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











Publication year range
2.
Dev Neurobiol ; 76(3): 287-97, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26037116

ABSTRACT

Fatty acid binding protein 7 (FABP7), abundant in the embryonic brain, binds with the highest affinity to docosahexaenoic acid (DHA) and is expressed in the early stages of embryogenesis. Here, we have examined the consequences of the exposure to different DHA levels and of the in utero depletion of FABP7 on early rat brain development. Neurodevelopment was evaluated through the contents of two proteins, connexin 43 (Cx43) and cyclin-dependent kinase 5 (CDK5), both involved in neuroblast proliferation, differentiation, and migration. The dams were fed with diets presenting different DHA contents, from deficiency to supplementation. DHA brain embryos contents already differed at embryonic day 11.5 and the differences kept increasing with time. Cx43 and CDK5 contents were positively associated with the brain DHA levels. When FABP7 was depleted in vivo by injections of siRNA in the telencephalon, the enhancement of the contents of both proteins was lost in supplemented animals, but FABP7 depletion did not modify phospholipid compositions regardless of the diets. Thus, FABP7 is a necessary mediator of the effect of DHA on these proteins synthesis, but its role in DHA uptake is not critical, although FABP7 is localized in phospholipid-rich areas. Our study shows that high contents of DHA associated with FABP7 are necessary to promote early brain development, which prompted us to recommend DHA supplementation early in pregnancy.


Subject(s)
Brain/growth & development , Brain/metabolism , Docosahexaenoic Acids/pharmacology , Fatty Acid-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Animals , Blotting, Western , Brain/drug effects , Brain Chemistry , Diet , Embryo, Mammalian , Fatty Acid-Binding Protein 7 , Female , Gene Knockdown Techniques , Immunohistochemistry , Maternal Exposure , Rats , Rats, Wistar , Spectroscopy, Fourier Transform Infrared
3.
Psychoneuroendocrinology ; 53: 82-93, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25614359

ABSTRACT

Epidemiological observations report an increase in fat consumption associated with low intake of n-3 relative to n-6 polyunsaturated fatty acids (PUFAs) in women of childbearing age. However, the impact of these maternal feeding habits on cognitive function in the offspring is unknown. This study aims to investigate the impact of early exposure to a high-fat diet (HFD) with an unbalanced n-6/n-3 PUFAs ratio on hippocampal function in adult rats. Furthermore, we explored the effects of perinatal HFD combined with exposure to HFD after weaning. Dams were fed a control diet (C, 12% of energy from lipids, n-6/n-3 PUFAs ratio: 5) or HFD (HF, 39% of energy from lipids, n-6/n-3 PUFAs ratio: 39) throughout gestation and lactation. At weaning, offspring were placed either on control (C-C, HF-C) or high-fat (HF-HF) diets. In adulthood, hippocampus-dependent memory was assessed using the water-maze task and potential hippocampal alterations were determined by studying PUFA levels, gene expression, neurogenesis and astrocyte morphology. Perinatal HFD induced long-lasting metabolic alterations and some changes in gene expression in the hippocampus, but had no effect on memory. In contrast, spatial memory was impaired in animals exposed to HFD during the perinatal period and maintained on this diet. HF-HF rats also exhibited low n-3 and high n-6 PUFA levels, decreased neurogenesis and downregulated expression of several plasticity-related genes in the hippocampus. To determine the contribution of the perinatal diet to the memory deficits reported in HF-HF animals, an additional experiment was conducted in which rats were only exposed to HFD starting at weaning (C-HF). Interestingly, memory performance in this group was similar to controls. Overall, our results suggest that perinatal exposure to HFD with an unbalanced n-6/n-3 ratio sensitizes the offspring to the adverse effects of subsequent high-fat intake on hippocampal function.


Subject(s)
Diet, High-Fat , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-6/pharmacology , Hippocampus/drug effects , Maze Learning/drug effects , Prenatal Exposure Delayed Effects , Spatial Memory/drug effects , Animals , Animals, Newborn , Female , Lactation , Pregnancy , Rats , Rats, Wistar , Weaning
4.
Article in English | MEDLINE | ID: mdl-25123062

ABSTRACT

Specific mechanisms for maintaining docosahexaenoic acid (DHA) concentration in brain cells but also transporting DHA from the blood across the blood-brain barrier (BBB) are not agreed upon. Our main objective was therefore to evaluate the level of gene expression of fatty acid transport and fatty acid binding proteins in the cerebral cortex and at the BBB level during the perinatal period of active brain DHA accretion, at weaning, and until the adult age. We measured by real time RT-PCR the mRNA expression of different isoforms of fatty acid transport proteins (FATPs), long-chain acyl-CoA synthetases (ACSLs), fatty acid binding proteins (FABPs) and the fatty acid transporter (FAT)/CD36 in cerebral cortex and isolated microvessels at embryonic day 18 (E18) and postnatal days 14, 21 and 60 (P14, P21 and P60, respectively) in rats receiving different n-3 PUFA dietary supplies (control, totally deficient or DHA-supplemented). In control rats, all the genes were expressed at the BBB level (P14 to P60), the mRNA levels of FABP5 and ACSL3 having the highest values. Age-dependent differences included a systematic decrease in the mRNA expressions between P14-P21 and P60 (2 to 3-fold), with FABP7 mRNA abundance being the most affected (10-fold). In the cerebral cortex, mRNA levels varied differently since FATP4, ACSL3 and ACSL6 and the three FABPs genes were highly expressed. There were no significant differences in the expression of the 10 genes studied in n-3 deficient or DHA-supplemented rats despite significant differences in their brain DHA content, suggesting that brain DHA uptake from the blood does not necessarily require specific transporters within cerebral endothelial cells and could, under these experimental conditions, be a simple passive diffusion process.


Subject(s)
Blood-Brain Barrier/metabolism , Cerebral Cortex/metabolism , Docosahexaenoic Acids/genetics , Fatty Acid Transport Proteins/biosynthesis , Fatty Acid-Binding Proteins/metabolism , Animals , Blood-Brain Barrier/growth & development , Cerebral Cortex/growth & development , Docosahexaenoic Acids/metabolism , Embryonic Development/drug effects , Embryonic Development/genetics , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/metabolism , Gene Expression Regulation , RNA, Messenger/biosynthesis , Rats
5.
Diabetes ; 63(5): 1624-36, 2014 May.
Article in English | MEDLINE | ID: mdl-24430437

ABSTRACT

Aberrations in gut microbiota are associated with metabolic disorders, including obesity. However, whether shifts in the microbiota profile during obesity are a characteristic of the phenotype or a consequence of obesogenic feeding remains elusive. Therefore, we aimed to determine differences in the gut microbiota of obese-prone (OP) and obese-resistant (OR) rats and examined the contribution of this microbiota to the behavioral and metabolic characteristics during obesity. We found that OP rats display a gut microbiota distinct from OR rats fed the same high-fat diet, with a higher Firmicutes-to-Bacteroidetes ratio and significant genera differences. Transfer of OP but not OR microbiota to germ-free (GF) mice replicated the characteristics of the OP phenotype, including reduced intestinal and hypothalamic satiation signaling, hyperphagia, increased weight gain and adiposity, and enhanced lipogenesis and adipogenesis. Furthermore, increased gut permeability through conventionalization resulted in inflammation by proinflammatory nuclear factor (NF)-κB/inhibitor of NF-κB kinase subunit signaling in adipose tissue, liver, and hypothalamus. OP donor and GF recipient animals harbored specific species from Oscillibacter and Clostridium clusters XIVa and IV that were completely absent from OR animals. In conclusion, susceptibility to obesity is characterized by an unfavorable microbiome predisposing the host to peripheral and central inflammation and promoting weight gain and adiposity during obesogenic feeding.


Subject(s)
Gastrointestinal Tract/microbiology , Hypothalamus/metabolism , Microbiota/physiology , Obesity/microbiology , Signal Transduction/physiology , Animals , Eating/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Rats
6.
J Steroid Biochem Mol Biol ; 138: 395-402, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23907015

ABSTRACT

Stress bears a negative impact on adult neurogenesis. High levels of corticoids have been shown to inhibit neural stem cell proliferation, and are considered responsible for the loss of neural precursors. Their effects on the differentiation of the glial and neuronal lineages have been less studied. We examined the effect of dexamethasone (Dex), a synthetic glucocorticoid, on the differentiation of rat neural stem cells in vitro. Dex had no effect on the differentiation of cells cultured under standard conditions. Since we previously determined that NSC, when cultured under classical conditions, were deprived of polyunsaturated fatty acids (PUFA), and displayed phospholipid compositions very different from the in vivo figures [1], we examined the effect of Dex under PUFA supplementation. Dex impaired neuron and oligodendrocyte maturation in PUFA-supplemented cells, demonstrated by the reduction of neurite lengths and oligodendrocyte sizes. This effect was mediated by the glucocorticoid receptor (GR), since it was eliminated by mifepristone, a GR antagonist, and could be relayed by a reduction of ERK phosphorylation. We determined that GR was associated with PPAR ß and α under basal conditions, and that this association was disrupted when PUFA were added in combination with Dex. We assumed that this effect on the receptor status enabled the effect of Dex on PUFA supplemented cells, since we determined that the binding to the glucocorticoid response element was higher in cells incubated with PUFA and Dex. In conclusion, corticoids can impair NSC differentiation, and consequently impact the entire process of neurogenesis.


Subject(s)
Dexamethasone/pharmacology , Fatty Acids, Unsaturated/pharmacology , Neuroglia/cytology , Neuroglia/drug effects , Neurons/cytology , Neurons/drug effects , Animals , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Immunoprecipitation , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Rats , Rats, Wistar
7.
J Nutr Biochem ; 24(1): 380-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23036607

ABSTRACT

We isolated neural stem cells/neural progenitors (NSC) from 1-day-old rat pups born to mothers fed diets that were deficient or supplemented with n-3 polyunsaturated fatty acids (PUFAs) and compared their proliferation and differentiation in vitro. The cells isolated from the n-3PUFA-deficient pups consistently proliferated more slowly than cells that were isolated from n-3PUFA-supplemented pups, despite the fact that both were cultured under the same conditions. The differences in the proliferation rates were evaluated up until 40 days of culture and were highly significant. When the cells were allowed to differentiate, the deficient cells exhibited a higher degree of neuronal maturation in response to the addition of PUFAs in the medium, as demonstrated by an increase in neurite length, whereas the neurons derived from the supplemented pups showed no change. This result was consistent, regardless of the age of the culture. The properties of the NSC were durably modified throughout the length of the culture, although the membrane phospholipid compositions were similar. We examined the differential expression of selected mRNAs and micro RNAs. We found significant differences in the gene expression of proliferating and differentiating cells, and a group of genes involved in neurogenesis was specifically modified by n-3 PUFA treatment. We conclude that n-3 PUFA levels in the maternal diet can induce persistent modifications of the proliferation and differentiation of NSCs and of their transcriptome. Therefore, the n-3 supply received in utero may condition on a long-term basis cell renewal in the brain.


Subject(s)
Cell Differentiation/drug effects , Fatty Acids, Unsaturated/pharmacology , Neural Stem Cells/drug effects , Prenatal Exposure Delayed Effects , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cells, Cultured , Docosahexaenoic Acids/pharmacology , Female , Gene Expression Regulation , Maternal Nutritional Physiological Phenomena , MicroRNAs , Neural Stem Cells/cytology , Pregnancy , RNA, Messenger , Rats , Rats, Wistar
8.
Cell Mol Life Sci ; 70(11): 2003-14, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23269438

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily and function as transcription factors that regulate gene expression in numerous biological processes. Although the PPARß/δ subtype is highly expressed in the brain, its physiological roles in neuronal function remain to be elucidated. In this study, we examined the presence of PPARß/δ in the master circadian clock of the Syrian hamster and investigated its putative functional role in this structure. In mammals, the central circadian clock, located in the suprachiasmatic nucleus (SCN), is entrained by the light-dark (LD) cycle via photic6 signals conveyed by a direct pathway whose terminals release glutamate. Using immunocytochemical and qRT-PCR analysis, we demonstrated that the rhythmic expression of PPAR ß/δ within the SCN of hamsters raised under an LD cycle was detectable only at the transcriptional level when the hamsters were maintained under constant darkness (DD). The increase in the number of immunoreactive PPARß/δ cells observed under DD after light stimulation during the early subjective night (CT14), but not during the subjective day (CT06), demonstrated that the expression of PPARß/δ can be up-regulated according to the photosensitive phase of the circadian clock. All of the PPARß/δ-positive cells in the SCN also expressed the glutamate receptor NMDAR1. Moreover, we demonstrated that at the photosensitive point (CT14), the administration of L-16504, a specific agonist of PPARß/δ, amplified the phase delay of the locomotor response induced by a light pulse. Taken together, these data suggest that PPARß/δ activation modulates glutamate release that mediates entrainment of the circadian clock by light.


Subject(s)
Glutamic Acid/metabolism , Light Signal Transduction , PPAR delta/physiology , PPAR-beta/physiology , Suprachiasmatic Nucleus/metabolism , Animals , Circadian Rhythm , Cricetinae , Darkness , Gene Expression Regulation , Immunohistochemistry , Light , Mesocricetus , PPAR delta/agonists , PPAR delta/metabolism , PPAR-beta/agonists , PPAR-beta/metabolism , Phenoxyacetates/pharmacology , Photoperiod , Real-Time Polymerase Chain Reaction , Suprachiasmatic Nucleus/radiation effects
9.
J Nutr Biochem ; 22(9): 820-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21129945

ABSTRACT

Hormonal and nutritional factors regulate the metabolism of long-chain polyunsaturated fatty acids (LC-PUFA). We aimed to determine whether ovarian hormones influence the capacity of rats to synthesize the end-products 22:6n-3 (DHA) and 22:5n-6 (n-6DPA) from their respective dietary precursors (18:3n-3 and 18:2n-6), and can regulate PUFA conversion enzymes gene transcription in brain and/or liver. Females born with a low DHA status were fed from weaning to 8 weeks of age a diet providing both essential precursors, and were concurrently submitted to sham-operated control (SOC) or ovariectomy (OVX) in combination with or without 17ß-estradiol (E2) dosed at 8 or 16 µg/day. Relative to SOC, OVX increased the hepatic Δ9-, Δ6- and Δ5-desaturase transcripts and cognate transcription factors (PPARα, PPARγ, RXRα, RARα), but it did not affect LC-PUFA contents in phospholipids. In comparison with SOC and OVX groups, both E2 doses prevented the increase of transcripts, while paradoxically augmenting DHA and n-6DPA in liver phospholipids. Thus, in the liver of rats undergoing ovariectomy, changes of LC-PUFA synthesizing enzyme transcripts and of LC-PUFA proportions were not correlated. In brain, ovariectomy did not modify the transcripts of lipid metabolism genes, but it decreased DHA (-15%) and n-6DPA (-28%). In comparison with SOC and OVX groups, ovariectomized females treated with E2 preserved their status of both LC-PUFA in brain and had increased transcripts of E2 receptor ß, PPARδ, RARα and LC-PUFA synthesizing enzymes. In conclusion, E2 sustained the transcription of lipid metabolism genes and proportions of neo-formed DHA and n-6DPA differently in brain and liver.


Subject(s)
Brain/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Animals , Brain/enzymology , Female , Organ Specificity , Ovariectomy , Rats , Rats, Wistar , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
10.
J Cell Biochem ; 110(6): 1356-64, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20564231

ABSTRACT

Rat neural stem cells/neural progenitors (NSC/NP) are generally grown in serum-free medium. In this study, NSC/NP were supplemented with the main long-chain polyunsaturated fatty acids (PUFAs) present in the brain, arachidonic acid (AA), or docosahexaenoic acid (DHA), and were monitored for their growth. Lipid and fatty acid contents of the cells were also determined. Under standard conditions, the cells were characterized by phospholipids displaying a highly saturated profile, and very low levels of PUFAs. When cultured in the presence of PUFAs, the cells easily incorporated them into the phospholipid fraction. We also compared the presence of three membrane proteins in the lipid raft fractions: GFR and connexin 43 contents in the rafts were increased by DHA supplementation, whereas Gbeta subunit content was not significantly modified. The restoration of DHA levels in the phospholipids could profoundly affect protein localization and, consequently, their functionalities.


Subject(s)
Fatty Acids, Unsaturated/pharmacology , Membrane Microdomains/drug effects , Membrane Proteins/metabolism , Phospholipids/metabolism , Stem Cells/drug effects , Animals , Animals, Newborn , Blotting, Western , Brain/cytology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Connexin 43/metabolism , Docosahexaenoic Acids/pharmacology , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Fatty Acids/analysis , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Membrane Microdomains/metabolism , Phospholipids/chemistry , Rats , Rats, Wistar , Stem Cells/cytology , Stem Cells/metabolism
11.
Neurochem Int ; 56(5): 703-10, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20153394

ABSTRACT

Several in vivo studies suggest that docosahexaenoic acid (22:6 n-3), the main n-3 long-chain polyunsaturated fatty acids (LC-PUFA) of brain membranes, could be an important regulator of brain energy metabolism by affecting glucose utilization and the density of the two isoforms of the glucose transporter-1 (GLUT1) (endothelial and astrocytic). This study was conducted to test the hypothesis that 22:6 n-3 in membranes may modulate glucose metabolism in brain endothelial cells. It compared the impact of 22:6 n-3 and the other two main LC-PUFA, arachidonic acid (20:4 n-6) and eicosapentaenoic acid (20:5 n-3), on fatty acid composition of membrane phospholipids, glucose uptake and expression of 55-kDa GLUT1 isoform in two models of rat brain endothelial cells (RBEC), in primary culture and in the immortalized rat brain endothelial cell line RBE4. Without PUFA supplementation, both types of cerebral endothelial cells were depleted in 22:6 n-3, RBE4 being also particularly low in 20:4 n-6. After exposure to supplemental 20:4 n-6, 20:5 n-3 or 22:6 n-3 (15microM, i.e. a physiological dose), RBEC and RBE4 avidly incorporated these PUFA into their membrane phospholipids thereby resembling physiological conditions, i.e. the PUFA content of rat cerebral microvessels. However, RBE4 were unable to incorporate physiological level of 20:4 n-6. Basal glucose transport in RBEC (rate of [(3)H]-3-o-methylglucose uptake) was increased after 20:5 n-3 or 22:6 n-3 supplementation by 50% and 35%, respectively, whereas it was unchanged with 20:4 n-6. This increase of glucose transport was associated with an increased GLUT1 protein, while GLUT1 mRNA was not affected. The different PUFA did not impact on glucose uptake in RBE4. Due to alterations in n-6 PUFA metabolism and weak expression of GLUT1, RBE4 seems to be less adequate than RBEC to study PUFA metabolism and glucose transport in brain endothelial cells. Physiological doses of n-3 LC-PUFA have a direct and positive effect on glucose transport and GLUT1 density in RBEC that could partly explain decreased brain glucose utilization in n-3 PUFA-deprived rats.


Subject(s)
Brain Chemistry/drug effects , Endothelial Cells/metabolism , Fatty Acids, Omega-3/pharmacology , Glucose/metabolism , 3-O-Methylglucose/metabolism , Animals , Blotting, Western , Capillaries/cytology , Capillaries/drug effects , Capillaries/metabolism , Cells, Cultured , DNA Primers , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Endothelial Cells/drug effects , Fatty Acids/analysis , Fatty Acids/metabolism , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 1/genetics , Glucose Transporter Type 3/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Male , Rats , Rats, Wistar
12.
Neurochem Int ; 56(1): 94-100, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19770016

ABSTRACT

Polyunsaturated fatty acids (PUFAs), particularly docosahexaenoic acid (DHA) and arachidonic acid (AA), are the main components of the phospholipids, in cerebral membranes. A dietary-induced cerebral DHA deficit results in altered behaviour and neurotransmission in rodents. To determine whether PUFA were acting on the neurotransmitter release machinery, we measured the release of [(3)H]-noradrenaline (NA) from SH-SY5Y neuroblastoma cells with modified PUFA membrane contents and from cells incubated with medium containing high DHA or AA. The membranes of cells incubated with 70 microM DHA for 3 days had 7.6-times more DHA in their ethanolamine glycerophospholipids, while the membranes of cells incubated with AA had 40% less. Incorporation of DHA enhanced basal [(3)H]-NA release (25%, p<0.05), but not KCl-evoked [(3)H]-NA release. Brief incubation with DHA during vesicle mobilization also strongly increased [3H]-NA release. AA had no effect. The genes encoding for the calcium sensor synaptotagmin 1, and for the two SNARE complex proteins syntaxin 1A and synaptobrevin 1 were not affected by PUFA incorporation, as indicated by assays for specific mRNAs and proteins. Thus both a high membrane DHA content and free DHA in the medium enhance the release of [(3)H]-NA from SH-SY5Y cells. This suggests that brain membrane DHA influences exocytosis, which then regulates neurotransmission.


Subject(s)
Cell Membrane/metabolism , Docosahexaenoic Acids/metabolism , Exocytosis/physiology , Membrane Lipids/metabolism , Neurons/metabolism , Norepinephrine/metabolism , Brain/metabolism , Brain Chemistry/drug effects , Brain Chemistry/physiology , Cell Line, Tumor , Cell Membrane/drug effects , Docosahexaenoic Acids/pharmacology , Exocytosis/drug effects , Glycerophospholipids/metabolism , Humans , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , Tritium , Up-Regulation/drug effects , Up-Regulation/physiology
13.
J Nutr Biochem ; 21(3): 180-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19157821

ABSTRACT

Dietary n-3 polyunsaturated fatty acids (PUFA) are major components of cell membranes and have beneficial effects on human health. Docosahexaenoic acid (DHA; 22:6n-3) is the most biologically important n-3 PUFA and can be synthesized from its dietary essential precursor, alpha-linolenic acid (ALA; 18:3n-3). Gender differences in the efficiency of DHA bioconversion have been reported, but underlying molecular mechanisms are unknown. We compared the capacity for DHA synthesis from ALA and the expression of related enzymes in the liver and cerebral cortex between male and female rats. Wistar rats, born with a low-DHA status, were supplied with a suboptimal amount of ALA from weaning to 8 weeks of age. Fatty acid composition was determined by gas chromatography, the mRNA expression of different genes involved in PUFA metabolism was determined by RT-PCR (low-density array) and the expression of proteins was determined by Western blot analysis. At 8 weeks, DHA content was higher (+20 to +40%) in each phospholipid class of female livers compared to male livers. The "Delta4," Delta5 and Delta6 desaturation indexes were 1.2-3 times higher in females than in males. The mRNA expression of Delta5- and Delta6-desaturase genes was 3.8 and 2.5 times greater, respectively, and the Delta5-desaturase protein was higher in female livers (+50%). No gender difference was observed in the cerebral cortex. We conclude that female rats replete their DHA status more readily than males, probably due to a higher expression of liver desaturases. Our results support the hypothesis on hormonal regulation of PUFA metabolism, which should be taken into account for specific nutritional recommendations.


Subject(s)
Fatty Acid Desaturases/metabolism , Fatty Acids, Omega-3/metabolism , Liver/enzymology , Stearoyl-CoA Desaturase/metabolism , Animals , Animals, Suckling , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Delta-5 Fatty Acid Desaturase , Dietary Fats, Unsaturated/administration & dosage , Docosahexaenoic Acids/blood , Docosahexaenoic Acids/metabolism , Fatty Acid Desaturases/genetics , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/blood , Female , Gene Expression Regulation , Liver/metabolism , Male , Maternal Nutritional Physiological Phenomena , Oligonucleotide Array Sequence Analysis , Organ Specificity , Phospholipids/metabolism , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sex Characteristics , Stearoyl-CoA Desaturase/genetics , Time Factors , alpha-Linolenic Acid/administration & dosage , alpha-Linolenic Acid/blood , alpha-Linolenic Acid/deficiency , alpha-Linolenic Acid/metabolism
14.
Lipids ; 43(1): 19-28, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17912567

ABSTRACT

Whether neurosteroids regulate the synthesis of long chain polyunsaturated fatty acids in brain cells is unknown. We examined the influence of 17-beta-estradiol (E2) on the capacity of SH-SY5Y cells supplemented with alpha-linolenic acid (ALA), to produce eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA). Cells were incubated for 24 or 72 h with ALA added alone or in combination with E2 (ALA + E2). Fatty acids were analyzed by gas chromatography of ethanolamine glycerophospholipids (EtnGpl) and phosphatidylcholine (PtdCho). Incubation for 24 h with ALA alone increased EPA and DPA in EtnGpl, by 330 and 430% compared to controls (P < 0.001) and DHA by only 10% (P < 0.05). Although DHA increased by 30% (P < 0.001) in ALA + E2-treated cells, the difference between the ALA and ALA + E2 treatments were not significant after 24 h (Anova-1, Fisher's test). After 72 h, EPA, DPA and DHA further increased in EtnGpl and PtdCho of cells supplemented with ALA or ALA + E2. Incubation for 72 h with ALA + E2 specifically increased EPA (+34% in EtnGpl, P < 0.001) and DPA (+15%, P < 0.001) compared to ALA alone. Thus, SH-SY5Y cells produced membrane EPA, DPA and DHA from supplemental ALA. The formation of DHA was limited, even in the presence of E2. E2 significantly favored EPA and DPA production in cells grown for 72 h. Enhanced synthesis of ALA-elongation products in neuroblastoma cells treated with E2 supports the hypothesis that neurosteroids could modulate the metabolism of PUFA.


Subject(s)
Eicosapentaenoic Acid/biosynthesis , Estradiol/pharmacology , Fatty Acids, Unsaturated/biosynthesis , Neuroblastoma/metabolism , alpha-Linolenic Acid/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/metabolism , Dose-Response Relationship, Drug , Eicosapentaenoic Acid/chemistry , Eicosapentaenoic Acid/metabolism , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/metabolism , Humans , Neuroblastoma/drug therapy , Phospholipids/metabolism , Tumor Cells, Cultured
15.
Lipids ; 40(7): 719-28, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16196423

ABSTRACT

Synthesis of docosahexaenoic acid (DHA) from its metabolic precursors contributes to membrane incorporation of this FA within the central nervous system. Although cultured neural cells are able to produce DHA, the membrane DHA contents resulting from metabolic conversion do not match the high values of those resulting from supplementation with preformed DHA. We have examined whether the DHA precursors down-regulate the incorporation of newly formed DHA within human neuroblastoma cells. SH-SY5Y cells were incubated with gradual doses of alpha-linolenic acid (alpha-LNA), EPA, or docosapentaenoic acid (DPA), and the incorporation of DHA into ethanolamine glycerophospholipids was analyzed as a reflection of synthesizing activity. The incorporation of EPA, DPA, and preformed DHA followed a dose-response saturating curve, whereas that of DHA synthesized either from alpha-LNA, EPA, or DPA peaked at concentrations of precursors below 15-30 microM and sharply decreased with higher doses. The mRNA encoding for six FA metabolism genes were quantified using real-time PCR. Two enzymes of the peroxisomal beta-oxidation, L-bifunctional protein and peroxisomal acyl-CoA oxidase, were expressed at lower levels than fatty acyl-CoA ligase 3 (FACL3) and delta6-desaturase (delta6-D). The delta6-D mRNA slightly increased between 16 and 48 h of culture, and this effect was abolished in the presence of 70 microM EPA. In contrast, the EPA treatment resulted in a time-dependent increase of FACL3 mRNA. The terminal step of DHA synthesis seems to form a "metabolic bottleneck," resulting in accretion of EPA and DPA when the precursor concentration exceeds a specific threshold value. We conclude that the critical precursor- concentration window of responsiveness may originate from the low basal expression level of peroxisomal enzymes.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Transcription, Genetic/drug effects , Base Sequence , Cell Division/drug effects , Cell Line, Tumor , DNA Primers , Dose-Response Relationship, Drug , Fatty Acids, Omega-3/metabolism , Humans , Polymerase Chain Reaction , RNA, Messenger/genetics
16.
J Nutr ; 135(9): 2241-6, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16140905

ABSTRACT

The altered neuron activity of rats deficient in (n-3) PUFAs may be due in part to a decrease in brain glucose utilization and glucose transport. We measured the glucose transporter protein GLUT1 isoforms at the blood-brain barrier (55-kDa) and in astrocytes (45-kDa) by Western immunoblotting and their mRNA by real time RT-PCR analysis in the cerebral cortex of adult male rats fed diets lacking (n-3) fatty acids (1st generation). The neuron glucose transporter GLUT3 was also assayed. The fatty acids in the phosphatidylcholine (PC), ethanolamine phosphoglycerolipid (EPG), and phosphatidylserine (PS) fractions of isolated microvessels and homogenates of the cerebral cortex were determined. The levels of (n-6) PUFAs [mainly arachidonic acid, 20:4(n-6)] in the phospholipid fractions of microvessels were higher and the levels of (n-3) PUFAs [mainly docosahexaenoic acid, 22:6(n-3)] were lower than in cerebral cortex homogenates. The microvessels and cortex of rats fed the (n-3) PUFA-deficient diet had 50% of the control 22:6(n-3) contents; 22:6(n-3) was replaced by 22:5(n-6). The 55-kDa GLUT1 immunoreactivity in (n-3) PUFA-deficient microvessels was decreased (down 25%, P < 0.01), as was the 45 kDa-GLUT1 in the homogenate (down 30%, P < 0.01). But the amount of immunoreactivity of GLUT3 did not change. The amount of GLUT1 mRNA was not affected by the (n-3) PUFA-deficient diet. These results suggest that the decreased glucose utilization in the cerebral cortex of (n-3) PUFA-deficient rats is due to reduced amounts of the 2 isoforms of GLUT1, indicating post-transcriptional regulation of GLUT1 synthesis.


Subject(s)
Brain/metabolism , Fatty Acids, Omega-3/metabolism , Animals , Blood Vessels/metabolism , Cerebral Cortex/blood supply , In Vitro Techniques , Lipid Metabolism , Male , Microcirculation , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats
17.
J Neurosci Res ; 75(1): 96-106, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14689452

ABSTRACT

Docosahexaenoic acid (DHA), the main n-3 polyunsaturated fatty acid (PUFA) in membranes, is particularly abundant in brain cells. Decreased cerebral concentrations of DHA, resulting from dietary n-3 deficiency, are associated with impaired cognitive function. Because the cellular causes of this impairment are still unknown, we need in vitro models that mimic the variations in n-3/n-6 PUFA seen in vivo. We have compared the PUFA profiles of hamster astrocytes cultured in medium supplemented with long-chain PUFA [DHA and/or arachidonic acid (AA)] with those of brain tissue from hamsters fed an n-6/n-3 PUFA-balanced diet or one lacking n-3 PUFA. Astrocytes were obtained from the brain cortex of newborn hamsters and cultured in minimum essential medium + 5% fetal calf serum (FCS) supplemented with DHA and/or AA for 10 days. The astrocytes cultured in medium + FCS had low n-3 PUFA contents, comparable to those of brain tissue from hamsters fed an n-3-deficient diet. We have shown that astrocytes grown in medium supplemented with DHA and/or AA, plus alpha-tocopherol to prevent lipid peroxidation, incorporated large amounts of these long-chain PUFA, so that the n-6/n-3 PUFA compositions of the phosphatidylethanolamine and phosphatidylcholine, the two main classes of membrane phospholipids, were greatly altered. Astrocytes cultured in medium plus DHA had a more physiological n-3 status, grew better, and retained their astrocyte phenotype. Thus astrocytes in culture are likely to be physiologically relevant only when provided with adequate DHA. This reliable method of altering membrane phospholipid composition promises to be useful for studying the influence of n-6/n-3 imbalance on astrocyte function.


Subject(s)
Astrocytes/metabolism , Cell Membrane/metabolism , Docosahexaenoic Acids/metabolism , Fatty Acids, Omega-6/metabolism , Fatty Acids, Unsaturated/metabolism , Phospholipids/metabolism , Triglycerides/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/drug effects , Blotting, Western , Brain Chemistry , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cricetinae , DNA/metabolism , Dietary Fats, Unsaturated/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Fatty Acids, Omega-3 , Glial Fibrillary Acidic Protein/metabolism , Male , Time Factors , alpha-Tocopherol/pharmacology
18.
Reprod Nutr Dev ; 44(6): 509-38, 2004.
Article in English | MEDLINE | ID: mdl-15762297

ABSTRACT

Docosahexaenoic acid (DHA, 22:6n-3) and arachidonic acid (AA, 20:4n-6) are the major polyunsaturated fatty acids in the membranes of brain and retinal cells. Animals specifically deficient in dietary n-3 fatty acids have low DHA content in their membranes, reduced visual acuity and impaired learning ability. Studies on bottle-fed human infants have shown that adding DHA and AA to milk replacer-formulas can bring their concentrations in the infant blood lipids to values as high as those produced by breast-feeding and significantly improves mental development and maturation of visual function. In older subjects, diverse neuropsychiatric and neurodegenerative diseases have been associated to decreased blood levels of n-3 PUFA. Low intakes of fish or of n-3 PUFA in populations have been associated with increased risks of depression and Alzheimer disease, and n-3 PUFA, especially eicosapentaenoic acid (EPA, 20:5n-3), have shown efficacy as adjunctive treatment - and in some cases as the only treatment--in several psychiatric disorders. The mechanisms by which polyunsaturated fatty acids have an impact on neuronal functions will be reviewed: the modulation of membrane biophysical properties, regulation of neurotransmitter release, synthesis of biologically active oxygenated derivatives, and nuclear receptor-mediated transcription of genes responsive to fatty acids or to their derivatives.


Subject(s)
Central Nervous System/physiology , Dietary Fats, Unsaturated/metabolism , Fatty Acids, Unsaturated/physiology , Visual Acuity/drug effects , Adolescent , Adult , Aged , Animals , Arachidonic Acid/administration & dosage , Arachidonic Acid/metabolism , Brain/metabolism , Brain/physiology , Central Nervous System/metabolism , Child , Child, Preschool , Dietary Fats, Unsaturated/administration & dosage , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/administration & dosage , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/physiology , Fatty Acids, Unsaturated/administration & dosage , Fatty Acids, Unsaturated/metabolism , Female , Food, Fortified , Humans , Infant , Infant Formula/chemistry , Infant Formula/standards , Infant, Newborn , Male , Middle Aged , Milk, Human/chemistry , Milk, Human/physiology , Nutritional Requirements , Pregnancy , Visual Acuity/physiology
19.
Am J Clin Nutr ; 78(4): 702-10, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14522727

ABSTRACT

BACKGROUND: Functional maturation of nervous tissues depends on membrane accretion of docosahexaenoic acid (DHA). Animal studies have shown that incorporation of dietary DHA into membrane phospholipids is dose dependent. The molecular effects of DHA are commonly studied in cultured cells, but questions remain about the physiologic connection between animal and cell models. OBJECTIVE: We developed a linear model for comparing the responses of rat nervous tissues to dietary DHA with the responses of human cell lines to DHA in medium. DESIGN: Rats were rendered chronically deficient in n-3 fatty acids by being reared on a peanut oil diet. DHA status was replenished in the F2 generation by using increasing supplements of a microalgal oil. Human retinoblastoma and neuroblastoma cells were dosed with unesterified DHA. DHA accumulation into phospholipids was defined by the plateau of the dose-response curve (DHA(max)) and by the supplement required to produce one-half the DHA(max) (DHA(50)). RESULTS: The DHA(max) values for 4 brain regions and 2 neuroblastoma lines were similar, and the value for the retinoblastoma line was similar to the retinal value. Expressing the DHA input as micro mol/10 g diet and as micro mol/L medium resulted in similar values for the ratio of DHA(max) to DHA(50) in the 4 brain regions and the 3 cell lines. The DHA(max)-DHA(50) ratios in the ethanolamine phosphoglyceride and phosphatidylcholine fractions in retinal phospholipids were 6 and 10 times, respectively, those in the brain and cultured cells. CONCLUSIONS: The dose-dependent responses of cells and the brain to DHA supplements can be compared by using DHA(max)-DHA(50) ratios. We propose a counting frame that allows the comparison of the dose responses of the brain and cells to exogenous DHA.


Subject(s)
Docosahexaenoic Acids/metabolism , Linear Models , Membranes/metabolism , Nerve Tissue/metabolism , Phospholipids/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cells, Cultured , Docosahexaenoic Acids/pharmacology , Female , Humans , Neuroblastoma/metabolism , Rats , Rats, Wistar , Retinoblastoma/metabolism
20.
J Neurosci Res ; 74(1): 134-41, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-13130515

ABSTRACT

The mRNA expression levels of acyl-CoA oxidase (AOX), a key enzyme in very-long-chain fatty acid peroxisomal oxidation, and of peroxisome proliferator-activated receptor-delta (PPAR-delta), a nuclear receptor possibly involved in the gene regulation of brain lipid metabolism, were determined in human Y79 retinoblastoma cells by using real-time quantitative polymerase chain reaction. Cells were dosed with alpha-linolenic acid (18:3n-3), the essential metabolic precursor of the n-3 polyunsaturated fatty acid series that normally gives rise through terminal peroxisomal oxidation to the synthesis of membrane docosahexaenoic acid (22:6n-3, or DHA). The AOX and PPAR-delta relative expression levels increased 2.3 and 3.4 times, respectively, upon dosing of cells with 7 microM 18:3n-3, whereas AOX cDNA abundance decreased by 50% upon dosing with 70 microM 18:3n-3. Concurrently, the DHA content increased by 23% in the membrane ethanolamine-phosphoglycerides from cells dosed with 7 microM 18:3n-3, whereas it decreased by 38% upon dosing with 70 microM 18:3n-3. The DHA's upstream precursors (20:5n-3 and 22:5n-3) both accumulated in cells dosed with 7 or 70 microM 18:3n-3. The 18:3n-3-induced changes in membrane phospholipid fatty acid composition support the hypothesis that the terminal peroxisomal step of n-3 conversion is rate limiting in the Y79 line. The concurrent 7 microM 18:3n-3-induced increase of mRNAs encoding for AOX and for PPAR-delta suggests that 18:3n-3 (or its metabolites) at low concentration could trigger its proper conversion to DHA, possibly through activation of PPAR-delta-mediated transcription of AOX. Decreased membrane DHA content and mRNA expression level of AOX in 70-microM 18:3n-3-dosed cells corroborated the relationship between AOX expression and DHA synthesis and suggested that simultaneous down-regulating events occurred at high concentrations of 18:3n-3.


Subject(s)
Docosahexaenoic Acids/metabolism , Oxidoreductases/biosynthesis , RNA, Messenger/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Retinoblastoma/metabolism , Transcription Factors/biosynthesis , alpha-Linolenic Acid/pharmacology , Acyl-CoA Oxidase , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/genetics , Docosahexaenoic Acids/analysis , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Oxidoreductases/analysis , Oxidoreductases/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Retinoblastoma/enzymology , Transcription Factors/analysis , Transcription Factors/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL