Docosahexaenoic and Arachidonic Acids as Neuroprotective Nutrients throughout the Life Cycle

Nutrients - Tập 13 Số 3 - Trang 986
Verónica Sambra1, Francisca Echeverría1, Alfonso Valenzuela2, Raphaël Chouinard‐Watkins3, Rodrigo Valenzuela1,3
1Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
2Faculty of Medicine, School of Nutrition, Universidad de Los Andes, Santiago 8380000, Chile
3Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada

Tóm tắt

The role of docosahexaenoic acid (DHA) and arachidonic acid (AA) in neurogenesis and brain development throughout the life cycle is fundamental. DHA and AA are long-chain polyunsaturated fatty acids (LCPUFA) vital for many human physiological processes, such as signaling pathways, gene expression, structure and function of membranes, among others. DHA and AA are deposited into the lipids of cell membranes that form the gray matter representing approximately 25% of the total content of brain fatty acids. Both fatty acids have effects on neuronal growth and differentiation through the modulation of the physical properties of neuronal membranes, signal transduction associated with G proteins, and gene expression. DHA and AA have a relevant role in neuroprotection against neurodegenerative pathologies such as Alzheimer’s disease and Parkinson’s disease, which are associated with characteristic pathological expressions as mitochondrial dysfunction, neuroinflammation, and oxidative stress. The present review analyzes the neuroprotective role of DHA and AA in the extreme stages of life, emphasizing the importance of these LCPUFA during the first year of life and in the developing/prevention of neurodegenerative diseases associated with aging.

Từ khóa


Tài liệu tham khảo

Svennerholm, 1968, Distribution and fatty acid composition of phosphoglycerides in normal human brain, J. Lipid Res., 9, 570, 10.1016/S0022-2275(20)42702-6

Crawford, 2012, The role of docosahexaenoic and the marine food web as determinants of evolution and hominid brain development: The challenge for human sustainability, Nutr. Health, 21, 17, 10.1177/0260106012437550

Carlson, 2016, Docosahexaenoic Acid and Arachidonic Acid Nutrition in Early Development, Adv. Pediatrics, 63, 453, 10.1016/j.yapd.2016.04.011

2008, Fatty acids and neurodevelopment, J. Pediatr. Gastroenterol. Nutr., 47, S7

Innis, 2008, Dietary omega 3 fatty acids and the developing brain, Brain Res., 1237, 35, 10.1016/j.brainres.2008.08.078

Weiser, M.J., Butt, C.M., and Mohajeri, M.H. (2016). Docosahexaenoic Acid and Cognition throughout the Lifespan. Nutrinets, 8.

Brenna, 2007, The influence of dietary docosahexaenoic acid and arachidonic acid on central nervous system polyunsaturated fatty acid composition, Prostaglandins Leukot. Essent. Fatty Acids, 77, 247, 10.1016/j.plefa.2007.10.016

Bourre, 2000, Specific phospholipid fatty acid composition of brain regions in mice: Effects of n–3 polyunsaturated fatty acid deficiency and phospholipid supplementation, J. Lipid Res., 41, 465, 10.1016/S0022-2275(20)34485-0

Garcia, 2002, Effect of Docosahexaenoic Acid on the Synthesis of Phosphatidylserine in Rat Brain Microsomes and C6 Glioma Cells, J. Neurochem., 70, 24, 10.1046/j.1471-4159.1998.70010024.x

Gawrisch, 2003, The structure of DHA in phospholipid membranes, Lipids, 38, 445, 10.1007/s11745-003-1082-0

Valenzuela, 2013, Docosahexaenoic acid (DHA), an essential fatty acid at the brain, Rev. Chil. Nutr., 40, 383

Katakura, 2009, Docosahexaenoic acid promotes neuronal differentiation by regulating basic helix–loop–helix transcription factors and cell cycle in neural stem cells, Neuroscience, 160, 651, 10.1016/j.neuroscience.2009.02.057

Jumpsen, 1997, Small changes of dietary (n-6) and (n-3)/fatty acid content ration alter phosphatidylethanolamine and phosphatidylcholine fatty acid composition during development of neuronal and glial cells in rats, J. Nutr., 127, 724, 10.1093/jn/127.5.724

Kyle, 1999, Low serum docosahexaenoic acid is a significant risk factor for alzheimer’s dementia, Lipids, 34, S245, 10.1007/BF02562306

Heude, 2003, Cognitive decline and fatty acid composition of erythrocyte membranes—The EVA Study, Am. J. Clin. Nutr., 77, 803, 10.1093/ajcn/77.4.803

Beydoun, 2007, Plasma n-3 fatty acids and the risk of cognitive decline in older adults: The Atherosclerosis Risk in Communities Study, Am. J. Clin. Nutr., 85, 1103, 10.1093/ajcn/85.4.1103

Conquer, 2000, Fatty acid analysis of blood plasma of patients with Alzheimer’s disease, other types of dementia, and cognitive impairment, Lipids, 35, 1305, 10.1007/s11745-000-0646-3

Schaefer, 2006, Plasma phosphatidylcholine docosahexaenoic acid content and risk of dementia and Alzheimer disease: The Framingham Heart Study, Arch. Neurol., 63, 1545, 10.1001/archneur.63.11.1545

Yurko-Mauro, K., Alexander, D.D., and van Elswyk, M.E. (2015). Docosahexaenoic acid and adult memory: A systematic review and meta-analysis. PLoS ONE, 10.

