Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 54(6): 4303-4315, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27339880

RESUMO

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.


Assuntos
Ácido Araquidônico/metabolismo , Encéfalo/metabolismo , Gorduras na Dieta/farmacologia , Ácido Linoleico/farmacologia , Lipopolissacarídeos/farmacologia , Animais , Ácido Araquidônico/sangue , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Isótopos de Carbono , Cinética , Masculino , Ratos Endogâmicos F344 , Fatores de Tempo
2.
BMC Neurosci ; 15: 113, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25359512

RESUMO

BACKGROUND: The polyunsaturated fatty acid, docosahexaenoic acid (DHA), participates in neurotransmission involving activation of calcium-independent phospholipase A2 (iPLA2), which is coupled to muscarinic, cholinergic and serotonergic neuroreceptors. Drug induced activation of iPLA2 can be measured in vivo with quantitative autoradiography using 14C-DHA as a probe. The present study used this approach to address whether a DHA signal is produced following dompaminergic (D)2-like receptor activation with quinpirole in rat brain. Unanesthetized rats were infused intravenously with 14C-DHA one minute after saline or quinpirole infusion, and serial blood samples were collected over a 20-minute period to obtain plasma. The animals were euthanized with sodium pentobarbital and their brains excised, coronally dissected and subjected to quantitative autoradiography to derive the regional incorporation coefficient, k*, a marker of DHA signaling. Plasma labeled and unlabeled unesterified DHA concentrations were measured. RESULTS: The incorporation coefficient (k*) for DHA did not differ significantly between quinpirole-treated and control rats in any of 81 identified brain regions. Plasma labeled DHA concentration over the 20-minute collection period (input function) and unlabeled unesterified DHA concentration did not differ significantly between the two groups. CONCLUSION: These findings demonstrate that D2-like receptor initiated signaling does not involve DHA as a second messenger, and likely does not involve iPLA2 activation.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Agonistas de Dopamina/farmacologia , Quimpirol/farmacologia , Animais , Autorradiografia , Análise Química do Sangue , Radioisótopos de Carbono , Ácidos Graxos/sangue , Masculino , Distribuição Aleatória , Ratos Endogâmicos F344 , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-24529827

RESUMO

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.


Assuntos
Ácido Araquidônico/metabolismo , Citocromo P-450 CYP4A/metabolismo , Fluoxetina/efeitos adversos , Proteínas do Tecido Nervoso/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Transmissão Sináptica/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Citocromo P-450 CYP4A/antagonistas & inibidores , Fluoxetina/farmacologia , Masculino , Camundongos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
4.
BMC Neurosci ; 13: 131, 2012 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23110484

RESUMO

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.


Assuntos
Ácido Araquidônico/metabolismo , Encéfalo/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Doenças Metabólicas/patologia , Regulação para Cima/fisiologia , Ácido 3-Hidroxibutírico/metabolismo , Animais , Glicemia/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Citoplasma/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Teste de Tolerância a Glucose , Insulina/sangue , Masculino , Doenças Metabólicas/sangue , Doenças Metabólicas/metabolismo , Fosfolipases A2/metabolismo , Radioisótopos , Ratos , Ratos Wistar , Triglicerídeos/sangue
5.
Artigo em Inglês | MEDLINE | ID: mdl-22939288

RESUMO

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.


Assuntos
Encéfalo/metabolismo , HIV-1/fisiologia , Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , Miocárdio/metabolismo , Animais , Colesterol/sangue , Colesterol/metabolismo , Ésteres do Colesterol/sangue , Ésteres do Colesterol/metabolismo , Ácidos Graxos Insaturados/sangue , HIV-1/genética , Humanos , Metabolismo dos Lipídeos/genética , Lipídeos , Masculino , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos , Triglicerídeos/sangue
6.
Artigo em Inglês | MEDLINE | ID: mdl-22841517

RESUMO

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.


Assuntos
Aminas/farmacologia , Ansiolíticos/farmacologia , Ácido Araquidônico/metabolismo , Transtorno Bipolar/metabolismo , Encéfalo/metabolismo , Ácidos Cicloexanocarboxílicos/farmacologia , Ácido gama-Aminobutírico/farmacologia , Animais , Biomarcadores/metabolismo , Transtorno Bipolar/sangue , Transtorno Bipolar/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Avaliação Pré-Clínica de Medicamentos , Ácidos Graxos/sangue , Frutose/análogos & derivados , Frutose/farmacologia , Gabapentina , Expressão Gênica , Fosfolipases A2 do Grupo VI/genética , Fosfolipases A2 do Grupo VI/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fosfolipases A2 Citosólicas/genética , Fosfolipases A2 Citosólicas/metabolismo , Fosfolipases A2 Secretórias/genética , Fosfolipases A2 Secretórias/metabolismo , Ratos , Ratos Endogâmicos F344 , Tromboxano B2/metabolismo , Topiramato
7.
J Neuroimmune Pharmacol ; 7(3): 701-13, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22760927

RESUMO

HIV-1 transgenic (Tg) rats, a model for human HIV-1 associated neurocognitive disorder (HAND), show upregulated markers of brain arachidonic acid (AA) metabolism with neuroinflammation after 7 months of age. Since lithium decreases AA metabolism in a rat lipopolysaccharide model of neuroinflammation, and may be useful in HAND, we hypothesized that lithium would dampen upregulated brain AA metabolism in HIV-1 Tg rats. Regional brain AA incorporation coefficients k* and rates J ( in ), markers of AA signaling and metabolism, were measured in 81 brain regions using quantitative autoradiography, after intravenous [1-(14) C]AA infusion in unanesthetized 10-month-old HIV-1 Tg and age-matched wildtype rats that had been fed a control or LiCl diet for 6 weeks. k* and J ( in ) for AA were significantly higher in HIV-1 Tg than wildtype rats fed the control diet. Lithium feeding reduced plasma unesterified AA concentration in both groups and J ( in ) in wildtype rats, and blocked increments in k* (19 of 54 regions) and J ( in ) (77 of 81 regions) in HIV-1 Tg rats. These in vivo neuroimaging data indicate that lithium treatment dampened upregulated brain AA metabolism in HIV-1 Tg rats. Lithium may improve cognitive dysfunction and be neuroprotective in HIV-1 patients with HAND through a comparable effect.


Assuntos
Ácido Araquidônico/antagonistas & inibidores , Ácido Araquidônico/biossíntese , Encéfalo/metabolismo , Infecções por HIV/metabolismo , HIV-1 , Cloreto de Lítio/administração & dosagem , Regulação para Cima/fisiologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/virologia , Infecções por HIV/tratamento farmacológico , Masculino , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Ratos Transgênicos , Regulação para Cima/efeitos dos fármacos
8.
Int J Neurosci ; 122(7): 373-80, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22376027

RESUMO

BACKGROUND: Dopamine transporter (DAT) homozygous knockout (DAT(-/-)) mice have a 10-fold higher extracellular (DA) concentration in the caudate-putamen and nucleus accumbens than do wildtype (DAT(+/+)) mice, but show reduced presynaptic DA synthesis and fewer postsynaptic D(2) receptors. One aspect of neurotransmission involves DA binding to postsynaptic D(2)-like receptors coupled to cytosolic phospholipase A(2) (cPLA(2)), which releases the second messenger, arachidonic acid (AA), from synaptic membrane phospholipid. We hypothesized that tonic overactivation of D(2)-like receptors in DAT(-/-) mice due to the excess DA would not increase brain AA signaling, because of compensatory downregulation of postsynaptic DA signaling mechanisms. METHODS: [1-(14)C]AA was infused intravenously for 3 min in unanesthetized DAT(+/+), heterozygous (DAT(+/-)), and DAT(-/-) mice. AA incorporation coefficients k* and rates J(in), markers of AA metabolism and signaling, were imaged in 83 brain regions using quantitative autoradiography; brain cPLA(2)-IV activity also was measured. RESULTS: Neither k* nor J(in) for AA in any brain region, or brain cPLA(2)-IV activity, differed significantly among DAT(-/-), DAT(+/-), and DAT(+/+) mice. CONCLUSIONS: These results differ from reported increases in k* and J(in) for AA, and in brain cPLA(2) expression, in serotonin reuptake transporter (5-HTT) knockout mice, and suggest that postsynaptic dopaminergic neurotransmission mechanisms involving AA are downregulated despite elevated DA in DAT(-/-) mice.


Assuntos
Ácido Araquidônico/metabolismo , Encéfalo/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/deficiência , Transdução de Sinais/genética , Análise de Variância , Animais , Autorradiografia , Peso Corporal/genética , Encéfalo/enzimologia , Mapeamento Encefálico , Isótopos de Carbono/metabolismo , Regulação para Baixo/genética , Ácidos Graxos/sangue , Masculino , Camundongos , Camundongos Knockout , Fosfolipases A2 , Potássio/metabolismo
9.
Int J Neuropsychopharmacol ; 15(7): 931-43, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21733229

RESUMO

An up-regulated brain arachidonic acid (AA) cascade and a hyperglutamatergic state characterize bipolar disorder (BD). Lamotrigine (LTG), a mood stabilizer approved for treating BD, is reported to interfere with glutamatergic neurotransmission involving N-methyl-d-aspartate receptors (NMDARs). NMDARs allow extracellular calcium into the cell, thereby stimulating calcium-dependent cytosolic phospholipase A2 (cPLA2) to release AA from membrane phospholipid. We hypothesized that LTG, like other approved mood stabilizers, would reduce NMDAR-mediated AA signalling in rat brain. An acute subconvulsant dose of NMDA (25 mg/kg) or saline was administered intraperitoneally to unanaesthetized rats that had been treated p.o. daily for 42 d with vehicle or a therapeutically relevant dose of LTG (10 mg/kg.d). Regional brain AA incorporation coefficients k* and rates J in, and AA signals, were measured using quantitative autoradiography after intravenous [1-14C]AA infusion, as were other AA cascade markers. In chronic vehicle-treated rats, acute NMDA compared to saline increased k* and J in in widespread regions of the brain, as well as prostaglandin (PG)E2 and thromboxane B2 concentrations. Chronic LTG treatment compared to vehicle reduced brain cyclooxygenase (COX) activity, PGE2 concentration, and DNA-binding activity of the COX-2 transcription factor, NF-κB. Pretreatment with chronic LTG blocked the acute NMDA effects on AA cascade markers. In summary, chronic LTG like other mood stabilizers blocks NMDA-mediated signalling involving the AA metabolic cascade. Since markers of the AA cascade and of NMDAR signalling are up-regulated in the post-mortem BD brain, mood stabilizers generally may be effective in BD by dampening NMDAR signalling and the AA cascade.


Assuntos
Ácido Araquidônico/sangue , Encéfalo/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/efeitos dos fármacos , Triazinas/farmacologia , Análise de Variância , Animais , Autorradiografia , Peso Corporal/efeitos dos fármacos , Encéfalo/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Eicosanoides/metabolismo , Agonistas de Aminoácidos Excitatórios/farmacologia , Lamotrigina , Masculino , N-Metilaspartato/farmacologia , NF-kappa B/metabolismo , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Tromboxano B2/metabolismo
10.
Brain Res Bull ; 87(2-3): 154-71, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22178644

RESUMO

The polyunsaturated fatty acids (PUFAs), arachidonic acid (AA, 20:4n-6) and docosahexaenoic acid (DHA, 22:6n-3), important second messengers in brain, are released from membrane phospholipid following receptor-mediated activation of specific phospholipase A(2) (PLA(2)) enzymes. We developed an in vivo method in rodents using quantitative autoradiography to image PUFA incorporation into brain from plasma, and showed that their incorporation rates equal their rates of metabolic consumption by brain. Thus, quantitative imaging of unesterified plasma AA or DHA incorporation into brain can be used as a biomarker of brain PUFA metabolism and neurotransmission. We have employed our method to image and quantify effects of mood stabilizers on brain AA/DHA incorporation during neurotransmission by muscarinic M(1,3,5), serotonergic 5-HT(2A/2C), dopaminergic D(2)-like (D(2), D(3), D(4)) or glutamatergic N-methyl-d-aspartic acid (NMDA) receptors, and effects of inhibition of acetylcholinesterase, of selective serotonin and dopamine reuptake transporter inhibitors, of neuroinflammation (HIV-1 and lipopolysaccharide) and excitotoxicity, and in genetically modified rodents. The method has been extended for the use with positron emission tomography (PET), and can be employed to determine how human brain AA/DHA signaling and consumption are influenced by diet, aging, disease and genetics.


Assuntos
Ácido Araquidônico/metabolismo , Mapeamento Encefálico , Encéfalo/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Transdução de Sinais/fisiologia , Animais , Antidepressivos/uso terapêutico , Autorradiografia , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Humanos , Modelos Biológicos , Neuroimagem , Fosfolipases A2/metabolismo , Cintilografia
11.
Neuropharmacology ; 61(8): 1256-64, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21839100

RESUMO

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.


Assuntos
Antimaníacos/farmacologia , Ácido Araquidônico/metabolismo , Encéfalo/efeitos dos fármacos , Receptores de Dopamina D2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ácido Valproico/farmacologia , Análise de Variância , Animais , Antimaníacos/sangue , Ácido Araquidônico/farmacocinética , Autorradiografia , Encéfalo/metabolismo , Radioisótopos de Carbono/farmacocinética , Dinoprostona/metabolismo , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Masculino , Ratos , Ratos Endogâmicos F344 , Tromboxano B2/metabolismo , Ácido Valproico/sangue
12.
Prostaglandins Other Lipid Mediat ; 96(1-4): 109-13, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21704722

RESUMO

Docosahexaenoic acid (DHA) is critical for maintaining normal brain structure and function, and is considered neuroprotective. Its brain concentration depends on dietary DHA content and hepatic conversion from its dietary derived n-3 precursor, α-linolenic acid (α-LNA). We have developed an in vivo method in rats using quantitative autoradiography and intravenously injected radiolabeled DHA to image net incorporation into the brain of unesterified plasma DHA, and showed with this method that the incorporation rate of DHA equals the rate of brain metabolic DHA consumption. The method has been extended for use in humans with positron emission tomography (PET). Furthermore, imaging in unanesthetized rats using DHA incorporation as a biomarker in response to acute N-methyl-D-aspartate administration confirms that regional DHA signaling is independent of extracellular calcium, and likely mediated by a calcium-independent phospholipase A(2) (iPLA(2)). Studies in mice in which iPLA(2)-VIA (ß) was knocked out confirmed that this enzyme is critical for baseline and muscarinic cholinergic signaling involving DHA. Thus, quantitative imaging of DHA incorporation from plasma into brain can be used as an in vivo biomarker of brain DHA metabolism and neurotransmission.


Assuntos
Autorradiografia/métodos , Química Encefálica , Ácidos Docosa-Hexaenoicos/metabolismo , Fosfolipases A2/metabolismo , Cintilografia/métodos , Transdução de Sinais/fisiologia , Transmissão Sináptica/fisiologia , Animais , Biomarcadores/análise , Ácidos Docosa-Hexaenoicos/farmacologia , Humanos , Injeções Intravenosas , Camundongos , Camundongos Knockout , N-Metilaspartato/metabolismo , N-Metilaspartato/farmacologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Tomografia por Emissão de Pósitrons , Ratos , Ratos Endogâmicos , Transdução de Sinais/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Ácido alfa-Linolênico/metabolismo
13.
Neurochem Res ; 36(1): 139-45, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20981485

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Ácido Araquidônico/metabolismo , Aspirina/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Animais , Dinoprostona/metabolismo , Humanos , Leucotrieno B4/metabolismo , Lipopolissacarídeos/farmacologia , Lipoxinas/metabolismo , Masculino , Ratos , Ratos Endogâmicos F344 , Tromboxano B2/metabolismo
14.
J Cereb Blood Flow Metab ; 31(2): 486-93, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20664612

RESUMO

Human immunodeficiency virus (HIV)-associated infection involves the entry of virus-bearing monocytes into the brain, followed by microglial activation, neuroinflammation, and upregulated arachidonic acid (AA) metabolism. The HIV-1 transgenic (Tg) rat, a noninfectious HIV-1 model, shows neurologic and behavioral abnormalities after 5 months of age. We hypothesized that brain AA metabolism would be elevated in older HIV-1 Tg rats in vivo. Arachidonic acid incorporation from the plasma into the brain of unanesthetized 7-to-9-month-old rats was imaged using quantitative autoradiography, after [1-(14)C]AA infusion. Brain phospholipase (PLA(2)) activities and eicosanoid concentrations were measured, and enzymes were localized by immunostaining. AA incorporation coefficients k* and rates J(in), measures of AA metabolism, were significantly higher in 69 of 81 brain regions in HIV-1 Tg than in control rats, as were activities of cytosolic (c)PLA(2)-IV, secretory (s)PLA(2), and calcium independent (i)PLA(2)-VI, as well as prostaglandin E(2) and leukotriene B(4) concentrations. Immunostaining of somatosensory cortex showed elevated cPLA(2)-IV, sPLA(2)-IIA, and cyclooxygenase-2 in neurons. Brain AA incorporation and other markers of AA metabolism are upregulated in HIV-1 Tg rats, in which neurologic changes and neuroinflammation have been reported. Positron emission tomography with [1-(11)C]AA could be used to test whether brain AA metabolism is upregulated in HIV-1-infected patients, in relation to cognitive and behavioral disturbances.


Assuntos
Ácido Araquidônico/biossíntese , HIV-1 , Algoritmos , Animais , Autorradiografia , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Ácidos Graxos/metabolismo , Ácidos Graxos não Esterificados/sangue , Humanos , Imuno-Histoquímica , Cinética , Leucotrieno B4/metabolismo , Masculino , Fosfolipases A2/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Ratos Transgênicos , Tromboxano B2/metabolismo , Regulação para Cima
15.
J Lipid Res ; 51(11): 3166-73, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20686114

RESUMO

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.


Assuntos
Encéfalo/citologia , Encéfalo/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Fosfolipases A2 do Grupo VI/deficiência , Imagem Molecular , Transdução de Sinais , Animais , Arecolina/administração & dosagem , Arecolina/farmacologia , Peso Corporal , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Artérias Cerebrais/fisiologia , Ácidos Docosa-Hexaenoicos/sangue , Fosfolipases A2 do Grupo VI/metabolismo , Cinética , Masculino , Camundongos
16.
J Lipid Res ; 51(8): 2334-40, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20388940

RESUMO

In vitro studies show that docosahexaenoic acid (DHA) can be released from membrane phospholipid by Ca(2+)-independent phospholipase A(2) (iPLA(2)), Ca(2+)-independent plasmalogen PLA(2) or secretory PLA(2 (sPLA2)), but not by Ca(2+)-dependent cytosolic PLA(2) (cPLA2), which selectively releases arachidonic acid (AA). Since glutamatergic NMDA (N-methyl-D-aspartate) receptor activation allows extracellular Ca(2+) into cells, we hypothesized that brain DHA signaling would not be altered in rats given NMDA, to the extent that in vivo signaling was mediated by Ca(2+)-independent mechanisms. Isotonic saline, a subconvulsive dose of NMDA (25 mg/kg), MK-801, or MK-801 followed by NMDA was administered i.p. to unanesthetized rats. Radiolabeled DHA or AA was infused intravenously and their brain incorporation coefficients k*, measures of signaling, were imaged with quantitative autoradiography. NMDA or MK-801 compared with saline did not alter k* for DHA in any of 81 brain regions examined, whereas NMDA produced widespread and significant increments in k* for AA. In conclusion, in vivo brain DHA but not AA signaling via NMDA receptors is independent of extracellular Ca(2+) and of cPLA(2). DHA signaling may be mediated by iPLA(2), plasmalogen PLA(2), or other enzymes insensitive to low concentrations of Ca(2+). Greater AA than DHA release during glutamate-induced excitotoxicity could cause brain cell damage.


Assuntos
Cálcio/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Espaço Extracelular/metabolismo , Transmissão Sináptica , Animais , Ácido Araquidônico/metabolismo , Artérias/efeitos dos fármacos , Artérias/fisiologia , Autorradiografia , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Maleato de Dizocilpina/farmacologia , Espaço Extracelular/efeitos dos fármacos , Masculino , Imagem Molecular , N-Metilaspartato/farmacologia , Ratos , Ratos Endogâmicos F344
17.
J Neurochem ; 89(3): 627-35, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15086519

RESUMO

The peptide neurotransmitter N-acetylaspartylglutamate is inactivated by extracellular peptidase activity following synaptic release. It is speculated that the enzyme, glutamate carboxypeptidase II (GCPII, EC 3.14.17.21), participates in this inactivation. However, CGCPII knockout mice appear normal in standard neurological tests. We report here the cloning and characterization of a mouse enzyme (tentatively identified as glutamate carboxypeptidase III or GCPIII) that is homologous to an enzyme identified in a human lung carcinoma. The mouse peptidase was cloned from two non-overlapping EST clones and mouse brain cDNA using PCR. The sequence (GenBank, AY243507) is 85% identical to the human carcinoma enzyme and 70% homologous to mouse GCPII. GCPIII sequence analysis suggests that it too is a zinc metallopeptidase. Northern blots revealed message in mouse ovary, testes and lung, but not brain. Mouse cortical and cerebellar neurons in culture expressed GCPIII message in contrast to the glial specific expression of GCPII. Message levels of GCPIII were similar in brains obtained from wild-type mice and mice that are null mutants for GCPII. Chinese hamster ovary (CHO) cells transfected with rat GCPII or mouse GCPIII expressed membrane bound peptidase activity with similar V(max) and K(m) values (1.4 micro m and 54 pmol/min/mg; 3.5 micro m and 71 pmol/min/mg, respectively). Both enzymes are activated by a similar profile of metal ions and their activities are blocked by EDTA. GCPIII message was detected in brain and spinal cord by RT-PCR with highest levels in the cerebellum and hippocampus. These data are consistent with the hypothesis that nervous system cells express at least two differentially distributed homologous enzymes with similar pharmacological properties and affinity for NAAG.


Assuntos
Encéfalo/enzimologia , Glutamato Carboxipeptidase II/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Membrana Celular/enzimologia , Células Cultivadas , Clonagem Molecular , Cricetinae , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Glutamato Carboxipeptidase II/genética , Concentração de Íons de Hidrogênio , Metais/farmacologia , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , RNA Mensageiro/biossíntese , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência , Medula Espinal/enzimologia , Transfecção
18.
J Neurophysiol ; 89(1): 128-34, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12522165

RESUMO

Deletion of the beta3 subunit of the GABA(A) receptor produces severe behavioral deficits and epilepsy. GABA(A) receptor-mediated miniature inhibitory postsynaptic currents (mIPSCs) in cortical neurons in cultures from beta3 -/- mice were significantly faster than those in beta3 +/+ mice and were more prolonged by zolpidem. Surface staining revealed that the number of beta2/3, alpha2, and alpha3 (but not of alpha1) subunit-expressing neurons and the intensity of subunit clusters were significantly reduced in beta3 -/- mice. Transfection of beta3 -/- neurons with beta3 cDNA restored beta2/3, alpha2, and alpha3 subunits immunostaining and slowed mIPSCs decay. We show that the deletion of the beta3 subunit causes the loss of a subset of GABA(A) receptors with alpha2 and alpha3 subunits while leaving a receptor population containing predominantly alpha1 subunit with fast spontaneous IPSC decay and increased zolpidem sensitivity.


Assuntos
Inibição Neural/fisiologia , Neurônios/fisiologia , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Feminino , Agonistas GABAérgicos/farmacologia , Deleção de Genes , Expressão Gênica/fisiologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutagênese/fisiologia , Neurônios/citologia , Técnicas de Patch-Clamp , Gravidez , Piridinas/farmacologia , Transfecção , Zolpidem
19.
J Neurochem ; 83(1): 20-9, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12358725

RESUMO

Glutamate carboxypeptidase II (GCPII, EC 3.14.17.21) is a membrane-bound enzyme found on the extracellular face ofglia. The gene for this enzyme is designated FOLH1 in humans and Folh1 in mice. This enzyme has been proposed to be responsible for inactivation of the neurotransmitter N-acetylaspartylglutamate (NAAG) following synaptic release. Mice harboring a disruption of the gene for GCPII/Folh1 were generated by inserting into the genome a targeting cassette in which the intron-exon boundary sequences of exons 1 and 2 were removed and stop codons were inserted in exons 1 and 2. Messenger RNA for GCPII was not detected by northern blotting or RT-PCR analysis of RNA from the brains of -/- mutant mice nor was GCPII protein detected on western blots of this tissue. These GCPII null mutant mice developed normally to adulthood and exhibited a normal range of neurologic responses and behaviors including mating, open field activity and retention of position in rotorod tests. No significant differences were observed among responses of wild type, heterozygous mutant and homozygous mutant mice on tail flick and hot plate latency tests. Glutamate, NAAG and mRNA for metabotropic glutamate receptor type 3 levels were not significantly altered in response to the deletion of glutamate carboxypeptidase II. A novel membrane-bound NAAG peptidase activity was discovered in brain, spinal cord and kidney of the GCPII knock out mice. The kinetic values for brain NAAG peptidase activity in the wild type and GCPII nullmutant were Vmax = 45 and 3 pmol/mg/min and Km = 2650 nm and 2494 nm, respectively. With the exception of magnesium and copper, this novel peptidase activity had a similar requirement for metal ions as GCPII. Two potent inhibitors of GCPII, 4,4'-phosphinicobis-(butane-1,3 dicarboxilic acid) (FN6) and 2-(phosphonomethyl)pentanedioic acid (2-PMPA) inhibited the residual activity. The IC50 value for 2-PMPA was about 1 nm for wild-type brain membrane NAAG peptidase activity consistent with its activity against cloned ratand human GCPII, and 88 nm for the activity in brain membranes of the null mutants.


Assuntos
Carboxipeptidases/deficiência , Dipeptídeos/metabolismo , Animais , Comportamento Animal/fisiologia , Western Blotting , Encéfalo/enzimologia , Química Encefálica , Células CHO , Carboxipeptidases/genética , Membrana Celular/enzimologia , Cricetinae , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Marcação de Genes , Genótipo , Glutamato Carboxipeptidase II , Hidrólise , Rim/química , Rim/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/biossíntese , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Medula Espinal/química , Medula Espinal/enzimologia , Sinapses
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA