Therapeutics

Docosahexaenoic acid (DHA)

Tools

Back to the Top

Overview

Name: Docosahexaenoic acid (DHA)
Synonyms: Omega-3 fatty acid
Therapy Type: Supplement, Dietary (timeline)
Target Type: Other (timeline)
Condition(s): Alzheimer's Disease
U.S. FDA Status: Alzheimer's Disease (Phase 4)
Company: Martek Biosciences Corporation, NeuroBioPharm, Inc.

Background

Docosahexaenoic acid (DHA) is one of the most abundant polyunsaturated fatty acids in the human brain. Epidemiological research has linked high DHA consumption with a lower risk of Alzheimer's disease (e.g., Morris et al., 2003, Zhang et al., 2016). People ingest DHA from foods such as fatty fish, walnuts, or flax seeds, or from dietary supplements. These are being marketed in different formulations of DHA, or DHA mixed with other omega-3 fatty acids such as eicosapentaenoic acid (EPA).

In early epidemiology work, elevated blood levels of DHA correlated with reduced dementia risk in some but not all studies (Schaefer et al., 2006; Laurin et al., 2003). Newer studies support the link between DHA intake or blood levels and a reduced risk of cognitive decline or AD (e.g. Wei et al., 2023; He et al., 2023; Shang et al., 2018). In people with AD, higher serum DHA was associated with slower cognitive decline (Chu et al., 2022). An analysis of ADNI data found associations between low omega-3 in red blood cells and greater Aβ accumulation and memory decline (Rouch et al., 2022). Data from the large Framingham Heart study correlated higher DHA or EPA in red blood cells with larger hippocampal volume and better abstract reasoning at midlife, and up to half the risk of incident AD in older adults (Satizabal et al., 2022; Sala-Vila et al., 2022). This and other studies (e.g., Sala-Vila et al., 2021; Coughlan et al., 2021) indicate that the relationship of DHA status to cognition and neurodegeneration biomarkers is affected by ApoE4 genotype (for review see Zhange et al., 2023).

In contrast, U.K. Biobank data indicate fish oil supplementation in older people reduced risk of all-cause dementia, but not specifically of AD (Huang et al., 2022; Liu et al., 2022). In the same population, total omega-3 status, but not DHA alone, correlated with lowered risk of dementia (Sala-Vila et al., 2023). A smaller study found no association of circulating DHA with incident dementia over 13 years (de Oliveira Otto et al., 2023).

Multiple autopsy studies have described deficits of DHA-containing phospholipids in brain tissue from people with Alzheimer's, which correlates with markers of the disease (reviewed in Heath and Wood, 2021). In samples from dementia with Lewy body cases, low levels of DHA-containing phospholipids were associated with lower soluble Aβ42, higher phosphorylated α-synuclein, and diminished synaptic proteins (Chong et al., 2023).

In animal studies, oral intake of DHA leads to a reduction of amyloid, tau, and neuritic pathology (Lim et al., 2005; Green et al., 2007; Calon et al., 2004). This appears to stem from beneficial effects on amyloid processing, inflammation, oxidative stress, and metabolic function (for review see Mett 2021). In APP/PS mice, omega-3 supplementation reversed some effects of a high-diet on Aβ peptide expression, insulin resistance, and inflammation (Ma et al., 2022). DHA reduced retinal amyloid pathology in 5XFAD mice, when delivered as a lysophosphatidylcholine form uniquely bioavailable to retinal cells (Sugasini et al., 2023). Nasal delivery of DHA reduced tau phosphorylation and improved cognitive function in J20 transgenic mice, or after brain injection of Aβ peptides in normal mice (Zussy et al., 2022). DHA supplementation also reversed cognitive and olfactory deficits due to ApoE4 expression in transgenic mice (González et al., 2023; Lessard-Beaudoin et al., 2021).

Findings

DHA has been tested in clinical trials by itself and as part of other food-supplement formulations.

In 2000, Karolinska University Hospital began the OmegAD trial, which evaluated a six-month course of treatment with a DHA-containing fish oil formulation called EPAX 1050 TG in 204 people with mild to moderate AD, of whom 174 completed the study. The treatment and placebo groups did not differ on either of the main outcome measures, decline on the MMSE and ADAS-Cog, or on neuropsychiatric symptoms overall. However, analysis of a small subgroup of the 32 mildest cases did suggest less decline on the MMSE, though not ADAS-Cog; a similar slowing of decline appeared to occur in the placebo group once switched to DHA after six months (Freund-Levi et al., 2006; see news and commentaryFreund-Levi et al., 2008). Substudies suggested that DHA treatment increased CSF levels of DHA and other fatty acids and decreased levels of tau, as well as changing expression of inflammation-related genes, and release of certain cytokines and inflammatory mediators in white blood cells (Freund-Levi et al., 2014; Vedin et al., 2012; Vedin et al., 2010Vedin et al., 2008; Wang et al., 2015). Other substudies reported no effect on CSF biomarkers of inflammation or amyloidosis, or urine biomarkers of oxidative stress (Freund-Levi et al., 2009; Freund-Levi et al., 2014). Supplementation appeared to increase plasma, but not CSF transthyretin, a cofactor for Aβ clearance (Faxen-Irving et al., 2013). The treatment caused a global DNA hypomethylation measured in blood cells, but the effect correlated more strongly with EPA blood concentrations than DHA (Karimi et al., 2017).

Additional post hoc analyses of this trial showed a dose response between the increase in omega-3 plasma levels after supplementation, and slowing of decline on the ADAS-Cog (Eriksdottir et al., 2015). Patients with better Vitamin B status showed more improvement after supplementation on the MMSE and CDR-SB, but not the global CDR or ADAS-Cog (Jerneren et al., 2019). Most recently, analysis of a panel of CSF biomarkers revealed an increase in YKL-40 and NfL after six months of supplementation, while other markers of Aβ, tau, and inflammation were stable (Tofiq et al., 2021).

A Dutch study conducted in 2006 reported no effect of six months of fish oil supplements in 302 cognitively normal older adults. The participants had to have an MMSE greater than 22, and took 1,800 mg or 400 mg EPA-DHA daily, or placebo. Treatment caused no change in a neuropsychiatric test battery of attention, sensorimotor speed, memory and executive function, in quality of life, or in mental well-being (van de Rest et al., 2008; van de Rest et al., 2008; van de Rest et al., 2009).

From 2007 to 2009, the Alzheimer's Disease Cooperative Study conducted a study at 51 centers in North America to evaluate an 18-month course of 2 grams per day of DHA in 402 patients with mild to moderate AD, of whom 295 completed the trial. DHA had no effect, relative to placebo, on the rate of decline on either the ADAS-Cog or the CDR-SOB clinical/functional assessment. Analysis by participants' ApoE genotype indicated a slower cognitive decline in ApoE4 noncarriers, who may have been relatively less advanced in their disease (Quinn et al., 2010Nov 2010 news). Subsequently, ApoE4 carriers were found to have smaller increases in plasma DHA and EPA, and less delivery to CSF after supplementation, than noncarriers (Tomaszewski et al., 2020; Yassine et al., 2016).

This pharmacogenetic hint of a differential effect prompted several bioavailability studies at the Université de Sherbrooke, Quebec, Canada, between 2009 and 2011, which analyzed the percentage of DHA in lipoproteins, incorporation into plasma lipids, and pharmacokinetics by ApoE genotype in healthy young adults and people with MCI (e.g., Chouinard-Watkins et al., 2013Plourde et al., 2014). The study confirmed lower plasma levels and higher oxidation rates, contributing to a shorter half-life of DHA in ApoE4 carriers.

The largest clinical trial of DHA is MAPT, conducted in four cities in France. This three-year, secondary prevention study in 1,680 participants began in 2008 and enrolled people 70 and older who reported a subjective memory complaint and a mild functional loss, were frail and walked slowly, but did not meet an Alzheimer's diagnosis (Carrié et al., 2012). MAPT compared three interventions—800 mg DHA and 225 mg EPA daily alone, DHA/EPA plus a multidomain behavioral intervention, multidomain behavioral intervention alone—to placebo (Gillette-Guyonnet et al., 2009). Neither intervention, alone or in combination, significantly slowed cognitive decline as measured by a composite score of four tests of recall, orientation, processing and verbal fluency (Andrieu et al., 2017). An amyloid PET substudy compared the effects of the multidomain intervention and DHA/EPA according to brain amyloid status. The multidomain intervention, with or without DHA/EPA, improved composite scores after 36 months compared to placebo in the amyloid-positive, but not -negative, subset; DHA/EPA alone had no effect in either subset (Delrieu et al., 2019; Delrieu et al., 2023). In another PET analysis, cortical amyloid was lower after two years in people who got the multidomain intervention, with or without DHA/EPA, but not in those who received DPA/EPA alone (Hooper et al., 2020). DPS/EPS had no effect on brain structure by MRI, global functional connectivity, or on a composite physical and cognitive endpoint of intrinsic capacity (Sivera et al., 2020; Perus et al, 2022; Giudici et al., 2020). As in the OmegAD study, results suggested that people with low Vitamin B status may benefit less from omega-3 supplementation (Maltais et al., 2022). In this population, low DHA/EPA was potentially associated with psychiatric disorders. Participants with low RBC DHA-EPA at baseline were more likely to be using psychotropic drugs, and those who increased DHA-EPA after treatment were less likely (Gallini et al., 2019).

