Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Clin Psychiatry ; 77(10): e1301-e1308, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27631140

ABSTRACT

OBJECTIVE: There is growing evidence that inflammation is an important mediator of pathophysiology in bipolar disorder. The omega-3 (n-3) and omega-6 (n-6) polyunsaturated fatty acid (PUFA) metabolic pathways participate in several inflammatory processes and have been linked through epidemiologic and clinical studies to bipolar disorder and its response to treatment. We review the data on PUFAs as biomarkers in bipolar disorder and n-3 PUFA used as treatment for bipolar disorder. DATA SOURCES: PubMed and CINAHL were searched for articles on PUFA and bipolar disorder published in the English language through November 6, 2013, with an updated search conducted on August 20, 2015. Keywords searched included omega 3 fatty acids and bipolar disorder, omega 3 fatty acids and bipolar mania, omega 3 fatty acids and bipolar depression, omega 3 fatty acids and mania, omega 3 fatty acids and cyclothymia, omega 3 fatty acids and hypomania, fatty acids and bipolar disorder, essential fatty acids and bipolar disorder, polyunsaturated fatty acids and bipolar disorder, DHA and bipolar disorder, and EPA and bipolar disorder. STUDY SELECTION: Studies selected measured PUFAs as biomarkers or introduced n-3 PUFA as treatment. RESULTS: We identified 17 relevant human clinical articles that either compared PUFA levels between a bipolar disorder group and a control group or used a PUFA intervention to treat depression or mania in bipolar disorder. Human studies suggest low n-3 red blood cell PUFA concentrations and correlations with clinical severity in studies of plasma concentrations in symptomatic bipolar disorder. Results of published n-3 PUFA dietary supplementation trials for bipolar disorder indicate efficacy in treatment for mania or depression in 5 of 5 open-label trials, efficacy in treatment of depression in 1 of 7 randomized controlled trials, and a signal for treatment of depression in 1 meta-analysis. CONCLUSIONS: Biomarker studies of PUFA and treatment studies of n-3 PUFA in bipolar disorder show promise for indicating a way forward in the study of PUFA in bipolar disorder. Investigation of the intake and metabolism of the n-3 and n-6 PUFA when supplementation is provided in treatment trials might offer clues for identification of when and how PUFA may be important for treatment in bipolar disorder.


Subject(s)
Biomarkers/blood , Bipolar Disorder/blood , Bipolar Disorder/drug therapy , Fatty Acids, Omega-3/blood , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/blood , Fatty Acids, Omega-6/therapeutic use , Bipolar Disorder/diagnosis , Bipolar Disorder/psychology , Humans , Randomized Controlled Trials as Topic , Reference Values
2.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27030719

ABSTRACT

BACKGROUND: Chronic idiopathic pain syndromes are major causes of personal suffering, disability, and societal expense. Dietary n-6 linoleic acid has increased markedly in modern industrialized populations over the past century. These high amounts of linoleic acid could hypothetically predispose to physical pain by increasing the production of pro-nociceptive linoleic acid-derived lipid autacoids and by interfering with the production of anti-nociceptive lipid autacoids derived from n-3 fatty acids. Here, we used a rat model to determine the effect of increasing dietary linoleic acid as a controlled variable for 15 weeks on nociceptive lipid autacoids and their precursor n-6 and n-3 fatty acids in tissues associated with idiopathic pain syndromes. RESULTS: Increasing dietary linoleic acid markedly increased the abundance of linoleic acid and its pro-nociceptive derivatives and reduced the abundance of n-3 eicosapentaenoic acid and docosahexaenoic acid and their anti-nociceptive monoepoxide derivatives. Diet-induced changes occurred in a tissue-specific manner, with marked alterations of nociceptive lipid autacoids in both peripheral and central tissues, and the most pronounced changes in their fatty acid precursors in peripheral tissues. CONCLUSIONS: The present findings provide biochemical support for the hypothesis that the high linoleic acid content of modern industrialized diets may create a biochemical susceptibility to develop chronic pain. Dietary linoleic acid lowering should be further investigated as part of an integrative strategy for the prevention and management of idiopathic pain syndromes.


Subject(s)
Autacoids/pharmacology , Dietary Fats/pharmacology , Linoleic Acid/pharmacology , Nociception/drug effects , Pain/pathology , Animals , Fatty Acids, Omega-3/pharmacology , Male , Organ Specificity/drug effects , Oxylipins/pharmacology , Rats, Inbred F344 , Syndrome
3.
Pain ; 156(4): 587-596, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25790451

