Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-19931440

ABSTRACT

Retinal pigment epithelial (RPE) cells, derived from the neuroectoderm, biosynthesize the novel lipid mediator neuroprotectin D1 (NPD1) from docosahexaenoic acid (DHA) in response to oxidative stress or to neurotrophins, and in turn, elicits cytoprotection. Here, we report the identification of a 16,17-epoxide-containing intermediate in the biosynthesis of NPD1 in ARPE-19 cells from 17S-hydro-(peroxy)-docosahexaenoic acid. We prepared and isolated tritium-labeled NPD1 ([(3)H]-NPD1) and demonstrate specific and high-affinity stereoselective binding to ARPE-19 cells (K(d)=31.3+/-13.1 pmol/mg of cell protein). The stereospecific NPD1 interactions with these cells in turn gave potent protection against oxidative stress-induced apoptosis, and other structurally related compounds were weak competitors of NPD1 specific binding. This [(3)H]-NPD1/PD1 also displayed specific and selective high affinity binding with isolated human neutrophils (K(d) approximately 25 nM). Neither resolvin E1 nor lipoxin A(4) competed for [(3)H]-NPD1/PD1 specific binding with human neutrophils. Together, these results provide evidence for stereoselective specific binding of NPD1/PD1 with retinal pigment epithelial cells as well as human neutrophils. Moreover, they suggest specific receptors for this novel mediator in both the immune and visual systems.


Subject(s)
Antioxidants/metabolism , Docosahexaenoic Acids/metabolism , Neuroprotective Agents/metabolism , Neutrophils/metabolism , Retinal Pigment Epithelium/metabolism , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/chemical synthesis , Antioxidants/pharmacology , Apoptosis/drug effects , Binding, Competitive , Cell Line , Docosahexaenoic Acids/chemical synthesis , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Epoxy Compounds/chemistry , Epoxy Compounds/metabolism , Humans , Hydrogen Peroxide/toxicity , Isomerism , Kinetics , Lipoxins/metabolism , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Neutrophils/drug effects , Oxidative Stress/drug effects , Retinal Pigment Epithelium/drug effects , Tumor Necrosis Factor-alpha/toxicity
2.
J Biol Chem ; 284(26): 17877-82, 2009 Jun 26.
Article in English | MEDLINE | ID: mdl-19403949

ABSTRACT

The integrity of the retinal pigment epithelial (RPE) cell is essential for the survival of rod and cone photoreceptor cells. Several stressors, including reactive oxygen species, trigger apoptotic damage in RPE cells preceded by an anti-inflammatory, pro-survival response, the formation of neuroprotectin D1 (NPD1), an oxygenation product derived from the essential omega-3 fatty acid family member docosahexaenoic acid. To define the ability of NPD1 and other endogenous novel lipid mediators in cell survival, we generated a stable knockdown human RPE (ARPE-19) cell line using short hairpin RNA to target 15-lipoxygenase-1. The 15-lipoxygenase-1-deficient cells exhibited 30% of the protein expression, and 15-lipoxygenase-2 remained unchanged, as compared with an ARPE-19 cell line control established using nonspecific short hairpin RNA transfected cells. NPD1 synthesis was stimulated by tumor necrosis factor alpha/H2O2-mediated oxidative stress in nonspecific cells (controls), whereas in silenced cells, negligible amounts of NPD1, 12(S)- and 15(S)-hydroxyeicosatetraenoic acid, and lipoxin A4 were found under these conditions. Neither control nor the deficient cells showed an increase in 15-lipoxygenase-1 protein content after 16 h of oxidative stress, suggesting that the increased activity of 15-lipoxygenase-1 is due to activation of pre-existing proteins. 15-Lipoxygenase-silenced cells also displayed an exacerbated sensitivity to oxidative stress-induced apoptosis when compared with the control cells. NPD1 selectively and potently rescued 15-lipoxygenase-silenced cells from oxidative stress-induced apoptosis. These results demonstrate that 15-lipoxygenase-1 is activated by oxidative stress in ARPE-19 cells and that NPD1 is part of an early survival signaling in RPE cells.