A subsequent analysis of MAPT data suggested higher odds of cognitive decline for people with low DHA/EPA levels (Bowman et al., 2019). Lower serum DHA was also correlated with more cerebral amyloidosis and atrophy (Yassine et al., 2016Aug 2016 news). In response to those findings, MAPT investigators in April 2018 began LO-MAPT. This 18-month study planned to enroll 400 older adults with low DHA/EPA status and randomize them to 1.53 g/day DHA/EPA or placebo. Participants had to have subjective memory complaints or a family history of AD. The primary outcome is change in a cognitive composite of scores on the Free Cued and Selective Reminding Test, MMSE, and Category Naming Test. The trial offered an 18-month open-label extension. It was completed in 2020, after enrolling 774 participants.

From 2009 to 2011, the company NeuroBioPharm Inc. ran a six-month trial at 14 different sites in Canada to compare soft-capsule formulations of fish oil and krill oil, each containing 100 mg DHA, to placebo in 175 people with mild to moderate Alzheimer's disease. Krill oil contains DHA and EPA in a form claimed to be more bioavailable than fish oil (e.g. Schuchardt et al., 2011). This trial used the Neuropsychological Test Battery as primary outcome; data have not been published.

Between 2013-2015, a study in Taiwan tested six months of daily DHA, EPA, or the combination, against placebo in 163 people with mild cognitive impairment or AD dementia. Doses of 0.7 g/day DHA and 1.6 g/day EPA had no effect on cognitive function or depressive symptoms (Lin et al., 2022).

One DHA study, at Oregon Health and Science University, started in May 2014. According to published baseline data, the trial recruited 102 people age 75 and older who were cognitively impaired but did not have dementia, and suboptimal blood levels of DHA and EPA of less than 110 μg/ml. Participants were randomized to receive a three-year course of 1.65 grams/day of EPA plus DHA or soybean oil placebo (Bowman et al., 2019). Seeking to understand DHA's effect on vascular cognitive aging, this trial used white-matter hyperintensity as primary outcome; secondary and other outcome measures included other brain imaging modalities as well as blood-based indicators of endothelial health and neuropsychological tests. Top-line results were presented at the 2020 CTAD. Supplementation failed to slow accumulation of white-matter hyperintensities in the 45 placebo and 42 treated participants who had a least one follow-up MRI. At the end of the study, 24 of the treatment group had plasma DHA+EPA levels greater than 110 μg/ml. In a prespecified analysis, this group had fewer white-matter hyperintensities and maintained better white-matter integrity than the placebo group. There were no differences in total brain volume, ventricular volume, medial temporal lobe atrophy, excessive cognitive functions, or adverse events.

In June 2016, a pilot study at the University of Southern California began evaluating how much of a DHA supplement enters the central nervous system, and whether CSF levels are influenced by ApoE4 status (for review, see Yassine et al., 2017). The trial enrolled 31 adults older than 55 with a family history of dementia, who took 2 g DHA per day or placebo for 26 weeks. The primary outcome was change in CSF DHA before and after supplementation. According to published results, this dose of DHA resulted in a 200 percent increase in plasma DHA compared to placebo, but only a 28 percent increase in CSF DHA (Arellanes et al., 2020). ApoE4 carriers had slightly lower CSF DHA. For EPA, CSF levels increased 43 percent, but were significantly lower in ApoE4 carriers than in noncarriers. Other endpoints including brain volume and cognitive scores did not differ between supplement and placebo groups.

In September 2018, the same group began a larger follow-up study in 368 healthy adults age 60-80, who have at least one dementia risk factor. Half are to be ApoE4 carriers. Participants take 2g DHA or placebo daily for two years. After six months of treatment, investigators will analyze CSF fatty acids in 168 participants, to determine the effect of their ApoE genotype on supplementation. Changes in MRI measures of structural and functional connectivity and cognition will be assessed in all participants at two years. The study will run through 2025. The trial protocol and baseline data are published (Yassine et al., 2023).

For details of clinical trials of DHA in Alzheimer's, see clinicaltrials.gov.

Last Updated: 12 Jan 2024

Comments

No Available Comments

Make a Comment

To make a comment you must login or register.

References

News Citations

  1. First Trial of Fish Fats Shows Promise for Early AD
  2. Paper Alert: Negative DHA Trial Fuels Soul-Searching in AD Field
  3. Less Salmon, More Plaques? Link Between Omega-3s and Aβ Reinvigorates Fish Oil Debate

Paper Citations

  1. . Omega-3 fatty acid treatment in 174 patients with mild to moderate Alzheimer disease: OmegAD study: a randomized double-blind trial. Arch Neurol. 2006 Oct;63(10):1402-8. PubMed.
  2. . Omega-3 supplementation in mild to moderate Alzheimer's disease: effects on neuropsychiatric symptoms. Int J Geriatr Psychiatry. 2008 Feb;23(2):161-9. PubMed.
  3. . Transfer of omega-3 fatty acids across the blood-brain barrier after dietary supplementation with a docosahexaenoic acid-rich omega-3 fatty acid preparation in patients with Alzheimer's disease: the OmegAD study. J Intern Med. 2014 Apr;275(4):428-36. Epub 2014 Jan 11 PubMed.
  4. . Effects of DHA-rich n-3 fatty acid supplementation on gene expression in blood mononuclear leukocytes: the OmegAD study. PLoS One. 2012;7(4):e35425. Epub 2012 Apr 24 PubMed.
  5. . Reduced prostaglandin F2 alpha release from blood mononuclear leukocytes after oral supplementation of omega3 fatty acids: the OmegAD study. J Lipid Res. 2010 May;51(5):1179-85. Epub 2009 Nov 24 PubMed.
  6. . Effects of docosahexaenoic acid-rich n-3 fatty acid supplementation on cytokine release from blood mononuclear leukocytes: the OmegAD study. Am J Clin Nutr. 2008 Jun;87(6):1616-22. PubMed.
  7. . Effects of n-3 FA supplementation on the release of proresolving lipid mediators by blood mononuclear cells: the OmegAD study. J Lipid Res. 2015 Mar;56(3):674-81. Epub 2015 Jan 23 PubMed.
  8. . Effects of omega-3 fatty acids on inflammatory markers in cerebrospinal fluid and plasma in Alzheimer's disease: the OmegAD study. Dement Geriatr Cogn Disord. 2009;27(5):481-90. PubMed.
  9. . Effects of supplementation with omega-3 fatty acids on oxidative stress and inflammation in patients with Alzheimer's disease: the OmegAD study. J Alzheimers Dis. 2014;42(3):823-31. PubMed.
  10. . Effects on Transthyretin in Plasma and Cerebrospinal Fluid by DHA-Rich n - 3 Fatty Acid Supplementation in Patients with Alzheimer's Disease: The OmegAD Study. J Alzheimers Dis. 2013 Jan 1;36(1):1-6. PubMed.
  11. . DHA-rich n-3 fatty acid supplementation decreases DNA methylation in blood leukocytes: the OmegAD study. Am J Clin Nutr. 2017 Oct;106(4):1157-1165. Epub 2017 Aug 30 PubMed.
  12. . Plasma Fatty Acid Profiles in Relation to Cognition and Gender in Alzheimer's Disease Patients During Oral Omega-3 Fatty Acid Supplementation: The OmegAD Study. J Alzheimers Dis. 2015;48(3):805-12. PubMed.
  13. . Homocysteine Status Modifies the Treatment Effect of Omega-3 Fatty Acids on Cognition in a Randomized Clinical Trial in Mild to Moderate Alzheimer's Disease: The OmegAD Study. J Alzheimers Dis. 2019;69(1):189-197. PubMed.
  14. . Effects of Peroral Omega-3 Fatty Acid Supplementation on Cerebrospinal Fluid Biomarkers in Patients with Alzheimer's Disease: A Randomized Controlled Trial-The OmegAD Study. J Alzheimers Dis. 2021;83(3):1291-1301. PubMed.
  15. . Effect of fish oil on cognitive performance in older subjects: a randomized, controlled trial. Neurology. 2008 Aug 5;71(6):430-8. PubMed.
  16. . Effect of fish-oil supplementation on mental well-being in older subjects: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr. 2008 Sep;88(3):706-13. PubMed.
  17. . Effect of fish oil supplementation on quality of life in a general population of older Dutch subjects: a randomized, double-blind, placebo-controlled trial. J Am Geriatr Soc. 2009 Aug;57(8):1481-6. Epub 2009 Jun 22 PubMed.
  18. . Docosahexaenoic acid supplementation and cognitive decline in Alzheimer disease: a randomized trial. JAMA. 2010 Nov 3;304(17):1903-11. PubMed.
  19. . Effect of APOE Genotype on Plasma Docosahexaenoic Acid (DHA), Eicosapentaenoic Acid, Arachidonic Acid, and Hippocampal Volume in the Alzheimer's Disease Cooperative Study-Sponsored DHA Clinical Trial. J Alzheimers Dis. 2020;74(3):975-990. PubMed.
  20. . The effect of APOE genotype on the delivery of DHA to cerebrospinal fluid in Alzheimer's disease. Alzheimers Res Ther. 2016 Jun 30;8:25. PubMed.
  21. . Disturbance in uniformly 13C-labelled DHA metabolism in elderly human subjects carrying the apoE ε4 allele. Br J Nutr. 2013 Apr 30;:1-9. PubMed.
  22. . Kinetics of 13C-DHA before and during fish-oil supplementation in healthy older individuals. Am J Clin Nutr. 2014 May 14;100(1):105-112. PubMed.
  23. . Recruitment strategies for preventive trials. The MAPT study (MultiDomain Alzheimer Preventive Trial). J Nutr Health Aging. 2012 Apr;16(4):355-9. PubMed.
  24. . Commentary on "A roadmap for the prevention of dementia II. Leon Thal Symposium 2008." The Multidomain Alzheimer Preventive Trial (MAPT): a new approach to the prevention of Alzheimer's disease. Alzheimers Dement. 2009 Mar;5(2):114-21. PubMed.
  25. . Effect of long-term omega 3 polyunsaturated fatty acid supplementation with or without multidomain intervention on cognitive function in elderly adults with memory complaints (MAPT): a randomised, placebo-controlled trial. Lancet Neurol. 2017 May;16(5):377-389. Epub 2017 Mar 27 PubMed.
  26. . Multidomain intervention and/or omega-3 in nondemented elderly subjects according to amyloid status. Alzheimers Dement. 2019 Nov;15(11):1392-1401. Epub 2019 Sep 23 PubMed.
  27. . Cognitive impact of multidomain intervention and omega 3 according to blood Aβ42/40 ratio: a subgroup analysis from the randomized MAPT trial. Alzheimers Res Ther. 2023 Oct 23;15(1):183. PubMed.
  28. . Cortical β-Amyloid in Older Adults Is Associated with Multidomain Interventions with and without Omega 3 Polyunsaturated Fatty Acid Supplementation. J Prev Alzheimers Dis. 2020;7(2):128-134. PubMed.
  29. . Voxel-based assessments of treatment effects on longitudinal brain changes in the Multidomain Alzheimer Preventive Trial cohort. Neurobiol Aging. 2020 Oct;94:50-59. Epub 2020 May 29 PubMed.
  30. . Impact of multidomain preventive strategies on functional brain connectivity in older adults with cognitive complaint: Subset from the Montpellier center of the ancillary MAPT-MRI study. Front Aging Neurosci. 2022;14:971220. Epub 2023 Jan 10 PubMed.
  31. . Effect of long-term omega-3 supplementation and a lifestyle multidomain intervention on intrinsic capacity among community-dwelling older adults: Secondary analysis of a randomized, placebo-controlled trial (MAPT study). Maturitas. 2020 Nov;141:39-45. Epub 2020 Jun 26 PubMed.
  32. . Omega-3 Supplementation for the Prevention of Cognitive Decline in Older Adults: Does It Depend on Homocysteine Levels?. J Nutr Health Aging. 2022;26(6):615-620. PubMed.
  33. . Red Blood Cell Omega-3 Fatty Acid Composition and Psychotropic Drug Use in Older Adults: Results from the MAPT Study. J Nutr Health Aging. 2019;23(9):805-812. PubMed.
  34. . A blood-based nutritional risk index explains cognitive enhancement and decline in the multidomain Alzheimer prevention trial. Alzheimers Dement (N Y). 2019;5:953-963. Epub 2019 Dec 28 PubMed. Correction.
  35. . Association of Serum Docosahexaenoic Acid With Cerebral Amyloidosis. JAMA Neurol. 2016 Oct 1;73(10):1208-1216. PubMed.
  36. . Incorporation of EPA and DHA into plasma phospholipids in response to different omega-3 fatty acid formulations--a comparative bioavailability study of fish oil vs. krill oil. Lipids Health Dis. 2011 Aug 22;10:145. PubMed.
  37. . Omega-3 fatty acids and blood-based biomarkers in Alzheimer's disease and mild cognitive impairment: A randomized placebo-controlled trial. Brain Behav Immun. 2022 Jan;99:289-298. Epub 2021 Oct 29 PubMed.
  38. . Randomized Trial of Marine n-3 Polyunsaturated Fatty Acids for the Prevention of Cerebral Small Vessel Disease and Inflammation in Aging (PUFA Trial): Rationale, Design and Baseline Results. Nutrients. 2019 Mar 29;11(4) PubMed.
  39. . Association of Docosahexaenoic Acid Supplementation With Alzheimer Disease Stage in Apolipoprotein E ε4 Carriers: A Review. JAMA Neurol. 2017 Mar 1;74(3):339-347. PubMed.
  40. . Brain delivery of supplemental docosahexaenoic acid (DHA): A randomized placebo-controlled clinical trial. EBioMedicine. 2020 Sep;59:102883. Epub 2020 Jul 17 PubMed.
  41. . Baseline Findings of PreventE4: A Double-Blind Placebo Controlled Clinical Trial Testing High Dose DHA in APOE4 Carriers before the Onset of Dementia. J Prev Alzheimers Dis. 2023;10(4):810-820. PubMed.
  42. . Consumption of fish and n-3 fatty acids and risk of incident Alzheimer disease. Arch Neurol. 2003 Jul;60(7):940-6. PubMed.
  43. . Intakes of fish and polyunsaturated fatty acids and mild-to-severe cognitive impairment risks: a dose-response meta-analysis of 21 cohort studies. Am J Clin Nutr. 2016 Feb;103(2):330-40. Epub 2015 Dec 30 PubMed.
  44. . Plasma phosphatidylcholine docosahexaenoic acid content and risk of dementia and Alzheimer disease: the Framingham Heart Study. Arch Neurol. 2006 Nov;63(11):1545-50. PubMed.
  45. . Omega-3 fatty acids and risk of cognitive impairment and dementia. J Alzheimers Dis. 2003 Aug;5(4):315-22. PubMed.
  46. . The Relationship of Omega-3 Fatty Acids with Dementia and Cognitive Decline: Evidence from Prospective Cohort Studies of Supplementation, Dietary Intake, and Blood Markers. Am J Clin Nutr. 2023 Jun;117(6):1096-1109. Epub 2023 Apr 5 PubMed.
  47. . Circulating polyunsaturated fatty acids, fish oil supplementation, and risk of incident dementia: a prospective cohort study of 440,750 participants. Geroscience. 2023 Jun;45(3):1997-2009. Epub 2023 Apr 12 PubMed.
  48. . Different Associations of Plasma Biomarkers in Alzheimer's Disease, Mild Cognitive Impairment, Vascular Dementia, and Ischemic Stroke. J Clin Neurol. 2018 Jan;14(1):29-34. PubMed.
  49. . Higher Serum DHA and Slower Cognitive Decline in Patients with Alzheimer's Disease: Two-Year Follow-Up. Nutrients. 2022 Mar 9;14(6) PubMed.
  50. . Associations of erythrocyte omega-3 fatty acids with cognition, brain imaging and biomarkers in the Alzheimer's disease neuroimaging initiative: cross-sectional and longitudinal retrospective analyses. Am J Clin Nutr. 2022 Dec 19;116(6):1492-1506. PubMed.
  51. . Association of Red Blood Cell Omega-3 Fatty Acids With MRI Markers and Cognitive Function in Midlife: The Framingham Heart Study. Neurology. 2022 Oct 5;99(23):e2572-82. PubMed.
  52. . Red Blood Cell DHA Is Inversely Associated with Risk of Incident Alzheimer's Disease and All-Cause Dementia: Framingham Offspring Study. Nutrients. 2022 Jun 9;14(12) PubMed.
  53. . DHA intake relates to better cerebrovascular and neurodegeneration neuroimaging phenotypes in middle-aged adults at increased genetic risk of Alzheimer disease. Am J Clin Nutr. 2021 Jun 1;113(6):1627-1635. PubMed.
  54. . APOE ε4 alters associations between docosahexaenoic acid and preclinical markers of Alzheimer's disease. Brain Commun. 2021;3(2):fcab085. Epub 2021 May 11 PubMed.
  55. . Effects of DHA on cognitive dysfunction in aging and Alzheimer's disease: The mediating roles of ApoE. Prog Lipid Res. 2024 Jan;93:101256. Epub 2023 Oct 27 PubMed.
  56. . Associations of fish oil supplementation with incident dementia: Evidence from the UK Biobank cohort study. Front Neurosci. 2022;16:910977. Epub 2022 Sep 7 PubMed.
  57. . Association of fish oil supplementation with risk of incident dementia: A prospective study of 215,083 older adults. Clin Nutr. 2022 Mar;41(3):589-598. Epub 2022 Jan 11 PubMed.
  58. . Plasma Omega-3 Fatty Acids and Risk for Incident Dementia in the UK Biobank Study: A Closer Look. Nutrients. 2023 Nov 23;15(23) PubMed.
  59. . Circulating Omega-3 and Omega-6 Fatty Acids, Cognitive Decline, and Dementia in Older Adults. J Alzheimers Dis. 2023;95(3):965-979. PubMed.
  60. . Why Have the Benefits of DHA Not Been Borne Out in the Treatment and Prevention of Alzheimer's Disease? A Narrative Review Focused on DHA Metabolism and Adipose Tissue. Int J Mol Sci. 2021 Oct 31;22(21) PubMed.
  61. . Decreased DHA-containing phospholipids in the neocortex of dementia with Lewy bodies are associated with soluble Aβ42 , phosphorylated α-synuclein, and synaptopathology. Brain Pathol. 2023 Nov;33(6):e13190. Epub 2023 Jul 18 PubMed.
  62. . A diet enriched with the omega-3 fatty acid docosahexaenoic acid reduces amyloid burden in an aged Alzheimer mouse model. J Neurosci. 2005 Mar 23;25(12):3032-40. PubMed.
  63. . Dietary docosahexaenoic acid and docosapentaenoic acid ameliorate amyloid-beta and tau pathology via a mechanism involving presenilin 1 levels. J Neurosci. 2007 Apr 18;27(16):4385-95. PubMed.
  64. . Docosahexaenoic acid protects from dendritic pathology in an Alzheimer's disease mouse model. Neuron. 2004 Sep 2;43(5):633-45. PubMed.
  65. . The Impact of Medium Chain and Polyunsaturated ω-3-Fatty Acids on Amyloid-β Deposition, Oxidative Stress and Metabolic Dysfunction Associated with Alzheimer's Disease. Antioxidants (Basel). 2021 Dec 14;10(12) PubMed.
  66. . Effects of distinct n-6 to n-3 polyunsaturated fatty acid ratios on insulin resistant and AD-like phenotypes in high-fat diets-fed APP/PS1 mice. Food Res Int. 2022 Dec;162(Pt B):112207. Epub 2022 Nov 19 PubMed.
  67. . Improvement of retinal function in Alzheimer disease-associated retinopathy by dietary lysophosphatidylcholine-EPA/DHA. Sci Rep. 2023 Jun 6;13(1):9179. PubMed.
  68. . Intranasal Administration of Nanovectorized Docosahexaenoic Acid (DHA) Improves Cognitive Function in Two Complementary Mouse Models of Alzheimer's Disease. Antioxidants (Basel). 2022 Apr 25;11(5) PubMed.
  69. . Amelioration of Cognitive and Olfactory System Deficits in APOE4 Transgenic Mice with DHA Treatment. Mol Neurobiol. 2023 Oct;60(10):5624-5641. Epub 2023 Jun 17 PubMed.
  70. . Diet enriched in omega-3 fatty acids alleviates olfactory system deficits in APOE4 transgenic mice. Eur J Neurosci. 2021 Nov;54(9):7092-7108. Epub 2021 Oct 13 PubMed.

External Citations

  1. clinicaltrials.gov

Further Reading

Papers

  1. . Ageing and apoE change DHA homeostasis: relevance to age-related cognitive decline. Proc Nutr Soc. 2013 Oct 9;:1-7. PubMed.
  2. . Nutrition and neurodegeneration: epidemiological evidence and challenges for future research. Br J Clin Pharmacol. 2013 Mar;75(3):738-55. PubMed.
  3. . ω-3 fatty acids in the prevention of cognitive decline in humans. Adv Nutr. 2013 Nov;4(6):672-6. Epub 2013 Nov 6 PubMed.
  4. . Omega-3 polyunsaturated fatty acids in Alzheimer's disease: key questions and partial answers. Curr Alzheimer Res. 2011 Aug;8(5):470-8. PubMed.
  5. . Quantitative Erythrocyte Omega-3 EPA Plus DHA Levels are Related to Higher Regional Cerebral Blood Flow on Brain SPECT. J Alzheimers Dis. 2017;58(4):1189-1199. PubMed.
  6. . Long-chain omega-3 fatty acids improve brain function and structure in older adults. Cereb Cortex. 2014 Nov;24(11):3059-68. Epub 2013 Jun 24 PubMed.
  7. . A Randomized Placebo-Controlled Pilot Trial of Omega-3 Fatty Acids and Alpha Lipoic Acid in Alzheimer's Disease. J Alzheimers Dis. 2014 Jan 1;38(1):111-20. PubMed.
  8. . Brain targeting with docosahexaenoic acid as a prospective therapy for neurodegenerative diseases and its passage across blood brain barrier. Biochimie. 2020 Mar;170:203-211. Epub 2020 Jan 31 PubMed.
  9. . Role of phosphatidylcholine-DHA in preventing APOE4-associated Alzheimer's disease. FASEB J. 2019 Feb;33(2):1554-1564. Epub 2018 Oct 5 PubMed.
  10. . Long-chain omega-3 fatty acids and the brain: a review of the independent and shared effects of EPA, DPA and DHA. Front Aging Neurosci. 2015;7:52. Epub 2015 Apr 21 PubMed.
  11. . Synthesis and Preclinical Evaluation of 22-[18F]Fluorodocosahexaenoic Acid as a Positron Emission Tomography Probe for Monitoring Brain Docosahexaenoic Acid Uptake Kinetics. ACS Chem Neurosci. 2023 Dec 20;14(24):4409-4418. Epub 2023 Dec 4 PubMed.
  12. . Potential Neuroprotective Effects of Dietary Omega-3 Fatty Acids on Stress in Alzheimer's Disease. Biomolecules. 2023 Jul 8;13(7) PubMed.
  13. . DHA/EPA (Omega-3) and LA/GLA (Omega-6) as Bioactive Molecules in Neurodegenerative Diseases. Int J Mol Sci. 2023 Jun 27;24(13) PubMed.
  14. . Efficacy and acceptability of anti-inflammatory eicosapentaenoic acid for cognitive function in Alzheimer's dementia: A network meta-analysis of randomized, placebo-controlled trials with omega-3 fatty acids and FDA-approved pharmacotherapy. Brain Behav Immun. 2023 Jul;111:352-364. Epub 2023 May 6 PubMed.
  15. . Associations of ApoE4 status and DHA supplementation on plasma and CSF lipid profiles and entorhinal cortex thickness. J Lipid Res. 2023 Jun;64(6):100354. Epub 2023 Mar 22 PubMed.
  16. . Diets with Higher ω-6/ω-3 Ratios Show Differences in Ceramides and Fatty Acid Levels Accompanied by Increased Amyloid-Beta in the Brains of Male APP/PS1 Transgenic Mice. Int J Mol Sci. 2021 Oct 9;22(20) PubMed.
  17. . Protective effect and mechanism of docosahexaenoic acid on the cognitive function in female APP/PS1 mice. Food Funct. 2021 Nov 15;12(22):11435-11448. PubMed.
  18. . The Cost-Effectiveness of Three Prevention Strategies in Alzheimer's Disease: Results from the Multidomain Alzheimer Preventive Trial (MAPT). J Prev Alzheimers Dis. 2021;8(4):425-435. PubMed.
  19. . Effects of Folic Acid Combined with DHA Supplementation on Cognitive Function and Amyloid-β-Related Biomarkers in Older Adults with Mild Cognitive Impairment by a Randomized, Double Blind, Placebo-Controlled Trial. J Alzheimers Dis. 2021;81(1):155-167. PubMed. RETRACTED
  20. . Associations of Omega-3 fatty acids with brain morphology and volume in cognitively healthy older adults: A narrative review. Ageing Res Rev. 2021 May;67:101300. Epub 2021 Feb 16 PubMed.
  21. . Association Between Dietary Fish and PUFA Intake in Midlife and Dementia in Later Life: The JPHC Saku Mental Health Study. J Alzheimers Dis. 2021;79(3):1091-1104. PubMed.
  22. . Eicosapentaenoic Acid Is Associated with Decreased Incidence of Alzheimer's Dementia in the Oldest Old. Nutrients. 2021 Jan 30;13(2) PubMed.
  23. . Omega-3 Fatty Acids Increase Amyloid-β Immunity, Energy, and Circadian Rhythm for Cognitive Protection of Alzheimer's Disease Patients Beyond Cholinesterase Inhibitors. J Alzheimers Dis. 2020;75(3):993-1002. PubMed.
  24. . Biological and Neuroimaging Markers as Predictors of 5-Year Incident Frailty in Older Adults: A Secondary Analysis of the MAPT Study. J Gerontol A Biol Sci Med Sci. 2021 Oct 13;76(11):e361-e369. PubMed.
  25. . Dietary fatty acids and risk of Alzheimer's disease and related dementias: Observations from the Washington Heights-Hamilton Heights-Inwood Columbia Aging Project (WHICAP). Alzheimers Dement. 2020 Jul 27; PubMed.
  26. . Omega-3 Fatty Acid-Type Docosahexaenoic Acid Protects against Aβ-Mediated Mitochondrial Deficits and Pathomechanisms in Alzheimer's Disease-Related Animal Model. Int J Mol Sci. 2020 May 29;21(11) PubMed.
  27. . Fish oil protects the blood-brain barrier integrity in a mouse model of Alzheimer's disease. Chin Med. 2020;15:29. Epub 2020 Mar 30 PubMed.
  28. . DHA brain uptake and APOE4 status: a PET study with [1-(11)C]-DHA. Alzheimers Res Ther. 2017 Mar 23;9(1):23. PubMed.
  29. . Structural and morphological characterization of aggregated species of α-synuclein induced by docosahexaenoic acid. J Biol Chem. 2011 Jun 24;286(25):22262-74. PubMed.