1
|
Oye Mintsa Mi-Mba MF, Lebbadi M, Alata W, Julien C, Emond V, Tremblay C, Fortin S, Barrow CJ, Bilodeau JF, Calon F. Differential impact of eicosapentaenoic acid and docosahexaenoic acid in an animal model of Alzheimer's disease. J Lipid Res 2024; 65:100682. [PMID: 39490923 DOI: 10.1016/j.jlr.2024.100682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Dietary supplementation with n-3 polyunsaturated fatty acids improves cognitive performance in several animal models of Alzheimer's disease (AD), an effect often associated with reduced amyloid-beta and/or tau pathologies. However, it remains unclear to what extent eicosapentaenoic (EPA) provides additional benefits compared to docosahexaenoic acid (DHA). Here, male and female 3xTg-AD mice were fed for 3 months (13-16 months of age) the following diets: (1) control (no DHA/EPA), (2) DHA (1.1g/kg) and low EPA (0.4g/kg), or (3) DHA (0.9g/kg) with high EPA (9.2g/kg). The DHA and DHA + EPA diets respectively increased DHA by 19% and 8% in the frontal cortex of 3xTg-AD mice, compared to controls. Levels of EPA, which were below the detection limit after the control diet, reached 0.14% and 0.29% of total brain fatty acids after the DHA and DHA + EPA diet, respectively. DHA and DHA + EPA diets lowered brain arachidonic acid levels and the n-6:n-3 docosapentaenoic acid ratio. Brain uptake of free 14C-DHA measured through intracarotid brain perfusion, but not of 14C-EPA, was lower in 3xTg-AD than in NonTg mice. DHA and DHA + EPA diets in 3xTg-AD mice reduced cortical soluble phosphorylated tau (pS202) (-34% high-DHA, -34% DHA + EPA, P < 0.05) while increasing p21-activated kinase (+58% and +83%, P < 0.001; respectively). High EPA intake lowered insoluble phosphorylated tau (-31% vs. DHA, P < 0.05). No diet effect on amyloid-beta levels was observed. In conclusion, dietary intake of DHA and EPA leads to differential changes in brain PUFA while altering cerebral biomarkers consistent with beneficial effects against AD-like neuropathology.
Collapse
Affiliation(s)
- Méryl-Farelle Oye Mintsa Mi-Mba
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Meryem Lebbadi
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Waël Alata
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Carl Julien
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Vincent Emond
- Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Cyntia Tremblay
- Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada
| | - Samuel Fortin
- Centre de recherche sur les biotechnologies marines, Rimouski, QC, Canada
| | - Colin J Barrow
- Centre for Sustainable Bioproducts, Deakin University Geelong, Victoria, Australia
| | - Jean-François Bilodeau
- Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada; Department of medicine, Faculty of Medecine, Laval University, Quebec, QC, Canada
| | - Frédéric Calon
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada; Centre Hospitalier de l'Université Laval (CHUL) Research Center, Quebec, QC, Canada.
| |
Collapse
|
2
|
Simard M, Mélançon K, Berthiaume L, Tremblay C, Pshevorskiy L, Julien P, Rajput AH, Rajput A, Calon F. Postmortem Fatty Acid Abnormalities in the Cerebellum of Patients with Essential Tremor. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2341-2359. [PMID: 39215908 PMCID: PMC11585516 DOI: 10.1007/s12311-024-01736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Fatty acids play many critical roles in brain function but have not been investigated in essential tremor (ET), a frequent movement disorder suspected to involve cerebellar dysfunction. Here, we report a postmortem comparative analysis of fatty acid profiles by gas chromatography in the cerebellar cortex from ET patients (n = 15), Parkinson's disease (PD) patients (n = 15) and Controls (n = 17). Phosphatidylcholine (PC), phosphatidylethanolamine (PE) and phosphatidylinositol (PI)/ phosphatidylserine (PS) were separated by thin-layer chromatography and analyzed separately. First, the total amounts of fatty acids retrieved from the cerebellar cortex were lower in ET patients compared with PD patients, including monounsaturated (MUFA) and polyunsaturated fatty acids (PUFA). The diagnosis of ET was associated with lower cerebellar levels of saturated fatty acids (SFA) and PUFA (DHA and ARA) in the PE fraction specifically, but with a higher relative content of dihomo-γ-linolenic acid (DGLA; 20:3 ω-6) in the PC fraction. In contrast, a diagnosis of PD was associated with higher absolute concentrations of SFA, MUFA and ω-6 PUFA in the PI + PS fractions. However, relative PI + PS contents of ω-6 PUFA were lower in both PD and ET patients. Finally, linear regression analyses showed that the ω-3:ω-6 PUFA ratio was positively associated with age of death, but inversely associated with insoluble α-synuclein. Although it remains unclear how these FA changes in the cerebellum are implicated in ET or PD pathophysiology, they may be related to an ongoing neurodegenerative process or to dietary intake differences. The present findings provide a window of opportunity for lipid-based therapeutic nutritional intervention.
Collapse
Affiliation(s)
- Mélissa Simard
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Koralie Mélançon
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Room T-2-67 (CHUL) 2705 boul. Laurier, Québec, QC, G1V 4G2, Canada
| | - Line Berthiaume
- Faculté de Médecine, Université Laval, Québec, QC, Canada
- Axe Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada
| | - Cyntia Tremblay
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Room T-2-67 (CHUL) 2705 boul. Laurier, Québec, QC, G1V 4G2, Canada
| | - Laura Pshevorskiy
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Room T-2-67 (CHUL) 2705 boul. Laurier, Québec, QC, G1V 4G2, Canada
| | - Pierre Julien
- Faculté de Médecine, Université Laval, Québec, QC, Canada
- Axe Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada
| | - Ali H Rajput
- Division of Neurology, Royal University Hospital, University of Saskatchewan, Saskatoon, SK, Canada
| | - Alex Rajput
- Division of Neurology, Royal University Hospital, University of Saskatchewan, Saskatoon, SK, Canada
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Room T-2-67 (CHUL) 2705 boul. Laurier, Québec, QC, G1V 4G2, Canada.
| |
Collapse
|
3
|
Stachowicz K. The role of polyunsaturated fatty acids in neuronal signaling in depression and cognitive processes. Arch Biochem Biophys 2023; 737:109555. [PMID: 36842491 DOI: 10.1016/j.abb.2023.109555] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/03/2023] [Accepted: 02/24/2023] [Indexed: 02/26/2023]
Abstract
This study aimed to evaluate research findings on the role of polyunsaturated fatty acids (PUFAs) in neuronal signaling. Polyunsaturated fatty acids (PUFAs) are the building blocks of the brain and are responsible for the proper functioning of neurons, synapses, and neuronal communication. The deficiency of a significant component, omega-3 (ω-3) FA, in favor of omega-6 (ω-6) FA can exacerbate the course of mental illness and be one of the triggers of the cascade of neurodegenerative changes. PUFAs play an essential role in transmitting neuronal signals, affecting brain homeostasis. Physicochemical parameters of lipid layers significantly affect their functioning; interactions between lipids and proteins in brain cells are critical for mechanical stability and maintaining adequate elasticity for vesicle budding and membrane fusion. This paper discusses the role of PUFA deficiency or inappropriate ratios in brain tissue. The deficiency is a crucial element in depressive disorders and cognitive impairment, while in Alzheimer's disease, there is some controversy.
Collapse
Affiliation(s)
- Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
4
|
Ablinger I, Dressel K, Rott T, Lauer AA, Tiemann M, Batista JP, Taddey T, Grimm HS, Grimm MOW. Interdisciplinary Approaches to Deal with Alzheimer's Disease-From Bench to Bedside: What Feasible Options Do Already Exist Today? Biomedicines 2022; 10:2922. [PMID: 36428494 PMCID: PMC9687885 DOI: 10.3390/biomedicines10112922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease is one of the most common neurodegenerative diseases in the western population. The incidence of this disease increases with age. Rising life expectancy and the resulting increase in the ratio of elderly in the population are likely to exacerbate socioeconomic problems. Alzheimer's disease is a multifactorial disease. In addition to amyloidogenic processing leading to plaques, and tau pathology, but also other molecular causes such as oxidative stress or inflammation play a crucial role. We summarize the molecular mechanisms leading to Alzheimer's disease and which potential interventions are known to interfere with these mechanisms, focusing on nutritional approaches and physical activity but also the beneficial effects of cognition-oriented treatments with a focus on language and communication. Interestingly, recent findings also suggest a causal link between oral conditions, such as periodontitis or edentulism, and Alzheimer's disease, raising the question of whether dental intervention in Alzheimer's patients can be beneficial as well. Unfortunately, all previous single-domain interventions have been shown to have limited benefit to patients. However, the latest studies indicate that combining these efforts into multidomain approaches may have increased preventive or therapeutic potential. Therefore, as another emphasis in this review, we provide an overview of current literature dealing with studies combining the above-mentioned approaches and discuss potential advantages compared to monotherapies. Considering current literature and intervention options, we also propose a multidomain interdisciplinary approach for the treatment of Alzheimer's disease patients that synergistically links the individual approaches. In conclusion, this review highlights the need to combine different approaches in an interdisciplinary manner, to address the future challenges of Alzheimer's disease.
Collapse
Affiliation(s)
- Irene Ablinger
- Speech and Language Therapy, Campus Bonn, SRH University of Applied Health Sciences, 53111 Bonn, Germany
| | - Katharina Dressel
- Speech and Language Therapy, Campus Düsseldorf, SRH University of Applied Health Sciences, 40210 Düsseldorf, Germany
| | - Thea Rott
- Interdisciplinary Periodontology and Prevention, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
| | - Anna Andrea Lauer
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
- Experimental Neurology, Saarland University, 66424 Homburg, Germany
| | - Michael Tiemann
- Sport Science, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
| | - João Pedro Batista
- Sport Science and Physiotherapy, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
| | - Tim Taddey
- Physiotherapy, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
| | - Heike Sabine Grimm
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
- Experimental Neurology, Saarland University, 66424 Homburg, Germany
| | - Marcus Otto Walter Grimm
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
- Experimental Neurology, Saarland University, 66424 Homburg, Germany
| |
Collapse
|
5
|
Arsenault D, Tremblay C, Emond V, Calon F. Sex-dependent alterations in the physiology of entorhinal cortex neurons in old heterozygous 3xTg-AD mice. Biol Sex Differ 2020; 11:63. [PMID: 33198813 PMCID: PMC7667843 DOI: 10.1186/s13293-020-00337-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/15/2020] [Indexed: 01/29/2023] Open
Abstract
While the higher prevalence of Alzheimer’s disease (AD) in women is clear, studies suggest that biological sex may also influence AD pathogenesis. However, mechanisms behind these differences are not clear. To investigate physiological differences between sexes at the cellular level in the brain, we investigated the intrinsic and synaptic properties of entorhinal cortex neurons in heterozygous 3xTg-AD mice of both sexes at the age of 20 months. This brain region was selected because of its early association with AD symptoms. First, we found physiological differences between male and female non-transgenic mice, providing indirect evidence of axonal alterations in old females. Second, we observed a transgene-dependent elevation of the firing activity, post-burst afterhyperpolarization (AHP), and spontaneous excitatory postsynaptic current (EPSC) activity, without any effect of sex. Third, the passive properties and the hyperpolarization-activated current (Ih) were altered by transgene expression only in female mice, whereas the paired-pulse ratio (PPR) of evoked EPSC was changed only in males. Fourth, both sex and transgene expression were associated with changes in action potential properties. Consistent with previous work, higher levels of Aβ neuropathology were detected in 3xTg-AD females, whereas tau deposition was similar. In summary, our results support the idea that aging and AD neuropathology differentially alter the physiology of entorhinal cortex neurons in males and females.
Collapse
Affiliation(s)
- Dany Arsenault
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada.,Neuroscience, Centre de Recherche du CHU de Québec (CHUQ), Quebec City, QC, Canada.,Physiotek, Quebec City, QC, Canada
| | - Cyntia Tremblay
- Neuroscience, Centre de Recherche du CHU de Québec (CHUQ), Quebec City, QC, Canada
| | - Vincent Emond
- Neuroscience, Centre de Recherche du CHU de Québec (CHUQ), Quebec City, QC, Canada
| | - Frédéric Calon
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada. .,Neuroscience, Centre de Recherche du CHU de Québec (CHUQ), Quebec City, QC, Canada.
| |
Collapse
|
6
|
Basak S, Vilasagaram S, Duttaroy AK. Maternal dietary deficiency of n-3 fatty acids affects metabolic and epigenetic phenotypes of the developing fetus. Prostaglandins Leukot Essent Fatty Acids 2020; 158:102109. [PMID: 32474355 DOI: 10.1016/j.plefa.2020.102109] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/13/2020] [Indexed: 02/07/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) play multiple physiological roles. They regulate the structure and function of cell membranes and cell growth and proliferation, and apoptosis. In addition, PUFAs are involved in cellular signaling, gene expression and serve as precursors to second messengers such as eicosanoids, docosanoids etc. and regulate several physiological processes including placentation, inflammation, immunity, angiogenesis, platelet function, synaptic plasticity, neurogenesis, bone formation, energy homeostasis, pain sensitivity, stress, and cognitive functions. Linoleic acid, 18:2n-6 (LA) and alpha-linolenic acid, 18:3n-3 (ALA) are the two essential fatty acids obtained from the diets and subsequently their long-chain polyunsaturated fatty acids (LCPUFAs) are accumulated in the body. The maternal plasma LCPUFAs especially accumulated in larger amounts in the brain during the third trimester of pregnancy via the placenta and postnatally from mother's breast milk. Various studies, including ours, suggest PUFA's important role in placentation, as well as in growth and development of the offspring. However, intakes of maternal n-3 PUFAs during pregnancy and lactation are much lower in India compared with the Western population. In India, n-3 fatty acid status is further reduced by higher intake of n-6 PUFA rich oils and trans fats. More data on the impacts of long term maternal n-3 PUFA deficiency on placental structure and function, gene expression, epigenetic changes and resultant cognitive function of fetus & infants are emerging. This review summarizes the impacts of n-3 PUFA deficiency in utero on fetal growth and development, adiposity, energy metabolism, musculoskeletal development, and epigenetic changes in feto-placental axis from the recently available pre-clinical and clinical data.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India.
| | - Srinivas Vilasagaram
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|
7
|
Abstract
The gut microbiota is directly influenced by dietary components, and it plays critical roles in chronic diseases. Excessive consumption of trans-fatty acids (TFA) is associated with obesity induced by alterations in gut microbiota, but the links between obesity and gut microbiota remain unclear. Therefore, studies examining the impact of TFA on intestinal microflora are essential. In our study, we performed 16S ribosomal RNA gene sequencing on faecal samples from Sprague-Dawley rats fed a basal diet (control (CON) group), high-fat (HF) diet (diet-induced obesity (DIO) group) or TFA diets (1 % TFA group and 8 % TFA group) for 8 weeks to investigate the effects of TFA/HF diets on obesity and gut microbiota composition. We found that the TFA/HF diets significantly induced obesity and changes in blood and brain physiological parameters of the rats. The relative abundance of the phyla Firmicutes and Bacteroidetes was inversely altered in the three test groups compared with the CON group. Proteobacteria increased slightly in the DIO, 1 % TFA and 8 % TFA groups. The genus Bacteroides increased in the DIO and 1 % TFA groups, but Muribaculaceae decreased in all experimental groups compared with the CON group. Moreover, significant differences were observed among clusters of orthologous group functional categories of the four dietary groups. Our observations suggested that the TFA/HF diets induced obesity and dysfunction of gut microbiota. Gut dysbiosis might mediate the obesity effects of TFA/HF diets.
Collapse
|
8
|
Bourassa P, Alata W, Tremblay C, Paris-Robidas S, Calon F. Transferrin Receptor-Mediated Uptake at the Blood-Brain Barrier Is Not Impaired by Alzheimer's Disease Neuropathology. Mol Pharm 2019; 16:583-594. [PMID: 30609376 DOI: 10.1021/acs.molpharmaceut.8b00870] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The transferrin receptor (TfR) is highly expressed by brain capillary endothelial cells (BCECs) forming the blood-brain barrier (BBB) and is therefore considered as a potential target for brain drug delivery. Monoclonal antibodies binding to the TfR, such as clone Ri7, have been shown to internalize into BCECs in vivo. However, since Alzheimer's disease (AD) is accompanied by a BBB dysfunction, it raises concerns about whether TfR-mediated transport becomes inefficient during the progression of the disease. Measurements of TfR levels using Western blot analysis in whole homogenates from human post-mortem parietal cortex and hippocampus did not reveal any significant difference between individuals with or without a neuropathological diagnosis of AD (respectively, n = 19 and 22 for the parietal cortex and n = 12 and 14 for hippocampus). Similarly, TfR concentrations in isolated human brain microvessels from parietal cortex were similar between controls and AD cases. TfR levels in isolated murine brain microvessels were not significantly different between groups of 12- and 18-month-old NonTg and 3xTg-AD mice, the latter modeling Aβ and τ neuropathologies. In situ brain perfusion assays were then conducted to measure the brain uptake and internalization of fluorolabeled Ri7 in BCECs upon binding. Consistently, TfR-mediated uptake in BCECs was similar between 3xTg-AD mice and nontransgenic controls (∼0.3 μL·g-1·s-1) at 12, 18, and 22 months of age. Fluorescence microscopy analysis following intravenous administration of fluorolabeled Ri7 highlighted that the signal from the antibody was widely distributed throughout the cerebral vasculature but not in neurons or astrocytes. Overall, our data suggest that both TfR protein levels and TfR-dependent internalization mechanisms are preserved in the presence of Aβ and τ neuropathologies, supporting the potential of TfR as a vector target for drug delivery into BCECs in AD.
Collapse
Affiliation(s)
- Philippe Bourassa
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Wael Alata
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Cyntia Tremblay
- Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Sarah Paris-Robidas
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Frédéric Calon
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| |
Collapse
|
9
|
Montoliu-Gaya L, Güell-Bosch J, Esquerda-Canals G, Roda AR, Serra-Mir G, Lope-Piedrafita S, Sánchez-Quesada JL, Villegas S. Differential effects of apoE and apoJ mimetic peptides on the action of an anti-Aβ scFv in 3xTg-AD mice. Biochem Pharmacol 2018; 155:380-392. [PMID: 30026023 DOI: 10.1016/j.bcp.2018.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/13/2018] [Indexed: 12/31/2022]
Abstract
Anti-Aβ immunotherapy has emerged as a promising approach to treat Alzheimer's disease (AD). The single-chain variable fragment scFv-h3D6 is an anti-Aβ antibody fragment that lacks the Fc region, which is associated with the induction of microglial reactivity by the full-length monoclonal antibody bapineuzumab. ScFv-h3D6 was previously shown to restore the levels of apolipoprotein E (apoE) and apolipoprotein J (apoJ) in a triple-transgenic-AD (3xTg-AD) mouse model. Since apoE and apoJ play an important role in the development of AD, we aimed to study the in vivo effect of the combined therapy of scFv-h3D6 with apoE and apoJ mimetic peptides (MPs). Four-and-a-half-month-old 3xTg-AD mice were treated for six weeks with scFv-h3D6, apoE-MP, apoJ-MP, or a combination of scFv-h3D6 with each of the MPs, or a vehicle, and then the results were compared to non-transgenic mice. Magnetic Resonance Imaging showed a general tendency of the different treatments to protect against the reduction in brain volume. Aβ burden decreased after treatment with scFv-h3D6, apoE-MP, or apoJ-MP, but the effect was not as evident with the combined therapies. In terms of glial reactivity, apoE-MP showed a potent anti-inflammatory effect that was eased by the presence of scFv-h3D6, whereas the combination of apoJ-MP and scFv-h3D6 was not detrimental. ScFv-h3D6 alone did not induce microglial reactivity, as full-length antibodies do; rather, it reduced it. Endogenous apoE and apoJ levels were decreased by scFv-h3D6, but the MPs lead to a simultaneous increase of both apolipoproteins. While apoE-MP and apoJ-MP demonstrated different effects in the combined therapies with scFv-h3D6, they did not improve the overall protective effect of scFv-h3D6 in reducing the Aβ burden, apolipoproteins levels or microglial reactivity.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Jofre Güell-Bosch
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Gisela Esquerda-Canals
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Alejandro R Roda
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Gabriel Serra-Mir
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Silvia Lope-Piedrafita
- Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Spain.
| | - Sandra Villegas
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| |
Collapse
|
10
|
ZHANG T, LIU P, SUN Y, REN N, NIE S. The Impairment of Trans Fatty Acids on Learning, Memory and Brain Amino Acid Neurotransmitters in Mice. J Nutr Sci Vitaminol (Tokyo) 2018; 64:63-67. [DOI: 10.3177/jnsv.64.63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Tiantian ZHANG
- Department of Hygiene Detection, College of Public Health, Shandong University
| | - Ping LIU
- Department of Hygiene Detection, College of Public Health, Shandong University
| | - Yu SUN
- Department of Hygiene Detection, College of Public Health, Shandong University
| | - Na REN
- Department of Hygiene Detection, College of Public Health, Shandong University
| | - Shuang NIE
- Department of Hygiene Detection, College of Public Health, Shandong University
| |
Collapse
|
11
|
Pan Y, Choy KHC, Marriott PJ, Chai SY, Scanlon MJ, Porter CJH, Short JL, Nicolazzo JA. Reduced blood-brain barrier expression of fatty acid-binding protein 5 is associated with increased vulnerability of APP/PS1 mice to cognitive deficits from low omega-3 fatty acid diets. J Neurochem 2017; 144:81-92. [DOI: 10.1111/jnc.14249] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/18/2017] [Accepted: 10/25/2017] [Indexed: 01/08/2023]
Affiliation(s)
- Yijun Pan
- Drug Delivery, Disposition and Dynamics; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Vic. Australia
| | - Kwok H. C. Choy
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Vic. Australia
| | - Philip J. Marriott
- Australian Centre for Research on Separation Science; School of Chemistry; Monash University; Vic. Australia
| | - Siew Y. Chai
- Department of Physiology; Biomedicine Discovery Institute; Monash University; Clayton Vic. Australia
| | - Martin J. Scanlon
- Medicinal Chemistry; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Vic. Australia
| | - Christopher J. H. Porter
- Drug Delivery, Disposition and Dynamics; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Vic. Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Vic. Australia
| | - Jennifer L. Short
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Vic. Australia
| | - Joseph A. Nicolazzo
- Drug Delivery, Disposition and Dynamics; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Vic. Australia
| |
Collapse
|
12
|
Kim D, Cho J, Lee I, Jin Y, Kang H. Exercise Attenuates High-Fat Diet-induced Disease Progression in 3xTg-AD Mice. Med Sci Sports Exerc 2017; 49:676-686. [PMID: 27875496 DOI: 10.1249/mss.0000000000001166] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Little is known regarding the therapeutic role of exercise against the risk of a high-fat diet (HFD) for Alzheimer's disease (AD) and AD-like cognitive deficits. This study aimed to investigate the therapeutic effect of treadmill running against HFD-induced progression in AD neuropathology and cognitive impairments in the triple-transgenic AD (3xTg-AD) mice. METHODS The 3xTg-AD mice were assigned to a chow diet (control, n = 10), an HFD (n = 10), or an HFD combined with exercise (HFD + EX, n = 10) group. Mice in the HFD were fed with a 60% fat diet for 20 wk. The HFD + EX mice were additionally subjected to treadmill running. RESULTS Compared with the control mice, the HFD mice had impaired brain insulin signaling, exacerbated AD neuropathology, defects in synaptic stability/plasticity, and apoptotic neuronal cell death in conjunction with exacerbated cognitive deficits in the affected brain regions, which were all significantly alleviated in the HFD + EX mice. CONCLUSION The current findings suggest that treadmill running protects against AD-like disease progression and cognitive deficits caused by an HFD in the 3xTg-AD mice.
Collapse
Affiliation(s)
- Donghyun Kim
- College of Sport Science, Sungkyunkwan University, Suwon, REPUBLIC OF KOREA
| | | | | | | | | |
Collapse
|
13
|
Kerdiles O, Layé S, Calon F. Omega-3 polyunsaturated fatty acids and brain health: Preclinical evidence for the prevention of neurodegenerative diseases. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
14
|
Impact of enriched environment on production of tau, amyloid precursor protein and, amyloid-β peptide in high-fat and high-sucrose-fed rats. Acta Neuropsychiatr 2017; 29:291-298. [PMID: 27923413 DOI: 10.1017/neu.2016.63] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The Western-type diet is associated with an elevated risk of Alzheimer's disease and other milder forms of cognitive impairment. The aim of the present study was to investigate the effects of the environmental enrichment on amyloid and tau pathology in high-fat and high-sucrose-fed rats. METHODS In total, 40 adult male rats were categorised into two main groups according to their housing conditions: enriched environment (EE, n=16) and standard housing condition (n=24). The groups were further divided into five subgroups that received standard diet, high-fat diet, and high-sucrose diet. We performed the analysis of amyloid β-peptide (Aβ) (1-40), Aβ(1-42), amyloid precursor protein (APP), and tau levels in the hippocampus of rats that were maintained under standard housing conditions or exposed to an EE. RESULTS The EE decreased the Aβ(1-40), Aβ(1-42), APP, and tau levels in high-fat and high-sucrose-fed rats. CONCLUSION This observation shows that EE may rescue diet-induced amyloid and tau pathology.
Collapse
|
15
|
Auguste S, Sharma S, Fisette A, Fernandes MF, Daneault C, Des Rosiers C, Fulton S. Perinatal deficiency in dietary omega-3 fatty acids potentiates sucrose reward and diet-induced obesity in mice. Int J Dev Neurosci 2017; 64:8-13. [PMID: 28919371 DOI: 10.1016/j.ijdevneu.2017.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 09/06/2017] [Accepted: 09/06/2017] [Indexed: 11/16/2022] Open
Abstract
Insufficient dietary intake of essential omega-3 polyunsaturated fatty acids (N-3), especially during critical stages of development, is well-associated with negative neurological and metabolic consequences. The increased availability and intake of foods rich in saturated fat coincides with reduced N-3 consumption, yet how N-3 dietary deficiency during perinatal development modulates motivation for palatable food and interacts with a high-fat diet to affect body weight and emotional states is not clear. Pregnant C57Bl6 mice and pups were subjected to diets either deficient or adequate (control) in N-3 until postnatal day 21. Adult male N-3 deficient or control offspring were tested in a progressive ratio operant task for sucrose motivated behavior or given prolonged access to a saturated high-fat diet or chow followed by measures of energy balance and anxiety-like behavior in the elevated-plus maze and open field test. Brain fatty acid profiles were measured via gas chromatography mass spectrometry. Perinatal dietary N-3 deficiency lowered brain N-3 levels, augmented the rewarding effects of sucrose, heightened diet-induced weight gain and fat mass accumulation and diminished spontaneous physical activity. Finally, perinatal N-3 deficiency increased anxiety-like behaviour independent of diet in the open field but not in the elevated-plus maze test. Insufficient dietary N-3 during critical periods of developmental can amplify the obesogenic effects of saturated fat intake, enhance motivated behaviour for palatable foods and may elicit negative emotional states that can perpetuate overeating and obesity.
Collapse
Affiliation(s)
- Stéphanie Auguste
- CRCHUM and Montreal Diabetes Research Center, QC, H3T 1J4, Canada; Department of Nutrition, Faculty of Medicine, Université de Montréal, QC, H3T 1J4, Canada
| | - Sandeep Sharma
- CRCHUM and Montreal Diabetes Research Center, QC, H3T 1J4, Canada
| | - Alexandre Fisette
- CRCHUM and Montreal Diabetes Research Center, QC, H3T 1J4, Canada; Department of Nutrition, Faculty of Medicine, Université de Montréal, QC, H3T 1J4, Canada
| | | | | | - Christine Des Rosiers
- Department of Nutrition, Faculty of Medicine, Université de Montréal, QC, H3T 1J4, Canada; Montreal Heart Institute, QC, H3T 1J4, Canada
| | - Stephanie Fulton
- CRCHUM and Montreal Diabetes Research Center, QC, H3T 1J4, Canada; Department of Nutrition, Faculty of Medicine, Université de Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
16
|
Yang BY, Tan JY, Liu Y, Liu B, Jin S, Guo HW, Kuang HX. A UPLC-TOF/MS-based metabolomics study of rattan stems ofSchisandra chinensiseffects on Alzheimer's disease rats model. Biomed Chromatogr 2017; 32. [DOI: 10.1002/bmc.4037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 06/07/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Bing-You Yang
- Key Laboratory of Chinese Materia Medica (Ministry of Education); Heilongjiang University of Chinese Medicine; Harbin People's Republic of China
| | - Jin-Yan Tan
- Key Laboratory of Chinese Materia Medica (Ministry of Education); Heilongjiang University of Chinese Medicine; Harbin People's Republic of China
| | - Yan Liu
- Key Laboratory of Chinese Materia Medica (Ministry of Education); Heilongjiang University of Chinese Medicine; Harbin People's Republic of China
| | - Bo Liu
- Key Laboratory of Chinese Materia Medica (Ministry of Education); Heilongjiang University of Chinese Medicine; Harbin People's Republic of China
| | - Shuang Jin
- Key Laboratory of Chinese Materia Medica (Ministry of Education); Heilongjiang University of Chinese Medicine; Harbin People's Republic of China
| | - Hong-Wei Guo
- Key Laboratory of Chinese Materia Medica (Ministry of Education); Heilongjiang University of Chinese Medicine; Harbin People's Republic of China
| | - Hai-Xue Kuang
- Key Laboratory of Chinese Materia Medica (Ministry of Education); Heilongjiang University of Chinese Medicine; Harbin People's Republic of China
| |
Collapse
|
17
|
Grimm MOW, Mett J, Grimm HS, Hartmann T. APP Function and Lipids: A Bidirectional Link. Front Mol Neurosci 2017; 10:63. [PMID: 28344547 PMCID: PMC5344993 DOI: 10.3389/fnmol.2017.00063] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 02/24/2017] [Indexed: 12/14/2022] Open
Abstract
Extracellular neuritic plaques, composed of aggregated amyloid-β (Aβ) peptides, are one of the major histopathological hallmarks of Alzheimer's disease (AD), a progressive, irreversible neurodegenerative disorder and the most common cause of dementia in the elderly. One of the most prominent risk factor for sporadic AD, carrying one or two aberrant copies of the apolipoprotein E (ApoE) ε4 alleles, closely links AD to lipids. Further, several lipid classes and fatty acids have been reported to be changed in the brain of AD-affected individuals. Interestingly, the observed lipid changes in the brain seem not only to be a consequence of the disease but also modulate Aβ generation. In line with these observations, protective lipids being able to decrease Aβ generation and also potential negative lipids in respect to AD were identified. Mechanistically, Aβ peptides are generated by sequential proteolytic processing of the amyloid precursor protein (APP) by β- and γ-secretase. The α-secretase appears to compete with β-secretase for the initial cleavage of APP, preventing Aβ production. All APP-cleaving secretases as well as APP are transmembrane proteins, further illustrating the impact of lipids on Aβ generation. Beside the pathological impact of Aβ, accumulating evidence suggests that Aβ and the APP intracellular domain (AICD) play an important role in regulating lipid homeostasis, either by direct effects or by affecting gene expression or protein stability of enzymes involved in the de novo synthesis of different lipid classes. This review summarizes the current literature addressing the complex bidirectional link between lipids and AD and APP processing including lipid alterations found in AD post mortem brains, lipids that alter APP processing and the physiological functions of Aβ and AICD in the regulation of several lipid metabolism pathways.
Collapse
Affiliation(s)
- Marcus O. W. Grimm
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
- Neurodegeneration and Neurobiology, Saarland UniversityHomburg/Saar, Germany
- Deutsches Institut für DemenzPrävention (DIDP), Saarland UniversityHomburg/Saar, Germany
| | - Janine Mett
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
| | - Heike S. Grimm
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
| | - Tobias Hartmann
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
- Neurodegeneration and Neurobiology, Saarland UniversityHomburg/Saar, Germany
- Deutsches Institut für DemenzPrävention (DIDP), Saarland UniversityHomburg/Saar, Germany
| |
Collapse
|
18
|
Hu X, Tanaka N, Guo R, Lu Y, Nakajima T, Gonzalez FJ, Aoyama T. PPARα protects against trans-fatty-acid-containing diet-induced steatohepatitis. J Nutr Biochem 2016; 39:77-85. [PMID: 27816763 DOI: 10.1016/j.jnutbio.2016.09.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/11/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
Consumption of trans-fatty acids (TFA), unsaturated fatty acids (FA) containing trans double bonds, is a risk factor for metabolic syndrome and steatohepatitis. Peroxisome proliferator-activated receptor α (PPARα) is a master regulator of hepatic lipid homeostasis. To examine the contribution of PPARα to changes in liver phenotypes induced by TFA, two diets were used: a purified control diet and an isocaloric diet in which most of the soybean oil, a major source of FA in the diet, was replaced with TFA-rich shortening. The diets were fed to wild-type and Ppara-null mice for 2 months. Ppara-null mice fed a TFA-containing diet showed more severe hepatic steatosis and liver damage compared with similarly treated wild-type mice, as revealed by increased hepatic triglyceride (TG) contents and serum alanine aminotransferase activities. While the TFA-rich diet increased the hepatic expression of enzymes involved in de novo FA synthesis and decreased TG-hydrolyzing enzymes in both genotypes, the expression of FA-catabolizing enzymes was decreased in Ppara-null mice, resulting in more severe hepatosteatosis. Additionally, the expression levels of key contributors to inflammation, such as osteopontin, were increased, and nuclear factor-kappa B was activated in TFA-containing diet-fed Ppara-null mice. Enhanced inflammatory signaling in these mice was presumably mediated by toll-like receptor 2, with no accompanying inflammasome activation. Collectively, these results suggest a protective role for PPARα in the pathological changes in the liver following TFA consumption. PPARα might prevent TFA-containing diet-induced steatohepatitis.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Japan.
| | - Ran Guo
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Yu Lu
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Takero Nakajima
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Toshifumi Aoyama
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| |
Collapse
|
19
|
Auguste S, Fisette A, Fernandes MF, Hryhorczuk C, Poitout V, Alquier T, Fulton S. Central Agonism of GPR120 Acutely Inhibits Food Intake and Food Reward and Chronically Suppresses Anxiety-Like Behavior in Mice. Int J Neuropsychopharmacol 2016; 19:pyw014. [PMID: 26888796 PMCID: PMC4966276 DOI: 10.1093/ijnp/pyw014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/09/2016] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND GPR120 (FFAR4) is a G-protein coupled receptor implicated in the development of obesity and the antiinflammatory and insulin-sensitizing effects of omega-3 (ω-3) polyunsaturated fatty acids. Increasing central ω-3 polyunsaturated fatty acid levels has been shown to have both anorectic and anxiolytic actions. Despite the strong clinical interest in GPR120, its role in the brain is largely unknown, and thus we sought to determine the impact of central GPR120 pharmacological activation on energy balance, food reward, and anxiety-like behavior. METHODS Male C57Bl/6 mice with intracerebroventricular cannulae received a single injection (0.1 or 1 µM) or continuous 2-week infusion (1 µM/d; mini-pump) of a GPR120 agonist or vehicle. Free-feeding intake, operant lever-pressing for palatable food, energy expenditure (indirect calorimetry), and body weight were measured. GPR120 mRNA expression was measured in pertinent brain areas. Anxiety-like behavior was assessed in the elevated-plus maze and open field test. RESULTS GPR120 agonist injections substantially reduced chow intake during 4 hours postinjection, suppressed the rewarding effects of high-fat/-sugar food, and blunted approach-avoidance behavior in the open field. Conversely, prolonged central GPR120 agonist infusions reduced anxiety-like behavior in the elevated-plus maze and open field, yet failed to affect free-feeding intake, energy expenditure, and body weight on a high-fat diet. CONCLUSION Acute reductions in food intake and food reward suggest that GPR120 could mediate the effects of central ω-3 polyunsaturated fatty acids to inhibit appetite. The anxiolytic effect elicited by GPR120 agonist infusions favors the testing of compounds that can enter the brain to activate GPR120 for the mitigation of anxiety.
Collapse
Affiliation(s)
- Stéphanie Auguste
- CRCHUM & Montreal Diabetes Research Center (Drs Auguste, Fisette, and Fernandes, Ms Hryhorczuk, and Drs Poitout, Alquier, and Fulton); Department of Nutrition (Drs Auguste, Fisette, and Fulton), Department of Physiology (Dr Fernandes and Ms Hryhorczuk), and Department of Medicine (Drs Poitout and Alquier), Faculty of Medicine, Université de Montréal, QC, Canada.
| | - Alexandre Fisette
- CRCHUM & Montreal Diabetes Research Center (Drs Auguste, Fisette, and Fernandes, Ms Hryhorczuk, and Drs Poitout, Alquier, and Fulton); Department of Nutrition (Drs Auguste, Fisette, and Fulton), Department of Physiology (Dr Fernandes and Ms Hryhorczuk), and Department of Medicine (Drs Poitout and Alquier), Faculty of Medicine, Université de Montréal, QC, Canada
| | - Maria F Fernandes
- CRCHUM & Montreal Diabetes Research Center (Drs Auguste, Fisette, and Fernandes, Ms Hryhorczuk, and Drs Poitout, Alquier, and Fulton); Department of Nutrition (Drs Auguste, Fisette, and Fulton), Department of Physiology (Dr Fernandes and Ms Hryhorczuk), and Department of Medicine (Drs Poitout and Alquier), Faculty of Medicine, Université de Montréal, QC, Canada
| | - Cécile Hryhorczuk
- CRCHUM & Montreal Diabetes Research Center (Drs Auguste, Fisette, and Fernandes, Ms Hryhorczuk, and Drs Poitout, Alquier, and Fulton); Department of Nutrition (Drs Auguste, Fisette, and Fulton), Department of Physiology (Dr Fernandes and Ms Hryhorczuk), and Department of Medicine (Drs Poitout and Alquier), Faculty of Medicine, Université de Montréal, QC, Canada
| | - Vincent Poitout
- CRCHUM & Montreal Diabetes Research Center (Drs Auguste, Fisette, and Fernandes, Ms Hryhorczuk, and Drs Poitout, Alquier, and Fulton); Department of Nutrition (Drs Auguste, Fisette, and Fulton), Department of Physiology (Dr Fernandes and Ms Hryhorczuk), and Department of Medicine (Drs Poitout and Alquier), Faculty of Medicine, Université de Montréal, QC, Canada
| | - Thierry Alquier
- CRCHUM & Montreal Diabetes Research Center (Drs Auguste, Fisette, and Fernandes, Ms Hryhorczuk, and Drs Poitout, Alquier, and Fulton); Department of Nutrition (Drs Auguste, Fisette, and Fulton), Department of Physiology (Dr Fernandes and Ms Hryhorczuk), and Department of Medicine (Drs Poitout and Alquier), Faculty of Medicine, Université de Montréal, QC, Canada
| | - Stephanie Fulton
- CRCHUM & Montreal Diabetes Research Center (Drs Auguste, Fisette, and Fernandes, Ms Hryhorczuk, and Drs Poitout, Alquier, and Fulton); Department of Nutrition (Drs Auguste, Fisette, and Fulton), Department of Physiology (Dr Fernandes and Ms Hryhorczuk), and Department of Medicine (Drs Poitout and Alquier), Faculty of Medicine, Université de Montréal, QC, Canada
| |
Collapse
|
20
|
Lavandera JV, Saín J, Fariña AC, Bernal CA, González MA. N-3 fatty acids reduced trans fatty acids retention and increased docosahexaenoic acid levels in the brain. Nutr Neurosci 2016; 20:424-435. [PMID: 27098669 DOI: 10.1080/1028415x.2016.1173343] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The levels of docosahexaenoic acid (DHA, 22:6n-3) and arachidonic acid (AA, 20:4n-6) are critical for the normal structure and function of the brain. Trans fatty acids (TFA) and the source of the dietary fatty acids (FA) interfere with long-chain polyunsaturated fatty acids (LC-PUFA) biosynthesis. OBJECTIVES The aim of this study was to investigate the effect of TFA supplementation in diets containing different proportions of n-9, n-6, and n-3 FA on the brain FA profile, including the retention of TFA, LC-PUFA levels, and n-6/n-3 PUFA ratios. These parameters were also investigated in the liver, considering that LC-PUFA are mainly bioconverted from their dietary precursors in this tissue and transported by serum to the brain. Also, stearoyl-CoA desaturase-1 (SCD1) and sterol regulatory element-binding protein-1c (SREBP-1c) gene expressions were evaluated. METHODS Male CF1 mice were fed (16 weeks) diets containing different oils (olive, corn, and rapeseed) with distinct proportions of n-9, n-6, and n-3 FA (55.2/17.2/0.7, 32.0/51.3/0.9, and 61.1/18.4/8.6), respectively, substituted or not with 0.75% of TFA. FA composition of the brain, liver, and serum was assessed by gas chromatography. RESULTS TFA were incorporated into, and therefore retained in the brain, liver, and serum. However, the magnitude of retention was dependent on the tissue and type of isomer. In the brain, total TFA retention was lower than 1% in all diets. DISCUSSION Dietary n-3 PUFA decreased TFA retention and increased DHA accretion in the brain. The results underscore the importance of the type of dietary FA on the retention of TFA in the brain and also on the changes of the FA profile.
Collapse
Affiliation(s)
- Jimena Verónica Lavandera
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina.,b Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Santa Fe , Argentina
| | - Juliana Saín
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina.,b Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Santa Fe , Argentina
| | - Ana Clara Fariña
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina
| | - Claudio Adrián Bernal
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina.,b Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Santa Fe , Argentina
| | - Marcela Aída González
- a Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas , Universidad Nacional del Litoral , Santa Fe , Argentina
| |
Collapse
|
21
|
Abstract
This manuscript summarizes the proceedings of the symposium entitled, "Stress, Palatable Food and Reward", that was chaired by Drs. Linda Rinaman and Yvonne Ulrich-Lai at the 2014 Neurobiology of Stress Workshop held in Cincinnati, OH. This symposium comprised research presentations by four neuroscientists whose work focuses on the biological bases for complex interactions among stress, food intake and emotion. First, Dr Ulrich-Lai describes her rodent research exploring mechanisms by which the rewarding properties of sweet palatable foods confer stress relief. Second, Dr Stephanie Fulton discusses her work in which excessive, long-term intake of dietary lipids, as well as their subsequent withdrawal, promotes stress-related outcomes in mice. Third, Dr Mark Wilson describes his group's research examining the effects of social hierarchy-related stress on food intake and diet choice in group-housed female rhesus macaques, and compared the data from monkeys to results obtained in analogous work using rodents. Finally, Dr Gorica Petrovich discusses her research program that is aimed at defining cortical-amygdalar-hypothalamic circuitry responsible for curbing food intake during emotional threat (i.e. fear anticipation) in rats. Their collective results reveal the complexity of physiological and behavioral interactions that link stress, food intake and emotional state, and suggest new avenues of research to probe the impact of genetic, metabolic, social, experiential and environmental factors on these interactions.
Collapse
Affiliation(s)
- Yvonne M. Ulrich-Lai
- Dept. of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45219
| | - Stephanie Fulton
- CRCHUM, Dept. of Nutrition, Université de Montréal, Montreal, QC, Canada, H1W 4A4
| | - Mark Wilson
- Division of Developmental and Cognitive Neuroscience, Emory, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322
| | | | - Linda Rinaman
- Dept. of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260
| |
Collapse
|
22
|
Trevizol F, Dias VT, Roversi K, Barcelos RC, Kuhn FT, Roversi K, Pase CS, Golombieski R, Veit JC, Piccolo J, Emanuelli T, Rocha JB, Bürger ME. Cross-generationaltransfat intake modifies BDNF mRNA in the hippocampus: Impact on memory loss in a mania animal model. Hippocampus 2014; 25:556-65. [DOI: 10.1002/hipo.22391] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Fabíola Trevizol
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria(UFSM)-RS; Brasil
| | | | | | - Raquel C.S. Barcelos
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria(UFSM)-RS; Brasil
| | - Fábio T. Kuhn
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria(UFSM)-RS; Brasil
| | - Karine Roversi
- Departamento de Fisiologia e Farmacologia- UFSM-RS; Brasil
| | - Camila S. Pase
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria(UFSM)-RS; Brasil
| | | | - Juliana C. Veit
- Programa de Pós-Graduação em Ciência e Tecnologia dos Alimentos-UFSM-RS; Brasil
| | - Jaqueline Piccolo
- Programa de Pós-Graduação em Ciência e Tecnologia dos Alimentos-UFSM-RS; Brasil
| | - Tatiana Emanuelli
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria(UFSM)-RS; Brasil
- Programa de Pós-Graduação em Ciência e Tecnologia dos Alimentos-UFSM-RS; Brasil
| | - João B.T. Rocha
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria(UFSM)-RS; Brasil
| | - Marilise E. Bürger
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria(UFSM)-RS; Brasil
- Programa de Pós-Graduação em Bioquímica Toxicológica-UFSM-RS; Brasil
| |
Collapse
|
23
|
Zendedel A, Habib P, Dang J, Lammerding L, Hoffmann S, Beyer C, Slowik A. Omega-3 polyunsaturated fatty acids ameliorate neuroinflammation and mitigate ischemic stroke damage through interactions with astrocytes and microglia. J Neuroimmunol 2014; 278:200-11. [PMID: 25468770 DOI: 10.1016/j.jneuroim.2014.11.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/04/2014] [Accepted: 11/07/2014] [Indexed: 01/10/2023]
Abstract
Omega-3 polyunsaturated fatty acids (PUFA n3) provide neuroprotection due to their anti-inflammatory and anti-apoptotic properties as well as their regulatory function on growth factors and neuronal plasticity. These qualities enable PUFA n3 to ameliorate stroke outcome and limit neuronal damage. Young adult male rats received transient middle cerebral artery occlusion (tMCAO). PUFA n3 were intravenously administered into the jugular vein immediately after stroke and 12h later. We analyzed stroke volume and behavioral performance as well as the regulation of functionally-relevant genes in the penumbra. The extent of ischemic damage was reduced and behavioral performance improved subject to applied PUFA n3. Expression of Tau and growth-associated protein-43 genes were likewise restored. Ischemia-induced increase of cytokine mRNA levels was abated by PUFA n3. Using an in vitro approach, we demonstrate that cultured astroglial and microglia directly respond to PUFA n3 administration by preventing ischemia-induced increase of cyclooxygenase 2, hypoxia-inducible factor 1alpha, inducible nitric oxide synthase, and interleukin 1beta. Cultured cortical neurons also appeared as direct targets, since PUFA n3 shifted the Bcl-2-like protein 4 (Bax)/B-cell lymphoma 2 (Bcl 2) ratio towards an anti-apoptotic constellation. Thus, PUFA n3 reveal a high neuroprotective and anti-inflammatory potential in an acute ischemic stroke model by targeting astroglial and microglial function as well as improving neuronal survival strategies. Our findings signify the potential clinical feasibility of PUFA n3 therapeutic treatment in stroke and other acute neurological diseases.
Collapse
Affiliation(s)
- Adib Zendedel
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Pardes Habib
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Jon Dang
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Leoni Lammerding
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Stefanie Hoffmann
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Medical School, RWTH Aachen University, 52074 Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| | - Alexander Slowik
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
24
|
Martin SAL, Jameson CH, Allan SM, Lawrence CB. Maternal high-fat diet worsens memory deficits in the triple-transgenic (3xTgAD) mouse model of Alzheimer's disease. PLoS One 2014; 9:e99226. [PMID: 24918775 PMCID: PMC4053375 DOI: 10.1371/journal.pone.0099226] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 05/12/2014] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is not normally diagnosed until later in life, although evidence suggests that the disease starts at a much earlier age. Risk factors for AD, such as diabetes, hypertension and obesity, are known to have their affects during mid-life, though events very early in life, including maternal over-nutrition, can predispose offspring to develop these conditions. This study tested whether over-nutrition during pregnancy and lactation affected the development of AD in offspring, using a transgenic AD mouse model. Female triple-transgenic AD dam mice (3xTgAD) were exposed to a high-fat (60% energy from fat) or control diet during pregnancy and lactation. After weaning (at 3 weeks of age), female offspring were placed on a control diet and monitored up until 12 months of age during which time behavioural tests were performed. A transient increase in body weight was observed in 4-week-old offspring 3xTgAD mice from dams fed a high-fat diet. However, by 5 weeks of age the body weight of 3xTgAD mice from the maternal high-fat fed group was no different when compared to control-fed mice. A maternal high-fat diet led to a significant impairment in memory in 2- and 12-month-old 3xTgAD offspring mice when compared to offspring from control fed dams. These effects of a maternal high-fat diet on memory were accompanied by a significant increase (50%) in the number of tau positive neurones in the hippocampus. These data demonstrate that a high-fat diet during pregnancy and lactation increases memory impairments in female 3xTgAD mice and suggest that early life events during development might influence the onset and progression of AD later in life.
Collapse
Affiliation(s)
- Sarah A. L. Martin
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Stuart M. Allan
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Catherine B. Lawrence
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
25
|
Barcelos RCS, Vey LT, Segat HJ, Roversi K, Roversi K, Dias VT, Trevizol F, Kuhn FT, Dolci GS, Pase CS, Piccolo J, Veit JC, Emanuelli T, Luz SCA, Bürger ME. Cross-generational trans fat intake exacerbates UV radiation-induced damage in rat skin. Food Chem Toxicol 2014; 69:38-45. [PMID: 24694906 DOI: 10.1016/j.fct.2014.03.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/28/2014] [Accepted: 03/25/2014] [Indexed: 01/11/2023]
Abstract
We evaluated the influence of dietary fats on ultraviolet radiation (UVR)-induced oxidative damage in skin of rats. Animals from two consecutive generations born of dams supplemented with fats during pregnancy and breastfeeding were maintained in the same supplementation: soybean-oil (SO, rich in n-6 FA, control group), fish-oil (FO, rich in n-3 FA) or hydrogenated-vegetable-fat (HVF, rich in TFA). At 90 days of age, half the animals from the 2nd generation were exposed to UVR (0.25 J/cm(2)) 3×/week for 12 weeks. The FO group presented higher incorporation of n-3 FA in dorsal skin, while the HVF group incorporated TFA. Biochemical changes per se were observed in skin of the HVF group: greater generation of reactive oxygen species (ROS), lower mitochondrial integrity and increased Na(+)K(+)-ATPase activity. UVR exposure increased skin wrinkles scores and ROS generation and decreased mitochondrial integrity and reduced-glutathione levels in the HVF group. In FO, UVR exposure was associated with smaller skin thickness and reduced levels of protein-carbonyl, together with increased catalase activity and preserved Na(+)K(+)-ATPase function. In conclusion, while FO may be protective, trans fat may be harmful to skin health by making it more vulnerable to UVR injury and thus more prone to develop photoaging and skin cancer.
Collapse
Affiliation(s)
- R C S Barcelos
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil
| | - L T Vey
- Departamento de Fisiologia e Farmacologia, UFSM, RS, Brazil
| | - H J Segat
- Departamento de Fisiologia e Farmacologia, UFSM, RS, Brazil
| | - K Roversi
- Departamento de Fisiologia e Farmacologia, UFSM, RS, Brazil
| | - Kr Roversi
- Departamento de Fisiologia e Farmacologia, UFSM, RS, Brazil
| | - V T Dias
- Departamento de Fisiologia e Farmacologia, UFSM, RS, Brazil
| | - F Trevizol
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil
| | - F T Kuhn
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil
| | - G S Dolci
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil
| | - C S Pase
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil
| | - J Piccolo
- Departamento de Tecnologia dos Alimentos, Programa de Pós-Graduação em Ciência Tecnologia dos Alimentos, UFSM, RS, Brazil
| | - J C Veit
- Departamento de Tecnologia dos Alimentos, Programa de Pós-Graduação em Ciência Tecnologia dos Alimentos, UFSM, RS, Brazil
| | - T Emanuelli
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil; Departamento de Tecnologia dos Alimentos, Programa de Pós-Graduação em Ciência Tecnologia dos Alimentos, UFSM, RS, Brazil
| | - S C A Luz
- Departamento de Patologia, UFSM, RS, Brazil
| | - M E Bürger
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil; Departamento de Fisiologia e Farmacologia, UFSM, RS, Brazil.
| |
Collapse
|
26
|
Knight EM, Martins IVA, Gümüsgöz S, Allan SM, Lawrence CB. High-fat diet-induced memory impairment in triple-transgenic Alzheimer's disease (3xTgAD) mice is independent of changes in amyloid and tau pathology. Neurobiol Aging 2014; 35:1821-32. [PMID: 24630364 PMCID: PMC4024197 DOI: 10.1016/j.neurobiolaging.2014.02.010] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/05/2014] [Accepted: 02/10/2014] [Indexed: 01/05/2023]
Abstract
Obesity and consumption of a high-fat diet are known to increase the risk of Alzheimer's disease (AD). Diets high in fat also increase disease neuropathology and/or cognitive deficits in AD mouse models. However, the effect of a high-fat diet on both the neuropathology and memory impairments in the triple-transgenic mouse model of AD (3xTgAD) is unknown. Therefore, groups of 2-month-old male 3xTgAD and control (non-Tg) mice were maintained on a high-fat or control diet and memory was assessed at the age of 3-4, 7-8, 11-12, and 15-16 months using a series of behavioral tests. A comparable increase in body weight was observed in non-Tg and 3xTgAD mice after high-fat feeding at all ages tested but a significantly greater increase in epididymal adipose tissue was observed in 3xTgAD mice at the age of 7-8, 11-12, and 15-16 months. A high-fat diet caused memory impairments in non-Tg control mice as early as the age of 3-4 months. In 3xTgAD mice, high-fat consumption led to a reduction in the age of onset and an increase in the extent of memory impairments. Some of these effects of high-fat diet on cognition in non-Tg and 3xTgAD mice were transient, and the age at which cognitive impairment was detected depended on the behavioral test. The effect of high-fat diet on memory in the 3xTgAD mice was independent of changes in AD neuropathology as no significant differences in (plaques, oligomers) or tau neuropathology were observed. An acute increase in microglial activation was seen in high-fat fed 3xTgAD mice at the age of 3-4 months but in non-Tg control mice microglial activation was not observed until the age of 15-16 months. These data indicate therefore that a high-fat diet has rapid and long-lasting negative effects on memory in both control and AD mice that are associated with neuroinflammation, but independent of changes in beta amyloid and tau neuropathology in the AD mice.
Collapse
Affiliation(s)
- Elysse M Knight
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | | | - Sarah Gümüsgöz
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | - Stuart M Allan
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | | |
Collapse
|
27
|
Loef M, Walach H. The omega-6/omega-3 ratio and dementia or cognitive decline: a systematic review on human studies and biological evidence. J Nutr Gerontol Geriatr 2013; 32:1-23. [PMID: 23451843 DOI: 10.1080/21551197.2012.752335] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
It has been suggested that the intake of certain fatty acids may influence the risk of dementia. However, current reviews have focused only on the therapeutic effects of omega-3 fatty acids, mostly as supplements. To date, the evidence for the relevance of the omega-6/omega-3 ratio has been neglected. Therefore, we searched the databases Alois, Medline, Biosis, Embase, Cochrane Central Register of Controlled Trials, and The Cochrane Database of Systematic Reviews for "essential fatty acids" and "dementia" and aimed to conduct a comprehensive review across study types. All studies that reported on the association between the n-6/n-3 ratio and dementia or cognitive decline were selected. In the 13 animal studies we examined, the dietary n-6/n-3 ratio was shown to affect brain composition, Alzheimer's disease pathology, and behavior. Our review of the 14 studies in humans that fulfilled the selection criteria (7 prospective studies, 3 cross-sectional studies, 1 controlled trial, 3 case-control studies) provided evidence, albeit limited, supporting an association between the n-6/n-3 ratio, cognitive decline, and incidence of dementia. This review supports growing evidence of a positive association between the dietary n-6/n-3 ratio and the risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Martin Loef
- Institute of Transcultural Health Studies, European University Viadrina, Frankfurt (Oder), Germany
| | | |
Collapse
|
28
|
Arsenault D, Dal-Pan A, Tremblay C, Bennett DA, Guitton MJ, De Koninck Y, Tonegawa S, Calon F. PAK inactivation impairs social recognition in 3xTg-AD Mice without increasing brain deposition of tau and Aβ. J Neurosci 2013; 33:10729-40. [PMID: 23804095 PMCID: PMC4019789 DOI: 10.1523/jneurosci.1501-13.2013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 05/13/2013] [Indexed: 11/21/2022] Open
Abstract
Defects in p21-activated kinase (PAK) are suspected to play a role in cognitive symptoms of Alzheimer's disease (AD). Dysfunction in PAK leads to cofilin activation, drebrin displacement from its actin-binding site, actin depolymerization/severing, and, ultimately, defects in spine dynamics and cognitive impairment in mice. To determine the role of PAK in AD, we first quantified PAK by immunoblotting in homogenates from the parietal neocortex of subjects with a clinical diagnosis of no cognitive impairment (n = 12), mild cognitive impairment (n = 12), or AD (n = 12). A loss of total PAK, detected in the cortex of AD patients (-39% versus controls), was correlated with cognitive impairment (r(2) = 0.148, p = 0.027) and deposition of total and phosphorylated tau (r(2) = 0.235 and r(2) = 0.206, respectively), but not with Aβ42 (r(2) = 0.056). Accordingly, we found a decrease of total PAK in the cortex of 12- and 20-month-old 3xTg-AD mice, an animal model of AD-like Aβ and tau neuropathologies. To determine whether PAK dysfunction aggravates AD phenotype, 3xTg-AD mice were crossed with dominant-negative PAK mice. PAK inactivation led to obliteration of social recognition in old 3xTg-AD mice, which was associated with a decrease in cortical drebrin (-25%), but without enhancement of Aβ/tau pathology or any clear electrophysiological signature. Overall, our data suggest that PAK decrease is a consequence of AD neuropathology and that therapeutic activation of PAK may exert symptomatic benefits on high brain function.
Collapse
Affiliation(s)
- Dany Arsenault
- Faculté de pharmacie, Université Laval, Quebec City, G1V 0A6, Quebec, Canada
- Centre Hospitalier de l'Université Laval, Research Center, Quebec City, Quebec, G1V 2L9, Canada
| | - Alexandre Dal-Pan
- Centre Hospitalier de l'Université Laval, Research Center, Quebec City, Quebec, G1V 2L9, Canada
| | - Cyntia Tremblay
- Centre Hospitalier de l'Université Laval, Research Center, Quebec City, Quebec, G1V 2L9, Canada
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612
| | - Matthieu J. Guitton
- Faculté de médecine, Université Laval, Quebec City, Quebec, G1V 0A6, Canada
- Institut Universitaire en Santé Mentale de Québec, Quebec City, Quebec, G1J 2G3, Canada, and
| | - Yves De Koninck
- Faculté de médecine, Université Laval, Quebec City, Quebec, G1V 0A6, Canada
- Institut Universitaire en Santé Mentale de Québec, Quebec City, Quebec, G1J 2G3, Canada, and
| | - Susumu Tonegawa
- Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Quebec City, G1V 0A6, Quebec, Canada
- Centre Hospitalier de l'Université Laval, Research Center, Quebec City, Quebec, G1V 2L9, Canada
| |
Collapse
|
29
|
de Souza AS, Rocha MS, Tavares do Carmo MDG. Effects of a normolipidic diet containing trans fatty acids during perinatal period on the growth, hippocampus fatty acid profile, and memory of young rats according to sex. Nutrition 2012; 28:458-64. [DOI: 10.1016/j.nut.2011.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/13/2011] [Accepted: 08/17/2011] [Indexed: 10/14/2022]
|
30
|
Grimm MOW, Rothhaar TL, Grösgen S, Burg VK, Hundsdörfer B, Haupenthal VJ, Friess P, Kins S, Grimm HS, Hartmann T. Trans fatty acids enhance amyloidogenic processing of the Alzheimer amyloid precursor protein (APP). J Nutr Biochem 2011; 23:1214-23. [PMID: 22209004 DOI: 10.1016/j.jnutbio.2011.06.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 06/22/2011] [Accepted: 06/29/2011] [Indexed: 01/20/2023]
Abstract
Hydrogenation of oils and diary products of ruminant animals leads to an increasing amount of trans fatty acids in the human diet. Trans fatty acids are incorporated in several lipids and accumulate in the membrane of cells. Here we systematically investigate whether the regulated intramembrane proteolysis of the amyloid precursor protein (APP) is affected by trans fatty acids compared to the cis conformation. Our experiments clearly show that trans fatty acids compared to cis fatty acids increase amyloidogenic and decrease nonamyloidogenic processing of APP, resulting in an increased production of amyloid beta (Aβ) peptides, main components of senile plaques, which are a characteristic neuropathological hallmark for Alzheimer's disease (AD). Moreover, our results show that oligomerization and aggregation of Aβ are increased by trans fatty acids. The mechanisms identified by this in vitro study suggest that the intake of trans fatty acids potentially increases the AD risk or causes an earlier onset of the disease.
Collapse
Affiliation(s)
- Marcus O W Grimm
- Deutsches Institut für DemenzPrävention (DIDP), Neurodegeneration and Neurobiology, 66421 Homburg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Bowman GL, Silbert LC, Howieson D, Dodge HH, Traber MG, Frei B, Kaye JA, Shannon J, Quinn JF. Nutrient biomarker patterns, cognitive function, and MRI measures of brain aging. Neurology 2011; 78:241-9. [PMID: 22205763 DOI: 10.1212/wnl.0b013e3182436598] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To examine the cross-sectional relationship between nutrient status and psychometric and imaging indices of brain health in dementia-free elders. METHODS Thirty plasma biomarkers of diet were assayed in the Oregon Brain Aging Study cohort (n = 104). Principal component analysis constructed nutrient biomarker patterns (NBPs) and regression models assessed the relationship of these with cognitive and MRI outcomes. RESULTS Mean age was 87 ± 10 years and 62% of subjects were female. Two NBPs associated with more favorable cognitive and MRI measures: one high in plasma vitamins B (B1, B2, B6, folate, and B12), C, D, and E, and another high in plasma marine ω-3 fatty acids. A third pattern characterized by high trans fat was associated with less favorable cognitive function and less total cerebral brain volume. Depression attenuated the relationship between the marine ω-3 pattern and white matter hyperintensity volume. CONCLUSION Distinct nutrient biomarker patterns detected in plasma are interpretable and account for a significant degree of variance in both cognitive function and brain volume. Objective and multivariate approaches to the study of nutrition in brain health warrant further study. These findings should be confirmed in a separate population.
Collapse
Affiliation(s)
- G L Bowman
- Department of Neurology, Oregon Health & Science University, Portland, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Tsai SJ, Liu WH, Yin MC. Trans Fatty Acids Enhanced Beta-Amyloid Induced Oxidative Stress in Nerve Growth Factor Differentiated PC12 Cells. Neurochem Res 2011; 37:786-94. [DOI: 10.1007/s11064-011-0673-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 12/07/2011] [Accepted: 12/15/2011] [Indexed: 11/29/2022]
|
33
|
Zhao Y, Calon F, Julien C, Winkler JW, Petasis NA, Lukiw WJ, Bazan NG. Docosahexaenoic acid-derived neuroprotectin D1 induces neuronal survival via secretase- and PPARγ-mediated mechanisms in Alzheimer's disease models. PLoS One 2011; 6:e15816. [PMID: 21246057 PMCID: PMC3016440 DOI: 10.1371/journal.pone.0015816] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 12/02/2010] [Indexed: 01/22/2023] Open
Abstract
Neuroprotectin D1 (NPD1) is a stereoselective mediator derived from the omega-3 essential fatty acid docosahexaenoic acid (DHA) with potent inflammatory resolving and neuroprotective bioactivity. NPD1 reduces Aβ42 peptide release from aging human brain cells and is severely depleted in Alzheimer's disease (AD) brain. Here we further characterize the mechanism of NPD1's neurogenic actions using 3xTg-AD mouse models and human neuronal-glial (HNG) cells in primary culture, either challenged with Aβ42 oligomeric peptide, or transfected with beta amyloid precursor protein (βAPP)(sw) (Swedish double mutation APP695(sw), K595N-M596L). We also show that NPD1 downregulates Aβ42-triggered expression of the pro-inflammatory enzyme cyclooxygenase-2 (COX-2) and of B-94 (a TNF-α-inducible pro-inflammatory element) and apoptosis in HNG cells. Moreover, NPD1 suppresses Aβ42 peptide shedding by down-regulating β-secretase-1 (BACE1) while activating the α-secretase ADAM10 and up-regulating sAPPα, thus shifting the cleavage of βAPP holoenzyme from an amyloidogenic into the non-amyloidogenic pathway. Use of the thiazolidinedione peroxisome proliferator-activated receptor gamma (PPARγ) agonist rosiglitazone, the irreversible PPARγ antagonist GW9662, and overexpressing PPARγ suggests that the NPD1-mediated down-regulation of BACE1 and Aβ42 peptide release is PPARγ-dependent. In conclusion, NPD1 bioactivity potently down regulates inflammatory signaling, amyloidogenic APP cleavage and apoptosis, underscoring the potential of this lipid mediator to rescue human brain cells in early stages of neurodegenerations.
Collapse
Affiliation(s)
- Yuhai Zhao
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Frederic Calon
- Molecular Endocrinology and Oncology Research Centre, Centre Hospitalier de l'Université Laval Research Centre (CHUL), Quebec, Canada
| | - Carl Julien
- Molecular Endocrinology and Oncology Research Centre, Centre Hospitalier de l'Université Laval Research Centre (CHUL), Quebec, Canada
| | - Jeremy W. Winkler
- Department of Chemistry, Loker Hydrocarbon Research Institute, University of Southern California, Los Angeles, California, United States of America
| | - Nicos A. Petasis
- Department of Chemistry, Loker Hydrocarbon Research Institute, University of Southern California, Los Angeles, California, United States of America
| | - Walter J. Lukiw
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Nicolas G. Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| |
Collapse
|
34
|
Hamilton LK, Aumont A, Julien C, Vadnais A, Calon F, Fernandes KJL. Widespread deficits in adult neurogenesis precede plaque and tangle formation in the 3xTg mouse model of Alzheimer's disease. Eur J Neurosci 2010; 32:905-20. [PMID: 20726889 DOI: 10.1111/j.1460-9568.2010.07379.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) affects cognitive modalities that are known to be regulated by adult neurogenesis, such as hippocampal- and olfactory-dependent learning and memory. However, the relationship between AD-associated pathologies and alterations in adult neurogenesis has remained contentious. In the present study, we performed a detailed investigation of adult neurogenesis in the triple transgenic (3xTg) mouse model of AD, a unique model that generates both amyloid plaques and neurofibrillary tangles, the hallmark pathologies of AD. In both neurogenic niches of the brain, the hippocampal dentate gyrus and forebrain subventricular zone, we found that 3xTg mice had decreased numbers of (i) proliferating cells, (ii) early lineage neural progenitors, and (iii) neuroblasts at middle age (11months old) and old age (18months old). These decreases correlated with major reductions in the addition of new neurons to the respective target areas, the dentate granule cell layer and olfactory bulb. Within the subventricular zone niche, cytological alterations were observed that included a selective loss of subependymal cells and the development of large lipid droplets within the ependyma of 3xTg mice, indicative of metabolic changes. Temporally, there was a marked acceleration of age-related decreases in 3xTg mice, which affected multiple stages of neurogenesis and was clearly apparent prior to the development of amyloid plaques or neurofibrillary tangles. Our findings indicate that AD-associated mutations suppress neurogenesis early during disease development. This suggests that deficits in adult neurogenesis may mediate premature cognitive decline in AD.
Collapse
Affiliation(s)
- Laura K Hamilton
- Département de pathologie et biologie cellulaire, Université de Montréal, C.P. 6128, succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
AD (Alzheimer's disease) is a progressive neurodegenerative disease of unknown origin. Despite questions as to the underlying cause(s) of this disease, shared risk factors for both AD and atherosclerotic cardiovascular disease indicate that vascular mechanisms may critically contribute to the development and progression of both AD and atherosclerosis. An increased risk of developing AD is linked to the presence of the apoE4 (apolipoprotein E4) allele, which is also strongly associated with increased risk of developing atherosclerotic cardiovascular disease. Recent studies also indicate that cardiovascular risk factors, including elevated blood cholesterol and triacylglycerol (triglyceride), increase the likelihood of AD and vascular dementia. Lipids and lipoproteins in the circulation interact intimately with the cerebrovasculature, and may have important effects on its constituent brain microvascular endothelial cells and the adjoining astrocytes, which are components of the neurovascular unit. The present review will examine the potential mechanisms for understanding the contributions of vascular factors, including lipids, lipoproteins and cerebrovascular Abeta (amyloid beta), to AD, and suggest therapeutic strategies for the attenuation of this devastating disease process. Specifically, we will focus on the actions of apoE, TGRLs (triacylglycerol-rich lipoproteins) and TGRL lipolysis products on injury of the neurovascular unit and increases in blood-brain barrier permeability.
Collapse
|
36
|
Current Opinion in Clinical Nutrition and Metabolic Care. Current world literature. Curr Opin Clin Nutr Metab Care 2010; 13:215-21. [PMID: 20145440 DOI: 10.1097/mco.0b013e32833643b4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
37
|
Long-term effects of high lipid and high energy diet on serum lipid, brain fatty acid composition, and memory and learning ability in mice. Int J Dev Neurosci 2009; 28:271-6. [PMID: 20015474 DOI: 10.1016/j.ijdevneu.2009.12.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 11/15/2009] [Accepted: 12/07/2009] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE It is well known that high lipid and high energy diet is harmful to health. But the different effects of high lipid diet composed of either saturated fatty acids or unsaturated fatty acids have not been distinguished. METHOD Eighteen pregnant C57BL/6j (22-25g) mice were randomly divided into three groups of six each and fed with chow or high lipid diet composed of either flaxseed oil (chow diet 84%, cholesterol 0.2%, flaxseed oil 15.8%) or lard fat (chow diet 84%, cholesterol 0.2%, lard fat 15.8%). After weaning, the offspring were fed the same diet as their mothers were fed during the experiment, and their spatial memory and learning ability were evaluated by Morris water maze when they were 8 weeks old. Next, the blood and tissues were sampled when they were 9 weeks old. Serum lipids were determined using kits, and brain fatty acids were measured using a gas chromatograph. RESULTS Compared to chow diet (control), high flaxseed oil diet (HFO) increased high density lipoprotein cholesterol level (HDL-C) in the mothers but not in offspring; high lard fat diet (HLF) increased serum total cholesterol level (TC) and low density lipoprotein cholesterol level (LDL-C) both in mothers and offspring. Brain fatty acids profile was altered by HLF compared with chow diet. Polyunsaturated fatty acids and long-chain polyunsaturated fatty acids content were significantly lower in the HLF group than in the control group, but saturated fatty acids content were significantly higher in HLF group than those in control group. The changed fatty acids composition affected the spatial memory and learning ability of adult offspring. CONCLUSIONS A long-term high lard diet increased offspring serum TC and LDL-C levels and affected the brain's fatty acid composition, and memory and learning ability. The polyunsaturated fatty acid content of the brain may be correlated with serum cholesterol levels.
Collapse
|