Morris, 2003, Consumption of Fish and n-3 Fatty Acids and Risk of Incident Alzheimer Disease, Arch. Neurol., 60, 940, 10.1001/archneur.60.7.940

Chang, 2012, Docosahexaenoic acid promotes dopaminergic differentiation in induced pluripotent stem cells and inhibits teratoma formation in rats with Parkinson-like pathology, Cell Transplant., 21, 313, 10.3727/096368911X580572

Ozkan, 2016, The protective mechanism of docosahexaenoic acid in mouse model of Parkinson: The role of heme oxygenase, Neurochem. Int., 101, 110, 10.1016/j.neuint.2016.10.012

Mori, 2018, Neuroprotective effect of omega-3 polyunsaturated fatty acids in the 6-OHDA model of Parkinson’s disease is mediated by a reduction of inducible nitric oxide synthase, Nutr. Neurosci., 21, 341, 10.1080/1028415X.2017.1290928

Derbyshire, E. (2018). Brain Health across the Lifespan: A Systematic Review on the Role of Omega-3 Fatty Acid Supplements. Nutrients, 10.

Lauritzen, L., Brambilla, P., Mazzocchi, A., Harsløf, L.B., Ciappolino, V., and Agostoni, C. (2016). DHA Effects in Brain Development and Function. Nutrients, 8.

Hadley, K.B., Ryan, A.S., Forsyth, S., Gautier, S., and Salem, N. (2016). The Essentiality of Arachidonic Acid in Infant Development. Nutrients, 8.

Gao, 2010, Quantifying conversion of linoleic to arachidonic and other n-6 polyunsaturated fatty acids in unanesthetized rats, J. Lipid Res., 51, 2940, 10.1194/jlr.M005595

Brenna, 2016, Arachidonic acid needed in infant formula when docosahexaenoic acid is present, Nutr. Rev., 74, 329, 10.1093/nutrit/nuw007

Harauma, 2017, Effects of arachidonic acid, eicosapentaenoic acid and docosahexaenoic acid on brain development using artificial rearing of delta-6-desaturase knockout mice, Prostaglandins Leukot. Essent. Fat. Acids, 127, 32, 10.1016/j.plefa.2017.10.001

Wilson, 1993, Lipid and fatty acid composition of brain tissue from adrenoleukodystrophy patients, J. Neurochem., 61, 290, 10.1111/j.1471-4159.1993.tb03567.x

Sampson, 1965, Fatty acid and fatty aldehyde composition of the major brain lipids in normal human gray matter, white matter, and myelin, J. Lipid Res., 6, 545, 10.1016/S0022-2275(20)39620-6

Jahn, 2006, SNAREs—Engines for membrane fusion, Nat. Rev. Mol. Cell Biol., 7, 631, 10.1038/nrm2002

Rickman, 2005, Arachidonic Acid Allows SNARE Complex Formation in the Presence of Munc18, Chem. Biol., 12, 545, 10.1016/j.chembiol.2005.03.004

Connell, 2007, Mechanism of arachidonic acid action on syntaxin–Munc18, EMBO Rep., 8, 414, 10.1038/sj.embor.7400935

Latham, 2006, Arachidonic acid potentiates exocytosis and allows neuronal SNARE complex to interact with Munc18a, J. Neurochem., 100, 1543, 10.1111/j.1471-4159.2006.04286.x

Tokuda, 2014, Differential effect of arachidonic acid and docosahexaenoic acid on age-related decreases in hippocampal neurogenesis, Neurosci. Res., 88, 58, 10.1016/j.neures.2014.08.002

Ho, 2001, Neuronal Cyclooxygenase 2 Expression in the Hippocampal Formation as a Function of the Clinical Progression of Alzheimer Disease, Arch. Neurol., 58, 487, 10.1001/archneur.58.3.487

Fujimi, 2007, Altered Expression of COX-2 in Subdivisions of the Hippocampus during Aging and in Alzheimer’s Disease: The Hisayama Study, Dement. Geriatr. Cogn. Disord., 23, 423, 10.1159/000101957

Ching, Y.K., Chin, Y.S., Appukutty, M., Ramanchadran, V., Yu, C.Y., Ang, G.Y., Gan, W.Y., Chan, Y.M., Teh, L.K., and Salleh, M.Z. (2019). Interaction of Dietary Linoleic Acid and α-Linolenic Acids with rs174547 in FADS1 Gene on Metabolic Syndrome Components among Vegetarians. Nutrinets, 11.

Valenzuela, 2016, Vegetable oils rich in alpha linolenic acid increment hepatic n-3 LCPUFA, modulating the fatty acid metabolism and antioxidant response in rats, Prostaglandins Leukot. Essent. Fat. Acids, 111, 25, 10.1016/j.plefa.2016.02.002

Gonzalez-Soto, M., and Mutch, D.M. (2020). Diet Regulation of Long-Chain PUFA Synthesis: Role of Macronutrients, Micronutrients, and Polyphenols on Δ-5/Δ-6 Desaturases and Elongases 2/5. Adv. Nutr., 142.

Kang, 2003, The Importance of Omega-6/Omega-3 Fatty Acid Ratio in Cell Function, World Rev. Nutr. Diet., 92, 23, 10.1159/000073790

Valenzuela, 2011, The importance of the long-chain polyunsaturated fatty acid n-6/n-3 ratio in development of non-alcoholic fatty liver associated with obesity, Food Funct., 2, 644, 10.1039/c1fo10133a

Food and Agriculture Organization (2008). Fatty Acids in Human Nutrition. Report of an Expert Consultation, Food and Agriculture Organization of the United Nations.

Brayner, B., Kaur, G., Keske, M.A., and Livingstone, K.M. (2018). FADS Polymorphism, Omega-3 Fatty Acids and Diabetes Risk: A Sys-tematic Review. Nutrients, 13.

Chen, 2019, Effects of the rs3834458 Single Nucleotide Polymorphism in FADS2 on Levels of n-3 Long-chain Polyunsaturated Fatty Acids: A Meta-analysis, Prostaglandins Leukot. Essent. Fat. Acids, 150, 1, 10.1016/j.plefa.2019.08.005

Xie, 2008, Genetic Variants of the FADS1 FADS2 Gene Cluster Are Associated with Altered (n-6) and (n-3) Essential Fatty Acids in Plasma and Erythrocyte Phospholipids in Women during Pregnancy and in Breast Milk during Lactation, J. Nutr., 138, 2222, 10.3945/jn.108.096156

Steer, C.D., Smith, G.D., Emmett, P.M., Hibbeln, J.R., and Golding, J. (2010). FADS2 Polymorphisms Modify the Effect of Breastfeeding on Child IQ. PLoS ONE, 5.

Miliku, 2019, Human milk fatty acid composition is associated with dietary, genetic, sociodemographic, and environmental factors in the CHILD Cohort Study, Am. J. Clin. Nutr., 110, 1370, 10.1093/ajcn/nqz229

Koletzko, 2019, FADS1 and FADS2 Polymorphisms Modulate Fatty Acid Metabolism and Dietary Impact on Health, Annu. Rev. Nutr., 39, 21, 10.1146/annurev-nutr-082018-124250

Xu, 2019, Association of Nonalcoholic Fatty Liver Disease and Coronary Artery Disease with FADS2 rs3834458 Gene Polymorphism in the Chinese Han Population, Gastroenterol. Res. Pr., 2019, 1

Valenzuela, 2014, N-3 long chain polyunsaturated fatty acids and cardiovascular disease, Rev. Chil. Nutr., 41, 319

Morales, 2012, New dietary sources of alpha-linolenic acid: A critical view, Rev. Chil. Nutr., 39, 79

Cholewski, M., Tomczykowa, M., and Tomczyk, M. (2018). A Comprehensive Review of Chemistry, Sources and Bioavailability of Omega-3 Fatty Acids. Nutrients, 10.

EFSA Panel on Dietetic Products (2010). Scientific Opinion on Dietary Reference Values for fats, including saturated fatty acids, pol-yunsaturated fatty acids, monounsaturated fatty acids, trans fatty acids, and cholesterol. EFSA J., 8, 1461.

Harris, 2009, Towards Establishing Dietary Reference Intakes for Eicosapentaenoic and Docosahexaenoic Acids, J. Nutr., 139, 804S, 10.3945/jn.108.101329

Butte, N.F., Lopez-Alarcon, M.G., and Garza, C. (2002). Nutrient Adequacy of Exclusive Breastfeeding for the Term Infant During the First Six Months of Life, World Health Organization.

Lauritzen, 2011, Maternal fatty acid status during pregnancy and lactation and relation to newborn and infant status, Matern. Child. Nutr., 7, 41, 10.1111/j.1740-8709.2011.00303.x

Koletzko, 2007, Placental transfer of long-chain polyunsaturated fatty acids (LC-PUFA), J. Peérinat. Med., 35, S5, 10.1515/JPM.2007.030

Demmelmair, 2015, Human lactation: Oxidation and maternal transfer of dietary 13C-labelled α-linolenic acid into human milk, Isot. Environ. Health Stud., 52, 270, 10.1080/10256016.2015.1071362

Koletzko, 2007, Dietary fat intakes for pregnant and lactating women, Br. J. Nutr., 98, 873, 10.1017/S0007114507764747

Yang, 2018, Nutritional composition of breast milk in Chinese women: A systematic review, Asia Pac. J. Clin. Nutr., 27, 491

Forsyth, 2017, The importance of dietary DHA and ARA in early life: A public health perspective, Proceedings of the Nutrition Society, Volume 76, 568, 10.1017/S0029665117000313

Hall, 1979, Uniformity of human milk, Am. J. Clin. Nutr., 32, 304, 10.1093/ajcn/32.2.304

Kim, 2017, Breast milk fatty acid composition and fatty acid intake of lactating mothers in South Korea, Br. J. Nutr., 117, 556, 10.1017/S0007114517000253

Brenna, 2007, Docosahexaenoic and arachidonic acid concentrations in human breast milk worldwide, Am. J. Clin. Nutr., 85, 1457, 10.1093/ajcn/85.6.1457

Koletzko, 2016, Human Milk Lipids, Ann. Nutr. Metab., 69, 28, 10.1159/000452819

Villalpando, 2001, Contribution of dietary and newly formed arachidonic acid to human milk lipids in women eating a low-fat diet, Am. J. Clin. Nutr., 74, 242, 10.1093/ajcn/74.2.242

Fu, 2016, An updated review of worldwide levels of docosahexaenoic and arachidonic acid in human breast milk by region, Public Health Nutr., 19, 2675, 10.1017/S1368980016000707

Lauritzen, 2001, The essentiality of long chain n-3 fatty acids in relation to development and function of the brain and retina, Prog. Lipid. Res., 40, 1, 10.1016/S0163-7827(00)00017-5

Elshani, 2021, Role of omega-3 polyunsaturated fatty acids in gestational diabetes, maternal and fetal insights: Current use and future directions, J. Matern. Neonatal. Med., 34, 124, 10.1080/14767058.2019.1593361

Álvarez, D., Muñoz, Y., Ortiz, M., Maliqueo, M., Chouinard-Watkins, R., and Valenzuela, R. (2020). Impact of Maternal Obesity on the Metabolism and Bioavailability of Polyunsaturated Fatty Acids during Pregnancy and Breastfeeding. Nutrients, 13.

Grote, 2016, Breast milk composition and infant nutrient intakes during the first 12 months of life, Eur. J. Clin. Nutr., 70, 250, 10.1038/ejcn.2015.162

Barrera, C., Valenzuela, R., Chamorro, R., Bascuñán, K., Sandoval, J., Sabag, N., Valenzuela, F., Valencia, M.P., Puigrredon, C., and Valenzuela, A. (2018). The Impact of Maternal Diet during Pregnancy and Lactation on the Fatty Acid Composition of Erythro-cytes and Breast Milk of Chilean Women. Nutrients, 10.

Devarshi, P.P., Grant, R.W., Ikonte, C.J., and Hazels-Mitmesser, S. (2019). Maternal Omega-3 Nutrition, Placental Transfer and Fetal Brain De-velopment in Gestational Diabetes and Preeclampsia. Nutrients, 11.

Mun, J.G., Legette, L.L., Ikonte, C.J., and Mitmesser, S.H. (2019). Choline and DHA in Maternal and Infant Nutrition: Synergistic Implications in Brain and Eye Health. Nutrients, 11.

Rodier, 2008, Chronology of Neuron Development: Animal Studies and their Clinical Implications, Dev. Med. Child. Neurol., 22, 525, 10.1111/j.1469-8749.1980.tb04363.x

Vasung, 2019, Exploring early human brain development with structural and physiological neuroimaging, NeuroImage, 187, 226, 10.1016/j.neuroimage.2018.07.041

Yamaguchi, 2012, How to form and close the brain: Insight into the mechanism of cranial neural tube closure in mammals, Cell. Mol. Life Sci., 70, 3171, 10.1007/s00018-012-1227-7

Wozniak, 2006, Advances in white matter imaging: A review of in vivo magnetic resonance methodologies and their applicability to the study of development and aging, Neurosci. Biobehav. Rev., 30, 762, 10.1016/j.neubiorev.2006.06.003

Lancaster, 2014, Generation of cerebral organoids from human pluripotent stem cells, Nat. Protoc., 9, 2329, 10.1038/nprot.2014.158

Kelava, 2016, Stem Cell Models of Human Brain Development, Cell Stem Cell, 18, 736, 10.1016/j.stem.2016.05.022

Tang, 2016, Maternal diet of polyunsaturated fatty acid altered the cell proliferation in the dentate gyrus of hippocampus and influenced glutamatergic and serotoninergic systems of neonatal female rats, Lipids Health Dis., 15, 1, 10.1186/s12944-016-0236-1

Derbyshire, E., and Obeid, R. (2020). Choline, Neurological Development and Brain Function: A Systematic Review Focusing on the First 1000 Days. Nutrients, 12.

Owji, 2020, The History of Discovery of Adult Neurogenesis, Clin. Anat., 33, 41, 10.1002/ca.23447

Dobbing, 1973, Quantitative growth and development of human brain, Arch. Dis. Child., 48, 757, 10.1136/adc.48.10.757

Kuipers, 2012, Fetal intrauterine whole body linoleic, arachidonic and docosahexaenoic acid contents and accretion rates, Prostaglandins Leukot. Essent. Fat. Acids, 86, 13, 10.1016/j.plefa.2011.10.012

Cocas, 2016, Cell Type-Specific Circuit Mapping Reveals the Presynaptic Connectivity of Developing Cortical Circuits, J. Neurosci., 36, 3378, 10.1523/JNEUROSCI.0375-15.2016

Lien, 2009, Dietary assessment of arachidonic acid and docosahexaenoic acid intake in 4–7 year-old children, J. Am. Coll. Nutr., 28, 7, 10.1080/07315724.2009.10719755

Lepping, 2019, Long-chain polyunsaturated fatty acid supplementation in the first year of life affects brain function, structure, and metabolism at age nine years, Dev. Psychobiol., 61, 5, 10.1002/dev.21780

Birch, 2010, The DIAMOND (DHA Intake and Measurement of Neural Development) Study: A double-masked, randomized controlled clinical trial of the maturation of infant visual acuity as a function of the dietary level of docosahexaenoic acid, Am. J. Clin. Nutr., 91, 848, 10.3945/ajcn.2009.28557

Colombo, 2017, Docosahexaenoic acid (DHA) and arachidonic acid (ARA) balance in developmental outcomes, Prostaglandins Leukot. Essent. Fat. Acids, 121, 52, 10.1016/j.plefa.2017.05.005

Koletzko, 2008, The roles of long-chain polyunsaturated fatty acids in pregnancy, lactation and infancy: Review of current knowledge and consensus recommendations, J. Peérinat. Med., 36, 5

Talamonti, 2019, Impairment of DHA synthesis alters the expression of neuronal plasticity markers and the brain inflammatory status in mice, FASEB J., 34, 2024, 10.1096/fj.201901890RR

Wainwright, 1997, Arachidonic Acid Offsets the Effects on Mouse Brain and Behavior of a Diet with a Low (n-6):(n-3) Ratio and Very High Levels of Docosahexaenoic Acid, J. Nutr., 127, 184, 10.1093/jn/127.1.184

Giusto, 2002, Age-associated changes in central nervous system glycerolipid composition and metabolism, Neurochem. Res., 27, 1513, 10.1023/A:1021604623208

Burrin, D.G. (2005). Biology of metabolism in growing animals. Essential Fatty Acid Metabolism During Early Development, Elsevier Science.

