neoplasms/metabolism , Receptors, Vitronectin/biosynthesis , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Apoptosis/physiology , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Blood Platelets/enzymology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Membrane/metabolism , Cell Survival/physiology , Flow Cytometry , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...