Subject(s)
Apoptosis/drug effects , Arachidonate 15-Lipoxygenase/metabolism , Docosahexaenoic Acids/pharmacology , Oxidative Stress/drug effects , Pigment Epithelium of Eye/metabolism , Arachidonate 15-Lipoxygenase/genetics , Blotting, Western , Humans , Hydrogen Peroxide/pharmacology , Immunoenzyme Techniques , Lipoxins/metabolism , Lipoxygenase Inhibitors , Oxidants/pharmacology , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/drug effects , RNA, Small Interfering/pharmacology , Retina/cytology , Retina/drug effects , Retina/metabolism , Tumor Necrosis Factor-alpha/pharmacology
3.
Proc Natl Acad Sci U S A ; 104(32): 13158-63, 2007 Aug 07.
Article in English | MEDLINE | ID: mdl-17670935

ABSTRACT

Photoreceptor cell (rods and cones) renewal is accompanied by intermittent shedding of the distal tips of the outer segment followed by their phagocytosis in the retinal pigment epithelial (RPE) cells. This renewal is essential for vision, and it is thought that it fosters survival of photoreceptors and of RPE cells. However, no specific survival messenger/mediators have as yet been identified. We show here that photoreceptor outer segment (POS) phagocytosis markedly attenuates oxidative stress-induced apoptosis in ARPE-19 cells in culture. This phenomenon does not seem to be a generalized outcome of phagocytosis because nonbiological (polystyrene microsphere) phagocytosis did not elicit protection. The free docosahexaenoic acid (DHA) pool size and neuroprotectin D1 (NPD1) content increased during POS phagocytosis but not during microspheres phagocytosis. We have also explored other lipid mediators [lipoxin A4 and 15(S)- and 12(S)-hydroxyeicosatetraenoic acids] under these conditions and found them unchanged upon POS phagocytosis. Moreover, oxidative stress challenge to RPE cells undergoing POS phagocytosis further increased DHA and NPD1 content. Under these conditions, NPD1 was found within the RPE cells as well as in the culture medium, suggesting autocrine and paracrine bioactivity. Furthermore, using deuterium-labeled DHA, we show that as the availability of free DHA increases during oxidative stress, NPD1 synthesis is augmented in ARPE-19 cells. Our data suggest a distinct signaling that promotes survival of photoreceptor and RPE cells by enhancing the synthesis of NPD1 during phagocytosis. Taken together, NPD1 may be a mediator that promotes homeostatic regulation of cell integrity during photoreceptor cell renewal.


Subject(s)
Apoptosis , Docosahexaenoic Acids/metabolism , Oxidative Stress , Phagocytosis , Rod Cell Outer Segment/physiology , Cell Line , Humans , Microspheres , Pigment Epithelium of Eye/metabolism , Polystyrenes
4.
Proc Natl Acad Sci U S A ; 104(32): 13152-7, 2007 Aug 07.
Article in English | MEDLINE | ID: mdl-17670936

ABSTRACT

Integrity of retinal pigment epithelial cells is necessary for photoreceptor survival and vision. The essential omega-3 fatty acid, docosahexaenoic acid, attains its highest concentration in the human body in photoreceptors and is assumed to be a target for lipid peroxidation during cell damage. We have previously shown, in contrast, that docosahexaenoic acid is also the precursor of neuroprotectin D1 (NPD1), which now we demonstrate, acts against apoptosis mediated by A2E, a byproduct of phototransduction that becomes toxic when it accumulates in aging retinal pigment epithelial (RPE) cells and in some inherited retinal degenerations. Furthermore, we show that neurotrophins, particularly pigment epithelium-derived factor, induce NPD1 synthesis and its polarized apical secretion. Moreover, docosahexaenoic acid (DHA) elicits a concentration-dependent and selective potentiation of pigment epithelial-derived factor-stimulated NPD1 synthesis and release through the apical RPE cell surface. The bioactivity of signaling activated by pigment epithelium-derived factor and DHA uncovered synergistic cytoprotection with concomitant NPD1 synthesis when cells were challenged with oxidative stress. Also, DHA and pigment epithelium-derived factor synergistically modify the expression of Bcl-2 family members, activating antiapoptotic proteins and decreasing proapoptotic proteins, and by attenuating caspase 3 activation during oxidative stress. Thus, our findings demonstrate that DHA-derived NPD1 protects against RPE cell damage mediated by aging/disease-induced A2E accumulation. Also, our results identify neurotrophins as regulators of NPD1 and of its polarized apical efflux from RPE cells. Taken together, these findings imply NPD1 may elicit autocrine actions on RPE cells and paracrine bioactivity in cells located in the proximity of the interphotoreceptor matrix.


Subject(s)
Docosahexaenoic Acids/pharmacology , Nerve Growth Factors/pharmacology , Pigment Epithelium of Eye/drug effects , Signal Transduction/physiology , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Docosahexaenoic Acids/metabolism , Eye Proteins/pharmacology , Humans , Neuroprotective Agents/pharmacology , Oxidative Stress , Pigment Epithelium of Eye/cytology , Pyridinium Compounds/toxicity , Retinoids/toxicity , Serpins/pharmacology
5.
Bioorg Med Chem ; 15(5): 2206-15, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-16919959

ABSTRACT

A series of acetaminophen (APAP) analogs, 2-(1,1-dioxido-3-oxo-1,2-benzisothiazol-2(3H)-yl)-N-(4-hydroxyphenyl)alkanecarboxamides, bearing a heterocyclic moiety linked to the p-acylaminophenol fragment, were prepared in a general project to develop APAP analogs with modulated pharmacokinetic profiles. Unexpectedly, the products described maintained the in vivo analgesic profile, while the characteristic hepatotoxicity of APAP was consistently reduced. One of the products, 5a, was studied in vivo in comparison with APAP. Compound 5a displayed an analgesic efficacy comparable to that of APAP. A relatively high acute oral dose of 5a (6 mmol/kg) produced no measurable toxicity, whereas the equimolar dose of APAP increased transaminase activity, depleted hepatic and renal glutathione, and resulted in mortality. In human hepatocytes (HEPG-2) and in human primary cultures of normal liver cells, APAP, but not 5a, was associated with apoptotic cell death, Fas-ligand up-regulation, and CAR (constitutive androstane receptor) activation, contributing to a favorable safety profile of 5a as an orally delivered analgesic.


Subject(s)
Acetaminophen/analogs & derivatives , Acetaminophen/chemical synthesis , Acetaminophen/pharmacology , Animals , Drug Evaluation, Preclinical , Liver/drug effects , Magnetic Resonance Spectroscopy , Male , Mice
6.
J Physiol ; 569(Pt 3): 751-60, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16239276

ABSTRACT

Exogenous cannabinoids have been shown to significantly alter neuroendocrine output, presaging the emergence of endogenous cannabinoids as important signalling molecules in the neuroendocrine control of homeostatic and reproductive functions, including the stress response, energy metabolism and gonadal regulation. We showed recently that magnocellular and parvocellular neuroendocrine cells of the hypothalamic paraventricular nucleus and supraoptic nucleus (SON) respond to glucocorticoids by releasing endocannabinoids as retrograde messengers to modulate the synaptic release of glutamate. Here we show directly for the first time that both of the main endocannabinoids, anandamide (AEA) and 2-arachidonoyl glycerol (2-AG), are released in an activity-dependent fashion from the soma/dendrites of SON magnocellular neurones and suppress synaptic glutamate release and postsynaptic spiking. Cannabinoid reuptake blockade increases activity-dependent endocannabinoid levels in the region of the SON, and results in the inhibition of synaptically driven spiking activity in magnocellular neurones. Together, these findings demonstrate an activity-dependent release of AEA and 2-AG that leads to the suppression of glutamate release and that is capable of shaping spiking activity in magnocellular neurones. This activity-dependent regulation of excitatory synaptic input by endocannabinoids may play a role in determining spiking patterns characteristic of magnocellular neurones under stimulated conditions.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Supraoptic Nucleus/metabolism , Animals , Arachidonic Acids/metabolism , Arachidonic Acids/pharmacology , Benzoxazines , Benzyl Compounds/pharmacology , Cannabinoids/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Glycerides/metabolism , In Vitro Techniques , Male , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/drug effects , Neurons/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , Receptors, Presynaptic/drug effects , Receptors, Presynaptic/metabolism , Supraoptic Nucleus/drug effects , Synaptic Transmission/drug effects
7.
Ann N Y Acad Sci ; 1053: 137-47, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16179516

ABSTRACT

Synaptic activity and ischemia/injury promote lipid messenger formation through phospholipase-mediated cleavage of specific phospholipids from membrane reservoirs. Lipid messengers modulate signaling cascades, contributing to development, differentiation, function (e.g., memory), protection, regeneration, and repair of neurons and overall regulation of neuronal, glial, and endothelial cell functional integrity. Oxidative stress disrupts lipid signaling and promotes lipid peroxidation and neurodegeneration. Lipid signaling at the neurovascular unit (neurons, astrocytes, oligodendrocytes, microglia, and cells of the microvasculature) is altered in early cerebrovascular and neurodegenerative disease. We discuss how lipid signaling regulates critical events in neuronal survival. Aberrant synaptic plasticity (e.g., epileptogenesis) is highlighted to show how gene expression may drive synaptic circuitry formation in the "wrong" direction. Docosahexaenoic acid has been implicated in memory, photoreceptor cell biogenesis and function, and neuroprotection. Free docosahexaenoic acid released in the brain during experimental stroke leads to the synthesis of stereospecific messengers through oxygenation pathways. One messenger, 10,17S-docosatriene (neuroprotectin D1; NPD1), counteracts leukocyte infiltration and proinflammatory gene expression in brain ischemia-reperfusion. In retina, photoreceptor survival depends on retinal pigment epithelial (RPE) cell integrity. NPD1 is synthesized in RPE cells undergoing oxidative stress, potently counteracts oxidative stress-triggered apoptotic DNA damage in RPE, upregulates antiapoptotic proteins Bcl-2 and Bcl-x(L), and decreases proapoptotic Bax and Bad expression. These findings expand our understanding of how the nervous system counteracts redox disturbances, mitochondrial dysfunction, and proinflammatory conditions. The specificity and potency of NPD1 indicate a potential target for therapeutic intervention for stroke, age-related macular degeneration, spinal cord injury, and other neuroinflammatory or neurodegenerative diseases.


Subject(s)
Brain Injuries/physiopathology , Brain/physiopathology , Neurodegenerative Diseases/physiopathology , Signal Transduction/physiology , Animals , Docosahexaenoic Acids/metabolism , Humans , Inflammation/physiopathology , Oxidative Stress/physiology , Phospholipids/physiology , Second Messenger Systems/physiology
8.
J Clin Invest ; 115(10): 2774-83, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16151530

ABSTRACT

Deficiency in docosahexaenoic acid (DHA), a brain-essential omega-3 fatty acid, is associated with cognitive decline. Here we report that, in cytokine-stressed human neural cells, DHA attenuates amyloid-beta (Abeta) secretion, an effect accompanied by the formation of NPD1, a novel, DHA-derived 10,17S-docosatriene. DHA and NPD1 were reduced in Alzheimer disease (AD) hippocampal cornu ammonis region 1, but not in the thalamus or occipital lobes from the same brains. The expression of key enzymes in NPD1 biosynthesis, cytosolic phospholipase A2 and 15-lipoxygenase, was altered in AD hippocampus. NPD1 repressed Abeta42-triggered activation of proinflammatory genes while upregulating the antiapoptotic genes encoding Bcl-2, Bcl-xl, and Bfl-1(A1). Soluble amyloid precursor protein-alpha stimulated NPD1 biosynthesis from DHA. These results indicate that NPD1 promotes brain cell survival via the induction of antiapoptotic and neuroprotective gene-expression programs that suppress Abeta42-induced neurotoxicity.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Docosahexaenoic Acids/metabolism , Up-Regulation , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/toxicity , Apoptosis/drug effects , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Minor Histocompatibility Antigens , Peptide Fragments/toxicity , Phospholipases A/metabolism , Phospholipases A2 , Proto-Oncogene Proteins c-bcl-2/genetics , Thalamus/metabolism , Thalamus/pathology , Up-Regulation/drug effects , bcl-X Protein/genetics , bcl-X Protein/metabolism
9.
Endocrinology ; 146(10): 4292-301, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15994343

