Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Neuropsychopharmacology ; 46(3): 579-602, 2021 02.
Article in English | MEDLINE | ID: mdl-32781459

ABSTRACT

Maternal immune activation (MIA) and poor maternal nutritional habits are risk factors for the occurrence of neurodevelopmental disorders (NDD). Human studies show the deleterious impact of prenatal inflammation and low n-3 polyunsaturated fatty acid (PUFA) intake on neurodevelopment with long-lasting consequences on behavior. However, the mechanisms linking maternal nutritional status to MIA are still unclear, despite their relevance to the etiology of NDD. We demonstrate here that low maternal n-3 PUFA intake worsens MIA-induced early gut dysfunction, including modification of gut microbiota composition and higher local inflammatory reactivity. These deficits correlate with alterations of microglia-neuron crosstalk pathways and have long-lasting effects, both at transcriptional and behavioral levels. This work highlights the perinatal period as a critical time window, especially regarding the role of the gut-brain axis in neurodevelopment, elucidating the link between MIA, poor nutritional habits, and NDD.


Subject(s)
Fatty Acids, Omega-3 , Prenatal Exposure Delayed Effects , Animals , Behavior, Animal , Brain , Female , Humans , Inflammation , Microglia , Pregnancy
2.
Brain Behav Immun ; 91: 716-730, 2021 01.
Article in English | MEDLINE | ID: mdl-32976934

ABSTRACT

Brain aging is characterized by a decline in cognitive functions, which can lead to the development of neurodegenerative pathologies. Age-related spatial learning and memory deficits are associated with a chronic low-grade inflammation. Anxiety disorders and stress response alterations, occurring for a part of the elderly, have also been linked to an increased neuroinflammation and thus, an accelerated cognitive decline. Nutrition is an innovative strategy to prevent age-related cognitive impairments. Among the nutrients, n-3 long chain polyunsaturated fatty acids (LC-PUFAs) and low molecular weight peptides from proteins, especially those from marine resources, are good candidates for their immunomodulatory, anxiolytic and neuroprotective properties. The aim of this study is to determine the combined effect of n-3 LC-PUFAs and low molecular weight peptides on cognitive functions, and their mechanism of action. We are the first to show that a dietary supplementation with a fish hydrolysate containing n-3 LC-PUFAs and low molecular weight peptides prevented the age-related spatial short-term memory deficits and modulated navigation strategies adopted during spatial learning. In addition, the fish hydrolysate displayed anxiolytic activities with the reduction of anxiety-like behaviour in aged mice, restored the plasmatic corticosterone levels similar to adult animals following an acute stress and modulated the hypothalamic stress response. These effects on behaviour can be explained by the immunomodulatory and neuroprotective properties of the fish hydrolysate that limited microgliosis in vivo, decreased LPS-induced expression of pro-inflammatory cytokines and increased the expression of growth factors such as BDNF and NGF in vitro. Thus, n-3 LC-PUFAs and low molecular weight peptides contained in the fish hydrolysate can play an important role in the limitation of neuroinflammation and stress response alterations during aging and represent a potential strategy for the prevention of age-related cognitive decline.


Subject(s)
Fatty Acids, Omega-3 , Memory, Short-Term , Animals , Diet , Dietary Supplements , Memory Disorders/prevention & control , Mice
3.
Nat Commun ; 11(1): 6133, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257673

ABSTRACT

Omega-3 fatty acids (n-3 PUFAs) are essential for the functional maturation of the brain. Westernization of dietary habits in both developed and developing countries is accompanied by a progressive reduction in dietary intake of n-3 PUFAs. Low maternal intake of n-3 PUFAs has been linked to neurodevelopmental diseases in Humans. However, the n-3 PUFAs deficiency-mediated mechanisms affecting the development of the central nervous system are poorly understood. Active microglial engulfment of synapses regulates brain development. Impaired synaptic pruning is associated with several neurodevelopmental disorders. Here, we identify a molecular mechanism for detrimental effects of low maternal n-3 PUFA intake on hippocampal development in mice. Our results show that maternal dietary n-3 PUFA deficiency increases microglia-mediated phagocytosis of synaptic elements in the rodent developing hippocampus, partly through the activation of 12/15-lipoxygenase (LOX)/12-HETE signaling, altering neuronal morphology and affecting cognitive performance of the offspring. These findings provide a mechanistic insight into neurodevelopmental defects caused by maternal n-3 PUFAs dietary deficiency.


Subject(s)
Brain/drug effects , Fatty Acids, Omega-3/pharmacology , Microglia/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/physiology , Phagocytosis/drug effects , Animals , Brain/growth & development , Dietary Supplements , Fatty Acids, Omega-3/deficiency , Fatty Acids, Omega-3/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Hippocampus/cytology , Hippocampus/growth & development , Homeostasis , Humans , Lipoxygenase , Male , Mice , Neurodevelopmental Disorders
4.
Brain Behav Immun ; 85: 162-169, 2020 03.
Article in English | MEDLINE | ID: mdl-31100369

ABSTRACT

Essential polyunsaturated fatty acids (PUFA) from the n-3 and n-6 series constitute the building blocks of brain cell membranes where they regulate most aspects of cell physiology. They are either biosynthesized from their dietary precursors or can be directly sourced from the diet. An overall increase in the dietary n-6/n-3 PUFA ratio, as observed in the Western diet, leads to reduced n-3 PUFAs in tissues that include the brain. Some clinical studies have shown a positive correlation between dietary n-3 PUFA intake and sleep quantity, yet evidence is still sparse. We here used a preclinical model of dietary n-3 PUFA deficiency to assess the precise relationship between dietary PUFA intake and sleep/wake activity. Using electroencephalography (EEG)/electromyography (EMG) recordings on n-3 PUFA deficient or sufficient mice, we showed that dietary PUFA deficiency affects the architecture of sleep-wake activity and the oscillatory activity of cortical neurons during sleep. In a second part of the study, and since PUFAs are a potent modulator of inflammation, we assessed the effect of dietary n-3 PUFA deficiency on the sleep response to an inflammatory stimulus known to modulate sleep/wake activity. We injected mice with the endotoxin lipopolysaccharide (LPS) and quantified the sleep response across the following 12 h. Our results revealed that n-3 PUFA deficiency affects the sleep response in basal condition and after a peripheral immune challenge. More studies are now required aimed at deciphering the molecular mechanisms underlying the intimate relationship between n-3 PUFAs and sleep/wake activity.


Subject(s)
Fatty Acids, Omega-3 , Fatty Acids, Omega-6 , Animals , Fatty Acids , Fatty Acids, Unsaturated , Mice , Sleep
5.
Behav Brain Res ; 379: 112348, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31711897

ABSTRACT

Dopamine (DA) is a critical neurotransmitter involved in motivational processes. Tetrahydrobiopterin (BH4) is an essential cofactor for tyrosine hydroxylase, the rate-limiting enzyme in DA synthesis. Decreases in BH4 levels are observed in several DA-related neuropsychiatric diseases involving impairment in motivation. Yet, whether BH4 could be used to treat motivational deficits has not been comprehensively investigated. To investigate the effects of exogenous BH4 administration on the dopaminergic system and related behaviors, we acutely injected mice with BH4 (50 mg/kg). Passage of BH4 through the blood brain barrier and accumulation in brain was measured using the in situ brain perfusion technique. DA release was then recorded using in-vivo micro-dialysis and motivation was evaluated through operant conditioning paradigms in basal condition and after an amphetamine (AMPH) injection. First, we showed that BH4 crosses the blood-brain barrier and that an acute peripheral injection of BH4 is sufficient to increase the concentrations of biopterins in the brain, without affecting BH4- and DA-related protein expression. Second, we report that this increase in BH4 enhanced AMPH-stimulated DA release in the nucleus accumbens. Finally, we found that BH4-induced DA release led to improved performance of a motivational task. Altogether, these findings suggest that BH4, through its action on the dopaminergic tone, could be used as a motivational enhancer.


Subject(s)
Amphetamine/pharmacology , Behavior, Animal/drug effects , Biopterins/analogs & derivatives , Brain/drug effects , Brain/metabolism , Conditioning, Operant/drug effects , Dopamine Agents/pharmacology , Dopamine/metabolism , Motivation/drug effects , Amphetamine/administration & dosage , Animals , Biopterins/administration & dosage , Biopterins/pharmacology , Dopamine Agents/administration & dosage , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism
6.
Neurosci Lett ; 708: 134348, 2019 08 24.
Article in English | MEDLINE | ID: mdl-31238131

ABSTRACT

Microglia are key players in brain function by maintaining brain homeostasis across lifetime. They participate to brain development and maturation through their ability to release neurotrophic factors, to remove immature synapses or unnecessary neural progenitors. They modulate neuronal activity in healthy adult brains and they also orchestrate the neuroinflammatory response in various pathophysiological contexts such as aging and neurodegenerative diseases. One of the main features of microglia is their high sensitivity to environmental factors, partly via the expression of a wide range of receptors. Recent data pinpoint that dietary fatty acids modulate microglia function. Both the quantity and the type of fatty acid are potent modulators of microglia physiology. The present review aims at dissecting the current knowledge on the direct and indirect mechanisms (focus on gut microbiota and hormones) through which fatty acids influence microglial physiology. We summarize main discoveries from in vitro and in vivo models on fatty acid-mediated microglial modulation. All these studies represent a promising field of research that could promote using nutrition as a novel therapeutic or preventive tool in diseases involving microglia dysfunctions.


Subject(s)
Fatty Acids/physiology , Microglia/physiology , Animals , Fatty Acids/pharmacology , Gastrointestinal Microbiome/physiology , Hormones/physiology , Humans , Lipid Metabolism , Microglia/drug effects
7.
Brain Behav Immun ; 76: 17-27, 2019 02.
Article in English | MEDLINE | ID: mdl-30086401

ABSTRACT

The brain is highly enriched in long chain polyunsaturated fatty acids (LC-PUFAs) that display immunomodulatory properties in the brain. At the periphery, the modulation of inflammation by LC-PUFAs occurs through lipid mediators called oxylipins which have anti-inflammatory and pro-resolving activities when derived from n-3 LC-PUFAs and pro-inflammatory activities when derived from n-6 LC-PUFAs. However, whether a diet rich in LC-PUFAs modulates oxylipins and neuroinflammation in the brain has been poorly investigated. In this study, the effect of a dietary n-3 LC-PUFA supplementation on oxylipin profile and neuroinflammation in the brain was analyzed. Mice were given diets deficient or supplemented in n-3 LC-PUFAs for a 2-month period starting at post-natal day 21, followed by a peripheral administration of lipopolysaccharide (LPS) at adulthood. We first showed that dietary n-3 LC-PUFA supplementation induced n-3 LC-PUFA enrichment in the hippocampus and subsequently an increase in n-3 PUFA-derived oxylipins and a decrease in n-6 PUFA-derived oxylipins. In response to LPS, n-3 LC-PUFA deficient mice presented a pro-inflammatory oxylipin profile whereas n-3 LC-PUFA supplemented mice displayed an anti-inflammatory oxylipin profile in the hippocampus. Accordingly, the expression of cyclooxygenase-2 and 5-lipoxygenase, the enzymes implicated in pro- and anti-inflammatory oxylipin synthesis, was induced by LPS in both diets. In addition, LPS-induced pro-inflammatory cytokine increase was reduced by dietary n-3 LC-PUFA supplementation. These results indicate that brain n-3 LC-PUFAs increase by dietary means and promote the synthesis of anti-inflammatory derived bioactive oxylipins. As neuroinflammation plays a key role in all brain injuries and many neurodegenerative disorders, the present data suggest that dietary habits may be an important regulator of brain cytokine production in these contexts.


Subject(s)
Fatty Acids, Omega-3/metabolism , Oxylipins/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cytokines/metabolism , Diet , Dietary Supplements , Fatty Acids , Fatty Acids, Omega-3/physiology , Fatty Acids, Omega-6 , Fatty Acids, Unsaturated/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation/metabolism , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred C57BL , Models, Animal
8.
Brain Behav Immun ; 73: 427-440, 2018 10.
Article in English | MEDLINE | ID: mdl-29879442

ABSTRACT

Maternal immune activation (MIA) is a common environmental insult on the developing brain and represents a risk factor for neurodevelopmental disorders. Animal models of in utero inflammation further revealed a causal link between maternal inflammatory activation during pregnancy and behavioural impairment relevant to neurodevelopmental disorders in the offspring. Accumulating evidence point out that proinflammatory cytokines produced both in the maternal and fetal compartments are responsible for social, cognitive and emotional behavioral deficits in the offspring. Polyunsaturated fatty acids (PUFAs) are essential fatty acids with potent immunomodulatory activities. PUFAs and their bioactive derivatives can promote or inhibit many aspects of the immune and inflammatory response. PUFAs of the n-3 series ('n-3 PUFAs', also known as omega-3) exhibit anti-inflammatory/pro-resolution properties and promote immune functions, while PUFAs of the n-6 series ('n-6 PUFAs' or omega-6) favor pro-inflammatory responses. The present study aimed at providing insight into the effects of n-3 PUFAs on the consequences of MIA on brain development. We hypothesized that a reduction in n-3 PUFAs exacerbates both maternal and fetal inflammatory responses to MIA and later-life defects in memory in the offspring. Based on a lipopolysaccharide (LPS) model of MIA (LPS injection at embryonic day 17), we showed that n-3 PUFA deficiency 1) alters fatty acid composition of the fetal and adult offspring brain; 2) exacerbates maternal and fetal inflammatory processes with no significant alteration of microglia phenotype, and 3) induces spatial memory deficits in the adult offspring. We also showed a strong negative correlation between brain content in n-3 PUFA and cytokine production in MIA-exposed fetuses. Overall, our study is the first to address the deleterious effects of n-3 PUFA deficiency on brain lipid composition, inflammation and memory performances in MIA-exposed animals and indicates that it should be considered as a potent environmental risk factor for the apparition of neurodevelopmental disorders.


Subject(s)
Fatty Acids, Omega-3/deficiency , Fatty Acids, Omega-3/metabolism , Spatial Memory/drug effects , Animals , Animals, Newborn , Behavior, Animal/drug effects , Brain/drug effects , Cytokines/drug effects , Dietary Supplements , Disease Models, Animal , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-6/metabolism , Fatty Acids, Omega-6/physiology , Female , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/metabolism , Social Behavior
10.
Prog Neuropsychopharmacol Biol Psychiatry ; 79(Pt A): 19-26, 2017 10 03.
Article in English | MEDLINE | ID: mdl-27392882

ABSTRACT

Within the central nervous system the traditional role of microglia has been in brain infection and disease, phagocytosing debris and secreting factors to modify disease progression. More recently, microglia have been found to be important for normal brain development, circuit refinement, and synaptic plasticity in ways that were previously unsuspected. Hence, the brain innate immune system appears to be key in all situations, ranging from physiology to pathology. This unique feature of microglia is established by the wide array of receptors it is equipped with to sense molecular patterns. This includes receptors to most if not all neurotransmitters, neuromodulators and purines. We here review novel, yet extensive literature on a new class of microglia modulators, namely bioactive fatty acids. These lipids are issued from metabolism of nutrients and can cross the blood brain barrier to reach the CNS. They appear to be direct modulators of microglial activity, triggering/inhibiting inflammatory processes or enhancing/inhibiting the ability of these cells to respond to hazardous agents.


Subject(s)
Brain/cytology , Fatty Acids/metabolism , Microglia/physiology , Neuroimmunomodulation/physiology , Animals , Brain/metabolism , Fatty Acids/pharmacology , Humans , Microglia/drug effects
11.
Int J Obes (Lond) ; 41(4): 502-509, 2017 04.
Article in English | MEDLINE | ID: mdl-28025577

ABSTRACT

BACKGROUND/OBJECTIVES: Energy-dense food exposure and stress during development have been suggested to contribute to obesity and metabolic disorders later in life. Although these factors are frequently associated, the effects of their combination have not yet been investigated. In this study, using an animal model, we examined the long-term impact of maternal high-fat diet (HFD) and early-life stress (ELS) on energy homoeostasis control and food motivation. METHODS: Body weight growth under HFD, adipose tissue, body weight control in response to fasting and refeeding, food-motivated behaviour and mesolimbic dopamine function were examined in adult male offspring exposed to maternal HFD (during gestation and lactation) and/or ELS (maternal separation 3 h per day from postnatal day 2 to 14). RESULTS: Maternal HFD or ELS alone had no significant effect on offspring body weight; however, the combination of these factors exacerbated body weight gain when animals were exposed to HFD after weaning. There are no other significant combinatory effects of these perinatal events. In contrast, independently of the maternal diet, ELS disrupted body weight control during a fasting-refeeding procedure, increased adipose tissue mass and altered lipid metabolism. Finally, maternal HFD and ELS both resulted in exacerbated food-motivated behaviour and blunted dopamine release in the nucleus accumbens during palatable food consumption. CONCLUSIONS: We report a synergistic effect of perinatal HFD exposure and stress on the susceptibility to gain weight under HFD. However, ELS has a stronger impact than maternal HFD exposure on energy homoeostasis and food motivation in adult offspring. Altogether, our results suggest a programming effect of stress and nutrition supporting the hypothesis of the developmental origin of health and disease.


Subject(s)
Diet, High-Fat/adverse effects , Dopamine/metabolism , Feeding Behavior/physiology , Obesity/metabolism , Prenatal Exposure Delayed Effects/metabolism , Stress, Psychological , Animals , Disease Models, Animal , Female , Male , Maternal Deprivation , Maternal Nutritional Physiological Phenomena , Pregnancy , Rats , Rats, Wistar , Signal Transduction/physiology
12.
Article in English | MEDLINE | ID: mdl-27914510

ABSTRACT

The aim of the study was to determine the effect of maternal diets administered since day 1 of gestation and containing dairy lipids or vegetable oils differing in the supply of n-3 polyunsaturated fatty acids (n-3 PUFAs) (equilibrated or deficient) and of Lactobacillus fermentum (L. fermentum) on the docosahexaenoic acid (DHA) accretion in the pups at postnatal day 14 in the prefrontal cortex (PFC) and hippocampus (HC) for brain structures and in the liver and adipose tissue for peripheral tissues. Maternal milk fatty acid composition was also assessed by analyzing the fatty acid composition of the gastric content of the pups. DHA was higher in mice supplemented with L. fermentum than in mice in the deficient group in HC and PFC and also in liver and adipose tissue. This increase could be linked to the slight but significant increase in C18:3n-3 in the maternal milk. This proportion was comparable in the dairy lipid group for which the brain DHA level was the highest. L. fermentum may have a key role in the protection of the brain during the perinatal period via the neuronal accretion of n-3 PUFAs, especially during n-3 PUFA deficiency.


Subject(s)
Adipose Tissue/metabolism , Brain/metabolism , Docosahexaenoic Acids/metabolism , Limosilactobacillus fermentum/physiology , Liver/metabolism , Milk/chemistry , Plant Oils/administration & dosage , Animals , Animals, Suckling , Dietary Supplements , Fatty Acids/analysis , Female , Hippocampus/metabolism , Maternal Nutritional Physiological Phenomena , Mice , Prefrontal Cortex/metabolism , Pregnancy , Tissue Distribution
13.
Transl Psychiatry ; 6(11): e966, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27898075

ABSTRACT

Anxiety disorders and depression are well-documented in subjects exposed to adverse childhood events. Recently, maternal obesity and/or maternal consumption of high-fat diets (HFD) have been also proposed as risk factors for offspring mental health. Here using an animal model in rats, we explored the combinatorial effects of a maternal HFD (40% of energy from fat without impact on maternal weight; during gestation and lactation) and maternal separation (MS) in offspring. In the prefrontal cortex (PFC) of pups, MS led to changes in the expression of several genes such as Bdnf (brain derived neurotrophic factor), 5HT-r1a (serotonin receptor 1a) and Rest4 (neuron-restrictive silencer element, repressor element 1, silencing transcription factor (Rest), splicing variant 4). Surprisingly, perinatal HFD strongly attenuated the developmental alterations induced by MS. Furthermore, maternal HFD totally prevented the endophenotypes (anxiety, spatial memory, social behavior, hypothalamic-pituitary-adrenal (HPA) axis response to stress, hippocampal neurogenesis and visceral pain) associated with MS at adulthood. Finally, we also demonstrated that HFD intake reduced anxiety and enhanced maternal care in stressed dams. Overall, our data suggest that a HFD restricted to gestation and lactation, which did not lead to overweight in dams, had limited effects in unstressed offspring, highlighting the role of maternal obesity, rather than fat exposure per se, on brain vulnerability during development.


Subject(s)
Disease Models, Animal , Life Change Events , Prenatal Exposure Delayed Effects , Animals , Animals, Newborn/genetics , Animals, Newborn/psychology , Anxiety/genetics , Anxiety/psychology , Body Weight , Brain-Derived Neurotrophic Factor/genetics , Diet, High-Fat , Female , Maternal Behavior , Maternal Deprivation , Prefrontal Cortex/metabolism , Pregnancy , Rats, Wistar , Receptor, Serotonin, 5-HT1A/genetics , Repressor Proteins/genetics
14.
Article in English | MEDLINE | ID: mdl-27720041

ABSTRACT

Polyunsaturated fatty acids (PUFAs) are essential fatty acids, which are critical for brain development and later life cognitive functions. The main brain PUFAs are docosahexaenoic acid (DHA) for the n-3 family and arachidonic acid (ARA) for the n-6 family, which are provided to the post-natal brain by breast milk or infant formula. Recently, the use of dairy lipids (DL) in replacement of vegetable lipids (VL) was revealed to potently promote the accretion of DHA in the developing brain. Brain DHA, in addition to be a key component of brain development, display potent anti-inflammatory activities, which protect the brain from adverse inflammatory events. In this work, we evaluated the protective effect of partial replacement of VL by DL, supplemented or not with DHA and ARA, on post-natal inflammation and its consequence on memory. Mice were fed with diets poor in vegetal n-3 PUFA (Def VL), balanced in vegetal n-3/n-6 PUFA (Bal VL), balanced in dairy lipids (Bal DL) or enriched in DHA and ARA (Supp VL; Supp DL) from the first day of gestation until adulthood. At post-natal day 14 (PND14), pups received a single administration of the endotoxin lipopolysaccharide (LPS) and brain cytokine expression, microglia phenotype and neurogenesis were measured. In a second set of experiments, memory and neurogenesis were measured at adulthood. Overall, our data showed that lipid quality of the diet modulates early life LPS effect on microglia phenotype, brain cytokine expression and neurogenesis at PND14 and memory at adulthood. In particular, Bal DL diet protects from the adverse effect of early life LPS exposure on PND14 neurogenesis and adult spatial memory.


Subject(s)
Dietary Fats/administration & dosage , Learning Disabilities/prevention & control , Lipopolysaccharides/adverse effects , Spatial Memory/drug effects , Animals , Animals, Newborn , Arachidonic Acid/metabolism , Brain/drug effects , Brain/growth & development , Brain/metabolism , Dietary Fats/pharmacology , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Learning Disabilities/chemically induced , Mice , Neurogenesis/drug effects , Plant Oils/administration & dosage , Plant Oils/pharmacology
15.
Article in English | MEDLINE | ID: mdl-27269711

ABSTRACT

Mimicking the breast milk lipid composition appears to be necessary for infant formula to cover the brain's needs in n-3 PUFA. In this study, we evaluated the impact of partial replacement of vegetable oil (VL) in infant formula by dairy fat (DL) on docosahexaenoic acid (DHA) brain level, neuroplasticity and corticosterone in mice. Mice were fed with balanced VL or balanced DL diets enriched or not in DHA and arachidonic acid (ARA) from the first day of gestation. Brain DHA level, microglia number, neurogenesis, corticosterone and glucocorticoid receptor expression were measured in the offsprings. DL diet increased DHA and neuroplasticity in the brain of mice at postnatal day (PND) 14 and at adulthood compared to VL. At PND14, ARA and DHA supplementation increased DHA in VL but not in DL mice brain. Importantly, DHA and ARA supplementation further improved neurogenesis and decreased corticosterone level in DL mice at adulthood. In conclusion, dairy lipids improve brain DHA level and neuroplasticity.


Subject(s)
Brain/growth & development , Corticosterone/metabolism , Dietary Fats/administration & dosage , Docosahexaenoic Acids/metabolism , Plant Oils/administration & dosage , Animals , Arachidonic Acid/metabolism , Brain/metabolism , Dairy Products , Dietary Fats/pharmacology , Gene Expression Regulation/drug effects , Humans , Infant , Infant Formula/chemistry , Mice , Models, Animal , Neuronal Plasticity/drug effects , Plant Oils/pharmacology , Receptors, Glucocorticoid/drug effects
16.
Brain Behav Immun ; 55: 249-259, 2016 07.
Article in English | MEDLINE | ID: mdl-26718448

ABSTRACT

Sustained inflammation in the brain together with microglia activation can lead to neuronal damage. Hence limiting brain inflammation and activation of microglia is a real therapeutic strategy for inflammatory disease. Resolvin D1 (RvD1) and resolvin E1 (RvE1) derived from n-3 long chain polyunsaturated fatty acids are promising therapeutic compounds since they actively turn off the systemic inflammatory response. We thus evaluated the anti-inflammatory activities of RvD1 and RvE1 in microglia cells in vitro. BV2 cells were pre-incubated with RvD1 or RvE1 before lipopolysaccharide (LPS) treatment. RvD1 and RvE1 both decreased LPS-induced proinflammatory cytokines (TNF-α, IL-6 and IL-1ß) gene expression, suggesting their proresolutive activity in microglia. However, the mechanisms involved are distinct as RvE1 regulates NFκB signaling pathway and RvD1 regulates miRNAs expression. Overall, our findings support that pro-resolving lipids are involved in the resolution of brain inflammation and can be considered as promising therapeutic agents for brain inflammation.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/analogs & derivatives , Inflammation/drug therapy , Interleukin-6/metabolism , MicroRNAs/metabolism , Microglia/drug effects , Tumor Necrosis Factor-alpha/drug effects , Animals , Eicosapentaenoic Acid/pharmacology , Mice
17.
Appetite ; 96: 333-346, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26407804

ABSTRACT

Providing well-being and maintaining good health are main objectives subjects seek from diet. This manuscript describes the development and preliminary validation of an instrument assessing well-being associated with food and eating habits in a general healthy population. Qualitative data from 12 groups of discussion (102 subjects) conducted with healthy subjects were used to develop the core of the Well-being related to Food Questionnaire (Well-BFQ). Twelve other groups of discussion with subjects with joint (n = 34), digestive (n = 32) or repetitive infection complaints (n = 30) were performed to develop items specific to these complaints. Five main themes emerged from the discussions and formed the modular backbone of the questionnaire: "Grocery shopping", "Cooking", "Dining places", "Commensality", "Eating and drinking". Each module has a common structure: items about subject's food behavior and items about immediate and short-term benefits. An additional theme - "Eating habits and health" - assesses subjects' beliefs about expected benefits of food and eating habits on health, disease prevention and protection, and quality of ageing. A preliminary validation was conducted with 444 subjects with balanced diet; non-balanced diet; and standard diet. The structure of the questionnaire was further determined using principal component analyses exploratory factor analyses, with confirmation of the sub-sections food behaviors, immediate benefits (pleasure, security, relaxation), direct short-term benefits (digestion and satiety, energy and psychology), and deferred long-term benefits (eating habits and health). Thirty-three subscales and 14 single items were further defined. Confirmatory analyses confirmed the structure, with overall moderate to excellent convergent and divergent validity and internal consistency reliability. The Well-BFQ is a unique, modular tool that comprehensively assesses the full picture of well-being related to food and eating habits in the general population.


Subject(s)
Feeding Behavior , Surveys and Questionnaires , White People , Adolescent , Adult , Aged , Body Mass Index , Female , France , Humans , Male , Middle Aged , Principal Component Analysis , Reproducibility of Results , Socioeconomic Factors , Young Adult
18.
Transl Psychiatry ; 4: e437, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25203168

ABSTRACT

Understanding how malnutrition contributes to depression is building momentum. In the present study we unravel molecular and cellular mechanisms by which nutritional disturbances lead to impaired emotional behaviour in mice. Here we report that nutritional n-3 polyunsaturated fatty acids (PUFA) deficiency induces a chronic stress state reflected by disrupted glucocorticoid receptor (GR)-mediated signalling pathway along with hypothalamic-pituitary-adrenal (HPA) axis hyperactivity. This hyperactivity in turn resulted in neuronal atrophy in the dorsolateral (dl)- and dorsomedial (dm)- prefrontal cortex (PFC) and subsequent mood-related behaviour alterations, similarly to chronic social defeat stress. Supplementation of n-3 PUFA prevented detrimental chronic social defeat stress-induced emotional and neuronal impairments by impeding HPA axis hyperactivity. These results indicate a role for dietary n-3 PUFA in the prevention of HPA axis dysfunction associated with the development of some neuropsychiatric disorders including depression.


Subject(s)
Depression/pathology , Depression/physiopathology , Disease Models, Animal , Emotions/physiology , Fatty Acids, Omega-3/physiology , Neurons/pathology , Neurons/physiology , Prefrontal Cortex/pathology , Prefrontal Cortex/physiopathology , Animals , Depression/psychology , Dominance-Subordination , Hypothalamo-Hypophyseal System/pathology , Hypothalamo-Hypophyseal System/physiopathology , Male , Mice , Mice, Inbred C57BL , Pituitary-Adrenal System/pathology , Pituitary-Adrenal System/physiopathology , Receptors, Glucocorticoid/physiology , Signal Transduction/physiology
19.
Article in English | MEDLINE | ID: mdl-24908517

ABSTRACT

Due to the implication of docosahexaenoic acid (DHA) in neurogenesis, synaptogenesis, neurite outgrowth and to its high incorporation into the brain, this n-3 long chain polyunsaturated fatty acid (LCPUFA) is considered as crucial in the development and maintenance of the learning memory performance throughout life. In the present chapter we aimed at reviewing data investigating the relation between DHA and cognition during the perinatal period, young adult- and adulthood and neurodegenerative diseases such as Alzheimer disease (AD). In Humans, dietary DHA supplementation from the perinatal period to adulthood does not reveal a clear and consistent memory improvement whereas it is the case in animal studies. The positive effects observed in animal models may have been enhanced by using n-3 PUFA deficient animal models as controls. In animal models of AD, a general consensus on the beneficial effects of n-3 LCPUFA in attenuating cognitive impairment was established. These studies make DHA a potential suitable micronutrient for the maintenance of cognitive performance at all periods of life.


Subject(s)
Cognition/drug effects , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Adult , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Brain/drug effects , Brain/physiology , Child , Cognition/physiology , Female , Fetus , Humans , Infant , Learning/drug effects , Learning/physiology , Male , Memory/drug effects , Memory/physiology , Pregnancy
20.
Brain Behav Immun ; 34: 151-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23994463

ABSTRACT

Within the central nervous system (CNS) the traditional role of microglia has been in brain infection and disease, phagocytosing debris and secreting factors to modify disease progression. This led to the concept of "resting" versus "activated" microglia. However, this is misleading because multiple phenotypic and morphological stages of microglia can influence neuronal structure and function in any condition and recent evidence extends their role to healthy brain homeostasis. The present work was thus aimed at reappraising the concept of morphofunctional activity of microglia in a context of peripheral acute immune challenge, where microglial activity is known to be modified, using the new state-of-the-art techniques available. To do so, mice were injected peripherally with lipopolysaccharide, a potent inducer of cerebral inflammation, and we assessed early cytokines production, phenotype, motility and morphology of microglial cells. Our results showed that LPS induced a widespread inflammatory response both peripherally and centrally, as revealed by the quantification of cytokines levels. We also found an alteration of microglial motility that was confirmed by in vivo studies showing an overall reduction of microglial processes length in the hippocampus of LPS-treated animals. Finally, analysis of various surface receptors expression revealed that LPS did not significantly impact microglial phenotype 2h after the injection but rather induced an increase of CD11b(+)/CD45(high) cells. These latter may be at the vasculature, at the CNS vicinity, or may have invaded the CNS.


Subject(s)
Cytokines/metabolism , Lipopolysaccharides/toxicity , Microglia/cytology , Microglia/physiology , Animals , Hippocampus/drug effects , Hippocampus/physiology , Inflammation/chemically induced , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...