Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Ageing Res Rev ; 62: 101079, 2020 09.
Article in English | MEDLINE | ID: mdl-32461136

ABSTRACT

The global increases in life expectancy and population have resulted in a growing ageing population and with it a growing number of people living with age-related neurodegenerative conditions and dementia, shifting focus towards methods of prevention, with lifestyle approaches such as nutrition representing a promising avenue for further development. This overview summarises the main themes discussed during the 3rd Symposium on "Nutrition for the Ageing Brain: Moving Towards Clinical Applications" held in Madrid in August 2018, enlarged with the current state of knowledge on how nutrition influences healthy ageing and gives recommendations regarding how the critical field of nutrition and neurodegeneration research should move forward into the future. Specific nutrients are discussed as well as the impact of multi-nutrient and whole diet approaches, showing particular promise to combatting the growing burden of age-related cognitive decline. The emergence of new avenues for exploring the role of diet in healthy ageing, such as the impact of the gut microbiome and development of new techniques (imaging measures of brain metabolism, metabolomics, biomarkers) are enabling researchers to approach finding answers to these questions. But the translation of these findings into clinical and public health contexts remains an obstacle due to significant shortcomings in nutrition research or pressure on the scientific community to communicate recommendations to the general public in a convincing and accessible way. Some promising programs exist but further investigation to improve our understanding of the mechanisms by which nutrition can improve brain health across the human lifespan is still required.


Subject(s)
Healthy Aging , Nutritional Status , Aging , Brain , Diet , Humans
2.
J Neurotrauma ; 36(1): 25-42, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29768974

ABSTRACT

Traumatic brain injury (TBI) leads to cellular loss, destabilization of membranes, disruption of synapses and altered brain connectivity, and increased risk of neurodegenerative disease. A significant and long-lasting decrease in phospholipids (PLs), essential membrane constituents, has recently been reported in plasma and brain tissue, in human and experimental TBI. We hypothesized that supporting PL synthesis post-injury could improve outcome post-TBI. We tested this hypothesis using a multi-nutrient combination designed to support the biosynthesis of PLs and available for clinical use. The multi-nutrient, Fortasyn® Connect (FC), contains polyunsaturated omega-3 fatty acids, choline, uridine, vitamins, cofactors required for PL biosynthesis, and has been shown to have significant beneficial effects in early Alzheimer's disease. Male C57BL/6 mice received a controlled cortical impact injury and then were fed a control diet or a diet enriched with FC for 70 days. FC led to a significantly improved sensorimotor outcome and cognition, reduced lesion size and oligodendrocyte loss, and it restored myelin. It reversed the loss of the synaptic protein synaptophysin and decreased levels of the axon growth inhibitor, Nogo-A, thus creating a permissive environment. It decreased microglia activation and the rise in ß-amyloid precursor protein and restored the depressed neurogenesis. The effects of this medical multi-nutrient suggest that support of PL biosynthesis post-TBI, a new treatment paradigm, has significant therapeutic potential in this neurological condition for which there is no satisfactory treatment. The multi-nutrient tested has been used in dementia patients and is safe and well tolerated, which would enable rapid clinical exploration in TBI.


Subject(s)
Brain Injuries, Traumatic/pathology , Brain/pathology , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Phospholipids/pharmacology , Recovery of Function , Animals , Disease Models, Animal , Male , Mice, Inbred C57BL
3.
Alzheimers Dement (N Y) ; 3(3): 416-431, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29067348

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) patients are at risk of nutritional insufficiencies because of physiological and psychological factors. Recently, we showed the results of the meta-analyses indicating lower plasma levels of vitamins A, B12, C, E, and folate in AD patients compared with cognitively intact elderly controls (controls). Now, additional and more extensive literature searches were performed selecting studies which compare blood and brain/cerebrospinal fluid (CSF) levels of vitamins, minerals, trace elements, micronutrients, and fatty acids in AD patients versus controls. METHODS: The literature published after 1980 in Cochrane Central Register of Controlled Trials, Medline, and Embase electronic databases was systematically analyzed using Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines to detect studies meeting the selection criteria. Search terms used are as follows: AD patients, Controls, vitamins, minerals, trace elements, micronutrients, and fatty acids. Random-effects meta-analyses using a linear mixed model with correction for age differences between AD patients and controls were performed when four or more publications were retrieved for a specific nutrient. RESULTS: Random-effects meta-analyses of 116 selected publications showed significant lower CSF/brain levels of docosahexaenoic acid (DHA), choline-containing lipids, folate, vitamin B12, vitamin C, and vitamin E. In addition, AD patients showed lower circulatory levels of DHA, eicosapentaenoic acid, choline as phosphatidylcholine, and selenium. CONCLUSION: The current data show that patients with AD have lower CSF/brain availability of DHA, choline, vitamin B12, folate, vitamin C, and vitamin E. Directionally, brain nutrient status appears to parallel the lower circulatory nutrient status; however, more studies are required measuring simultaneously circulatory and central nutrient status to obtain better insight in this observation. The brain is dependent on nutrient supply from the circulation, which in combination with nutrient involvement in AD-pathophysiological mechanisms suggests that patients with AD may have specific nutritional requirements. This hypothesis could be tested using a multicomponent nutritional intervention.

4.
Nutrients ; 8(4): 185, 2016 Mar 26.
Article in English | MEDLINE | ID: mdl-27102170

ABSTRACT

The inherited metabolic disease phenylketonuria (PKU) is characterized by increased concentrations of phenylalanine in the blood and brain, and as a consequence neurotransmitter metabolism, white matter, and synapse functioning are affected. A specific nutrient combination (SNC) has been shown to improve synapse formation, morphology and function. This could become an interesting new nutritional approach for PKU. To assess whether treatment with SNC can affect synapses, we treated PKU mice with SNC or an isocaloric control diet and wild-type (WT) mice with an isocaloric control for 12 weeks, starting at postnatal day 31. Immunostaining for post-synaptic density protein 95 (PSD-95), a post-synaptic density marker, was carried out in the hippocampus, striatum and prefrontal cortex. Compared to WT mice on normal chow without SNC, PKU mice on the isocaloric control showed a significant reduction in PSD-95 expression in the hippocampus, specifically in the granular cell layer of the dentate gyrus, with a similar trend seen in the cornus ammonis 1 (CA1) and cornus ammonis 3 (CA3) pyramidal cell layer. No differences were found in the striatum or prefrontal cortex. PKU mice on a diet supplemented with SNC showed improved expression of PSD-95 in the hippocampus. This study gives the first indication that SNC supplementation has a positive effect on hippocampal synaptic deficits in PKU mice.


Subject(s)
Animal Feed/analysis , Gene Expression Regulation/drug effects , Guanylate Kinases/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Phenylalanine/administration & dosage , Phenylketonurias/metabolism , Animals , Diet , Disks Large Homolog 4 Protein , Female , Genotype , Guanylate Kinases/genetics , Male , Membrane Proteins/genetics , Mice , Random Allocation
5.
Alzheimers Dement ; 12(6): 633-44, 2016 06.
Article in English | MEDLINE | ID: mdl-26776762

ABSTRACT

INTRODUCTION: Loss of synapses best correlates to cognitive deficits in Alzheimer's disease (AD) in which oligomeric neurotoxic species of amyloid-ß appears to contribute synaptic pathology. Although a number of clinical pathologic studies have been performed with limited sample size, there are no systematic studies encompassing large samples. Therefore, we performed a meta-analysis study. METHODS: We identified 417 publications reporting postmortem synapse and synaptic marker loss from AD patients. Two meta-analyses were performed using a single database of subselected publications and calculating the standard mean differences. RESULTS: Meta-analysis confirmed synaptic loss in selected brain regions is an early event in AD pathogenesis. The second meta-analysis of 57 synaptic markers revealed that presynaptic makers were affected more than postsynaptic markers. DISCUSSION: The present meta-analysis study showed a consistent synaptic loss across brain regions and that molecular machinery including endosomal pathways, vesicular assembly mechanisms, glutamate receptors, and axonal transport are often affected.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Synapses/metabolism , Vesicular Transport Proteins/metabolism , Clinical Trials as Topic/statistics & numerical data , Diagnosis , Humans , MEDLINE/statistics & numerical data , Synapses/pathology
6.
J Alzheimers Dis ; 38(3): 459-79, 2014.
Article in English | MEDLINE | ID: mdl-23985420

ABSTRACT

Synapse loss and synaptic dysfunction are pathological processes already involved in the early stages of Alzheimer's disease (AD). Synapses consist principally of neuronal membranes, and the neuronal and synaptic losses observed in AD have been linked to the degeneration and altered composition and structure of these membranes. Consequently, synapse loss and membrane-related pathology provide viable targets for intervention in AD. The specific nutrient combination Fortasyn Connect (FC) is designed to ameliorate synapse loss and synaptic dysfunction in AD by addressing distinct nutritional needs believed to be present in these patients. This nutrient combination comprises uridine, docosahexaenoic acid, eicosapentaenoic acid, choline, phospholipids, folic acid, vitamins B12, B6, C, and E, and selenium, and is present in Souvenaid, a medical food intended for use in early AD. It has been hypothesized that FC counteracts synaptic loss and reduces membrane-related pathology in AD by providing nutritional precursors and cofactors that act together to support neuronal membrane formation and function. Preclinical studies formed the basis of this hypothesis which is being validated in a broad clinical study program investigating the potential of this nutrient combination in AD. Memory dysfunction is one key early manifestation in AD and is associated with synapse loss. The clinical studies to date show that the FC-containing medical food improves memory function and preserves functional brain network organization in mild AD compared with controls, supporting the hypothesis that this intervention counteracts synaptic dysfunction. This review provides a comprehensive overview of basic scientific studies that led to the creation of FC and of its effects in various preclinical models.


Subject(s)
Alzheimer Disease/diet therapy , Alzheimer Disease/pathology , Brain/pathology , Dietary Supplements , Synapses/physiology , Animals , Humans , Nutritional Status , Synapses/pathology
7.
J Alzheimers Dis ; 33(1): 177-90, 2013.
Article in English | MEDLINE | ID: mdl-22914588

ABSTRACT

Diet is an important lifestyle factor implicated in the etiology of Alzheimer's disease (AD), but so far it is not fully elucidated to which nutrients the suggested protective effect of diet can be attributed. Recent evidence obtained in the amyloid-ß 1-42 (Aß(42)) infusion model in rats has shown that a multi-nutrient intervention known as Fortasyn™ Connect (FC) may protect the central cholinergic system against Aß(42)-induced toxicity. FC comprises the nutritional precursors and cofactors for membrane synthesis, viz. docosahexaenoic acid (DHA), eicosapentaenoic acid, uridine-mono-phosphate (UMP), choline, phospholipids, folic acid, vitamins B6, B12, C, E, and selenium. In order to investigate whether the combined administration of these nutrients may also affect AD-like pathology, we now evaluated the effects of the FC diet intervention in the transgenic AßPP(swe)/PS1(dE9) mouse model with endogenous Aß production. In addition we evaluated the effects of diets containing the individual nutrients DHA and UMP and their combination in this model. Between the age of 3 and 6 months, FC diet decreased brain Aß levels and amyloid plaque burden in the hippocampus of AßPP/PS1 mice. The FC diet also reduced ongoing disintegrative degeneration in the neocortex, as indicated by Amino Cupric Silver staining. Although all three DHA-containing diets were equally effective in changing brain fatty acid profiles, diets differentially affected amyloid-related measures, indicating that effects of DHA may depend on its dietary context. The current data, showing that dietary enrichment with FC reduces AD-like pathology in AßPP/PS1 mice, confirm and extend our previous findings in the Aß(42) infusion model and favor the combined administration of relevant nutrients.


Subject(s)
Alzheimer Disease/diet therapy , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Diet/methods , Food , Presenilin-1/genetics , Alzheimer Disease/genetics , Animals , Female , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic
8.
J Alzheimers Dis ; 27(2): 327-39, 2011.
Article in English | MEDLINE | ID: mdl-21811020

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia in the elderly. Substantial evidence suggests a role for nutrition in the management of AD and especially suggests that interventions with combinations of nutrients are more effective than single-nutrient interventions. The specific multi-nutrient combination Fortasyn™Connect (FC), shown to improve memory in AD, provides phosphatide precursors and cofactors and is designed to stimulate the formation of phospholipids, neuronal membranes, and synapses. The composition comprises nucleotides, omega-3 polyunsaturated fatty acids (n3 PUFA), choline, B-vitamins, phospholipids, and antioxidants. The current study explored the protective properties of FC in a membrane toxicity model of AD, the amyloid-ß 1-42 (Aß42) infused rat, which shows reduced exploratory behavior in an Open Field and impaired cholinergic functioning. To this end, rats were fed an FC enriched diet or a control diet and five weeks later infused with vehicle or Aß42 into the lateral ventricle. Ten weeks post-infusion Aß42-rats fed the FC diet showed increased membrane n3 PUFA and phosphatidylcholine content while they did not show the reductions in exploratory behavior or in choline acetyltransferase (ChAT) and vesicular acetylcholine transporter (VAChT) immunoreactivity that were seen in Aß42-rats fed the control diet. We conclude that FC protects the cholinergic system against Aß42-induced toxicity and speculate that the effects of FC on membrane formation and composition might be supportive for this protective effect. Based on these data a long-term intervention study was started in the prodromal stages of AD (NTR1705, LipiDiDiet, EU FP7).


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Food, Fortified , Neuroprotective Agents/administration & dosage , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/toxicity , Alzheimer Disease/etiology , Alzheimer Disease/physiopathology , Alzheimer Disease/prevention & control , Animals , Food , Male , Rats , Rats, Sprague-Dawley
9.
Eur J Pharmacol ; 668 Suppl 1: S59-69, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21816137

ABSTRACT

Alzheimer's disease (AD) is a multi-factorial neurodegenerative disorder and the leading cause of dementia, wherein synapse loss is the strongest structural correlate with cognitive impairment. Basic research has shown that dietary supply of precursors and co-factors for synthesis of neuronal membranes enhances the formation of synapses. Daily intake of a medical food containing a mix of these nutrients for 12 weeks in humans improved memory, measured as immediate and delayed verbal recall by the Wechsler Memory Scale-revised, in patients with very mild AD (MMSE 24-26). An improvement of immediate verbal recall was noted following 24 weeks of intervention in an exploratory extension of the study. These data suggest that the intervention may improve synaptic formation and function in early AD. Here we review emerging technologies that help identify changes in pathological hallmarks in AD, including synaptic function and loss of connectivity in the early stages of AD, before cognitive and behavioural symptoms are observable. These techniques include the detection of specific biomarkers in the cerebrospinal fluid, as well as imaging procedures such as fluorine-18 fluorodeoxyglucose positron emission tomography (FDG-PET), amyloid PET, structural/functional magnetic resonance imaging, diffusion tensor imaging, magnetoencephalography (MEG) and electroencephalography (EEG). Such techniques can provide new insights into the functional and structural changes in the brain over time, and may therefore help to develop more effective AD therapies. In particular, nutritional intervention studies that target synapse formation and function may benefit from these techniques, especially FDG-PET and EEG/MEG employed in the preclinical or early stages of the disease.


Subject(s)
Alzheimer Disease/diet therapy , Alzheimer Disease/diagnosis , Neuroimaging/methods , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/metabolism , Animals , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Functional Neuroimaging , Humans
10.
J Biol Chem ; 286(16): 14028-39, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21324907

ABSTRACT

Alzheimer disease is characterized by accumulation of the ß-amyloid peptide (Aß) generated by ß- and γ-secretase processing of the amyloid precursor protein (APP). The intake of the polyunsaturated fatty acid docosahexaenoic acid (DHA) has been associated with decreased amyloid deposition and a reduced risk in Alzheimer disease in several epidemiological trials; however, the exact underlying molecular mechanism remains to be elucidated. Here, we systematically investigate the effect of DHA on amyloidogenic and nonamyloidogenic APP processing and the potential cross-links to cholesterol metabolism in vivo and in vitro. DHA reduces amyloidogenic processing by decreasing ß- and γ-secretase activity, whereas the expression and protein levels of BACE1 and presenilin1 remain unchanged. In addition, DHA increases protein stability of α-secretase resulting in increased nonamyloidogenic processing. Besides the known effect of DHA to decrease cholesterol de novo synthesis, we found cholesterol distribution in plasma membrane to be altered. In the presence of DHA, cholesterol shifts from raft to non-raft domains, and this is accompanied by a shift in γ-secretase activity and presenilin1 protein levels. Taken together, DHA directs amyloidogenic processing of APP toward nonamyloidogenic processing, effectively reducing Aß release. DHA has a typical pleiotropic effect; DHA-mediated Aß reduction is not the consequence of a single major mechanism but is the result of combined multiple effects.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Docosahexaenoic Acids/pharmacology , ADAM Proteins/metabolism , ADAM17 Protein , Amyloid Precursor Protein Secretases/metabolism , Animal Feed , Animals , Aspartic Acid Endopeptidases/metabolism , Cell Line , Cell Membrane/metabolism , Cholesterol/metabolism , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Presenilin-1/biosynthesis , Risk
11.
J Alzheimers Dis ; 21(4): 1271-81, 2010.
Article in English | MEDLINE | ID: mdl-21504136

ABSTRACT

The effect of supplementation with the omega 3 polyunsaturated fatty acid (n3 PUFA) docosahexaenoic acid (DHA) on membrane composition and amyloid-ß1₋42 (Aß42) secretion was studied in human amyloid-ß protein precursor-transfected Chinese Hamster Ovary (CHO) cells. Twenty-four hour incubation with a range of DHA concentrations resulted in a dose-dependent increase in membrane DHA and eicosapentaenoic acid content and a decrease in arachidonic acid content. In addition, DHA supplementation caused a dose-dependent reduction in the secreted Aß42 levels and resulted in a 4-8 fold decrease in extracellular prostaglandin E2 (PGE2) levels. Tocopherol, which was added to DHA to prevent oxidation, may have contributed to the effect of DHA, since it slightly decreased extracellular Aß42 and PGE2 levels when given alone. The addition of selective COX2 inhibitors Celebrex and curcumin to the culture medium resulted in a significant and comparable inhibition of PGE2 release, but did not inhibit Aß42 secretion, and even significantly increased Aß42 production in this cell system. Together, the present data show that, whereas both DHA and COX2 inhibitors may reduce PGE2 production, only DHA in the presence of tocopherol significantly reduced Aß42 production and concurrently changed membrane lipid composition in CHO cells. It is concluded that in this in vitro setting DHA reduced Aß42 secretion through membrane-related, but not PGE2-related mechanisms.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Dinoprostone/biosynthesis , Docosahexaenoic Acids/pharmacology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , CHO Cells , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Dinoprostone/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Transfection/methods
12.
Brain Res ; 988(1-2): 9-19, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-14519522

ABSTRACT

The beneficial effect of dietary n-3 polyunsaturated fatty acids (PUFAs) on developing hypertension has been repeatedly demonstrated. However, related changes in brain membrane composition and its cognitive correlates have remained unclear. Our study aimed at a comprehensive analysis of behavior and cerebral fatty acid concentration in hypertension after long-term PUFA-rich dietary treatment. Hypertensive and normotensive rats were provided a placebo, or one of two PUFA-enriched diets with a reduced (n-6)/(n-3) ratio for 75 weeks. Exploratory behavior and spatial learning capacity were tested. Systolic blood pressure (BP) was repeatedly measured. Finally, brain fatty acid composition was analyzed by gas chromatography. Hypertensive rats exhibited more active exploration but impaired spatial learning compared to normotensives. Both diets reduced BP, increased PUFA and monounsaturated fatty acid (MUFA) concentration, and reduced saturated fatty acid content in brain. The level of cerebral PUFAs and MUFAs was lower in hypertensive than in normotensive rats. Furthermore, BP positively, while spatial learning negatively correlated with cerebral (n-6)/(n-3) PUFA ratio. We concluded that regular n-3 PUFA consumption could prevent the development of hypertension, but reached only a very delicate improvement in spatial learning. Furthermore, we consider a potential role of metabolically generated MUFAs in the beneficial effects of PUFA supplementation.


Subject(s)
Behavior, Animal/drug effects , Blood Pressure/drug effects , Brain/metabolism , Cell Membrane/metabolism , Fatty Acids, Unsaturated/pharmacology , Hypertension/drug therapy , Hypolipidemic Agents/pharmacology , Learning/drug effects , Triglycerides/pharmacology , Animals , Body Weight/drug effects , Cerebral Cortex/metabolism , Chromatography, Gas , Cognition/drug effects , Dietary Fats/pharmacology , Fatty Acids, Omega-3 , Fatty Acids, Unsaturated/administration & dosage , Fatty Acids, Unsaturated/metabolism , Hypertension/metabolism , Hypertension/psychology , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/metabolism , Male , Neurons/metabolism , Phospholipids/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Space Perception/drug effects , Triglycerides/administration & dosage , Triglycerides/metabolism
13.
Neurosci Lett ; 344(1): 29-32, 2003 Jun 19.
Article in English | MEDLINE | ID: mdl-12781914

ABSTRACT

Natural hypothermia during hibernation results in physiological and behavioral deficits. These changes may be traced at the level of hippocampal signal transduction. We investigated synaptophysin immunoreactivity (SYN-ir) in the hippocampus after short and long periods of hypothermia and short and long periods of euthermy in hibernating ground squirrels. SYN-ir in the stratum lucidum of the hippocampus was transiently reduced during natural hypothermia. Natural hypothermia thus reduces synaptic efficacy. This may play a role in the reduced neuronal connectivity of CA3 pyramidal cell dendrites observed in hibernating ground squirrels.


Subject(s)
Hibernation/physiology , Hippocampus/metabolism , Hypothermia , Synaptophysin/biosynthesis , Animals , Blotting, Western , Immunohistochemistry , Sciuridae , Signal Transduction/physiology , Synapses/metabolism
14.
Ann N Y Acad Sci ; 977: 77-86, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12480735

ABSTRACT

Dietary supplementation with long-chain polyunsaturated fatty acids (PUFAs) has become an attractive possibility to alleviate or prevent cerebrovascular pathophysiology. To characterize the potentially beneficial cerebrovascular action of n-3 PUFAs that predominantly occur in fish oil, we set up an experimental paradigm where rats with chronic cerebral hypoperfusion were supplied with n-3 PUFA-enriched diets. Cerebral hypoperfusion was created by a permanent, bilateral occlusion of the common carotid arteries (2VO) of rats at the age of 4 months, with a survival of 3 months. Simultaneously, the rats were provided with experimental diets from the time of weaning until the termination of the experiments. The control diet was comparable to standard rat chow, while diet 1 contained additional n-3 PUFAs and diet 2 was further enriched with structural phospholipids and neurotransmitter precursors. In summary, the data show that diet 2 improved spatial learning of 2VO rats in the Morris water maze. Both diet 1 and diet 2 augmented blood-brain barrier parameters and increased the density of the M1-type muscarinic cholinergic receptors in the hippocampus independent of the rate of cerebral perfusion. In addition to an overview of these results, changes that were supportive or accompanying those described in the CNS are also presented. Briefly, plasma corticosterone concentration was elevated most explicitly by 2VO, while the relative weight of the liver and spleen increased due to the diets. The data draw attention to changes not only in the CNS, but also in the periphery as a consequence of chronic supplementation with n-3 PUFA-enriched diets.


Subject(s)
Cerebrovascular Circulation/physiology , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Maze Learning/drug effects , Animals , Blood-Brain Barrier/drug effects , Cerebrovascular Circulation/drug effects , Corticosterone/blood , Male , Rats , Rats, Wistar
15.
Brain Res ; 954(1): 32-41, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12393230

ABSTRACT

The chronic dietary intake of essential polyunsaturated fatty acids (PUFAs) can modulate learning and memory by being incorporated into neuronal plasma membranes. Representatives of two PUFA families, the n-3 and n-6 types become integrated into membrane phospholipids, where the actual (n-6)/(n-3) ratio can determine membrane fluidity and thus the function of membrane-bound proteins. In the present experiment we studied hippocampal neurotransmitter receptors after chronic administration of n-3 PUFA enriched diets in a brain hypoperfusion model, which mimics decreased cerebral perfusion as it occurs in ageing and dementia. Male Wistar rats received experimental diets with a decreased (n-6)/(n-3) ratio from weaning on. Chronic experimental cerebral hypoperfusion was imposed by a permanent, bilateral occlusion of the common carotid arteries (2VO) at the age of 4 months. The experiment was terminated when the rats were 7 months old. Three receptor types, the muscarinic 1, serotonergic 1A and the glutaminergic NMDA receptors were labeled in hippocampal slices by autoradiographic methods. Image analysis demonstrated that 2VO increased muscarinic 1 and NMDA receptor density, specifically in the dentate gyrus and the CA3 region, respectively. The increased ratio of n-3 fatty acids in combination with additional dietary supplements enhanced the density of the serotonergic 1A and muscarinic 1 receptors, while n-3 fatty acids alone increased binding only to the muscarinic 1 receptors. Since the examined receptor types reacted differently to the diets, we concluded that besides changes in membrane fluidity, the biochemical regulation of receptor sensitivity might also play a role in increasing hippocampal receptor density.


Subject(s)
Cerebrovascular Circulation/drug effects , Dietary Fats, Unsaturated/administration & dosage , Fatty Acids, Essential/administration & dosage , Receptors, Muscarinic/metabolism , Receptors, Serotonin/metabolism , Animals , Brain Ischemia/metabolism , Carotid Artery, Common/surgery , Cerebral Cortex/blood supply , Cerebrovascular Disorders , Hippocampus/metabolism , Male , Perfusion , Rats , Rats, Wistar , Receptor, Muscarinic M1 , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Serotonin, 5-HT1
16.
Brain Res ; 947(2): 166-73, 2002 Aug 30.
Article in English | MEDLINE | ID: mdl-12176157

ABSTRACT

Western diets consist to a large part of n-6 polyunsaturated fatty acids (PUFAs). These n-6 PUFAs and their conversion products favor immune and inflammatory reactions and compromise vasoregulation, which can contribute to the development of dementia. Recent epidemiological studies associated dementia, particularly the type accompanied by a vascular component, with high, saturated dietary fat intake. Conversely, high fish consumption (a source of long chain n-3 PUFAs) was related to a reduced risk for cognitive decline. Therefore we studied the effects of long chain n-3 PUFAs in rats with bilateral occlusion of the common carotid arteries (2VO), which mimics cerebral hypoperfusion, a risk factor for dementia. Male Wistar rats received experimental diets with a decreased (n-6)/(n-3) ratio from weaning on. At the age of 3 months, the animals underwent 2VO surgery. The rats were tested in the elevated plus maze, an active avoidance paradigm and the Morris water maze (at different survival times). Following behavioral testing, the animals were sacrificed at the age of 7 months. The frontoparietal cortex was analyzed for capillary ultrastructure with electron microscopy. No effects of cerebral hypoperfusion or diet were found on elevated plus maze and active avoidance, while spatial memory in the Morris maze was compromised due to cerebral hypoperfusion under placebo dietary conditions. n-3 PUFA supplementation in combination with extra additives improved the performance of the 2VO animals. The number of endothelial mitochondria, as well as the ratio of microvessels with degenerative pericytes appeared to be lower due to long chain n-3 PUFAs. These results may indicate an improved condition of the blood-brain barrier.


Subject(s)
Brain Ischemia/diet therapy , Cerebrovascular Circulation/drug effects , Cognition Disorders/diet therapy , Diet , Fatty Acids, Omega-3/pharmacology , Frontal Lobe/blood supply , Parietal Lobe/blood supply , Animals , Avoidance Learning , Brain Ischemia/complications , Brain Ischemia/physiopathology , Capillaries/pathology , Carotid Artery, Common/surgery , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Male , Maze Learning , Memory , Microscopy, Electron , Mitochondria/pathology , Rats , Rats, Wistar , Water
17.
Drugs Today (Barc) ; 38(5): 365-76, 2002 May.
Article in English | MEDLINE | ID: mdl-12532171

ABSTRACT

Neurodegenerative disorders, and dementia in particular, have been shown to have a cerebrovascular pathogenic component often in the form of reduced cerebral blood flow. The debate whether such a reduced brain perfusion is a primary trigger or a secondary symptom in the neuropathological progression of dementia has not been conclusively decided yet. However, compelling experimental evidence has been collected to demonstrate an initiating role of reduced cerebral blood flow in neurodegenerative processes. Along these lines, experimental cerebral hypoperfusion in rodents was shown to impair spatial learning and to generate neuronal damage and associated gliosis in sensitive brain regions like the hippocampus and frontoparietal cortex. Since suboptimal cerebral blood supply was thus identified as a potential trigger of cognitive decline, the improvement of cerebral blood flow in cognitive disorders has emerged as an alternative treatment to moderate the symptoms and to delay the onset of advanced dementia. Various drugs, such as cholinergic compounds, hemorheologic agents and vascular smooth muscle relaxants, have already been tested in some instances for their efficacy to increase brain perfusion. In this respect, both clinical and preclinical trials delivered positive data. Furthermore, not only the treatment but also the prevention of the development of cognitive deficiency can target the cerebrovascular system. For this purpose, long-chain polyunsaturated fatty acids derived from fish oil (also known as n-3 PUFAs) have been considered as dietary supplements. These fatty acids appeared particularly effective in the prevention of hypertension-associated vascular pathology. The present review provides an overview of the actions of these compounds focusing on cerebral blood flow, neurodegeneration and cognitive decline.


Subject(s)
Brain Ischemia/pathology , Brain Ischemia/psychology , Brain/pathology , Cognition Disorders/pathology , Cognition Disorders/psychology , Cognition/physiology , Animals , Anticoagulants/therapeutic use , Brain Ischemia/drug therapy , Cholinergic Agents/therapeutic use , Cognition/drug effects , Cognition Disorders/drug therapy , Diet , Humans , Muscle, Smooth, Vascular/drug effects , Vasodilator Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...