ABSTRACT

Omega-3 and omega-6 fatty acids are precursors of bioactive lipid mediators posited to modulate both physical pain and psychological distress. In a randomized trial of 67 subjects with severe headaches, we recently demonstrated that targeted dietary manipulation-increasing omega-3 fatty acids with concurrent reduction in omega-6 linoleic acid (the H3-L6 intervention)-produced major reductions in headache compared with an omega-6 lowering (L6) intervention. Because chronic pain is often accompanied by psychological distress and impaired health-related quality of life (HRQOL), we used data from this trial to examine whether the H3-L6 intervention favorably impacted these domains. Additionally, we examined the effect of the interventions on the number of cases with substantial physical or mental impairments as defined by cutoff values in the Brief Symptom Inventory (BSI-18), Medical Outcomes Study Short Forms 12 (SF-12), Headache Impact Test (HIT-6), and the number of headache days per month. In the intention-to-treat analysis, participants in the H3-L6 group experienced statistically significant reductions in psychological distress (BSI-18 mean difference: -6.56; 95% confidence interval [CI]: -11.43 to -1.69) and improvements in SF-12 mental (mean difference: 6.01; 95% CI: 0.57 to 11.45) and physical (mean difference: 6.65; 95% CI: 2.14 to 11.16) health summary scores. At 12 weeks, the proportion of subjects experiencing substantial impairment according to cutoff values in the BSI-18, SF-12 physical, HIT-6, and headache days per month was significantly lower in the H3-L6 group. Dietary manipulation of n-3 and n-6 fatty acids, previously shown to produce major improvements in headache, was found to also reduce psychological distress and improve HRQOL and function.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/therapeutic use , Headache Disorders/complications , Quality of Life , Stress, Psychological/diet therapy , Stress, Psychological/etiology , Adult , Algorithms , Fatty Acids, Omega-3/blood , Fatty Acids, Omega-6/blood , Female , Follow-Up Studies , Headache Disorders/blood , Humans , Male , Mental Disorders/etiology , Middle Aged , Psychiatric Status Rating Scales
4.
PLoS One ; 9(5): e95318, 2014.
Article in English | MEDLINE | ID: mdl-24798187

ABSTRACT

BACKGROUND: Dietary long-chain n-3 polyunsaturated fatty acid (PUFA) supplementation may be beneficial for chronic brain illnesses, but the issue is not agreed on. We examined effects of dietary n-3 PUFA deprivation or supplementation, compared with an n-3 PUFA adequate diet (containing alpha-linolenic acid [18:3 n-3] but not docosahexaenoic acid [DHA, 22:6n-3]), on brain markers of lipid metabolism and excitotoxicity, in rats treated chronically with NMDA or saline. METHODS: Male rats after weaning were maintained on one of three diets for 15 weeks. After 12 weeks, each diet group was injected i.p. daily with saline (1 ml/kg) or a subconvulsive dose of NMDA (25 mg/kg) for 3 additional weeks. Then, brain fatty acid concentrations and various markers of excitotoxicity and fatty acid metabolism were measured. RESULTS: Compared to the diet-adequate group, brain DHA concentration was reduced, while n-6 docosapentaenoic acid (DPA, 22:5n-6) concentration was increased in the n-3 deficient group; arachidonic acid (AA, 20:4n-6) concentration was unchanged. These concentrations were unaffected by fish oil supplementation. Chronic NMDA increased brain cPLA2 activity in each of the three groups, but n-3 PUFA deprivation or fish oil did not change cPLA2 activity or protein compared with the adequate group. sPLA2 expression was unchanged in the three conditions, whereas iPLA2 expression was reduced by deprivation but not changed by supplementation. BDNF protein was reduced by NMDA in N-3 PUFA deficient rats, but protein levels of IL-1ß, NGF, and GFAP did not differ between groups. CONCLUSIONS: N-3 PUFA deprivation significantly worsened several pathological NMDA-induced changes produced in diet adequate rats, whereas n-3 PUFA supplementation did not affect NMDA induced changes. Supplementation may not be critical for this measured neuropathology once the diet has an adequate n-3 PUFA content.


Subject(s)
Brain Diseases/metabolism , Dietary Fats/adverse effects , Excitatory Amino Acid Agonists/adverse effects , Fatty Acids, Omega-3/adverse effects , Lipid Metabolism/drug effects , N-Methylaspartate/adverse effects , Animals , Brain Chemistry/drug effects , Brain Diseases/chemically induced , Brain Diseases/pathology , Chronic Disease , Dietary Fats/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Fatty Acids, Omega-3/pharmacology , Group IV Phospholipases A2/metabolism , Interleukin-1beta/metabolism , Male , N-Methylaspartate/pharmacology , Nerve Growth Factor/metabolism , Rats
5.
Article in English | MEDLINE | ID: mdl-24209500

ABSTRACT

Eicosapentaenoic acid (EPA, 20:5n-3), a precursor of docosahexaenoic acid (DHA), may benefit cardiovascular and brain health. Quantifying EPA's in vivo kinetics might elucidate these effects. [1-(14)C]EPA was infused i.v. for 5min in unanesthetized male rats fed a standard EPA-DHA diet. Plasma and microwaved tissue were analyzed. Kinetic parameters were calculated using our compartmental model. At 5min, 31-48% of labeled EPA in brain and heart was oxidized, 7% in liver. EPA incorporation rates from brain and liver precursor EPA-CoA pools into lipids, mainly phospholipids, were 36 and 2529nmol/s/g×10(-4), insignificant for heart. Deacylation-reacylation half-lives were 22h and 38-128min. Conversion rates to DHA equaled 0.65 and 25.1nmol/s/g×10(-4), respectively. The low brain concentration and incorporation rate and high oxidation of EPA suggest that, if EPA has a beneficial effect in brain, it might result from its suppression of peripheral inflammation and hepatic conversion to bioactive DHA.


Subject(s)
Brain/metabolism , Eicosapentaenoic Acid/pharmacokinetics , Fatty Acids, Omega-3/administration & dosage , Liver/metabolism , Myocardium/metabolism , Acyl Coenzyme A/analysis , Animals , Brain Chemistry , Carbon Radioisotopes , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/analysis , Eicosapentaenoic Acid/blood , Half-Life , Kinetics , Liver/chemistry , Male , Myocardium/chemistry , Oxidation-Reduction , Rats , Rats, Inbred F344
6.
J Neurochem ; 125(1): 63-73, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23336521

ABSTRACT

Aging is a risk factor for Alzheimer's disease (AD) and is associated with cognitive decline. However, underlying molecular mechanisms of brain aging are not clear. Recent studies suggest epigenetic influences on gene expression in AD, as DNA methylation levels influence protein and mRNA expression in postmortem AD brain. We hypothesized that some of these changes occur with normal aging. To test this hypothesis, we measured markers of the arachidonic acid (AA) cascade, neuroinflammation, pro- and anti-apoptosis factors, and gene specific epigenetic modifications in postmortem frontal cortex from nine middle-aged [41 ± 1 (SEM) years] and 10 aged subjects (70 ± 3 years). The aged compared with middle-aged brain showed elevated levels of neuroinflammatory and AA cascade markers, altered pro and anti-apoptosis factors and loss of synaptophysin. Some of these changes correlated with promoter hypermethylation of brain derived neurotrophic factor (BDNF), cyclic AMP responsive element binding protein (CREB), and synaptophysin and hypomethylation of BCL-2 associated X protein (BAX). These molecular alterations in aging are different from or more subtle than changes associated with AD pathology. The degree to which they are related to changes in cognition or behavior during normal aging remains to be evaluated.


Subject(s)
Aging/metabolism , Arachidonic Acid/metabolism , Epigenesis, Genetic , Frontal Lobe/metabolism , Synapses/metabolism , Adult , Aged , Aged, 80 and over , Aging/immunology , Apoptosis , Biomarkers/metabolism , DNA Methylation , Humans , Inflammation/metabolism , Middle Aged
7.
BMC Neurosci ; 13: 131, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23110484

ABSTRACT

BACKGROUND: In animal models, the metabolic syndrome elicits a cerebral response characterized by altered phospholipid and unesterified fatty acid concentrations and increases in pro-apoptotic inflammatory mediators that may cause synaptic loss and cognitive impairment. We hypothesized that these changes are associated with phospholipase (PLA2) enzymes that regulate arachidonic (AA, 20:4n-6) and docosahexaenoic (DHA, 22:6n-6) acid metabolism, major polyunsaturated fatty acids in brain. Male Wistar rats were fed a control or high-sucrose diet for 8 weeks. Brains were assayed for markers of AA metabolism (calcium-dependent cytosolic cPLA2 IVA and cyclooxygenases), DHA metabolism (calcium-independent iPLA2 VIA and lipoxygenases), brain-derived neurotrophic factor (BDNF), and synaptic integrity (drebrin and synaptophysin). Lipid concentrations were measured in brains subjected to high-energy microwave fixation. RESULTS: The high-sucrose compared with control diet induced insulin resistance, and increased phosphorylated-cPLA2 protein, cPLA2 and iPLA2 activity and 12-lipoxygenase mRNA, but decreased BDNF mRNA and protein, and drebrin mRNA. The concentration of several n-6 fatty acids in ethanolamine glycerophospholipids and lysophosphatidylcholine was increased, as was unesterified AA concentration. Eicosanoid concentrations (prostaglandin E2, thromboxane B2 and leukotriene B4) did not change. CONCLUSION: These findings show upregulated brain AA and DHA metabolism and reduced BDNF and drebrin, but no changes in eicosanoids, in an animal model of the metabolic syndrome. These changes might contribute to altered synaptic plasticity and cognitive impairment in rats and humans with the metabolic syndrome.


Subject(s)
Arachidonic Acid/metabolism , Brain/metabolism , Docosahexaenoic Acids/metabolism , Metabolic Diseases/pathology , Up-Regulation/physiology , 3-Hydroxybutyric Acid/metabolism , Animals , Blood Glucose/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cytoplasm/metabolism , Dietary Supplements , Disease Models, Animal , Fatty Acids/metabolism , Glucose Tolerance Test , Insulin/blood , Male , Metabolic Diseases/blood , Metabolic Diseases/metabolism , Phospholipases A2/metabolism , Radioisotopes , Rats , Rats, Wistar , Triglycerides/blood
8.
Article in English | MEDLINE | ID: mdl-22841517

ABSTRACT

In rats, FDA-approved mood stabilizers used for treating bipolar disorder (BD) selectively downregulate brain markers of the arachidonic acid (AA) cascade, which are upregulated in postmortem BD brain. Phase III clinical trials show that the anticonvulsant gabapentin (GBP) is ineffective in treating BD. We hypothesized that GBP would not alter the rat brain AA cascade. Chronic GBP (10 mg/kg body weight, injected i.p. for 30 days) compared to saline vehicle did not significantly alter brain expression or activity of AA-selective cytosolic phospholipase A(2) (cPLA(2)) IVA or secretory (s)PLA(2) IIA, activity of cyclooxygenase-2, or prostaglandin E(2) or thromboxane B(2) concentrations. Plasma esterified and unesterified AA concentration was unaffected. These results, taken with evidence of an upregulated AA cascade in the BD brain and that approved mood stabilizers downregulate the rat brain AA cascade, support the hypothesis that effective anti-BD drugs act by targeting the brain AA cascade whereas ineffective drugs (such as GBP) do not target this pathway, and suggest that the rat model might be used for screening new anti-BD drugs.


Subject(s)
Amines/pharmacology , Anti-Anxiety Agents/pharmacology , Arachidonic Acid/metabolism , Bipolar Disorder/metabolism , Brain/metabolism , Cyclohexanecarboxylic Acids/pharmacology , gamma-Aminobutyric Acid/pharmacology , Animals , Biomarkers/metabolism , Bipolar Disorder/blood , Bipolar Disorder/drug therapy , Brain/drug effects , Brain/enzymology , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Drug Evaluation, Preclinical , Fatty Acids/blood , Fructose/analogs & derivatives , Fructose/pharmacology , Gabapentin , Gene Expression , Group VI Phospholipases A2/genetics , Group VI Phospholipases A2/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phospholipases A2, Cytosolic/genetics , Phospholipases A2, Cytosolic/metabolism , Phospholipases A2, Secretory/genetics , Phospholipases A2, Secretory/metabolism , Rats , Rats, Inbred F344 , Thromboxane B2/metabolism , Topiramate
9.
BMC Neurosci ; 13: 50, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22621398

ABSTRACT

BACKGROUND: Neuroinflammation, caused by six days of intracerebroventricular infusion of bacterial lipopolysaccharide (LPS), stimulates rat brain arachidonic acid (AA) metabolism. The molecular changes associated with increased AA metabolism are not clear. We examined effects of a six-day infusion of a low-dose (0.5 ng/h) and a high-dose (250 ng/h) of LPS on neuroinflammatory, AA cascade, and pre- and post-synaptic markers in rat brain. We used artificial cerebrospinal fluid-infused brains as controls. RESULTS: Infusion of low- or high-dose LPS increased brain protein levels of TNFα, and iNOS, without significantly changing GFAP. High-dose LPS infusion upregulated brain protein and mRNA levels of AA cascade markers (cytosolic cPLA2-IVA, secretory sPLA2-V, cyclooxygenase-2 and 5-lipoxygenase), and of transcription factor NF-κB p50 DNA binding activity. Both LPS doses increased cPLA2 and p38 mitogen-activated protein kinase levels, while reducing protein levels of the pre-synaptic marker, synaptophysin. Post-synaptic markers drebrin and PSD95 protein levels were decreased with high- but not low-dose LPS. CONCLUSIONS: Chronic LPS infusion has differential effects, depending on dose, on inflammatory, AA and synaptic markers in rat brain. Neuroinflammation associated with upregulated brain AA metabolism can lead to synaptic dysfunction.


Subject(s)
Arachidonic Acid/metabolism , Brain/metabolism , Brain/pathology , Encephalitis/pathology , Gene Expression Regulation/drug effects , Synapses/metabolism , Analysis of Variance , Animals , Body Weight/drug effects , Brain/drug effects , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalitis/chemically induced , Infusions, Intraventricular , Lipopolysaccharides/toxicity , Lipoxygenases/genetics , Lipoxygenases/metabolism , Male , Microfilament Proteins/metabolism , Molecular Weight , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Article in English | MEDLINE | ID: mdl-21880477

ABSTRACT

Knowing threshold changes in brain lipids and lipid enzymes during dietary n-3 polyunsaturated fatty acid deprivation may elucidate dietary regulation of brain lipid metabolism. To determine thresholds, rats were fed for 15 weeks DHA-free diets having graded reductions of α-linolenic acid (α-LNA). Compared with control diet (4.6% α-LNA), plasma DHA fell significantly at 1.7% dietary α-LNA while brain DHA remained unchanged down to 0.8% α-LNA, when plasma and brain docosapentaenoic acid (DPAn-6) were increased and DHA-selective iPLA(2) and COX-1 activities were downregulated. Brain AA was unchanged by deprivation, but AA selective-cPLA(2), sPLA(2) and COX-2 activities were increased at or below 0.8% dietary α-LNA, possibly in response to elevated brain DPAn-6. In summary, homeostatic mechanisms appear to maintain a control brain DHA concentration down to 0.8% dietary DHA despite reduced plasma DHA, when DPAn-6 replaces DHA. At extreme deprivation, decreased brain iPLA(2) and COX-1 activities may reduce brain DHA loss.


Subject(s)
Brain/metabolism , Dietary Fats/metabolism , Docosahexaenoic Acids/deficiency , Fatty Acids, Unsaturated/metabolism , alpha-Linolenic Acid/deficiency , Animals , Brain/enzymology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Fatty Acids, Unsaturated/blood , Female , Gene Expression , Lipoxygenase/genetics , Lipoxygenase/metabolism , Liver/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phospholipases A2/genetics , Phospholipases A2/metabolism , Rats , Rats, Inbred F344 , Weight Gain
11.
Biochim Biophys Acta ; 1811(7-8): 484-9, 2011.
Article in English | MEDLINE | ID: mdl-21651989

ABSTRACT

The long-chain polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid (EPA, 20:5n-3), docosahexaenoic acid (DHA, 22:6n-3), and arachidonic acid (AA, 20:4n-6), are critical for health. These PUFAs can be synthesized in liver from their plant-derived precursors, α-linolenic acid (α-LNA, 18:3n-3) and linoleic acid (LA, 18:2n-6). Vegetarians and vegans may have suboptimal long-chain n-3 PUFA status, and the extent of the conversion of α-LNA to EPA and DHA by the liver is debatable. We quantified liver conversion of DHA and other n-3 PUFAs from α-LNA in rats fed a DHA-free but α-LNA (n-3 PUFA) adequate diet, and compared results to conversion of LA to AA. [U-(13)C]LA or [U-(13)C]α-LNA was infused intravenously for 2h at a constant rate into unanesthetized rats fed a DHA-free α-LNA adequate diet, and published equations were used to calculate kinetic parameters. The conversion coefficient k(⁎) of DHA from α-LNA was much higher than for AA from LA (97.2×10(-3) vs. 10.6×10(-3)min(-1)), suggesting that liver elongation-desaturation is more selective for n-3 PUFA biosynthesis on a per molecule basis. The net daily secretion rate of DHA, 20.3µmol/day, exceeded the reported brain DHA consumption rate by 50-fold, suggesting that the liver can maintain brain DHA metabolism with an adequate dietary supply solely of α-LNA. This infusion method could be used in vegetarians or vegans to determine minimal daily requirements of EPA and DHA in humans.


Subject(s)
Arachidonic Acid/metabolism , Dietary Fats, Unsaturated/administration & dosage , Docosahexaenoic Acids/metabolism , Liver/metabolism , Animals , Brain/metabolism , Diet, Vegetarian/adverse effects , Docosahexaenoic Acids/administration & dosage , Fatty Acids, Omega-3/administration & dosage , Infusions, Intravenous , Kinetics , Male , Nutritional Requirements , Rats , Rats, Inbred F344 , alpha-Linolenic Acid/administration & dosage
12.
Article in English | MEDLINE | ID: mdl-21664114

ABSTRACT

Diets given for 30 days with various mono-(MUFA) and poly-(PUFA) unsaturated fatty acid contents were evaluated for brain protection in magnesium-deficient mice: a commercial and three synthetic diets (n-6PUFA, n-3PUFA and MUFA-based chows enriched with 5% corn/sunflower oils 1:3, with 5% rapeseed oil and with 5% high oleic acid sunflower oil/sunflower oil 7:3, respectively). Unlike magnesium deprivation, they induced significant differences in brain and erythrocyte membrane phospholipid fatty acid compositions. n-3PUFA but not other diets protected magnesium-deficient mice against hyperactivity and moderately towards maximal electroshock- and NMDA-induced seizures. This diet also inhibited audiogenic seizures by 50%, preventing animal deaths. Because, like n-6PUFA diet, matched control MUFA diet failed to induce brain protections, alpha-linolenate (ALA) rather than reduced n-6 PUFA diet content is concluded to cause n-3PUFA neuroprotection. Present in vivo data also corroborate literature in vitro inhibition of T type calcium channels by n-3 PUFA, adding basis to ALA supplementation in human anti-epileptic/neuroprotective strategies.


Subject(s)
Brain/drug effects , Dietary Fats, Unsaturated/administration & dosage , Erythrocyte Membrane/drug effects , Magnesium Deficiency/drug therapy , Plant Oils/administration & dosage , Animals , Brain/cytology , Brain/metabolism , Dietary Fats, Unsaturated/pharmacology , Erythrocyte Membrane/metabolism , Fatty Acids, Monounsaturated/administration & dosage , Fatty Acids, Monounsaturated/pharmacology , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/pharmacology , Female , Humans , Magnesium Deficiency/metabolism , Mice , Models, Animal , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Phospholipids/metabolism , Plant Oils/pharmacology , Rapeseed Oil
13.
Neurochem Res ; 36(1): 139-45, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20981485

ABSTRACT

Pro-inflammatory and anti-inflammatory mediators derived from arachidonic acid (AA) modulate peripheral inflammation and its resolution. Aspirin (ASA) is a unique non-steroidal anti-inflammatory drug, which switches AA metabolism from prostaglandin E2 (PGE2) and thromboxane B2 (TXB2) to lipoxin A4 (LXA4) and 15-epi-LXA4. However, it is unknown whether chronic therapeutic doses of ASA are anti-inflammatory in the brain. We hypothesized that ASA would dampen increases in brain concentrations of AA metabolites in a rat model of neuroinflammation, produced by a 6-day intracerebroventricular infusion of bacterial lipopolysaccharide (LPS). In rats infused with LPS (0.5 ng/h) and given ASA-free water to drink, concentrations in high-energy microwaved brain of PGE2, TXB2 and leukotriene B4 (LTB4) were elevated. In rats infused with artificial cerebrospinal fluid, 6 weeks of treatment with a low (10 mg/kg/day) or high (100 mg/kg/day) ASA dose in drinking water decreased brain PGE2, but increased LTB4, LXA4 and 15-epi-LXA4 concentrations. Both doses attenuated the LPS effects on PGE2, and TXB2. The increments in LXA4 and 15-epi-LXA4 caused by high-dose ASA were significantly greater in LPS-infused rats. The ability of ASA to increase anti-inflammatory LXA4 and 15-epi-LXA4 and reduce pro-inflammatory PGE2 and TXB2 suggests considering aspirin further for treating clinical neuroinflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arachidonic Acid/metabolism , Aspirin/pharmacology , Brain/drug effects , Brain/metabolism , Animals , Dinoprostone/metabolism , Humans , Leukotriene B4/metabolism , Lipopolysaccharides/pharmacology , Lipoxins/metabolism , Male , Rats , Rats, Inbred F344 , Thromboxane B2/metabolism
14.
J Lipid Res ; 51(5): 1049-56, 2010 May.
Article in English | MEDLINE | ID: mdl-20040630

ABSTRACT

Neuroinflammation, caused by 6 days of intracerebroventricular infusion of a low dose of lipopolysaccharide (LPS; 0.5 ng/h), stimulates brain arachidonic acid (AA) metabolism in rats, but 6 weeks of lithium pretreatment reduces this effect. To further understand this action of lithium, we measured concentrations of eicosanoids and docosanoids generated from AA and docosahexaenoic acid (DHA), respectively, in high-energy microwaved rat brain using LC/MS/MS and two doses of LPS. In rats fed a lithium-free diet, low (0.5 ng/h)- or high (250 ng/h)-dose LPS compared with artificial cerebrospinal fluid increased brain unesterified AA and prostaglandin E(2) concentrations and activities of AA-selective Ca(2+)-dependent cytosolic phospholipase A(2) (cPLA(2))-IV and Ca(2+)-dependent secretory sPLA(2). LiCl feeding prevented these increments. Lithium had a significant main effect by increasing brain concentrations of lipoxygenase-derived AA metabolites, 5- hydroxyeicosatetraenoic acid (HETE), 5-oxo-eicosatetranoic acid, and 17-hydroxy-DHA by 1.8-, 4.3- and 1.9-fold compared with control diet. Lithium also increased 15-HETE in high-dose LPS-infused rats. Ca(2+)-independent iPLA(2)-VI activity and unesterified DHA and docosapentaenoic acid (22:5n-3) concentrations were unaffected by LPS or lithium. This study demonstrates, for the first time, that lithium can increase brain 17-hydroxy-DHA formation, indicating a new and potentially important therapeutic action of lithium.


Subject(s)
Arachidonic Acid/metabolism , Brain/drug effects , Brain/metabolism , Docosahexaenoic Acids/metabolism , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Lithium/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Bipolar Disorder/drug therapy , Body Weight/drug effects , Brain/radiation effects , Catheterization , Dietary Fats/analysis , Dose-Response Relationship, Drug , Esterification , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/physiopathology , Lithium/therapeutic use , Male , Microwaves , Rats , Time Factors
15.
J Psychiatr Res ; 44(3): 177-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19767014

ABSTRACT

Reduced concentrations of docosahexaenoic acid (DHA, 22:6n-3) and arachidonic acid (AA, 20:4n-6) have been reported in the postmortem bipolar disorder (BD) brain. Additionally, an increased prevalence of BD has been related to low dietary intake of fish, and dietary supplements containing fish products or DHA have been reported to ameliorate BD symptoms. These observations suggest that brain lipid metabolism, particularly involving DHA, is disturbed in BD. To test this suggestion, concentrations of different lipids were measured using internal standards in postmortem frontal cortex from eight BD patients and six matched controls. Compared with control cortex, the BD cortex showed no statistically significant difference in mean concentrations (per gram wet weight) of "stable" lipids (total lipid, total phospholipid, individual phospholipids, or cholesterol), of unesterified fatty acids, or of esterified DHA or AA within stable lipids. Fractional esterified AA and DHA concentrations also did not differ significantly between groups. Some fatty acid concentration differences were found in low-abundant cholesteryl ester. These results do not support the hypothesis of disturbed brain lipid concentrations, including concentrations of AA and DHA, in BD. Positron emission tomography might be used, however, to see if brain AA or DHA kinetics are disturbed in the disease.