Poling, 1995, Time- and voltage-dependent block of delayed rectifier potassium channels by docosahexaenoic acid, Mol. Pharmacol., 47, 381

Strokin, 2006, Neuroprotection of rat hippocampal slices exposed to oxygen-glucose dep-rivation by enrichment with docosahexaenoic acid and by inhibition of hydrolysis of docosahexaenoic acid-containing phos-pholipids by calcium independent phospholipase A2, Neuroscience, 140, 547, 10.1016/j.neuroscience.2006.02.026

Chalon, 2002, α-Linolenic Acid Dietary Deficiency Alters Age-Related Changes of Dopaminergic and Serotoninergic Neurotransmission in the Rat Frontal Cortex, J. Neurochem., 66, 1582

Cheng, 2020, DHA reduces hypothalamic inflammation and improves central leptin signaling in mice, Life Sci., 257, 118036, 10.1016/j.lfs.2020.118036

Chalon, 1998, Dietary fish oil affects mon-oaminergic neurotransmission and behavior in rats, J. Nutr., 128, 2512, 10.1093/jn/128.12.2512

Feltham, 2020, Docosahexaenoic Acid: Outlining the Therapeutic Nutrient Potential to Combat the Prenatal Alcohol-Induced Insults on Brain Development, Adv. Nutr., 11, 724, 10.1093/advances/nmz135

Singh, 2012, Metabolic syndrome: A brain disease, Can. J. Physiol. Pharmacol., 90, 1171, 10.1139/y2012-122

Masliah, 2006, Synaptic remodeling during aging and in Alzheimer’s disease, J. Alzheimers Dis., 9, 91, 10.3233/JAD-2006-9S311

Martinez, 1992, Tissue levels of polyunsaturated fatty acids during early human development, J. Pediatr., 120, S129, 10.1016/S0022-3476(05)81247-8

Hsieh, 2007, The Influence of Moderate and High Dietary Long Chain Polyunsaturated Fatty Acids (LCPUFA) on Baboon Neonate Tissue Fatty Acids, Pediatr. Res., 61, 537, 10.1203/pdr.0b013e318045bec9

Wijendran, 2002, Significant utilization of dietary arachidonic acid is for brain adrenic acid in baboon neonates, J. Lipid Res., 43, 762, 10.1016/S0022-2275(20)30118-8

Alashmali, 2017, Maternal dietary n-6 polyunsaturated fatty acid deprivation does not exacerbate post-weaning reductions in arachidonic acid and its mediators in the mouse hippocampus, Nutr. Neurosci., 22, 223, 10.1080/1028415X.2017.1372160

Yang, 2012, Axonal Gradient of Arachidonic Acid-containing Phosphatidylcholine and Its Dependence on Actin Dynamics, J. Biol. Chem., 287, 5290, 10.1074/jbc.M111.316877

Darios, 2006, Omega-3 and omega-6 fatty acids stimulate cell membrane expansion by acting on syntaxin 3, Nat. Cell Biol., 440, 813

Novak, 2008, High dietary omega-6 fatty acids contribute to reduced docosahexaenoic acid in the developing brain and inhibit secondary neurite growth, Brain Res., 1237, 136, 10.1016/j.brainres.2008.07.107

Buckley, 2014, Proresolving lipid mediators and mechanisms in the resolution of acute inflam-mation, Immunity, 40, 315, 10.1016/j.immuni.2014.02.009

Dyall, 2017, Interplay between n-3 and n-6 Long-Chain Polyunsaturated Fatty Acids and the Endocannabinoid System in Brain Protection and Repair, Lipids, 52, 885, 10.1007/s11745-017-4292-8

Hammels, 2019, Novel CB1-ligands maintain homeostasis of the endocannabinoid system in ω3- and ω6-long-chain-PUFA deficiency, J. Lipid Res., 60, 1396, 10.1194/jlr.M094664

Watson, 2019, Emerging class of omega-3 fatty acid endocannabinoids & their derivatives, Prostaglandins Other Lipid Mediat., 143, 106337, 10.1016/j.prostaglandins.2019.106337

Simopoulos, 2002, The importance of the ratio of omega-6/omega-3 essential fatty acids, Biomed. Pharmacother., 56, 365, 10.1016/S0753-3322(02)00253-6

Wood, 2010, Dietary docosahexaenoic acid supplementation alters select physiological endocannabinoid-system metabolites in brain and plasma, J. Lipid Res., 51, 1416, 10.1194/jlr.M002436

Dauer, 2003, Parkinson’s disease: Mechanisms and models, Neuron, 11, 889, 10.1016/S0896-6273(03)00568-3

Camilleri, 2014, The Centrality of Mitochondria in the Pathogenesis and Treatment of Parkinson’s Disease, CNS Neurosci. Ther., 20, 591, 10.1111/cns.12264

Tentillier, 2015, The relation between α-synuclein and microglia in Parkinson’s disease: Recent developments, Neuroscience, 302, 47, 10.1016/j.neuroscience.2015.02.008

Xie, 2014, Shared Mechanisms of Neurodegeneration in Alzheimer’s Disease and Parkinson’s Disease, BioMed Res. Int., 2014, 1

Zhang, 2016, Design, synthesis and evaluation of novel tacrine-multialkoxybenzene hybrids as multi-targeted compounds against Alzheimer’s disease, Eur. J. Med. Chem., 116, 200, 10.1016/j.ejmech.2016.03.077

Alzheimer’s Disease International (2019). World Alzheimer Report 2019: Attitudes to Dementia, Alzheimer’s Disease International.

Shalini, 2018, Distribution of Alox15 in the Rat Brain and Its Role in Prefrontal Cortical Resolvin D1 Formation and Spatial Working Memory, Mol. Neurobiol., 55, 1537, 10.1007/s12035-017-0413-x

Sun, 2018, Docosahex-aenoic acid (DHA): An essential nutrient and a nutraceutical for brain health and diseases, Prostaglandins Leukot. Essent Fatty Acids., 136, 3, 10.1016/j.plefa.2017.03.006

Ishikado, A., Morino, K., Nishio, Y., Nakagawa, F., Mukose, A., Sono, Y., Yoshioka, N., Kondo, K., Sekine, O., and Yoshizaki, T. (2013). 4-Hydroxy Hexenal Derived from Docosahexaenoic Acid Protects Endothelial Cells via Nrf2 Activation. PLoS ONE, 8.

Ostermann, 2019, Plasma oxylipins respond in a linear dose-response manner with increased intake of EPA and DHA: Results from a randomized controlled trial in healthy humans, Am. J. Clin. Nutr., 109, 1251, 10.1093/ajcn/nqz016

Lin, 2020, mTOR drives cerebrovascular, synaptic, and cognitive dysfunction in normative aging, Aging Cell., 19, e13057, 10.1111/acel.13057

Tisserand, 2002, Regional frontal cortical volumes decrease differentially in aging: An MRI study to compare volumetric approaches and voxel-based morphometry, Neuroimage, 17, 657, 10.1006/nimg.2002.1173

Marin, 2016, Lipid Raft Alterations in Aged-Associated Neuropathologies, Curr. Alzheimer Res., 13, 973, 10.2174/1567205013666160314150017

Sonnino, 2014, Lipid Rafts in Neurodegeneration and Neuroprotection, Mol. Neurobiol., 50, 130, 10.1007/s12035-013-8614-4

Santos, 2016, Lipid Raft Size and Lipid Mobility in Non-raft Domains Increase during Aging and Are Exac-erbated in APP/PS1 Mice Model of Alzheimer’s Disease. Predictions from an Agent-Based Mathematical Model, Front Physiol., 7, 90, 10.3389/fphys.2016.00090

Talesa, 2001, Acetylcholinesterase in Alzheimer’s disease, Mech. Ageing. Dev., 122, 1961, 10.1016/S0047-6374(01)00309-8

Hardy, 2009, The amyloid hypothesis for Alzheimer’s disease: A critical reappraisal, J. Neurochem., 110, 1129, 10.1111/j.1471-4159.2009.06181.x

Fabelo, 2010, Lipid alterations in lipid rafts from Alzheimer’s disease human brain cortex, J. Alzheimers Dis., 19, 489, 10.3233/JAD-2010-1242

Small, 2011, Revisiting the role of acetylcholinesterase in Alzheimer’s disease: Cross-talk with P-tau and β-amyloid, Front. Mol. Neurosci., 4, 22

Gao, 2018, Tau in Alzheimer’s Disease: Mechanisms and Therapeutic Strategies, Curr. Alzheimer Res., 15, 283, 10.2174/1567205014666170417111859

Gauthier, 2016, Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer’s disease: A randomised, controlled, double-blind, parallel-arm, phase 3 trial, Lancet, 388, 2873, 10.1016/S0140-6736(16)31275-2

Trushina, 2017, Oxidative Stress, Synaptic Dysfunction, and Alzheimer’s Disease, J. Alzheimer’s Dis., 57, 1105, 10.3233/JAD-161088

Robert, 2020, Evolution of the Research Literature and the Scientific Community of Alzheimer’s Disease from 1983–2017: A 35-Year Survey, J. Alzheimer’s Dis., 75, 1105, 10.3233/JAD-191281

Jellinger, 2016, Accuracy of clinical diagnosis of Parkinson disease: A systematic review and meta-analysis. Author Response, Neurology, 87, 237, 10.1212/WNL.0000000000002876

Muller, 2007, Metabolic syndrome and dementia risk in a multiethnic elderly cohort, Dement. Geriatr. Cogn. Disord., 24, 185, 10.1159/000105927

Twelves, 2003, Systematic review of incidence studies of Parkinson’s disease, Mov. Disord., 18, 19, 10.1002/mds.10305

Poewe, 2017, Parkinson disease, Nat. Rev. Dis. Primers., 23, 17013, 10.1038/nrdp.2017.13

Kim, 2014, Alpha-synuclein biology in Lewy body diseases, Alzheimer’s Res. Ther., 6, 73, 10.1186/s13195-014-0073-2

Przedborski, 2017, The two-century journey of Parkinson disease research, Nat. Rev. Neurosci., 18, 251, 10.1038/nrn.2017.25

Warner, 2003, Genetic and Environmental Factors in the Cause of Parkinson’s Disease, Ann. Neurol., 53, S16, 10.1002/ana.10487

Bose, 2016, Mitochondrial dysfunction in Parkinson’s disease, J. Neurochem., 139, 216, 10.1111/jnc.13731

Keeney, 2006, Parkinson’s disease brain mitochondrial complex I has oxidatively damaged subunits and is functionally impaired and misassembled, J. Neurosci., 10, 5256, 10.1523/JNEUROSCI.0984-06.2006

Bolam, 2012, Living on the edge with too many mouths to feed: Why dopamine neurons die, Mov. Disord., 27, 1478, 10.1002/mds.25135

Pissadaki, 2013, The energy cost of action potential propagation in dopamine neurons: Clues to susceptibility in Parkinson’s disease, Front Comput. Neurosci., 7, 13, 10.3389/fncom.2013.00013

Surmeier, 2011, The role of calcium and mitochondrial oxidant stress in the loss of substantia nigra pars compacta dopaminergic neurons in Parkinson’s disease, Neuroscience, 198, 221, 10.1016/j.neuroscience.2011.08.045

Mosharov, 2009, Interplay between cytosolic dopamine, calcium, and alpha-synuclein causes selective death of substantia nigra neurons, Neuron, 62, 218, 10.1016/j.neuron.2009.01.033

Hirsch, 2009, Neuroinflammation in Parkinson’s disease: A target for neuroprotection?, Lancet Neurol., 8, 382, 10.1016/S1474-4422(09)70062-6

Ransohoff, 2016, How neuroinflammation contributes to neurodegeneration, Science, 353, 777, 10.1126/science.aag2590

Rodrigues, 2016, Pharmacological interventions for daytime sleepiness and sleep disorders in Parkinson’s disease: Systematic review and meta-analysis, Parkinsonism Relat Disord., 27, 25, 10.1016/j.parkreldis.2016.03.002

Wissel, 2018, Functional neurological disorders in Parkinson disease, J. Neurol. Neurosurg. Psychiatry, 89, 566, 10.1136/jnnp-2017-317378

Pfeiffer, 2018, Gastrointestinal Dysfunction in Parkinson’s Disease, Curr. Treat. Options Neurol., 20, 54, 10.1007/s11940-018-0539-9

Fasano, 2015, Gastrointestinal dysfunction in Parkinson’s disease, Lancet Neurol., 14, 625, 10.1016/S1474-4422(15)00007-1

Cole, 2009, Omega-3 fatty acids and dementia, Prostaglandins Leukot. Essent. Fat. Acids, 81, 213, 10.1016/j.plefa.2009.05.015

Wu, 2008, Docosahexaenoic acid dietary supplementation enhances the effects of exercise on synaptic plasticity and cognition, Neuroscience, 155, 751, 10.1016/j.neuroscience.2008.05.061

Lim, 2005, A Diet Enriched with the Omega-3 Fatty Acid Docosahexaenoic Acid Reduces Amyloid Burden in an Aged Alzheimer Mouse Model, J. Neurosci., 25, 3032, 10.1523/JNEUROSCI.4225-04.2005

Hjorth, 2013, Omega-3 fatty acids enhance phagocytosis of Alzheimer’s disease-related amyloid-β42 by human microglia and decrease inflammatory markers, J. Alzheimers Dis., 35, 697, 10.3233/JAD-130131

Thornton, 2006, Soluble amyloid precursor protein α reduces neuronal injury and improves functional outcome following diffuse traumatic brain injury in rats, Brain Res., 1094, 38, 10.1016/j.brainres.2006.03.107

Yang, 2011, Effects of fatty acid unsaturation numbers on membrane fluidity and α-secretase-dependent amyloid precursor protein processing, Neurochem. Int., 58, 321, 10.1016/j.neuint.2010.12.004

Hashimoto, 2002, Docosahexaenoic acid provides protection from impairment of learning ability in Alzheimer’s disease model rats, J. Neurochem., 81, 1084, 10.1046/j.1471-4159.2002.00905.x

Geng, 2020, Effects of Docosahexaenoic Acid and Its Peroxidation Product on Amyloid-β Peptide-Stimulated Microglia, Mol. Neurobiol., 57, 1085, 10.1007/s12035-019-01805-4

Yuan, 2020, DHA attenuates Aβ-induced necroptosis through the RIPK1/RIPK3 signaling pathway in THP-1 monocytes, Biomed. Pharmacother., 126, 110102, 10.1016/j.biopha.2020.110102

Wang, 2015, Effects of n-3 FA supplementation on the release of proresolving lipid mediators by blood mononuclear cells: The OmegAD study, J. Lipid Res., 56, 674, 10.1194/jlr.P055418

Vedin, 2010, Reduced prostaglandin F2α release from blood mononuclear leukocytes after oral supplementation of ω3 fatty acids: The OmegAD study, J. Lipid Res., 51, 1179, 10.1194/jlr.M002667

Vedin, 2008, Effects of docosahexaenoic acid–rich n−3 fatty acid supplementation on cytokine release from blood mononuclear leukocytes: The OmegAD study, Am. J. Clin. Nutr., 87, 1616, 10.1093/ajcn/87.6.1616

Yassine, 2017, Association of Do-cosahexaenoic Acid Supplementation with Alzheimer Disease Stage in Apolipoprotein E ε4 Carriers: A Review, JAMA Neurol., 74, 339, 10.1001/jamaneurol.2016.4899

Quinn, 2010, Docosahexaenoic acid supplementation and cognitive decline in Alzheimer disease: A randomized trial, JAMA, 304, 1903, 10.1001/jama.2010.1510

Tanriover, 2010, The effects of docosahexaenoic acid on glial derived neurotrophic factor and neurturin in bilateral rat model of Parkinson’s disease, Folia Histochem. Cytobiol., 48, 434, 10.2478/v10042-010-0047-6

2016, Role of docosahexaenoic acid in the modulation of glial cells in Alzheimer’s disease, J. Neuroinflammat., 13, 61, 10.1186/s12974-016-0525-7

Oguro, 2021, Contribution of DHA diols (19,20-DHDP) produced by cytochrome P450s and soluble epoxide hydrolase to the beneficial effects of DHA supplementation in the brains of rotenone-induced rat models of Parkinson’s disease, Biochim. Biophys. Acta Mol. Cell Biol. Lipids, 1866, 158858, 10.1016/j.bbalip.2020.158858

Wurtman, 2010, Nutritional modifiers of aging brain function: Use of uridine and other phosphatide precursors to increase formation of brain synapses, Nutr. Rev., 68, S88, 10.1111/j.1753-4887.2010.00344.x

Okaichi, 2005, Arachidonic acid improves aged rats’ spatial cognition, Physiol. Behav., 84, 617, 10.1016/j.physbeh.2005.02.008

Wang, 2005, Dietary uridine-5′-monophosphate supplementation increases potassi-um-evoked dopamine release and promotes neurite outgrowth in aged rats, J. Mol. Neurosci., 27, 137, 10.1385/JMN:27:1:137

Dodiya, 2018, Gut-brain and brain-gut axis in Parkinson’s disease models: Effects of a uridine and fish oil diet, Nutr. Neurosci., 21, 391, 10.1080/1028415X.2017.1294555

Hacioglu, 2012, Docosahexaenoic acid provides pro-tective mechanism in bilaterally MPTP-lesioned rat model of Parkinson’s disease, Folia Histochem. Cytobiol., 50, 228, 10.5603/FHC.2012.0032

Ozsoy, 2011, The influence and the mechanism of do-cosahexaenoic acid on a mouse model of Parkinson’s disease, Neurochem. Int., 59, 664, 10.1016/j.neuint.2011.06.012

Hosono, 2015, Arachidonic or Docosahexaenoic Acid Diet Prevents Memory Impairment in Tg2576 Mice, J. Alzheimer’s Dis., 48, 149, 10.3233/JAD-150341

Schrag, 2000, Dyskinesias and motor fluctuations in Parkinson’s disease. A community-based study, Brain, 123, 2297, 10.1093/brain/123.11.2297

Lee, 2015, Many Faces of Parkinson’s Disease: Non-Motor Symptoms of Parkinson’s Disease, J. Mov. Disord., 8, 92, 10.14802/jmd.15003

Hosono, 2015, Arachidonic acid diet attenuates brain Aβ deposition in Tg2576 mice, Brain Res., 1613, 92, 10.1016/j.brainres.2015.04.005

Amtul, 2012, Detrimental effects of arachidonic acid and its metabolites in cellular and mouse models of Alzheimer’s disease: Structural insight, Neurobiol. Aging., 33, 831.e21, 10.1016/j.neurobiolaging.2011.07.014

Lukiw, 1997, Cyclooxygenase 2 RNA message abundance, stability, and hypervariability in sporadic alzheimer neocortex, J. Neurosci. Res., 50, 937, 10.1002/(SICI)1097-4547(19971215)50:6<937::AID-JNR4>3.0.CO;2-E

Mohri, 2007, Hematopoietic Prostaglandin D Synthase and DP1 Receptor Are Selectively Upregulated in Microglia and Astrocytes Within Senile Plaques From Human Patients and in a Mouse Model of Alzheimer Disease, J. Neuropathol. Exp. Neurol., 66, 469, 10.1097/01.jnen.0000240472.43038.27

Thomas, 2016, Dietary arachidonic acid as a risk factor for age-associated neurodegenerative diseases: Potential mechanisms, Biochimistry, 130, 168

Fukaya, 2007, Arachidonic acid preserves hip-pocampal neuron membrane fluidity in senescent rats, Neurobiol. Aging., 28, 1179, 10.1016/j.neurobiolaging.2006.05.023

Kotani, 2003, Synaptic plasticity preserved with arachidonic acid diet in aged rats, Neurosci. Res., 46, 453, 10.1016/S0168-0102(03)00123-8

Clements, 1991, Increase in arachidonic acid concentration in a postsynaptic membrane fraction following the induction of long-term potentiation in the dentate gyrus, Neuroscience, 45, 379, 10.1016/0306-4522(91)90235-G

Kotani, 2006, Dietary supplementation of arachidonic and docosahexaenoic acids improves cognitive dysfunction, Neurosci. Res., 56, 159, 10.1016/j.neures.2006.06.010

Mucke, 2010, Phospholipase A2 and arachidonic acid in Alzheimer’s disease, Biochim. Biophys. Acta., 1801, 784, 10.1016/j.bbalip.2010.05.013

Roseth, 1998, The effect of arachidonic acid and free fatty acids on vesicular uptake of glutamate and gamma-aminobutyric acid, Eur. J. Pharmacol., 341, 281, 10.1016/S0014-2999(97)01449-0

Carta, 2014, Membrane lipids tune synaptic transmission by direct modulation of presynaptic potassium channels, Neuron, 81, 787, 10.1016/j.neuron.2013.12.028

Angelova, 2009, Arachidonic acid potently inhibits both postsynaptic-type Kv4.2 and presynaptic-type Kv1.4 IApotassium channels, Eur. J. Neurosci., 29, 1943, 10.1111/j.1460-9568.2009.06737.x

Lakkappa, 2016, Possible role of Epoxyeicosatrienoic acid in prevention of oxidative stress mediated neuroinflammation in Parkinson disorders, Med. Hypotheses, 93, 161, 10.1016/j.mehy.2016.06.003

Lakkappa, 2019, Soluble epoxide hydrolase inhibitor, APAU, protects dopaminergic neurons against rotenone induced neurotoxicity: Implications for Parkinson’s disease, NeuroToxicology, 70, 135, 10.1016/j.neuro.2018.11.010

Iljina, 2016, Arachidonic acid mediates the formation of abundant al-pha-helical multimers of alpha-synuclein, Sci. Rep., 6, 1, 10.1038/srep33928

Cheng, 2019, Development of FABP3 ligands that inhibit arachidonic ac-id-induced α-synuclein oligomerization, Brain Res., 1707, 190, 10.1016/j.brainres.2018.11.036

Julien, 2006, Postmortem brain fatty acid profile of levo-dopa-treated Parkinson disease patients and parkinsonian monkeys, Neurochem. Int., 48, 404, 10.1016/j.neuint.2005.12.002

Sun, 2019, Relationship between high dietary fat intake and Parkinson’s disease risk: A meta-analysis, Neural. Regen. Res., 14, 2156, 10.4103/1673-5374.262599