Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Neurochem Res ; 39(8): 1522-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24919816

ABSTRACT

Calcium-independent phospholipase A2 group VIa (iPLA2ß) preferentially releases docosahexaenoic acid (DHA) from the sn-2 position of phospholipids. Mutations of its gene, PLA2G6, are found in patients with several progressive motor disorders, including Parkinson disease. At 4 months, PLA2G6 knockout mice (iPLA2ß(-/-)) show minimal neuropathology but altered brain DHA metabolism. By 1 year, they develop motor disturbances, cerebellar neuronal loss, and striatal α-synuclein accumulation. We hypothesized that older iPLA2ß(-/-) mice also would exhibit inflammatory and other neuropathological changes. Real-time polymerase chain reaction and Western blotting were performed on whole brain homogenate from 15 to 20-month old male iPLA2ß(-/-) or wild-type (WT) mice. These older iPLA2ß(-/-) mice compared with WT showed molecular evidence of microglial (CD-11b, iNOS) and astrocytic (glial fibrillary acidic protein) activation, disturbed expression of enzymes involved in arachidonic acid metabolism, loss of neuroprotective brain derived neurotrophic factor, and accumulation of cytokine TNF-α messenger ribonucleic acid, consistent with neuroinflammatory pathology. There was no evidence of synaptic loss, of reduced expression of dopamine active reuptake transporter, or of accumulation of the Parkinson disease markers Parkin or Pink1. iPLA2γ expression was unchanged. iPLA2ß deficient mice show evidence of neuroinflammation and associated neuropathology with motor dysfunction in later life. These pathological biomarkers could be used to assess efficacy of dietary intervention, antioxidants or other therapies on disease progression in this mouse model of progressive human motor diseases associated with a PLA2G6 mutation.


Subject(s)
Aging/metabolism , Disease Models, Animal , Disease Progression , Group VI Phospholipases A2/deficiency , Motor Skills Disorders/metabolism , Aging/genetics , Animals , Group VI Phospholipases A2/genetics , Male , Mice , Mice, Knockout , Motor Skills Disorders/genetics , Motor Skills Disorders/pathology
2.
Biochim Biophys Acta ; 1821(9): 1278-86, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22349267

ABSTRACT

Calcium-independent phospholipase A(2) group VIA (iPLA(2)ß) releases docosahexaenoic acid (DHA) from phospholipids in vitro. Mutations in the iPLA(2)ß gene, PLA2G6, are associated with dystonia-parkinsonism and infantile neuroaxonal dystrophy. To understand the role of iPLA(2)ß in brain, we applied our in vivo kinetic method using radiolabeled DHA in 4 to 5-month-old wild type (iPLA(2)ß(+/+)) and knockout (iPLA(2)ß(-/-)) mice, and measured brain DHA kinetics, lipid concentrations, and expression of PLA(2), cyclooxygenase (COX), and lipoxygenase (LOX) enzymes. Compared to iPLA(2)ß(+/+) mice, iPLA(2)ß(-/-) mice showed decreased rates of incorporation of unesterified DHA from plasma into brain phospholipids, reduced concentrations of several fatty acids (including DHA) esterified in ethanolamine- and serine-glycerophospholipids, and increased lysophospholipid fatty acid concentrations. DHA turnover in brain phospholipids did not differ between genotypes. In iPLA(2)ß(-/-) mice, brain levels of iPLA(2)ß mRNA, protein, and activity were decreased, as was the iPLA(2)γ (Group VIB PLA(2)) mRNA level, while levels of secretory sPLA(2)-V mRNA, protein, and activity and cytosolic cPLA(2)-IVA mRNA were increased. Levels of COX-1 protein were decreased in brain, while COX-2 protein and mRNA were increased. Levels of 5-, 12-, and 15-LOX proteins did not differ significantly between genotypes. Thus, a genetic iPLA(2)ß deficiency in mice is associated with reduced DHA metabolism, profound changes in lipid-metabolizing enzyme expression (demonstrating lack of redundancy) and of phospholipid fatty acid content of brain (particularly of DHA), which may be relevant to neurologic abnormalities in humans with PLA2G6 mutations.


Subject(s)
Brain/metabolism , Docosahexaenoic Acids/metabolism , Group VI Phospholipases A2 , Lipid Metabolism , Nerve Tissue Proteins/metabolism , Phospholipids/metabolism , Animals , Brain/pathology , Brain Chemistry/genetics , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 1/genetics , Docosahexaenoic Acids/genetics , Gene Expression Regulation, Enzymologic/genetics , Humans , Lipoxygenase/biosynthesis , Lipoxygenase/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Phospholipases A2, Secretory/biosynthesis , Phospholipases A2, Secretory/genetics , Phospholipids/genetics
3.
J Neurochem ; 124(3): 376-87, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23121637

ABSTRACT

Chronic administration of mood stabilizers to rats down-regulates the brain arachidonic acid (AA) cascade. This down-regulation may explain their efficacy against bipolar disorder (BD), in which brain AA cascade markers are elevated. The atypical antipsychotics, olanzapine (OLZ) and clozapine (CLZ), also act against BD. When given to rats, both reduce brain cyclooxygenase activity and prostaglandin E(2) concentration; OLZ also reduces rat plasma unesterified and esterified AA concentrations, and AA incorporation and turnover in brain phospholipid. To test whether CLZ produces similar changes, we used our in vivo fatty acid method in rats given 10 mg/kg/day i.p. CLZ, or vehicle, for 30 days; or 1 day after CLZ washout. [1-(14) C]AA was infused intravenously for 5 min, arterial plasma was collected and high-energy microwaved brain was analyzed. CLZ increased incorporation coefficients ki * and decreased [corrected] rates J(in,i) of plasma unesterified AA into brain phospholipids. [corrected]. These effects disappeared after washout. Thus, CLZ and OLZ similarly down-regulated kinetics and cyclooxygenase expression of the brain AA cascade, likely by reducing plasma unesterified AA availability. Atypical antipsychotics and mood stabilizers may be therapeutic in BD by down-regulating, indirectly or directly respectively, the elevated brain AA cascade of that disease.


Subject(s)
Antipsychotic Agents/administration & dosage , Arachidonic Acid/blood , Arachidonic Acid/metabolism , Brain/drug effects , Brain/metabolism , Clozapine/administration & dosage , Animals , Antipsychotic Agents/blood , Antipsychotic Agents/pharmacokinetics , Arachidonic Acid/antagonists & inhibitors , Clozapine/blood , Clozapine/pharmacokinetics , Down-Regulation/genetics , Male , Rats , Rats, Inbred F344
4.
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
5.
J Neurochem ; 119(2): 364-76, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21812779

ABSTRACT

The atypical antipsychotic, olanzapine (OLZ), is used to treat bipolar disorder, but its therapeutic mechanism of action is not clear. Arachidonic acid (AA, 20:4n-6) plays a critical role in brain signaling and an up-regulated AA metabolic cascade was reported in postmortem brains from bipolar disorder patients. In this study, we tested whether, similar to the action of the mood stabilizers lithium, carbamazepine and valproate, chronic OLZ treatment would reduce AA turnover in rat brain. We administered OLZ (6 mg/kg/day) or vehicle i.p. to male rats once daily for 21 days. A washout group received 21 days of OLZ followed by vehicle on day 22. Two hours after the last injection, [1-¹4C]AA was infused intravenously for 5 min, and timed arterial blood samples were taken. After the rat was killed at 5 min, its brain was microwaved, removed and analyzed. Chronic OLZ decreased plasma unesterified AA concentration, AA incorporation rates and AA turnover in brain phospholipids. These effects were absent after washout. Consistent with reduced AA turnover, OLZ decreased brain cyclooxygenase activity and the brain concentration of the proinflammatory AA-derived metabolite, prostaglandin E2, In view of up-regulated brain AA metabolic markers in bipolar disorder, the abilities of OLZ and the mood stabilizers to commonly decrease prostaglandin E2, and AA turnover in rat brain phospholipids, albeit by different mechanisms, may be related to their efficacy against the disease.


Subject(s)
Antipsychotic Agents/pharmacology , Arachidonic Acid/metabolism , Benzodiazepines/pharmacology , Brain Chemistry/drug effects , Dinoprostone/metabolism , Acyl Coenzyme A/metabolism , Algorithms , Animals , Blood Pressure/drug effects , Blotting, Western , Body Weight/drug effects , Choline/metabolism , Chromatography, Gas , Cytosol/drug effects , Cytosol/metabolism , Half-Life , Heart Rate/drug effects , Kinetics , Lipid Metabolism/drug effects , Male , Olanzapine , Phospholipases A2/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction
6.
J Lipid Res ; 51(11): 3166-73, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20686114

ABSTRACT

Ca(2+)-independent phospholipase A(2)ß (iPLA(2)ß) selectively hydrolyzes docosahexaenoic acid (DHA, 22:6n-3) in vitro from phospholipid. Mutations in the PLA2G6 gene encoding this enzyme occur in patients with idiopathic neurodegeneration plus brain iron accumulation and dystonia-parkinsonism without iron accumulation, whereas mice lacking PLA2G6 show neurological dysfunction and neuropathology after 13 months. We hypothesized that brain DHA metabolism and signaling would be reduced in 4-month-old iPLA(2)ß-deficient mice without overt neuropathology. Saline or the cholinergic muscarinic M(1,3,5) receptor agonist arecoline (30 mg/kg) was administered to unanesthetized iPLA(2)ß(-/-), iPLA(2)ß(+/-), and iPLA(2)ß(+/+) mice, and [1-(14)C]DHA was infused intravenously. DHA incorporation coefficients k* and rates J(in), representing DHA metabolism, were determined using quantitative autoradiography in 81 brain regions. iPLA(2)ß(-/-) or iPLA(2)ß(+/-) compared with iPLA(2)ß(+/+) mice showed widespread and significant baseline reductions in k* and J(in) for DHA. Arecoline increased both parameters in brain regions of iPLA(2)ß(+/+) mice but quantitatively less so in iPLA(2)ß(-/-) and iPLA(2)ß(+/-) mice. Consistent with iPLA(2)ß's reported ability to selectively hydrolyze DHA from phospholipid in vitro, iPLA(2)ß deficiency reduces brain DHA metabolism and signaling in vivo at baseline and following M(1,3,5) receptor activation. Positron emission tomography might be used to image disturbed brain DHA metabolism in patients with PLA2G6 mutations.


Subject(s)
Brain/cytology , Brain/metabolism , Docosahexaenoic Acids/metabolism , Group VI Phospholipases A2/deficiency , Molecular Imaging , Signal Transduction , Animals , Arecoline/administration & dosage , Arecoline/pharmacology , Body Weight , Brain/blood supply , Brain/drug effects , Cerebral Arteries/physiology , Docosahexaenoic Acids/blood , Group VI Phospholipases A2/metabolism , Kinetics , Male , Mice
7.
Mol Neurobiol ; 54(6): 4303-4315, 2017 08.
Article in English | MEDLINE | ID: mdl-27339880

ABSTRACT

Linoleic acid (LA, 18:2n-6) is a precursor to arachidonic acid (AA, 20:4n-6), which can be converted by brain lipoxygenase and cyclooxygenase (COX) enzymes into various lipid mediators involved in the regulation of brain immunity. Brain AA metabolism is activated in rodents by the bacterial endotoxin, lipopolysaccharide (LPS). This study tested the hypothesis that dietary LA lowering, which limits plasma supply of AA to the brain, reduces LPS-induced upregulation in brain AA metabolism. Male Fischer CDF344 rats fed an adequate LA (5.2 % energy (en)) or low LA (0.4 % en) diet for 15 weeks were infused with LPS (250 ng/h) or vehicle into the fourth ventricle for 2 days using a mini-osmotic pump. The incorporation rate of intravenously infused unesterified 14C-AA into brain lipids, eicosanoids, and activities of phospholipase A2 and COX-1 and 2 enzymes were measured. Dietary LA lowering reduced the LPS-induced increase in prostaglandin E2 concentration and COX-2 activity (P < 0.05 by two-way ANOVA) without altering phospholipase activity. The 14C-AA incorporation rate into brain lipids was decreased by dietary LA lowering (P < 0.05 by two-way ANOVA). The present findings suggest that dietary LA lowering reduced LPS-induced increase in brain markers of AA metabolism. The clinical utility of LA lowering in brain disorders should be explored in future studies.


Subject(s)
Arachidonic Acid/metabolism , Brain/metabolism , Dietary Fats/pharmacology , Linoleic Acid/pharmacology , Lipopolysaccharides/pharmacology , Animals , Arachidonic Acid/blood , Body Weight/drug effects , Brain/drug effects , Brain/enzymology , Carbon Isotopes , Kinetics , Male , Rats, Inbred F344 , Time Factors
8.
Lipids ; 39(11): 1077-83, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15726822

ABSTRACT

Fatty acids (FA) regulate the expression of genes involved in lipid and energy metabolism. In particular, two transcription factors, sterol regulatory element binding protein-1c (SREBP-1c) and peroxisome proliferator activated receptor alpha (PPARalpha), have emerged as key mediators of gene regulation by FA. SREBP-1c induces a set of lipogenic enzymes in liver. Polyunsaturated fatty acids (PUFA), but not saturated or monounsaturated FA, suppress the induction of lipogenic genes by inhibiting the expression and processing of SREBP-1c. This unique effect of PUFA suggests that SREBP-1c may regulate the synthesis of unsaturated FA for incorporation into glycerolipids and cholesteryl esters. PPARalpha plays an essential role in metabolic adaptation to fasting by inducing the genes for mitochondrial and peroxisomal FA oxidation as well as those for ketogenesis in mitochondria. FA released from adipose tissue during fasting are considered as ligands of PPARalpha. Dietary PUFA, except for 18:2 n-6, are likely to induce FA oxidation enzymes via PPARalpha as a "feed-forward " mechanism. PPARalpha is also required for regulating the synthesis of highly unsaturated FA, indicating pleiotropic functions of PPARalpha in the regulation of lipid metabolic pathways. It is yet to be determined whether FA regulate other transcription factors such as liver-X receptor, hepatocyte nuclear factor 4, and carbohydrate response element binding protein.


Subject(s)
Fatty Acids/metabolism , Gene Expression Regulation , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Diet , Fatty Acids/pharmacology , Gene Expression Regulation/drug effects , Humans , Oxidation-Reduction , PPAR alpha/metabolism , Sterol Regulatory Element Binding Protein 1 , Transcription Factors/metabolism
9.
Article in English | MEDLINE | ID: mdl-24529827

ABSTRACT

Fetal and perinatal exposure to selective serotonin (5-HT) reuptake inhibitors (SSRIs) has been reported to alter childhood behavior, while transient early exposure in rodents is reported to alter their behavior and decrease brain extracellular 5-HT in adulthood. Since 5-HT2A/2C receptor-mediated neurotransmission can involve G-protein coupled activation of cytosolic phospholipase A2 (cPLA2), releasing arachidonic acid (ARA) from synaptic membrane phospholipid, we hypothesized that transient postnatal exposure to fluoxetine would alter brain ARA metabolism in adult mice. Brain ARA incorporation coefficients k* and rates Jin were quantitatively imaged following intravenous [1-(14)C]ARA infusion of unanesthetized adult mice that had been injected daily with fluoxetine (10mg/kg i.p.) or saline during postnatal days P4-P21. Expression of brain ARA metabolic enzymes and other relevant markers also was measured. On neuroimaging, k* and Jin was decreased widely in early fluoxetine- compared to saline-treated adult mice. Of the enzymes measured, cPLA2 activity was unchanged, while Ca(2+)-independent iPLA2 activity was increased. There was a significant 74% reduced protein level of cytochrome P450 (CYP) 4A, which can convert ARA to 20-HETE. Reduced brain ARA metabolism in adult mice transiently exposed to postnatal fluoxetine, and a 74% reduction in CYP4A protein, suggest long-term effects independent of drug presence in brain ARA metabolism, and in CYP4A metabolites. These changes might contribute to reported altered behavior following early SSRI in rodents.


Subject(s)
Arachidonic Acid/metabolism , Cytochrome P-450 CYP4A/metabolism , Fluoxetine/adverse effects , Nerve Tissue Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/adverse effects , Synaptic Transmission/drug effects , Animals , Behavior, Animal/drug effects , Calcium/metabolism , Calcium Signaling/drug effects , Cytochrome P-450 CYP4A/antagonists & inhibitors , Fluoxetine/pharmacology , Male , Mice , Selective Serotonin Reuptake Inhibitors/pharmacology
10.
Article in English | MEDLINE | ID: mdl-24675168

ABSTRACT

BACKGROUND: Dietary linoleic acid (LA, 18:2n-6) lowering in rats reduces n-6 polyunsaturated fatty acid (PUFA) plasma concentrations and increases n-3 PUFA (eicosapentaenoic (EPA) and docosahexaenoic acid (DHA)) concentrations. OBJECTIVE: To evaluate the extent to which 12 weeks of dietary n-6 PUFA lowering, with or without increased dietary n-3 PUFAs, alters unesterified and esterified plasma n-6 and n-3 PUFA concentrations in subjects with chronic headache. DESIGN: Secondary analysis of a randomized trial. Subjects with chronic headache were randomized for 12 weeks to (1) average n-3, low n-6 (L6) diet; or (2) high n-3, low n-6 LA (H3-L6) diet. Esterified and unesterified plasma fatty acids were quantified at baseline (0 weeks) and after 12 weeks on a diet. RESULTS: Compared to baseline, the L6 diet reduced esterified plasma LA and increased esterified n-3 PUFA concentrations (nmol/ml), but did not significantly change plasma arachidonic acid (AA, 20:4n-6) concentration. In addition, unesterified EPA concentration was increased significantly among unesterified fatty acids. The H3-L6 diet decreased esterified LA and AA concentrations, and produced more marked increases in esterified and unesterified n-3 PUFA concentrations. CONCLUSION: Dietary n-6 PUFA lowering for 12 weeks significantly reduces LA and increases n-3 PUFA concentrations in plasma, without altering plasma AA concentration. A concurrent increase in dietary n-3 PUFAs for 12 weeks further increases n-3 PUFA plasma concentrations and reduces AA.


Subject(s)
Chronic Pain , Dietary Supplements , Fatty Acids, Omega-3/blood , Fatty Acids, Omega-6 , Fatty Acids/blood , Headache , Adult , Animals , Chronic Pain/blood , Chronic Pain/diet therapy , Fatty Acids, Omega-6/administration & dosage , Fatty Acids, Omega-6/pharmacokinetics , Female , Headache/blood , Headache/diet therapy , Humans , Male , Middle Aged , Rats , Time Factors
11.
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
12.
Biol Psychiatry ; 75(1): 38-46, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23890734

ABSTRACT

BACKGROUND: Understanding the nature of environmental factors that contribute to behavioral health is critical for successful prevention strategies in individuals at risk for psychiatric disorders. These factors are typically experiential in nature, such as stress and urbanicity, but nutrition--in particular dietary deficiency of omega-3 polyunsaturated fatty acids (n-3 PUFAs)-has increasingly been implicated in the symptomatic onset of schizophrenia and mood disorders, which typically occurs during adolescence to early adulthood. Thus, adolescence might be the critical age range for the negative impact of diet as an environmental insult. METHODS: A rat model involving consecutive generations of n-3 PUFA deficiency was developed on the basis of the assumption that dietary trends toward decreased consumption of these fats began 4-5 decades ago when the parents of current adolescents were born. Behavioral performance in a wide range of tasks as well as markers of dopamine-related neurotransmission was compared in adolescents and adults fed n-3 PUFA adequate and deficient diets. RESULTS: In adolescents, dietary n-3 PUFA deficiency across consecutive generations produced a modality-selective and task-dependent impairment in cognitive and motivated behavior distinct from the deficits observed in adults. Although this dietary deficiency affected expression of dopamine-related proteins in both age groups in adolescents but not adults, there was an increase in tyrosine hydroxylase expression that was selective to the dorsal striatum. CONCLUSIONS: These data support a nutritional contribution to optimal cognitive and affective functioning in adolescents. Furthermore, they suggest that n-3 PUFA deficiency disrupts adolescent behaviors through enhanced dorsal striatal dopamine availability.


Subject(s)
Aging , Behavior, Animal/physiology , Brain/growth & development , Dopamine/metabolism , Fatty Acids, Omega-3/metabolism , Adolescent , Animals , Animals, Newborn , Conditioning, Operant/physiology , Exploratory Behavior/physiology , Extinction, Psychological , Female , Humans , Male , Maze Learning , Rats , Rats, Sprague-Dawley , Recognition, Psychology
13.
J Psychiatr Res ; 47(5): 636-43, 2013 May.
Article in English | MEDLINE | ID: mdl-23428160

ABSTRACT

BACKGROUND: Disturbances in prefrontal cortex phospholipid and fatty acid composition have been reported in patients with schizophrenia (SCZ), often as an incomplete lipid profile or a percent of total lipid concentration. In this study, we quantified absolute concentrations (nmol/g wet weight) and fractional concentrations (i.e. percent of total fatty acids) of several lipid classes and their constituent fatty acids in postmortem prefrontal cortex of SCZ patients (n = 10) and age-matched controls (n = 10). METHODS: Lipids were extracted, fractionated with thin layer chromatography and assayed. RESULTS: Mean total lipid, phospholipid, individual phospholipids, plasmalogen, triglyceride and cholesteryl ester concentrations did not differ significantly between the groups. Compared to controls, SCZ brains showed significant increases in several monounsaturated and polyunsaturated fatty acid absolute concentrations in cholesteryl ester. Significant increases or decreases occurred in palmitoleic, linoleic, γ-linolenic and n-3 docosapentaenoic acid absolute concentrations in total lipids, triglycerides or phospholipids. Changes in fractional concentrations did not consistently reflect absolute concentration changes. CONCLUSION: These findings suggest disturbed prefrontal cortex fatty acid absolute concentrations, particularly within cholesteryl esters, as a pathological aspect of schizophrenia.


Subject(s)
Fatty Acids/metabolism , Prefrontal Cortex/metabolism , Schizophrenia/pathology , Adult , Aged , Cholesterol Esters/metabolism , Chromatography, Thin Layer , Female , Humans , Male , Middle Aged , Phospholipids/metabolism , Plasmalogens/metabolism , Postmortem Changes , Triglycerides/metabolism
14.
Psychopharmacology (Berl) ; 222(4): 663-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22414961

ABSTRACT

BACKGROUND: The mode of action of clozapine, an atypical antipsychotic approved for treating schizophrenia (SZ) and used for bipolar disorder (BD) mania, remains unclear. We tested for overlap with the actions of the mood stabilizers, lithium, carbamazepine and valproate, which downregulate arachidonic acid (AA) cascade markers in rat brain and upregulate BDNF. AA cascade markers are upregulated in BD and SZ postmortem BD brain in association with neuroinflammation and synaptic loss, while BDNF is decreased. METHODS: Rats were injected intraperitoneally with a therapeutically relevant dose of clozapine (10 mg/kg/day) or with saline for 30 days, and AA cascade and synaptic markers and BDNF were measured in the brain. RESULTS: Compared with saline-injected rats, chronic clozapine increased brain activity, mRNA and protein levels of docosahexaenoic acid (DHA)-selective calcium-independent phospholipase A2 type VIA (iPLA2), mRNA and protein levels of BDNF and of the postsynaptic marker, drebrin, while decreasing cyclooxygenase (COX) activity and concentration of prostaglandin E2 (PGE2), a proinflammatory AA metabolite. Activity and expression of AA-selective calcium-dependent cytosolic cPLA2 type IVA and of secretory sPLA2 Type II were unchanged. CONCLUSIONS: These results show overlap with effects of mood stabilizers with regard to downregulation of COX activity and PGE2 and to increased BDNF and suggest a common action against the reported neuropathology of BD and SZ. The increased iPLA2 expression following clozapine suggests increased production of anti-inflammatory DHA metabolites, and, with increased BDNF and drebrin, clear neuroprotective action.


Subject(s)
Arachidonic Acid/metabolism , Clozapine/pharmacology , Docosahexaenoic Acids/metabolism , Down-Regulation/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects , Animals , Antipsychotic Agents/pharmacology , Biomarkers/metabolism , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Clozapine/administration & dosage , Cytoplasm/drug effects , Cytoplasm/metabolism , Dinoprostone/metabolism , Drug Administration Schedule , Male , Neuropeptides/metabolism , Phospholipases A2/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Inbred F344 , Synaptic Membranes/drug effects , Synaptic Membranes/metabolism
15.
J Nucl Med ; 53(9): 1383-91, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22851635

ABSTRACT

UNLABELLED: Arachidonic acid (AA) is found in high concentrations in brain phospholipids and is released as a second messenger during neurotransmission and much more so during neuroinflammation and excitotoxicity. Upregulated brain AA metabolism associated with neuroinflammation has been imaged in rodents using [1-(14)C]AA and with PET in Alzheimer disease patients using [1-(11)C]AA. Radiotracer brain AA uptake is independent of cerebral blood flow, making it an ideal tracer despite altered brain functional activity. However, the 20.4-min radioactive half-life of (11)C-AA and challenges of routinely synthesizing (11)C fatty acids limit their translational utility as PET biomarkers. METHODS: As a first step to develop a clinically useful (18)F-fluoroarachidonic acid ((18)F-FAA) with a long radioactive half-life of 109.8 min, we report here a high-yield stereoselective synthetic method of nonradioactive 20-(19)F-FAA. We tested its in vivo pharmacokinetics by infusing purified nonradioactive (19)F-FAA intravenously for 5 min at 2 doses in unanesthetized mice and measured its plasma and brain distribution using gas chromatography-mass spectrometry. RESULTS: Incorporation coefficients of injected (19)F-FAA into brain phospholipids (ratio of brain (19)F-FAA concentration to plasma input function) were 3- to 29-fold higher for choline glycerophospholipid and phosphatidylinositol than for ethanolamine glycerophospholipid and phosphatidylserine at each of the 2 tested doses. The selectivities and values of incorporation coefficients were comparable to those reported after [1-(14)C]AA (the natural arachidonate) infusion in mice. CONCLUSION: These results suggest that it would be worthwhile to translate our stereoselective synthetic method for (19)F-FAA to synthesize positron-emitting (18)F-FAA for human brain AA metabolism in neuroinflammatory disorders such as Alzheimer disease.


Subject(s)
Arachidonic Acid/chemical synthesis , Arachidonic Acid/pharmacokinetics , Halogenation , Molecular Imaging/methods , Nervous System Diseases/diagnosis , Animals , Arachidonic Acid/chemistry , Carbon Radioisotopes , Chemistry Techniques, Synthetic , Inflammation/diagnosis , Male , Mice
16.
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
17.
Article in English | MEDLINE | ID: mdl-22939288

ABSTRACT

Disturbed lipid metabolism has been reported in antiretroviral-naive HIV-1-infected patients suggesting a direct effect of the virus on lipid metabolism. To test that the HIV-1 virus alone could alter lipid concentrations, we measured these concentrations in an HIV-1 transgenic (Tg) rat model of human HIV-1 infection, which demonstrates peripheral and central pathology by 7-9 months of age. Concentrations were measured in high-energy microwaved heart, brain and liver from 7-9 month-old HIV-1 Tg and wildtype rats, and in plasma from non-microwaved rats. Plasma triglycerides and liver cholesteryl ester and total cholesterol concentrations were significantly higher in HIV-1 Tg rats than controls. Heart and plasma fatty acid concentrations reflected concentration differences in liver, which showed higher n-3 and n-6 polyunsaturated fatty acid (PUFA) concentrations in multiple lipid compartments. Fatty acid concentrations were increased or decreased in heart and liver phospholipid subfractions. Brain fatty acid concentrations differed significantly between the groups for minor fatty acids such as linoleic acid and n-3 docosapentaenoic acid. The profound changes in heart, plasma and liver lipid concentrations suggest a direct effect of chronic exposure to the HIV-1 virus on peripheral lipid (including PUFA) metabolism.


Subject(s)
Brain/metabolism , HIV-1/physiology , Lipid Metabolism/physiology , Liver/metabolism , Myocardium/metabolism , Animals , Cholesterol/blood , Cholesterol/metabolism , Cholesterol Esters/blood , Cholesterol Esters/metabolism , Fatty Acids, Unsaturated/blood , HIV-1/genetics , Humans , Lipid Metabolism/genetics , Lipids , Male , Rats , Rats, Inbred F344 , Rats, Transgenic , Triglycerides/blood
18.
Article in English | MEDLINE | ID: mdl-22959954

ABSTRACT

Linoleic acid (LA) is the most abundant polyunsaturated fatty acid in human diets, a major component of human tissues, and the direct precursor to the bioactive oxidized LA metabolites (OXLAMs), 9- and 13 hydroxy-octadecadienoic acid (9- and 13-HODE) and 9- and 13-oxo-octadecadienoic acid (9- and 13-oxoODE). These four OXLAMs have been mechanistically linked to pathological conditions ranging from cardiovascular disease to chronic pain. Plasma OXLAMs, which are elevated in Alzheimer's dementia and non-alcoholic steatohepatitis, have been proposed as biomarkers useful for indicating the presence and severity of both conditions. Because mammals lack the enzymatic machinery needed for de novo LA synthesis, the abundance of LA and OXLAMs in mammalian tissues may be modifiable via diet. To examine this issue in humans, we measured circulating LA and OXLAMs before and after a 12-week LA lowering dietary intervention in chronic headache patients. Lowering dietary LA significantly reduced the abundance of plasma OXLAMs, and reduced the LA content of multiple circulating lipid fractions that may serve as precursor pools for endogenous OXLAM synthesis. These results show that lowering dietary LA can reduce the synthesis and/or accumulation of oxidized LA derivatives that have been implicated in a variety of pathological conditions. Future studies evaluating the clinical implications of diet-induced OXLAM reductions are warranted.


Subject(s)
Linoleic Acid/blood , Linoleic Acid/metabolism , Adult , Dietary Fats/blood , Dietary Fats/metabolism , Female , Headache/blood , Headache/diet therapy , Headache/metabolism , Humans , Linoleic Acid/administration & dosage , Linoleic Acids/blood , Linoleic Acids/metabolism , Linoleic Acids, Conjugated/blood , Linoleic Acids, Conjugated/metabolism , Male , Middle Aged , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Young Adult
19.
Neuropharmacology ; 61(8): 1256-64, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21839100

ABSTRACT

BACKGROUND AND OBJECTIVE: Hyperdopaminergic signaling and an upregulated brain arachidonic acid (AA) cascade may contribute to bipolar disorder (BD). Lithium and carbamazepine, FDA-approved for the treatment of BD, attenuate brain dopaminergic D(2)-like (D(2), D(3), and D(4)) receptor signaling involving AA when given chronically to awake rats. We hypothesized that valproate (VPA), with mood-stabilizing properties, would also reduce D(2)-like-mediated signaling via AA. METHODS: An acute dose of quinpirole (1 mg/kg) or saline was administered to unanesthetized rats that had been treated for 30 days with a therapeutically relevant dose of VPA (200 mg/kg/day) or vehicle. Regional brain AA incorporation coefficients, k*, and incorporation rates, J(in), markers of AA signaling and metabolism, were measured by quantitative autoradiography after intravenous [1-(14)C]AA infusion. Whole brain concentrations of prostaglandin (PG)E(2) and thromboxane (TX)B(2) also were measured. RESULTS: Quinpirole compared to saline significantly increased k* in 40 of 83 brain regions, and increased brain concentrations of PGE(2) in chronic vehicle-treated rats. VPA treatment by itself reduced concentrations of plasma unesterified AA and whole brain PGE(2) and TXB(2), and blocked the quinpirole-induced increments in k* and PGE(2). CONCLUSION: These results further provide evidence that mood stabilizers downregulate brain dopaminergic D(2)-like receptor signaling involving AA.


Subject(s)
Antimanic Agents/pharmacology , Arachidonic Acid/metabolism , Brain/drug effects , Receptors, Dopamine D2/metabolism , Signal Transduction/drug effects , Valproic Acid/pharmacology , Analysis of Variance , Animals , Antimanic Agents/blood , Arachidonic Acid/pharmacokinetics , Autoradiography , Brain/metabolism , Carbon Radioisotopes/pharmacokinetics , Dinoprostone/metabolism , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Male , Rats , Rats, Inbred F344 , Thromboxane B2/metabolism , Valproic Acid/blood
20.
Am J Physiol Regul Integr Comp Physiol ; 288(6): R1525-35, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15650118

ABSTRACT

Peroxisome proliferator-activated receptor alpha (PPARalpha), a key regulator of fatty acid oxidation, is essential for adaptation to fasting in rats and mice. However, physiological functions of PPARalpha in other species, including humans, are controversial. A group of PPARalpha ligands called peroxisome proliferators (PPs) causes peroxisome proliferation and hepatocarcinogenesis only in rats and mice. To elucidate the role of PPARalpha in adaptation to fasting in nonproliferating species, we compared gene expressions in pig liver from fasted and clofibric acid (a PP)-fed groups against a control diet-fed group. As in rats and mice, fasting induced genes involved with mitochondrial fatty acid oxidation and ketogenesis in pigs. Those genes were also induced by clofibric acid feeding, indicating that PPARalpha mediates the induction of these genes. In contrast to rats and mice, little or no induction of genes for peroxisomal or microsomal fatty acid oxidation was observed in clofibric acid-fed pigs. Histology showed no significant hyperplasia or hepatomegaly in the clofibric acid-fed pigs, whereas it showed a reduction of glycogen by clofibric acid, an effect of PPs also observed in rats. Copy number of PPARalpha mRNA was higher in pigs than in mice and rats, suggesting that peroxisomal proliferation and hyperresponse of several genes to PPs seen only in rats and mice are unrelated to the abundance of PPARalpha. In conclusion, PPARalpha is likely to play a central role in adaptation to fasting in pig liver as in rats and mice.


Subject(s)
Fasting/physiology , Gene Expression Regulation/physiology , PPAR alpha/genetics , PPAR alpha/physiology , Animals , Cholesterol/metabolism , Clofibric Acid/pharmacology , DNA/genetics , DNA/isolation & purification , DNA Probes , Fatty Acids/metabolism , Gene Library , Ketone Bodies/metabolism , Liver/metabolism , Liver Glycogen/metabolism , Mice , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Swine
SELECTION OF CITATIONS
SEARCH DETAIL