Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Neurobiol Aging ; 113: 118-130, 2022 05.
Article in English | MEDLINE | ID: mdl-35334439

ABSTRACT

In Alzheimer's disease (AD), hyper-phosphorylation and aggregation of tau correlate with clinical progression and represent a valid therapeutic target. A recent 20-year prospective study revealed an association between moderate to high frequency of Finnish sauna bathing and a lower incidence of dementia and AD, but the molecular mechanisms underlying these benefits remain uncertain. Here, we tested the hypothesis that sauna-like conditions could lower tau phosphorylation by increasing body temperature. We observed a decrease in tau phosphorylation in wild-type and hTau mice as well as in neuron-like cells when exposed to higher temperatures. These effects were correlated with specific changes in phosphatase and kinase activities, but not with inflammatory or heat shock responses. We also used a drug strategy to promote thermogenesis: topical application of menthol, which led to a sustained increase in body temperature in hTau mice, concomitant with a significant decrease in tau phosphorylation. Our results suggest that sauna-like conditions or menthol treatment could lower tau pathology through mild hyperthermia, and may provide promising therapeutic strategies for AD and other tauopathies.


Subject(s)
Alzheimer Disease , Steam Bath , Tauopathies , Alzheimer Disease/pathology , Animals , Menthol , Mice , Phosphorylation , Prospective Studies , Tauopathies/pathology , tau Proteins/metabolism
2.
Nutrients ; 13(12)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34959768

ABSTRACT

The aggregation of alpha-synuclein protein (αSyn) is a hallmark of Parkinson's disease (PD). Considerable evidence suggests that PD involves an early aggregation of αSyn in the enteric nervous system (ENS), spreading to the brain. While it has previously been reported that omega-3 polyunsaturated fatty acids (ω-3 PUFA) acts as neuroprotective agents in the brain in murine models of PD, their effect in the ENS remains undefined. Here, we studied the effect of dietary supplementation with docosahexaenoic acid (DHA, an ω-3 PUFA), on the ENS, with a particular focus on enteric dopaminergic (DAergic) neurons. Thy1-αSyn mice, which overexpress human αSyn, were fed ad libitum with a control diet, a low ω-3 PUFA diet or a diet supplemented with microencapsulated DHA and then compared with wild-type littermates. Our data indicate that Thy1-αSyn mice showed a lower density of enteric dopaminergic neurons compared with non-transgenic animals. This decrease was prevented by dietary DHA. Although we found that DHA reduced microgliosis in the striatum, we did not observe any evidence of peripheral inflammation. However, we showed that dietary intake of DHA promoted a build-up of ω-3 PUFA-derived endocannabinoid (eCB)-like mediators in plasma and an increase in glucagon-like peptide-1 (GLP-1) and the redox regulator, Nrf2 in the ENS. Taken together, our results suggest that DHA exerts neuroprotection of enteric DAergic neurons in the Thy1-αSyn mice, possibly through alterations in eCB-like mediators, GLP-1 and Nrf2.


Subject(s)
Dietary Supplements , Docosahexaenoic Acids/pharmacology , Enteric Nervous System/drug effects , Neuroprotective Agents/pharmacology , Synucleinopathies/drug therapy , Animals , Diet , Disease Models, Animal , Dopaminergic Neurons/drug effects , Mice , Mice, Transgenic , Thy-1 Antigens/metabolism , alpha-Synuclein/metabolism
3.
Neurobiol Dis ; 161: 105542, 2021 12.
Article in English | MEDLINE | ID: mdl-34737043

ABSTRACT

BACKGROUND: Vitamin A (VitA), via its active metabolite retinoic acid (RA), is critical for the maintenance of memory function with advancing age. Although its role in Alzheimer's disease (AD) is not well understood, data suggest that impaired brain VitA signaling is associated with the accumulation of ß-amyloid peptides (Aß), and could thus contribute to the onset of AD. METHODS: We evaluated the protective action of a six-month-long dietary VitA-supplementation (20 IU/g), starting at 8 months of age, on the memory and the neuropathology of the 3xTg-AD mouse model of AD (n = 11-14/group; including 4-6 females and 7-8 males). We also measured protein levels of Retinoic Acid Receptor ß (RARß) and Retinoid X Receptor γ (RXRγ) in homogenates from the inferior parietal cortex of 60 participants of the Religious Orders study (ROS) divided in three groups: no cognitive impairment (NCI) (n = 20), mild cognitive impairment (MCI) (n = 20) and AD (n = 20). RESULTS: The VitA-enriched diet preserved spatial memory of 3xTg-AD mice in the Y maze. VitA-supplementation affected hippocampal RXR expression in an opposite way according to sex by tending to increase in males and decrease in females their mRNA expression. VitA-enriched diet also reduced the amount of hippocampal Aß40 and Aß42, as well as the phosphorylation of tau protein at sites Ser396/Ser404 (PHF-1) in males. VitA-supplementation had no effect on tau phosphorylation in females but worsened their hippocampal Aß load. However, the expression of Rxr-ß in the hippocampus was negatively correlated with the amount of both soluble and insoluble Aß in both males and females. Western immunoblotting in the human cortical samples of the ROS study did not reveal differences in RARß levels. However, it evidenced a switch from a 60-kDa-RXRγ to a 55-kDa-RXRγ in AD, correlating with ante mortem cognitive decline and the accumulation of neuritic plaques in the brain cortex. CONCLUSION: Our data suggest that (i) an altered expression of RXRs receptors is a contributor to ß-amyloid pathology in both humans and 3xTg-AD mice, (ii) a chronic exposure of 3xTg-AD mice to a VitA-enriched diet may be protective in males, but not in females.


Subject(s)
Alzheimer Disease , Vitamin A , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Diet , Disease Models, Animal , Female , Hippocampus/metabolism , Humans , Male , Mice , Mice, Transgenic , Retinoid X Receptors/metabolism , tau Proteins/metabolism
4.
Eur J Neurosci ; 54(9): 7092-7108, 2021 11.
Article in English | MEDLINE | ID: mdl-34549475

ABSTRACT

Olfactory dysfunction is observed in several neurological disorders including Mild Cognitive Impairment (MCI) and Alzheimer disease (AD). These deficits occur early and correlate with global cognitive performance, depression and degeneration of olfactory regions in the brain. Despite extensive human studies, there has been little characterization of the olfactory system in models of AD. In order to determine if olfactory structural and/or molecular phenotypes are observed in a model expressing a genetic risk factor for AD, we assessed the olfactory bulb (OB) in APOE4 transgenic mice. A significant decrease in OB weight was observed at 12 months of age in APOE4 mice concurrent with inflammation and decreased NeuN expression. In order to determine if a diet rich in omega-3s may alleviate the olfactory system phenotypes observed, we assessed WT and APOE4 mice on a docosahexaenoic acid (DHA) diet. APOE4 mice on a DHA diet did not present with atrophy of the OB, and the alterations in NeuN and IBA-1 expression were alleviated. Furthermore, alterations in caspase mRNA and protein expression in the APOE4 OB were not observed with a DHA diet. Similar to the human AD condition, OB atrophy is an early phenotype in the APOE4 mice and concurrent with inflammation. These data support a link between the structural olfactory brain region atrophy and the olfactory dysfunction observed in AD and suggest that inflammation and cell death pathways may contribute to the olfactory deficits observed. Furthermore, the results suggest that diets enriched in DHA may provide benefit to APOE4 allele carriers.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Docosahexaenoic Acids/physiology , Olfaction Disorders/diet therapy , Olfactory Bulb , Alzheimer Disease/complications , Alzheimer Disease/genetics , Animals , Apolipoprotein E4/genetics , Atrophy , Diet , Disease Models, Animal , Mice , Mice, Transgenic , Olfaction Disorders/etiology , Olfaction Disorders/genetics , Olfactory Bulb/growth & development , Olfactory Bulb/metabolism , Olfactory Bulb/pathology
5.
Alzheimers Dement (N Y) ; 4: 677-687, 2018.
Article in English | MEDLINE | ID: mdl-30560200

ABSTRACT

INTRODUCTION: High levels of plasmatic branched-chain amino acids (BCAA), commonly used as dietary supplements, are linked to metabolic risk factors for Alzheimer's disease (AD). BCAA directly influence amino acid transport to the brain and, therefore, neurotransmitter levels. We thus investigated the impact of BCAA on AD neuropathology in a mouse model. METHODS: 3xTg-AD mice were fed either a control diet or a high-fat diet from 6 to 18 months of age. For the last 2 months, dietary BCAA content was adjusted to high (+50%), normal (+0%), or low (-50%). RESULTS: Mice fed a BCAA-supplemented high-fat diet displayed higher tau neuropathology and only four out of 13 survived. Mice on the low-BCAA diet showed higher threonine and tryptophan cortical levels while performing better on the novel object recognition task. DISCUSSION: These preclinical data underscore a potential risk of combining high-fat and high BCAA consumption, and possible benefits from BCAA restriction in AD.

6.
Exp Neurol ; 301(Pt A): 39-49, 2018 03.
Article in English | MEDLINE | ID: mdl-29229294

ABSTRACT

Polyunsaturated fatty acids omega-3 (n-3 PUFA), such as docosahexaenoic acid (DHA), have been shown to prevent, and partially reverse, neurotoxin-induced nigrostriatal denervation in animal models of Parkinson's disease (PD). However, the accumulation of α-synuclein (αSyn) in cerebral tissues is equally important to the pathophysiology. To determine whether DHA intake improves various aspects related to synucleinopathy, ninety male mice overexpressing human αSyn under the Thy-1 promoter (Thy1-αSyn) were fed one of three diets (specially formulated control, low n-3 PUFA or high DHA) and compared to non-transgenic C57/BL6 littermate mice exposed to a control diet. Thy1-αSyn mice displayed impaired motor skills, lower dopaminergic neuronal counts within the substantia nigra (-13%) in parallel to decreased levels of the striatal dopamine transporter (DAT) (-24%), as well as reduced NeuN (-41%) and synaptic proteins PSD-95 (-51%), synaptophysin (-80%) and vesicular acetylcholine transporter (VChAT) (-40%) in the cerebral cortex compared to C57/BL6 mice. However, no significant difference in dopamine concentrations was observed by HPLC analysis between Thy1-αSyn and non-transgenic C57/BL6 littermates under the control diet. The most striking finding was a favorable effect of DHA on the survival/longevity of Thy1-αSyn mice (+51% survival rate at 12months of age). However, dietary DHA supplementation did not have a significant effect on other parameters examined in this study, despite increased striatal dopamine concentrations. While human αSyn monomers and oligomers were detected in the cortex of Thy1-αSyn mice, the effects of the diets were limited to a small increase of 42kDa oligomers in insoluble protein fractions upon n-3 PUFA deprivation. Overall, our data indicate that a diet rich in n-3 PUFA has a beneficial effect on the longevity of a murine model of α-synucleinopathy without a major impact on the dopamine system and motor impairments, nor αSyn levels.


Subject(s)
Brain/drug effects , Brain/pathology , Docosahexaenoic Acids/pharmacology , Parkinsonian Disorders/pathology , alpha-Synuclein/genetics , Animals , Dietary Supplements , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
Neurobiol Aging ; 57: 28-35, 2017 09.
Article in English | MEDLINE | ID: mdl-28595105

ABSTRACT

At a population level, dietary consumption of fish rich in docosahexaenoic acid (DHA) is associated with prevention of cognitive decline but this association is not clear in carriers of the apolipoprotein E epsilon 4 allele (E4). Plasma and liver DHA concentrations show significant alterations in E4 carriers, in part corrected by DHA supplementation. However, whether DHA sufficiency in E4 carriers has consequences on cognition is unknown. Mice expressing human E4 or apolipoprotein E epsilon 3 allele (E3) were fed either a control diet or a diet containing DHA for 8 months and cognitive performance was tested using the object recognition test and the Barnes maze test. In E4 mice fed the control diet, impaired memory was detected and arachidonic acid concentrations were elevated in the hippocampus compared to E3 mice fed the control diet. DHA consumption prevented memory decline and restored arachidonic acid concentrations in the hippocampus of E4 mice. Our results suggest that long-term high-dose DHA intake may prevent cognitive decline in E4 carriers.


Subject(s)
Apolipoprotein E4 , Cognitive Dysfunction/genetics , Cognitive Dysfunction/prevention & control , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Animals , Arachidonic Acid/metabolism , Cognitive Dysfunction/metabolism , Female , Hippocampus , Male , Mice, Inbred C57BL
8.
Aging (Albany NY) ; 9(5): 1386-1403, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28522792

ABSTRACT

Defects in p21-activated kinase (PAK) lead to dendritic spine abnormalities and are sufficient to cause cognition impairment. The decrease in PAK in the brain of Alzheimer's disease (AD) patients is suspected to underlie synaptic and dendritic disturbances associated with its clinical expression, particularly with symptoms related to frontal cortex dysfunction. To investigate the role of PAK combined with Aß and tau pathologies (3xTg-AD mice) in the frontal cortex, we generated a transgenic model of AD with a deficit in PAK activity (3xTg-AD-dnPAK mice). PAK inactivation had no effect on Aß40 and Aß42 levels, but increased the phosphorylation ratio of tau in detergent-insoluble protein fractions in the frontal cortex of 18-month-old heterozygous 3xTg-AD mice. Morphometric analyses of layer II/III pyramidal neurons in the frontal cortex showed that 3xTg-AD-dnPAK neurons exhibited significant dendritic attrition, lower spine density and longer spines compared to NonTg and 3xTg-AD mice. Finally, behavioral assessments revealed that 3xTg-AD-dnPAK mice exhibited pronounced anxious traits and disturbances in social behaviors, reminiscent of fronto-dependent symptoms observed in AD. Our results substantiate a critical role for PAK in the genesis of neuronal abnormalities in the frontal cortex underlying the emergence of psychiatric-like symptoms in AD.


Subject(s)
Alzheimer Disease/enzymology , Behavior, Animal , Frontal Lobe/enzymology , Pyramidal Cells/enzymology , p21-Activated Kinases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Anxiety/enzymology , Anxiety/psychology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Disease Models, Animal , Exploratory Behavior , Frontal Lobe/pathology , Frontal Lobe/physiopathology , Genetic Predisposition to Disease , Interpersonal Relations , Locomotion , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/metabolism , Phenotype , Phosphorylation , Presenilin-1/genetics , Promoter Regions, Genetic , Pyramidal Cells/pathology , Synaptic Transmission , p21-Activated Kinases/genetics , tau Proteins/genetics , tau Proteins/metabolism
9.
Acta Neuropathol ; 133(1): 101-119, 2017 01.
Article in English | MEDLINE | ID: mdl-27752775

ABSTRACT

Soluble oligomers of amyloid-ß (Aß) impair synaptic plasticity, perturb neuronal energy homeostasis, and are implicated in Alzheimer's disease (AD) pathogenesis. Therefore, significant efforts in AD drug discovery research aim to prevent the formation of Aß oligomers or block their neurotoxicity. The eukaryotic elongation factor-2 kinase (eEF2K) plays a critical role in synaptic plasticity, and couples neurotransmission to local dendritic mRNA translation. Recent evidence indicates that Aß oligomers activate neuronal eEF2K, suggesting a potential link to Aß induced synaptic dysfunction. However, a detailed understanding of the role of eEF2K in AD pathogenesis, and therapeutic potential of eEF2K inhibition in AD, remain to be determined. Here, we show that eEF2K activity is increased in postmortem AD patient cortex and hippocampus, and in the hippocampus of aged transgenic AD mice. Furthermore, eEF2K inhibition using pharmacological or genetic approaches prevented the toxic effects of Aß42 oligomers on neuronal viability and dendrite formation in vitro. We also report that eEF2K inhibition promotes the nuclear factor erythroid 2-related factor (NRF2) antioxidant response in neuronal cells, which was crucial for the beneficial effects of eEF2K inhibition in neurons exposed to Aß42 oligomers. Accordingly, NRF2 knockdown or overexpression of the NRF2 inhibitor, Kelch-Like ECH-Associated Protein-1 (Keap1), significantly attenuated the neuroprotection associated with eEF2K inhibition. Finally, genetic deletion of the eEF2K ortholog efk-1 reduced oxidative stress, and improved chemotaxis and serotonin sensitivity in C. elegans expressing human Aß42 in neurons. Taken together, these findings highlight the potential utility of eEF2K inhibition to reduce Aß-mediated oxidative stress in AD.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Elongation Factor 2 Kinase/deficiency , Peptide Fragments/metabolism , Alzheimer Disease/enzymology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Elongation Factor 2 Kinase/antagonists & inhibitors , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/metabolism , Enzyme Inhibitors/pharmacology , Female , Frontal Lobe/drug effects , Frontal Lobe/enzymology , Hippocampus/drug effects , Hippocampus/enzymology , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Male , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptide Fragments/genetics , Peptide Fragments/toxicity , Reactive Oxygen Species
10.
J Alzheimers Dis ; 55(1): 115-135, 2017.
Article in English | MEDLINE | ID: mdl-27662290

ABSTRACT

No effective preventive treatment is available for age-related cognitive decline and Alzheimer's disease (AD). Epidemiological studies indicate that a diet rich in fruit is associated with cognitive improvement. It was thus proposed that high polyphenol concentrations found in berries can prevent cognitive impairment associated with aging and AD. Therefore, the Neurophenols project aimed at investigating the effects of a polyphenolic extract from blueberries and grapes (PEBG) in the triple-transgenic (3xTg-AD) mouse model of AD, which develops AD neuropathological markers, including amyloid-ß plaques and neurofibrillary tangles, leading to memory deficits. In this study, 12-month-old 3xTg-AD and NonTg mice were fed a diet supplemented with standardized PEBG (500 or 2500 mg/kg) for 4 months (n = 15-20/group). A cognitive evaluation with the novel object recognition test was performed at 15 months of age and mice were sacrificed at 16 months of age. We observed that PEBG supplementation with doses of 500 or 2500 mg/kg prevented the decrease in novel object recognition observed in both 15-month-old 3xTg-AD mice and NonTg mice fed a control diet. Although PEBG treatment did not reduce Aß and tau pathologies, it prevented the decrease in mature BDNF observed in 16-month-old 3xTg-AD mice. Finally, plasma concentrations of phenolic metabolites, such as dihydroxyphenyl valerolactone, a microbial metabolite of epicatechin, positively correlated with memory performances in supplemented mice. The improvement in object recognition observed in 3xTg-AD mice after PEBG administration supports the consumption of polyphenols-rich extracts to prevent memory impairment associated with age-related disease, without significant effects on classical AD neuropathology.


Subject(s)
Alzheimer Disease/drug therapy , Blueberry Plants , Nootropic Agents/pharmacology , Plant Extracts/pharmacology , Polyphenols/pharmacology , Vitis , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fruit , Humans , Male , Mice, Transgenic , Presenilin-1/genetics , Presenilin-1/metabolism , Recognition, Psychology/drug effects , tau Proteins/genetics , tau Proteins/metabolism
11.
Food Funct ; 7(8): 3421-30, 2016 Aug 10.
Article in English | MEDLINE | ID: mdl-27443888

ABSTRACT

The low bioavailability of dietary phenolic compounds, resulting from poor absorption and high rates of metabolism and excretion, is a concern as it can limit their potential beneficial effects on health. Targeted metabolomic profiling in plasma and feces of mice supplemented for 15 days with a blueberry extract, a grape extract or their combination revealed significantly increased plasma concentrations (3-5 fold) of blueberry phenolic metabolites in the presence of a co-ingested grape extract, associated with an equivalent decrease in their appearance in feces. Additionally, the repeated daily administration of the blueberry-grape combination significantly increased plasma phenolic concentrations (2-3-fold) compared to animals receiving only a single acute dose, with no such increase being observed with individual extracts. These findings highlight a positive interaction between blueberry and grape constituents, in which the grape extract enhanced the absorption of blueberry phenolic compounds. This study provides for the first time in vivo evidence of such an interaction occurring between co-ingested phenolic compounds from fruit extracts leading to their improved bioavailability.


Subject(s)
Blueberry Plants/chemistry , Feces/chemistry , Phenol/blood , Phenol/pharmacokinetics , Animals , Biological Availability , Dietary Supplements , Grape Seed Extract/blood , Grape Seed Extract/pharmacokinetics , Male , Metabolomics , Mice , Mice, Inbred C57BL , Phytochemicals/blood , Phytochemicals/pharmacokinetics , Vitis/chemistry
12.
Article in English | MEDLINE | ID: mdl-25457546

ABSTRACT

A brief overview of the evidence for omega-3 fatty acids and, in particular, of docosahexaenoic acid (DHA), involvement in cognition and in dementia is given. Two studies are presented in this regard in which the key intervention is a DHA supplement. The fist, the MIDAS Study demonstrated that DHA can be of benefit for episodic memory in healthy adults with a mild memory complaint. The second, the ADCS AD trial found no benefit of DHA in the primary outcomes but found an intriguing benefit for cognitive score in ApoE4 negative allele patients. This leads to a consideration of the mechanisms of action and role of ApoE and its modulation by DHA. Given the fundamental role of ApoE in cellular lipid transport and metabolism in the brain and periphery, it is no surprise that ApoE affects n-3 PUFA brain function as well. It remains to be seen to what extent ApoE4 deleterious effect in AD is associated with n-3 PUFA-related cellular mechanisms in the brain and, more specifically, whether ApoE4 directly impairs the transport of DHA into the brain, as has been suggested.


Subject(s)
Aging , Brain/physiopathology , Dementia/drug therapy , Docosahexaenoic Acids/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Brain/drug effects , Cognition/drug effects , Docosahexaenoic Acids/pharmacology , Humans , Neuroprotective Agents/pharmacology , Randomized Controlled Trials as Topic
13.
J Neurochem ; 122(2): 427-43, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22551210

ABSTRACT

Dietary lipids modify brain fatty acid profile, but evidence of their direct effect on neuronal function is sparse. The enthorinal cortex (EC) neurons connecting to the hippocampus play a critical role in learning and memory. Here, we have exposed mice to diets based on canola:soybean oils (40 : 10, g/kg) or safflower : corn oils (25 : 25, g/kg) to investigate the relationship between the lipid profile of brain fatty acids and the intrinsic properties of EC neurons. Consumption of canola : soybean oil-enriched diet led to the increase of the monounsaturated fatty acid oleic acid and to a decrease of arachidonic acid in ethanolamine glycerophospholipids of the white matter. We also found an important rise in docosahexaenoic acid (DHA) within ethanolamine glycerophospholipids and phosphatidylserine of gray matter. The canola:soybean oil treatment led to a shorter duration of action potential (-21%), a reduction in the duration of postsynaptic response (-21%) and increased firing activity (+43%). Data from additional experiments with animals fed DHA alone or DHA with canola oil suggested that dietary monounsaturated fatty acid may have contributed to these effects on EC neuron physiology. Since neuronal function within the enthorhinal-hippocampal loop is critical to learning and memory processes, the present data may provide a functional basis for the beneficial cognitive effects of canola oil-based diets.


Subject(s)
Diet , Entorhinal Cortex/cytology , Entorhinal Cortex/drug effects , Fatty Acids, Unsaturated/pharmacology , Neurons/drug effects , Action Potentials/drug effects , Animals , Brain Chemistry/drug effects , Data Interpretation, Statistical , Excitatory Postsynaptic Potentials/drug effects , Fatty Acids, Monounsaturated/analysis , Fatty Acids, Monounsaturated/pharmacology , Flame Ionization , Lipids/analysis , Mice , Mice, Inbred C57BL , Nerve Net/drug effects , Patch-Clamp Techniques , Phospholipids/analysis , Prefrontal Cortex/chemistry , Rapeseed Oil , Soybean Oil/analysis
14.
J Alzheimers Dis ; 27(4): 853-69, 2011.
Article in English | MEDLINE | ID: mdl-21914946

ABSTRACT

Dietary supplementation with n-3 polyunsaturated fatty acids (n-3 PUFA) reduces amyloid-ß (Aß) and tau pathology and improves cognitive performance in animal models of Alzheimer's disease (AD). To exclude confounding variables associated with the diet, we crossed 3 × Tg-AD mice (modeling AD neuropathology) with transgenic Fat-1 mice that express the fat-1 gene encoding a PUFA desaturase, which endogenously produces n-3 PUFA from n-6 PUFA. The expression of fat-1 shifted the n-3:n-6 PUFA ratio upward in the brain (+11%, p < 0.001), including docosahexaenoic acid (DHA; +5%, p < 0.001) in 20 month-old mice. The expression of fat-1 decreased the levels of soluble Aß42 (-41%, p < 0.01) at 20 months without reducing the level of insoluble forms of Aß40 and Aß42 in the brain of 3 × Tg-AD mice. The 3 × Tg-AD/Fat-1 mice exhibited lower cortical levels of both soluble (-25%, p < 0.05) and insoluble phosphorylated tau (-55%, p < 0.05) compared to 3 × Tg-AD mice, but only in 20 month-old animals. Whereas a decrease of calcium/calmodulin-dependent protein kinase II was observed in 3 × Tg-AD/Fat-1 mice (-039%, p < 0.05), altered tau phosphorylation could not be related to changes in glycogen synthase kinase 3ß, cyclin-dependent kinase 5, or protein phosphatase type 2A enzymatic activity. In addition, the expression of the fat-1 transgene prevented the increase of glial fibrillary acidic protein (-37%, p < 0.01) observed in 20 month-old 3 × Tg-AD mice. In conclusion, the expression of fat-1 in 3 × Tg-AD mice increases brain DHA and induces biomarker changes that are consistent with a beneficial effect against an AD-like neuropathology.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cofilin 1/metabolism , Cyclin-Dependent Kinase 5 , Disease Models, Animal , Docosahexaenoic Acids , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Presenilin-1/genetics , Presenilin-1/metabolism , Signal Transduction/genetics , Statistics as Topic , tau Proteins/genetics , tau Proteins/metabolism
15.
Stroke ; 42(10): 2903-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21852616

ABSTRACT

BACKGROUND AND PURPOSE: Consumption of fish has been shown to reduce risk of coronary heart disease and, possibly, of ischemic stroke. Because docosahexaenoic acid (DHA) is the most likely neuroactive component within fish oil, we hypothesized that exposing mice to a DHA-enriched diet may reduce inflammation and protect neurons against ischemic injury. METHODS: To visualize the effects of DHA on neuroinflammation after stroke, TLR2-fluc-GFP transgenic mice were exposed to either a control diet, a diet depleted in n-3 polyunsaturated fatty acid, or a diet enriched in DHA during 3 months. Real-time biophotonic/bioluminescence imaging of the TLR2 response was performed before and after middle cerebral artery occlusion, whereas cytokines concentrations and stroke area analyses were performed at 3 and 7 days after middle cerebral artery occlusion, respectively. RESULTS: We show that a 3-month DHA treatment prevented microglial activation after ischemic injury, reduced the ischemic lesion size, and increased levels of the antiapoptotic molecule Bcl-2 in the brain. Additional analysis revealed a significant decrease in the levels of COX2 and IL-1ß, but not in other proinflammatory cytokines. Importantly, long-term DHA supplementation significantly changed the n-3:n-6 polyunsaturated fatty acid ratio in the brain. CONCLUSIONS: Collectively, these data indicate that diet-induced accumulation of DHA in the brain protects against postischemic inflammation and injury. Because DHA is widely available at low cost and has an excellent safety profile, our data suggest that increased DHA intake may provide protection against acute immune response/brain damage in ischemic stroke.


Subject(s)
Brain Ischemia/immunology , Brain/immunology , Docosahexaenoic Acids/administration & dosage , Immunity, Active/immunology , Neurons/immunology , Animals , Brain/drug effects , Brain/metabolism , Brain Ischemia/metabolism , Docosahexaenoic Acids/metabolism , Inflammation/immunology , Mice , Mice, Transgenic , Neurons/metabolism , Toll-Like Receptor 2/metabolism
16.
Ageing Res Rev ; 10(4): 453-63, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21414422

ABSTRACT

Current epidemiological, preclinical and clinical data suggest that omega-3 polyunsaturated fatty acids (n-3 PUFAs) may constitute therapeutic strategy for several disorders of the central nervous system, including Parkinson's disease (PD). PD is a neurodegenerative disorder primarily characterized by motor symptoms but which also includes several other pathological features such as autonomic system failures, mood disorders, and cognitive deficits. Current pharmacological options for the disease are limited to symptom management and their long-term use leads to important side effects. In this review, we discuss the evidence for the effects of n-3 PUFAs in PD both from an epidemiological perspective as well as in light of data gathered on various pathological features of the disease. Effects of n-3 PUFAs on the dopaminergic system, α-synucleinopathy, their possible mechanisms of action as well as their therapeutic potential for PD patients are also reviewed. n-3 PUFAs are inexpensive, readily transferable to the clinical setting and their use could represent a neuroprotective strategy or a disease-modifying option to delay the appearance of symptoms. It could also be beneficial as a symptomatologic treatment or serve as an add-on therapy to current pharmacological approaches. Review of the current literature as well as the undertaking of future clinical trials will shed light on these possibilities.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/diet therapy , Parkinson Disease/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Fatty Acids, Omega-3/pharmacology , Humans , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Parkinson Disease/pathology , Reactive Oxygen Species/metabolism
17.
PLoS One ; 6(2): e17397, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21383850

ABSTRACT

Defects in neuronal activity of the entorhinal cortex (EC) are suspected to underlie the symptoms of Alzheimer's disease (AD). Whereas neuroprotective effects of docosahexaenoic acid (DHA) have been described, the effects of DHA on the physiology of EC neurons remain unexplored in animal models of AD. Here, we show that DHA consumption improved object recognition (↑12%), preventing deficits observed in old 3xTg-AD mice (↓12%). Moreover, 3xTg-AD mice displayed seizure-like akinetic episodes, not detected in NonTg littermates and partly prevented by DHA (↓50%). Patch-clamp recording revealed that 3xTg-AD EC neurons displayed (i) loss of cell capacitance (CC), suggesting reduced membrane surface area; (ii) increase of firing rate versus injected current (F-I) curve associated with modified action potentials, and (iii) overactivation of glutamatergic synapses, without changes in synaptophysin levels. DHA consumption increased CC (↑12%) and decreased F-I slopes (↓21%), thereby preventing the opposite alterations observed in 3xTg-AD mice. Our results indicate that cognitive performance and basic physiology of EC neurons depend on DHA intake in a mouse model of AD.


Subject(s)
Alzheimer Disease/physiopathology , Cognition Disorders/prevention & control , Cognition/drug effects , Docosahexaenoic Acids/pharmacology , Entorhinal Cortex/drug effects , Neurons/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Animals , Arachidonic Acid/analysis , Arachidonic Acid/metabolism , Brain Chemistry , Cognition Disorders/genetics , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Cytoprotection/drug effects , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Entorhinal Cortex/chemistry , Entorhinal Cortex/metabolism , Entorhinal Cortex/physiology , Mice , Mice, Transgenic , Neurons/chemistry , Neurons/metabolism , Neurons/physiology , Osmolar Concentration
18.
J Lipid Res ; 52(2): 263-71, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21115966

ABSTRACT

We have recently identified a neuroprotective role for omega-3 polyunsaturated fatty acids (n-3 PUFAs) in a toxin-induced mouse model of Parkinson's disease (PD). Combined with epidemiological data, these observations suggest that low n-3 PUFA intake is a modifiable environmental risk factor for PD. In order to strengthen these preclinical findings as prerequisite to clinical trials, we further investigated the neuroprotective role of n-3 PUFAs in Fat-1 mice, a transgenic model expressing an n-3 fatty acid desaturase converting n-6 PUFAs into n-3 PUFAs. Here, we report that the expression of the fat-1 transgene increased cortical n-3:n-6 PUFA ratio (+28%), but to a lesser extent than dietary supplementation (92%). Such a limited endogenous production of n-3 PUFAs in the Fat-1 mouse was insufficient to confer neuroprotection against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity as assessed by dopamine levels, tyrosine hydroxylase (TH)-positive neurons and fibers, as well as nigral Nurr1 and dopamine transporter (DAT) mRNA expression. Nevertheless, higher cortical docosahexaenoic acid (DHA) concentrations were positively correlated with markers of nigral dopaminergic neurons such as the number of TH-positive cells, in addition to Nurr1 and DAT mRNA levels. These associations are consistent with the protective role of DHA in a mouse model of PD. Taken together, these data suggest that dietary intake of a preformed DHA supplement is more effective in reaching the brain and achieving neuroprotection in an animal model of PD.


Subject(s)
Parkinson Disease, Secondary/metabolism , Animals , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Fatty Acids, Omega-3/metabolism , Mice , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/chemically induced , Transgenes
19.
Neurobiol Aging ; 31(9): 1516-31, 2010 Sep.
Article in English | MEDLINE | ID: mdl-18926603

ABSTRACT

To investigate potential dietary risk factors of Alzheimer's disease (AD), triple transgenic (3xTg-AD) mice were exposed from 4 to 13 months of age to diets with a low n-3:n-6 polyunsaturated fatty acid (PUFA) ratio incorporated in either low-fat (5% w/w) or high-fat (35% w/w) formulas and compared with a control diet. The n-3:n-6 PUFA ratio was decreased independently of the dietary treatments in the frontal cortex of 3xTg-AD mice compared to non-transgenic littermates. Consumption of a high-fat diet with a low n-3:n-6 PUFA ratio increased amyloid-beta (Abeta) 40 and 42 concentrations in detergent-insoluble extracts of parieto-temporal cortex homogenates from 3xTg-AD mice. Low n-3:n-6 PUFA intake ratio increased insoluble tau regardless of total fat consumption, whereas high-fat diet incorporating a low n-3:n-6 PUFA ratio also increased soluble tau compared to controls. Moreover, the high-fat diet decreased cortical levels of the postsynaptic marker drebrin, while leaving presynaptic proteins synaptophysin, SNAP-25 and syntaxin 3 unchanged. Overall, these results suggest that high-fat consumption combined with low n-3 PUFA intake promote AD-like neuropathology.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Dietary Fats/pharmacology , Disease Models, Animal , Fatty Acids, Omega-3/pharmacology , tau Proteins/metabolism , Alzheimer Disease/chemically induced , Animals , Brain/drug effects , Dietary Fats/adverse effects , Dietary Fats/metabolism , Fatty Acids, Omega-3/adverse effects , Humans , Mice , Mice, Transgenic
20.
Article in English | MEDLINE | ID: mdl-18037281

ABSTRACT

Studies in animals clearly show that oral intake of docosahexaenoic acid (DHA) can alter brain DHA concentrations and thereby modify brain functions. This provides us with an opportunity to use DHA as a nutraceutical or pharmaceutical tool in brain disorders such as Alzheimer disease (AD) and Parkinson disease (PD). Most of the published epidemiological studies are consistent with a positive association between high reported DHA consumption or high DHA blood levels and a lower risk of developing AD later in life. Such observations have prompted the investigation of DHA in three different transgenic models of AD. These analyses show that animal models of AD are more vulnerable to DHA depletion than controls and that DHA exerts a beneficial effect against pathological signs of AD, including A beta accumulation, cognitive impairment, synaptic marker loss, and hyperphosphorylation of tau. Multiple mechanisms of action can be associated with the neuroprotective effects of DHA and include antioxidant properties and activation of distinct cell signaling pathways. Although the first randomized clinical assays have yet failed to demonstrate convincing beneficial effects of DHA for AD patients, the knowledge gathered in recent years holds out a hope for prevention and suggests that the elderly and people bearing a genetic risk for AD should at least avoid DHA deficiency.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/pharmacology , Fatty Acids, Omega-3/administration & dosage , Humans , Models, Biological , Neurodegenerative Diseases/pathology , Neuroprotective Agents/administration & dosage , Parkinson Disease/drug therapy , Parkinson Disease/pathology
SELECTION OF CITATIONS
SEARCH DETAIL