ABSTRACT

Glucocorticoids secreted in response to stress activation of the hypothalamic-pituitary-adrenal axis feed back onto the brain to rapidly suppress neuroendocrine activation, including oxytocin and vasopressin secretion. Here we show using whole-cell patch clamp recordings that glucocorticoids elicit a rapid, opposing action on synaptic glutamate and gamma-aminobutyric acid (GABA) release onto magnocellular neurons of the hypothalamic supraoptic nucleus and paraventricular nucleus, suppressing glutamate release and facilitating GABA release by activating a putative membrane receptor. The glucocorticoid effect on both glutamate and GABA release was blocked by inhibiting postsynaptic G protein activity, suggesting a dependence on postsynaptic G protein signaling and the involvement of a retrograde messenger. Biochemical analysis of hypothalamic slices treated with dexamethasone revealed a glucocorticoid-induced rapid increase in the levels of the endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG). The glucocorticoid suppression of glutamate release was blocked by the type I cannabinoid receptor cannabinoid receptor antagonist, AM251, and was mimicked and occluded by AEA and 2-AG, suggesting it was mediated by retrograde endocannabinoid release. The glucocorticoid facilitation of GABA release was also blocked by AM251 but was not mimicked by AEA, 2-AG, or a synthetic cannabinoid, WIN 55,212-2, nor was it blocked by vanilloid or ionotropic glutamate receptor antagonists, suggesting that it was mediated by a retrograde messenger acting at an AM251-sensitive, noncannabinoid/nonvanilloid receptor at presynaptic GABA terminals. The combined, opposing actions of glucocorticoids mediate a rapid inhibition of the magnocellular neuroendocrine cells, which in turn should mediate rapid feedback inhibition of the secretion of oxytocin and vasopressin by glucocorticoids during stress activation of the hypothalamic-pituitary-adrenal axis.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Cholesterol/pharmacology , Corticosterone/pharmacology , Endocannabinoids , Glutamic Acid/pharmacology , Hypothalamus/physiology , Neurons/physiology , gamma-Aminobutyric Acid/pharmacology , Animals , Dexamethasone/pharmacology , Excitatory Postsynaptic Potentials , Hypothalamus/drug effects , In Vitro Techniques , Male , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
10.
Stroke ; 36(1): 118-23, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15569878

ABSTRACT

BACKGROUND AND PURPOSE: High-dose human albumin therapy is strongly neuroprotective in models of brain ischemia and trauma and is currently being studied in a pilot-phase clinical stroke trial. Among its actions in ischemia, albumin induces the systemic mobilization of n-3 polyunsaturated fatty acids and may help to replenish polyunsaturated fatty acids lost from neural membranes. METHODS: We complexed 25% human albumin to docosahexaenoic acid (DHA; 22:6n-3) and compared its neuroprotective efficacy with that of native albumin in rats with 2-hour focal ischemia produced by intraluminal suture-occlusion of the middle cerebral artery. RESULTS: In animals treated with DHA-albumin, 0.63 g/kg, the improvement in neurobehavioral scores at 72 hours significantly exceeded that of other treatment groups, and the extent of histological protection (86% reduction in cortical infarction) was highly significant and tended to surpass the degree of cortical protection produced by native albumin at 1.25 g/kg (65%). DHA-albumin 0.63 g/kg, but not native albumin, also significantly reduced subcortical infarction and markedly diminished brain swelling. Lipidomic analysis of DHA-albumin-treated postischemic brains revealed a large accumulation of the neuroprotective DHA metabolite, 10,17S-docosatriene, in the ipsilateral hemisphere. CONCLUSIONS: The high-grade neuroprotection afforded by the DHA-albumin complex at relatively low albumin doses is clinically advantageous in that it might reduce the likelihood of acute intravascular volume overload and congestive heart failure sometimes induced when patients with compromised cardiovascular function are treated with high-dose albumin.


Subject(s)
Brain Ischemia/prevention & control , Docosahexaenoic Acids/therapeutic use , Neuroprotective Agents/therapeutic use , Serum Albumin/therapeutic use , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacokinetics , Male , Neuroprotective Agents/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reflex , Serum Albumin/pharmacokinetics
11.
Proc Natl Acad Sci U S A ; 101(22): 8491-6, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15152078

ABSTRACT

Docosahexaenoic acid (DHA) is a lipid peroxidation target in oxidative injury to retinal pigment epithelium (RPE) and retina. Photoreceptor and synaptic membranes share the highest content of DHA of all cell membranes. This fatty acid is required for RPE functional integrity; however, it is not known whether specific mediators generated from DHA contribute to its biological significance. We used human ARPE-19 cells and demonstrated the synthesis of 10,17S-docosatriene [neuroprotectin D1 (NPD1)]. This synthesis was enhanced by the calcium ionophore A-23187, by IL-1beta, or by supplying DHA. Under these conditions, there is a time-dependent release of endogenous free DHA followed by NPD1 formation, suggesting that phospholipase A(2) releases the mediator's precursor. Added NPD1 potently counteracted H(2)O(2)/tumor necrosis factor alpha oxidative-stress-triggered apoptotic RPE DNA damage. NPD1 also up-regulated the antiapoptotic proteins Bcl-2 and Bcl-x(L) and decreased proapoptotic Bax and Bad expression. Moreover, NPD1 (50 nM) inhibited oxidative-stress-induced caspase-3 activation. NPD1 also inhibited IL-1beta-stimulated expression of cyclooxygenase 2 promoter transfected into ARPE-19 cells. Overall, NPD1 protected RPE cells from oxidative-stress-induced apoptosis, and we predict that it will similarly protect neurons. This lipid mediator therefore may indirectly contribute to photoreceptor cell survival as well. Because both RPE and photoreceptor cells die in retinal degenerations, our findings contribute to the understanding of retinal cell survival signaling and potentially to the development of new therapeutic strategies.


Subject(s)
Docosahexaenoic Acids/metabolism , Epithelial Cells/metabolism , Oxidative Stress , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/metabolism , Apoptosis/physiology , Caspase 3 , Caspases/metabolism , Cell Line , Cyclooxygenase 2 , DNA Fragmentation , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/pharmacology , Epithelial Cells/drug effects , Humans , Interleukin-1/metabolism , Isoenzymes/metabolism , Lipid Peroxidation , Membrane Proteins , Molecular Structure , Pigment Epithelium of Eye/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
12.
J Biol Chem ; 278(44): 43807-17, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12923200

ABSTRACT

Ischemic stroke triggers lipid peroxidation and neuronal injury. Docosahexaenoic acid released from membrane phospholipids during brain ischemia is a major source of lipid peroxides. Leukocyte infiltration and pro-inflammatory gene expression also contribute to stroke damage. In this study using lipidomic analysis, we have identified stereospecific messengers from docosahexaenoate-oxygenation pathways in a mouse stroke model. Aspirin, widely used to prevent cerebrovascular disease, activates an additional pathway, which includes the 17R-resolvins. The newly discovered brain messenger 10,17S-docosatriene potently inhibited leukocyte infiltration, NFkappaB, and cyclooxygenase-2 induction in experimental stroke and elicited neuroprotection. In addition, in neural cells in culture, this lipid messenger also inhibited both interleukin 1-beta-induced NFkappaB activation and cyclooxygenase-2 expression. Thus, the specific novel bioactive docosanoids generated in vivo counteract leukocyte-mediated injury as well as pro-inflammatory gene induction. These results challenge the view that docosahexaenoate only participates in brain damage and demonstrate that this fatty acid is also the endogenous precursor to a neuroprotective signaling response to ischemia-reperfusion.


Subject(s)
Brain/pathology , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/pharmacology , Leukocytes/metabolism , Reperfusion Injury , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Cells, Cultured , Cerebral Arteries/pathology , Cyclooxygenase 2 , Hippocampus/metabolism , Humans , Immunohistochemistry , Interleukin-1/metabolism , Isoenzymes/metabolism , Leukocytes/pathology , Lipid Peroxidation , Membrane Proteins , Mice , Microscopy, Fluorescence , Models, Chemical , NF-kappa B/metabolism , Neurons/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Signal Transduction , Stem Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...