Subject(s)
Arachidonic Acid/metabolism , Bipolar Disorder/metabolism , Docosahexaenoic Acids/metabolism , Frontal Lobe/metabolism , Adult , Aged , Aged, 80 and over , Bipolar Disorder/physiopathology , Cholesterol/metabolism , Female , Humans , Lipid Metabolism , Male , Middle Aged , Phospholipids/metabolism
16.
BMC Neurosci ; 10: 123, 2009 Sep 28.
Article in English | MEDLINE | ID: mdl-19785755

ABSTRACT

BACKGROUND: Chronic N-Methyl-d-aspartate (NMDA) administration to rats is reported to increase arachidonic acid signaling and upregulate neuroinflammatory markers in rat brain. These changes may damage brain cells. In this study, we determined if chronic NMDA administration (25 mg/kg i.p., 21 days) to rats would alter expression of pro- and anti-apoptotic factors in frontal cortex, compared with vehicle control. RESULTS: Using real time RT-PCR and Western blotting, chronic NMDA administration was shown to decrease mRNA and protein levels of anti-apoptotic markers Bcl-2 and BDNF, and of their transcription factor phospho-CREB in the cortex. Expression of pro-apoptotic Bax, Bad, and 14-3-3zeta was increased, as well as Fluoro-Jade B (FJB) staining, a marker of neuronal loss. CONCLUSION: This alteration in the balance between pro- and anti-apoptotic factors by chronic NMDA receptor activation in this animal model may contribute to neuronal loss, and further suggests that the model can be used to examine multiple processes involved in excitotoxicity.


Subject(s)
Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Frontal Lobe/metabolism , N-Methylaspartate/administration & dosage , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Blotting, Western , Excitatory Amino Acid Agonists/administration & dosage , Frontal Lobe/drug effects , Injections, Intraperitoneal , Male , Phosphorylation/drug effects , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions
17.
Biochim Biophys Acta ; 1791(2): 132-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19073280

ABSTRACT

Few studies have examined effects of feeding animals a diet deficient in n-6 polyunsaturated fatty acids (PUFAs) but with an adequate amount of n-3 PUFAs. To do this, we fed post-weaning male rats a control n-6 and n-3 PUFA adequate diet and an n-6 deficient diet for 15 weeks, and measured stable lipid and fatty acid concentrations in different organs. The deficient diet contained nutritionally essential linoleic acid (LA,18:2n-6) as 2.3% of total fatty acids (10% of the recommended minimum LA requirement for rodents) but no arachidonic acid (AA, 20:4n-6), and an adequate amount (4.8% of total fatty acids) of alpha-linolenic acid (18:3n-3). The deficient compared with adequate diet did not significantly affect body weight, but decreased testis weight by 10%. AA concentration was decreased significantly in serum (-86%), brain (-27%), liver (-68%), heart (-39%), testis (-25%), and epididymal adipose tissue (-77%). Eicosapentaenoic (20:5n-3) and docosahexaenoic acid (22:6n-3) concentrations were increased in all but adipose tissue, and the total monounsaturated fatty acid concentration was increased in all organs. The concentration of 20:3n-9, a marker of LA deficiency, was increased by the deficient diet, and serum concentrations of triacylglycerol, total cholesterol and total phospholipid were reduced. In summary, 15 weeks of dietary n-6 PUFA deficiency with n-3 PUFA adequacy significantly reduced n-6 PUFA concentrations in different organs of male rats, while increasing n-3 PUFA and monounsaturated fatty acid concentrations. This rat model could be used to study metabolic, functional and behavioral effects of dietary n-6 PUFA deficiency.


Subject(s)
Arachidonic Acid/metabolism , Dietary Fats, Unsaturated , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/deficiency , Animal Feed , Animals , Body Weight , Brain/metabolism , Chromatography, Gas , Lipids , Male , Organ Size , Rats , Rats, Inbred F344 , Weaning
18.
Neurochem Res ; 33(11): 2318-23, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18500552

ABSTRACT

Chronic N-Methyl-D: -aspartate (NMDA) administration, a model of excitotoxicity, and chronic intracerebroventricular lipopolysaccharide infusion, a model of neuroinflammation, are reported to upregulate arachidonic acid incorporation and turnover in rat brain phospholipids as well as enzymes involved in arachidonic acid metabolism. This suggests cross-talk between signaling pathways of excitotoxicity and of neuroinflammation, involving arachidonic acid. To test whether chronic NMDA administrations to rats can upregulate brain markers of neuroinflammation, NMDA (25 mg/kg i.p.) or vehicle (1 ml saline/kg i.p.) was administered daily to adult male rats for 21 days. Protein and mRNA levels of cytokines and other inflammatory markers were measured in the frontal cortex using immunoblot and real-time PCR. Compared with chronic vehicle, chronic NMDA significantly increased protein and mRNA levels of interleukin-1beta, tumor necrosis factor alpha, glial fibrillary acidic protein and inducible nitric oxide synthase. Chronic NMDA receptor overactivation results in increased levels of neuroinflammatory markers in the rat frontal cortex, consistent with cross-talk between excitotoxicity and neuroinflammation. As both processes have been reported in a number of human brain diseases, NMDA receptor inhibitors might be of use in treating neuroinflammation in these diseases.


Subject(s)
Biomarkers/metabolism , Cytokines/metabolism , Frontal Lobe/drug effects , Inflammation/metabolism , N-Methylaspartate/administration & dosage , Animals , Blotting, Western , Frontal Lobe/metabolism , Frontal Lobe/pathology , Male , N-Methylaspartate/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
19.
Neurochem Res ; 33(5): 861-6, 2008 May.
Article in English | MEDLINE | ID: mdl-18080190

ABSTRACT

Chronic administration to rats of mood-stabilizers that are effective against mania in bipolar disorder, is reported to downregulate markers of the brain arachidonic acid cascade. We hypothesized that chronic administration of lamotrigine, which is used to treat depression and rapid cycling in bipolar disorder, might do so as well. Male CDF rats were administered a therapeutically relevant dose of lamotrigine (10 mg/kg) or vehicle intragastrically once daily for 42 days. Protein levels of isoforms of phospholipase A(2) (PLA(2)) and of cyclooxygenase (COX), and the mRNA level of COX-2, were quantified in the frontal cortex using immunoblotting and RT-PCR, respectively. Compared to vehicle-treated rats, chronic lamotrigine significantly decreased frontal cortex protein and mRNA levels of COX-2 without altering protein levels of the PLA(2) isoforms. Consistent with the hypothesis, lamotrigine and other mood-stabilizers have a common downregulatory action on COX-2 expression in rat brain, which may account in part for their efficacy in bipolar disorder.


Subject(s)
Cyclooxygenase 2/metabolism , Down-Regulation/drug effects , Frontal Lobe/drug effects , RNA, Messenger/genetics , Triazines/pharmacology , Animals , Blotting, Western , Cyclooxygenase 2/genetics , Frontal Lobe/enzymology , Lamotrigine , Male , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Triazines/administration & dosage
20.
Article in English | MEDLINE | ID: mdl-18060754

ABSTRACT

Plasma alpha-linolenic acid (alpha-LNA, 18:3n-3) and linoleic acid (LA, 18:2n-6) do not contribute significantly to the brain content of docosahexaenoic acid (DHA, 22:6n-3) or arachidonic acid (AA, 20:4n-6), respectively, and neither DHA nor AA can be synthesized de novo in vertebrate tissue. Therefore, measured rates of incorporation of circulating DHA and AA into brain exactly represent their rates of consumption by brain. Positron emission tomography (PET) has been used to show, based on this information, that the adult human brain consumes AA and DHA at rates of 17.8 and 4.6 mg/day, respectively, and that AA consumption does not change significantly with age. In unanesthetized adult rats fed an n-3 PUFA "adequate" diet containing 4.6% alpha-LNA (of total fatty acids) as its only n-3 PUFA, the rate of liver synthesis of DHA was more than sufficient to maintain brain DHA, whereas the brain's rate of DHA synthesis is very low and unable to do so. Reducing dietary alpha-LNA in the DHA-free diet led to upregulation of liver but not brain coefficients of alpha-LNA conversion to DHA and of liver expression of elongases and desaturases that catalyze this conversion. Concurrently, brain DHA loss slowed due to downregulation of several of its DHA-metabolizing enzymes. Dietary alpha-LNA deficiency also promoted accumulation of brain docosapentaenoic acid (22:5n-6), and upregulated expression of AA-metabolizing enzymes, including cytosolic and secretory phospholipases A(2) and cyclooxygenase-2. These changes, plus reduced levels of brain derived neurotrophic factor (BDNF) and cAMP response element-binding protein (CREB) in n-3 PUFA diet deficient rats, likely render their brain more vulnerable to neuropathological insults.


Subject(s)
Brain/metabolism , Dietary Fats/metabolism , Fatty Acids, Essential/metabolism , Fatty Acids, Unsaturated/metabolism , Liver/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dietary Fats/administration & dosage , Docosahexaenoic Acids/metabolism , Fatty Acids, Omega-3/metabolism , Humans , Models, Biological , Phospholipids/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL