Abstract

Omega-6 (n-6) and omega-3 (n-3) polyunsaturated fatty acids (PUFA) are precursors of potent lipid mediators, termed eicosanoids, which play an important role in the regulation of inflammation. Eicosanoids derived from n-6 PUFAs (e.g., arachidonic acid) have proinflammatory and immunoactive functions, whereas eicosanoids derived from n-3 PUFAs [e.g., eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)] have anti-inflammatory properties, traditionally attributed to their ability to inhibit the formation of n-6 PUFA-derived eicosanoids. While the typical Western diet has a much greater ratio of n-6 PUFAs compared with n-3 PUFAs, research has shown that by increasing the ratio of n-3 to n-6 fatty acids in the diet, and consequently favoring the production of EPA in the body, or by increasing the dietary intake of EPA and DHA through consumption of fatty fish or fish-oil supplements, reductions may be achieved in the incidence of many chronic diseases that involve inflammatory processes; most notably, these include cardiovascular diseases, inflammatory bowel disease (IBD), cancer, and rheumatoid arthritis, but psychiatric and neurodegenerative illnesses are other examples.

INTRODUCTION

Polyunsaturated fatty acids (PUFAs) contain two or more double bonds that are classified as omega-3 (n-3) and omega-6 (n-6) based on the location of the last double bond relative to the terminal methyl end of the molecule. PUFA constitute an important component of all cell membranes and influence membrane fluidity and the behavior of membrane-bound enzymes and receptors. PUFAs regulate a wide range of functions in the body, including blood pressure, blood clotting, and correct development and functioning of the brain and nervous systems.1 Furthermore, PUFAs have a role in regulating inflammatory responses through the production of inflammatory mediators termed eicosanoids.1,2 The human body can produce all but two of the fatty acids it requires. Thus, linoleic acid (LA, C18:2n-6) (precursor of the n-6 series of fatty acids) and α-linolenic acid (ALA, C18:3n-3) (precursor of the n-3 series of fatty acids) are essential in the human diet; a dietary (n-6):(n-3) ratio of 4:1 is recommended as optimal. However, actual dietary intakes of these fatty acids are in excess of 15–16:1, particularly in Western countries, due to increased consumption of LA-rich vegetable oils.3 As an example, in Europe, consumption of LA has increased by 50% during the last two decades.4 In parallel with increased LA intakes, increased rates of many diseases that involve inflammatory processes have occurred, most notably cardiovascular diseases, inflammatory diseases, obesity, cancer, and certain psychiatric diseases such as depression.5 This increasing inflammatory disease incidence is associated with excessive production of the proinflammatory eicosanoids, prostaglandin E2 (PGE2) and leukotriene B4 (LTB4), which are derived from the n-6 fatty acid arachidonic acid (C20:4n-6) that is maintained at high cellular concentrations by the high n-6 and low n-3 PUFA content of the modern Western diet.6 However, this imbalance can be corrected by dietary supplementation with n-3 fatty acids such as EPA (C20:5n-3) and DHA (C22:6n-3) or by eating fish rich in EPA and DHA. EPA and DHA in the diet partially replace arachidonic acid as an eicosanoid substrate in cell membranes; this probably occurs in all cells, but it is especially true in the membranes of erythrocytes, neutrophils, monocytes, and liver cells and thus suppresses the production of n-6 proinflammatory eicosanoids.7 Marine fish are the principal sources of EPA and DHA.8 However, the content of marine n-3 fatty acids varies greatly according to the species of fish. For example, fish such as salmon, trout, and herring are higher in EPA and DHA than others (e.g., cod, haddock, and catfish). It is acknowledged that EPA and DHA can prevent the development of inflammatory diseases by directly or indirectly affecting different stages of the immune response. In addition, EPA and DHA can alleviate inflammatory processes that already exist, thus highlighting the therapeutic importance of these fatty acids.

METABOLISM OF OMEGA-6 AND OMEGA-3 POLYUNSATURATED FATTY ACIDS IN HUMANS

The main dietary sources of LA include plant oils, such as sunflower, safflower, and corn oils, cereals, animal fat, and wholegrain bread, while ALA is abundant in green leafy vegetables, flaxseed, and rapeseed oils. Although mammalian cells cannot synthesize LA and ALA, they can metabolize them into more physiologically active compounds by the introduction of further double bonds (desaturation) via Δ5 and Δ6 desaturases and by lengthening the acyl chain (elongation) via elongases. Consequently, LA is converted into γ-linolenic acid (C18:3n-6) via the action of Δ6 desaturase, and γ-linolenic acid is elongated via elongase to dihomo-γ-linolenic acid (C20:3n-6). Dihomo-γ-linolenic acid is further converted to arachidonic acid (C20:4n-6) via the action of Δ5 desaturase. Arachidonic acid is either metabolized to docosatetraenoic acid (C22:4n-6) via elongase or to eicosanoids via cyclooxygenase (COX) and lipoxygenase (LOX) (Figure 1).

Figure 1

Metabolism of n-6 and n-3 PUFA.

Using the same series of enzymes as those used to metabolize n-6 PUFAs (elongases, Δ5, and Δ6 desaturases), ALA is converted to stearidonic acid (C18:4n-3) by Δ6 desaturase; stearidonic acid is then elongated to eicosatetraenoic acid (C20:4n-3), which is converted to EPA via Δ5 desaturase. EPA is either metabolized to DHA or to eicosanoids via COX and LOX. Conversion of EPA into DHA involves the following processes: the addition of two carbons via elongase to form docosapentaenoic acid (C22:5n-3), the addition of two more carbons via elongase to produce tetracosapentaenoic acid (C24:5n-3), desaturation using Δ6 desaturase to form tetracosahexaenoic acid (C24:6n-3), and removal of two carbons by limited β-oxidation to yield DHA (C22:6n-3)9 (Figure 1). In mammals, the pathway of desaturation and elongation of n-6 and n-3 fatty acids occurs mainly in the liver. Since LA and ALA are metabolized by the same set of enzymes, competition exists between these two fatty acids, with an excess of one causing a decrease in the metabolism of the other. Normally, Δ5 and Δ6 desaturases and elongases exhibit affinity to metabolize n-3 over n-6 PUFAs, provided that both exist in a physiological ratio of 1:1–4.1,3 However, the higher concentrations of LA typically found in the Western diet results in a greater conversion of LA to arachidonic acid.10,11 Nonetheless, increasing ALA intake, such that total intake exceeds 4.5 g/day, appears to result in significant enhancements in the EPA content of plasma phospholipids.12,14 Furthermore, decreasing the LA content of the diet has been shown to result in increased metabolism of ALA to its longer chain derivatives. For example, a recent study showed that decreasing LA in the diet to an optimal LA : ALA ratio of 4:1 resulted in higher plasma phospholipid EPA and a 40% lower arachidonic acid : EPA ratio than a diet containing a LA : ALA ratio of 10:1.15 Approximately 8–20% of ALA is converted to EPA in humans, while conversion of ALA to DHA is less and estimated to be around 0.5–9%.16,17 Consequently, this route is unlikely to provide sufficient levels of EPA and DHA for optimal human health, which emphasizes the importance of dietary intake of EPA and DHA. Indeed, dietary recommendations now include not only LA and ALA but also EPA and DHA for optimal nutrition and for improving health outcomes. These recommendations are in the range of 0.5–1.8 g n-3 fatty acids per day (or the consumption of at least two servings of fish per week).18

Production of n-3 PUFAs in marine bacteria and enrichment in fish

PUFAs were once thought to be absent in bacterial membranes,19 but numerous bacterial species of marine origin, such as Shewanella spp. and Photobacterium, have now been shown to produce long-chain n-3 PUFAs such as EPA and DHA. Such isolates inhabit relatively unusual environments including low-temperature deep-sea environments and the intestines of sea fish.20,22 The enrichment of PUFA-producing strains from these environments has led to speculation that PUFA synthesis is an important adaptation for countering the effects of elevated hydrostatic pressure and low temperature on membrane fluidity. However, it is interesting that the PUFAs detected in these bacteria are mostly EPA and DHA and not C18-PUFAs such as LA and ALA, which are most common in animals, plants, fungi, and cyanobacteria.

In marine bacteria, EPA and DHA are synthesized de novo by polyunsaturated fatty acid synthase pfa genes following the polyketide biosynthesis pathway that forms PUFAs from C2-compounds (possibly acetyl-CoA) rather than by chain elongation and oxygen-dependent desaturation of existing fatty acids.23,25 Various techniques have recently been presented in order to metabolically enhance the production of EPA and DHA by bacteria that inherently synthesize them26,27 or by host bacteria transformed with pfa genes.28,29 Expressing EPA and DHA biosynthesis gene clusters in various host organisms is an alternative way of providing these beneficial fatty acids, especially since the contamination of fish due to pollution has received much attention in recent years.

The content of marine n-3 fatty acids varies greatly according to the species of fish, the total fat content of the fish, and the geographical location of waters they inhabit.30 However, as a general rule, deep water fish such as tuna, salmon, mackerel, herring, and sardines (“oily” fish) from colder temperatures have the highest content of EPA and DHA (Table 1) since they store lipids in the flesh, whereas lean fish that store lipids in the liver (e.g., cod) contain less EPA and DHA. As an example, one portion of cod provides approximately 0.3 g of EPA and DHA, one portion of salmon provides approximately 1.5 g of EPA and DHA, whereas one portion of mackerel provides approximately 3 g of these fatty acids.31 The oil obtained from the flesh of oily fish or livers of lean fish is rich in EPA and DHA, and one fish oil capsule from these sources consists of approximately 30% of these fatty acids. Thus, consumption of a typical 1 g fish oil capsule provides approximately 300 mg of EPA and DHA, which is equivalent to the consumption of one portion of cod. However, the intake of n-3 fatty acids in the absence of oily fish or fish oil supplements is likely to be <100 mg/day.32,33 Thus, an individual who consumes little or no fish could increase their daily intake of n-3 fatty acids 5-fold (or more) by taking a single standard fish oil capsule per day.34

Table 1

EPA and DHA content of fish and the amount of dietary fish required to provide approximately 1 g of EPA and DHA per day.

Fish speciesn-3 (EPA+DHA) content (g) per 100 g of fishAmount of fish (g) required to provide 1 g EPA+DHA
Atlantic herring2.0150
Atlantic salmon1.28–2.1542.5–70.9
Sardines1.15–250–87
Rainbow trout1.1587
Mackerel0.4–1.8554–250
Halibut0.47–1.1885–213
Tuna0.28–1.5166–357
Oyster0.44227
Flounder or Sole0.4204
Shrimp0.32313
Tuna (canned)0.31323
Cod0.28357
Haddock0.24417
Catfish0.18556
Scallop0.2500
Fish speciesn-3 (EPA+DHA) content (g) per 100 g of fishAmount of fish (g) required to provide 1 g EPA+DHA
Atlantic herring2.0150
Atlantic salmon1.28–2.1542.5–70.9
Sardines1.15–250–87
Rainbow trout1.1587
Mackerel0.4–1.8554–250
Halibut0.47–1.1885–213
Tuna0.28–1.5166–357
Oyster0.44227
Flounder or Sole0.4204
Shrimp0.32313
Tuna (canned)0.31323
Cod0.28357
Haddock0.24417
Catfish0.18556
Scallop0.2500

Based on data from the USDA Nutrient Data Laboratory, adapted from Kris-Etherton117. (The intakes of fish provided are rough estimates because oil content can vary markedly depending on species, season, diet, and packaging and cooking methods).

Table 1

EPA and DHA content of fish and the amount of dietary fish required to provide approximately 1 g of EPA and DHA per day.

Fish speciesn-3 (EPA+DHA) content (g) per 100 g of fishAmount of fish (g) required to provide 1 g EPA+DHA
Atlantic herring2.0150
Atlantic salmon1.28–2.1542.5–70.9
Sardines1.15–250–87
Rainbow trout1.1587
Mackerel0.4–1.8554–250
Halibut0.47–1.1885–213
Tuna0.28–1.5166–357
Oyster0.44227
Flounder or Sole0.4204
Shrimp0.32313
Tuna (canned)0.31323
Cod0.28357
Haddock0.24417
Catfish0.18556
Scallop0.2500
Fish speciesn-3 (EPA+DHA) content (g) per 100 g of fishAmount of fish (g) required to provide 1 g EPA+DHA
Atlantic herring2.0150
Atlantic salmon1.28–2.1542.5–70.9
Sardines1.15–250–87
Rainbow trout1.1587
Mackerel0.4–1.8554–250
Halibut0.47–1.1885–213
Tuna0.28–1.5166–357
Oyster0.44227
Flounder or Sole0.4204
Shrimp0.32313
Tuna (canned)0.31323
Cod0.28357
Haddock0.24417
Catfish0.18556
Scallop0.2500

Based on data from the USDA Nutrient Data Laboratory, adapted from Kris-Etherton117. (The intakes of fish provided are rough estimates because oil content can vary markedly depending on species, season, diet, and packaging and cooking methods).

POLYUNSATURATED FATTY ACIDS AND EICOSANOID PRODUCTION

Eicosanoids, including prostaglandins (PGs), thromboxanes (TXs), leukotrienes (LTs), and hydroxyeicosatetraenoic acids (HETEs), are derived from 20-carbon PUFAs, mainly arachidonic acid and EPA, and are key mediators and regulators of inflammation.35 These mediators are involved in modulating the intensity and duration of inflammatory responses.35,36 However, the overall physiological outcome of this response depends upon the cells present, the nature of the stimulus, the timing of eicosanoid generation, the concentrations of different eicosanoids generated, and the sensitivity of target cells and tissues to the eicosanoids generated.37 Competition between n-6 and n-3 fatty acids occurs in the production of eicosanoids by COX and LOX enzymes. Since inflammatory cells typically contain a high proportion of arachidonic acid and low proportions of EPA, arachidonic acid is usually the major substrate for eicosanoid synthesis.38,39 Arachidonic acid in the cell membrane is released by phospholipases, most notably phospholipase A2, and the free acid subsequently acts as a substrate for COX and LOX. Metabolism of arachidonic acid by COX gives rise to the 2-series PGs and the 2-series TXs. Monocytes and macrophages produce large amounts of prostaglandin E2 (PGE2) and prostaglandin F2α (PGF2α), neutrophils produce moderate amounts of PGE2, and mast cells produce prostaglandin D2 (PGD2). Metabolism of arachidonic acid by the 5-lipoxygenase (5-LOX) pathway gives rise to hydroxy and hydroperoxy derivatives, such as 5-HETE and 5-hydroperoxyeicosatetraenoic acid (5-HPETE), and the 4-series LTs; leukotriene A4 (LTA4), leukotriene B4 (LTB4), leukotriene C4 (LTC4), leukotriene D4 (LTD4), and leukotriene E4 (LTE4). Neutrophils, monocytes and macrophages produce LTB4, while LTC4, LTD4, and LTE4 are produced by mast cells, basophils, and eosinophils. The eicosanoids derived from arachidonic acid are generally ascribed to be proinflammatory.40 For example, PGE2 induces production of the proinflammatory cytokine interleukin (IL)-6 (IL-6) in macrophages and causes pain and vasodilation.35,40 LTB4 is a potent chemotactic agent for leukocytes and an activator of neutrophils. It also leads to the production of inflammatory cytokines such as tumor necrosis factor alpha (TNFα), IL-1 beta (IL-1β), and IL-6 by macrophages.35 Although the arachidonic acid-derived eicosanoids are generally categorized as proinflammatory mediators, they play an important modulatory role in the immune response through complex interactions with leukocytes; they also have a crucial role in the early phase of inflammation.35,39 However, in excessive concentrations, they can cause damage to host tissues and contribute to the formation of thrombi and the development of inflammatory disorders.7 Indeed, a common characteristic of chronic inflammatory diseases such as inflammatory bowel disease (IBD) and rheumatoid arthritis is excessive production of arachidonic acid-derived eicosanoids.38,42

EPA acts as a substrate for COX and LOX enzymes and gives rise to a different family of eicosanoids, the 3-series PGs and TXs, the 5-series LTs, and the hydroxy-EPAs. The eicosanoids derived from EPA are considered to be less inflammatory or even anti-inflammatory, compared to eicosanoids derived from arachidonic acid.40,43 The best example of differential inflammatory potencies of eicosanoids produced from arachidonic acid and EPA is that of LTB4 versus LTB5. LTB5, derived from EPA, is 10- to 100-fold less potent as a neutrophil chemotactic agent than LTB4, derived from arachidonic acid, and thus a much weaker inducer of inflammation.44,45 Moreover, Bagga et al. 40 demonstrated that PGE3 was a less potent inducer of IL-6 production by macrophages than PGE2.

DHA can be metabolized to resolvins via LOX-initiated mechanisms.46 Resolvins are endogenous, local-acting mediators possessing potent anti-inflammatory and immunoregulatory properties.47,49 At the cellular level, these include reducing neutrophil infiltration and regulating the cytokine-chemokine axis and reactive oxygen species, as well as lowering the magnitude of the inflammatory response.47,50 As an example, the DHA-derived resolvin E1 was shown to protect from experimental colitis in animal models.51

Increased consumption of n-3 fatty acids such as EPA and DHA results in increased proportions of these fatty acids in inflammatory cell phospholipids.52,53 The incorporation of EPA and DHA into human inflammatory cells occurs in a dose-response fashion54,55 and partly at the expense of arachidonic acid. Indeed, consumption of fish oil has been reported to increase the concentrations of EPA and DHA in inflammatory cells53,56 and subsequently decrease the production of arachidonic acid-derived eicosanoids, such as PGE2,55,58 thromboxane B2,57 LTB4,45,59 and LTE4.60

n-3 PUFAs and inflammatory gene expression

Another key anti-inflammatory effect of n-3 PUFAs is mediated at the level of altered inflammatory gene expression through their effects on transcription factors such as nuclear factor kappa B (NFκB) and peroxisome proliferator-activated receptors (PPARs).2,61 NFκB is a transcription factor that plays an important role in various inflammatory signaling pathways. It controls several cytokines (e.g., IL-1, IL-2, IL-6, IL-12, TNF-α), chemokines (e.g., IL-8, monocyte chemoattractant protein-1), adhesion molecules (intercellular adhesion molecule, vascular cell adhesion molecule, and E-selectin), and inducible effector enzymes (e.g., inducible nitric oxide synthase and COX-2).62 EPA has been shown to block the activity of NFκB through decreased degradation of the inhibitory subunit of NFκB, IκB, in cultured pancreatic cells and human monocytes.63,65 This has also been supported by the finding that transgenic mice that endogenously biosynthesize n-3 PUFAs from n-6 PUFAs are protected from colitis through a decrease in NFκB activity.66 PPARs (α, β/δ, and γ) are ligand-activated nuclear transcription factors that play important roles in cellular differentiation, cancer, inflammation, insulin sensitization, atherosclerosis, and several metabolic diseases.67 Ligands for PPARs are PUFAs, especially those of the n-3 family and their eicosanoid derivatives.68 When activated, PPARs bind to the PPAR-response element and repress or induce the transcription of target genes. PPARs have been shown to inhibit NFκB and therefore play an important role in several inflammatory processes.69,70 As an example, both EPA and DHA downregulate lipopolysaccharide-induced activation of NFκB via a PPAR-γ-dependent pathway in human kidney-2 cells.71

OMEGA-3 PUFAS AND CHRONIC INFLAMMATORY DISORDERS

A large number of clinical investigations with long-chain n-3 PUFAs in chronic inflammatory disorders have been conducted, particularly in patients with rheumatoid arthritis (reviewed in Calder72) and IBD (reviewed in Calder37). In these studies, n-3 PUFAs have been demonstrated to exhibit immunomodulatory effects by changing the profiles of the eicosanoids produced and decreasing the levels of proinflammatory cytokines, via both lipid-mediator-related and nonlipid-mediator-related mechanisms.2,73 The increased incidence of IBD in humans correlates with an increased dietary intake of n-6 fatty acids.74,75 Thus, increasing the ratio of n-3 to n-6 fatty acids in the diet may be of therapeutic importance for IBD. Indeed, it has been reported that n-3 PUFAs are incorporated into the gut mucosal tissue of patients with IBD who supplement their diet with fish oil, subsequently resulting in an anti-inflammatory effect with decreased production of LTB4 by neutrophils and colonic mucosa76,77 and decreased production of PGE2 and IFN-γ by blood mononuclear cells.78 A recent study using IL-10 knockout mice (mice that spontaneously develop colitis) demonstrated significantly reduced colonic inflammation of mice that were fed fish oil compared with mice that were fed n-6 PUFA-rich corn oil.79 Furthermore, in a 1-year study, patients with Crohn's disease in remission were randomized to receive either placebo or 2.7 g of n-3 PUFAs (administered as fish oil capsules) per day. The primary outcome of the study was relapse. After 12 months, there was a significant difference in the proportion of patients who relapsed over this time: 11/39 (28%) in the fish-oil group versus 27/39 (69%) in the placebo group. There was also a significant difference in the proportion of patients who remained in remission at 12 months: 59% in the fish-oil group compared to 26% in the placebo group.80 Ferrucci et al. 81 examined the relationship between relative concentrations of fatty acids in fasting plasma and levels of inflammatory markers in 1,123 persons aged 20–98 years. The total concentrations of n-3 PUFAs were associated with lower levels of proinflammatory markers (IL-6, TNF-α, C-reactive protein) and higher concentrations of anti-inflammatory markers (IL-10, transforming growth factor-β). Thus, the authors concluded that n-3 fatty acids are beneficial in patients affected by diseases characterized by active inflammation.81

Uncontrolled inflammation also governs the pathogenesis of many prevalent diseases, such as cardiovascular disease, cancer, obesity, and Alzheimer's disease.82 For example, there is a strong association between systemic inflammation and coronary artery disease. This association is thought to be casual, i.e., inflammation increases the risk of the disease, rather than simply marking the presence of atherosclerosis, which is an inflammatory process.83 In the last decade it has been increasingly recognized that n-3 PUFAs can modulate the mechanisms of development and progression of atherosclerosis. Early studies in Greenland Eskimos, a population consuming a high-fat diet rich in n-3 PUFAs, demonstrated that the ingestion of EPA and DHA protected them from cardiovascular disease.84 Similarly, the Japanese population eats more fish than North Americans and presents a lower rate of acute myocardial infarction and atherosclerosis.85,86

Since the early observation in the Greenland Eskimos, many supplementation studies have been completed that have demonstrated associations between n-3 PUFAs and decreased risks of cardiovascular disease87,89 and reduced mortality.90,91 However, some studies have shown no significant associations.92,93 Two major randomized trials have documented the effects of n-3 PUFAs in the prevention of cardiovascular disease.90,94 The GISSI-Prevenzione study randomized 11,323 patients with recent myocardial infarction to n-3 PUFAs (1 g/day), vitamin E (300 mg/day), both, or none for 3.5 years. Treatment with n-3 PUFAs significantly reduced cardiac death by 32% and sudden death by 45%, whereas vitamin E showed no significant benefit.94 In the more recent Japanese JELIS trial, 18,645 patients with hypercholesterolemia were randomized to receive statin alone or statin and highly purified EPA (1.8 g/day). At the end of the 5-year study, it was found that those patients who were randomized to EPA had a 19% significant reduction in major coronary events.90 Furthermore, Matsuzaki et al. 95 examined whether EPA is effective for the secondary prevention of coronary artery disease. Patients with established coronary artery disease were randomly assigned to receive either 1.8 g of EPA in combination with statin or statin alone. After 4.6 years, the incidence of major coronary events was significantly lower in the patients receiving EPA, suggesting that EPA is effective for secondary prevention of coronary artery disease.95

Although there are some inconsistent results between n-3 PUFA intake and cardiovascular health, which may be explained by variations in such factors as dosage, sample size, and follow-up period, the majority of experimental and observational studies have shown that intake of n-3 PUFAs is associated with reduced risk of cardiovascular diseases.96 Numerous organizations and health agencies are therefore recommending consumption of EPA and DHA for general cardiovascular health. It is not yet clear how much n-3 PUFA is required to significantly reduce the risk of cardiovascular diseases or what the optimal dose is for primary prevention. However, the typical recommendation for secondary prevention of myocardial infarction is a minimum intake of 1 g/day.97,98

Alzheimer's disease (AD) is a neurodegenerative disease that commonly affects the elderly. Currently, there is no known cure for AD. However, there is growing interest in the role of diet in the prevention of AD. DHA is one of the major fatty acids in the brain, representing between 12 and 16% of total fatty acids in grey-matter lipids.99 DHA is required for fetal brain development and is held to be critical for appropriate development and intelligence as well as the maintenance of cognitive capacity.100,101 Quite remarkably, the main source of DHA for the brain is diet. A number of animal studies have revealed that the dietary intake of DHA can significantly alter levels of DHA in the brain.102,104 Two independent prospective studies have shown that lower plasma DHA levels are associated with increased risk of developing AD later in life.105,106 As an example, patients with blood DHA concentrations in the highest quartile had a lower risk of developing dementia compared with the lowest three quartiles during a follow-up period of 9 years.106 This suggests that dietary intake of DHA can alter the risk of developing AD over the long-term and that low blood DHA concentrations might be an important risk factor for AD. Studies have also shown that DHA provides support to learning and memory events in animal models of AD and protection against AD.107,109 Moreover, several epidemiological studies have shown a protective effect associated with increased fish consumption and intake of unsaturated fats leading to low n-6/n-3 fatty acid ratios.110,112 For example, van Gelder et al.113 studied the association between fish consumption and cognitive decline in patients aged 70–89 years. Fish consumers exhibited significantly less cognitive decline after 5 years compared with nonconsumers. Thus, n-3 fatty acids, particularly DHA, may have therapeutic utility for the prevention and treatment of AD.

Recent studies have also demonstrated that administration of n-3 fatty acids decreases the length of hospital stay in patients undergoing surgery by modulating the immune response.114 For example, parenteral infusion with a fish oil containing emulsion to patients following major abdominal surgery lead to a rapid incorporation of n-3 fatty acids (EPA and DHA) into plasma phospholipids of cell membranes of leukocytes and platelets and increased EPA : AA and LTB5 : LTB4 ratios.115 Moreover, the administration of n-3 fatty acid-supplemented oil to patients undergoing radical colorectal cancer resection decreased serum IL-6 concentrations and TNF-α concentrations and reduced the hospital stay for these patients.116 Thus, postoperative supplementation of n-3 PUFAs lower the magnitude of inflammatory responses and leads to shorter hospital and intensive care unit stays in surgical patients.

CONCLUSION

Increased consumption of fatty fish or fish oil supplements containing n-3 PUFAs increases the amount of these fatty acids and their metabolites in human immune cells and consequently changes the production of important mediators and regulators of inflammation and immune responses towards an anti-inflammatory profile. Since excessive intake of n-6 PUFAs, which is characteristic of Western diets, could potentiate inflammatory processes and consequently predispose to, or exacerbate, inflammatory diseases, increasing intake of fatty acids that elicit anti-inflammatory effects, such as n-3 PUFAs, could decrease the risk of many chronic diseases like arthritis, diabetes, obesity, inflammation, cancer, and cardiovascular disease as well as improve mental health. Based on the recognized health effects of n-3 PUFAs, recommendations have been made to increase dietary intake of these fatty acids; this can be achieved by increasing consumption of oily fish or by consuming fish oil supplements.

Acknowledgments

Funding

The authors are funded by Alimentary Pharmabiotic Centre through the Science Foundation Ireland (SFI), NUTRMARA, funded by the Marine Institute, Co. Galway, the Irish Ministry for Food and Agriculture, the Higher Education Authority, and the Health Research Board of Ireland.

Declaration of interest

The authors have no relevant interests to declare.

REFERENCES

1

Das
UN
.
Biological significance of essential fatty acids
.
J Assoc Physicians India
.
2006
;
54
:
309
319
.

2

Calder
PC
.
n–3 polyunsaturated fatty acids, inflammation, and inflammatory diseases
.
Am J Clin Nutr
.
2006
;
83
:
1505
1519
.

3

Simopoulos
AP
.
Importance of the ratio of omega-6/omega-3 essential fatty acids: evolutionary aspects
.
World Rev Nutr Diet
.
2003
;
92
:
1
22
.

4

Sanders
TA
.
Polyunsaturated fatty acids in the food chain in Europe
.
Am J Clin Nutr
.
2000
;
71(Suppl)
:
S176
S178
.

5

Simopoulos
AP
Leaf
A
Salem
N
.
Workshop statement on the essentiality of and recommended dietary intakes for omega-6 and omega-3 fatty acids
.
Prostaglandins Leukot Essent Fatty Acids
.
2000
;
63
:
119
121
.

6

James
MJ
Gibson
RA
Cleland
LG
.
Dietary polyunsaturated fatty acids and inflammatory mediator production
.
Am J Clin Nutr
.
2000
;
71(Suppl)
:
S343
S348
.

7

Simopoulos
AP
.
Essential fatty acids in health and chronic diseases
.
Forum Nutr
.
2003
;
56
:
67
70
.

8

Shahidi
F
Miraliakbari
H
.
Omega-3 (n-3) fatty acids in health and disease: part 1 – cardiovascular disease and cancer
.
J Med Food
.
2004
;
7
:
387
401
.

9

Sprecher
H
.
Metabolism of highly unsaturated n-3 and n-6 fatty acids
.
Biochim Biophys Acta
.
2000
;
1486
:
219
231
.

10

Simopoulos
AP
.
n-3 fatty acids and human health: defining strategies for public policy
.
Lipids
.
2001
;
36(Suppl)
:
S83
S89
.

11

Kris-Etherton
P
Hecker
M
Binkoski
KD
.
Polyunsaturated fatty acids and cardiovascular health
.
Nutr Rev
.
2004
;
62
:
414
426
.

12

Mantzioris
E
James
MJ
Gibson
RA
Cleland
LG
.
Dietary substitution with an alpha-linolenic acid rich vegetable oil increases eicosapentaenoic acid concentrations in tissues
.
Am J Clin Nutr
.
1994
;
59
:
1304
1309
.

13

Wallace
FA
Miles
EA
Calder
PC
.
Comparison of the effects of linseed oil and different doses of fish oil on mononuclear cell function in healthy human subjects
.
Br J Nutr
.
2003
;
89
:
679
689
.

14

Finnegan
YE
Minihane
AM
Leigh-Firbank
EC
, et al.
Plant- and marine-derived n-3 polyunsaturated fatty acids have differential effects on fasting and postprandial blood lipid concentrations and on the susceptibility of LDL to oxidative modification in moderately hyperlipidemic subjects
.
Am J Clin Nutr
.
2003
;
77
:
783
795
.

15

Liou
YA
King
DJ
Zibrik
D
Innis
SM
.
Decreasing linoleic acid with constant alpha-linolenic acid in dietary fats increases (n-3) eicosapentaenoic acid in plasma phospholipids in healthy men
.
J Nutr
.
2007
;
137
:
945
952
.

16

Burdge
GC
.
Metabolism of alpha-linolenic acid in humans
.
Prostaglandins Leukot Essent Fatty Acids
.
2006
;
75
:
161
168
.

17

DeFilippis
AP
Sperling
LS
.
Understanding omega-3s
.
Am Heart J
.
2006
;
151
:
564
570
.

18

Kris-Etherton
PM
Griegera
JA
Etherton
TB
.
Dietary reference intakes for DHA, EPA
.
Prostaglandins Leukot Essent Fatty Acids
.
2009
;
81
:
99
104
.

19

Erwin
J
Bloch
K
.
Biosynthesis of unsaturated fatty acids in microorganisms
.
Science
.
1964
;
143
:
1006
1012
.

20

Wirsen
CO
Jannaschm
HW
Waleham
SG
Canuel
EA
.
Membrane lipids of a psychrophilic and barophilic deep-sea bacterium
.
Curr Microbiol
.
1987
;
14
:
312
322
.

21

Singh
A
Ward
OP
.
Microbial production of docosahexaenoic acid (DHA, C22:6)
.
Adv Appl Microbiol
.
1997
;
45
:
271
312
.

22

Yanu
Y
Nakayama
A
Saito
H
Ishihara
K
.
Production of docosahexaenoic acid by marine bacteria isolated from deep-sea fish
.
Lipids
.
1994
;
29
:
527
528
.

23

Metz
JG
Roessler
P
Facciotti
D
, et al.
Production of polyunsaturated fatty acids by polyketide synthases in both prokaryotes and eukaryotes
.
Science
.
2001
;
293
:
290
293
.

24

Ootaki
M
Morita
N
Nishida
T
, et al.
Genes and pathways involved in biosynthesis of eicosapentaenoic and docosahexaenoic acids in bacteria
. In:
Murata
N
Yamada
M
Nishida
I
Okuyama
H
Sekiya
J
Wada
H
, eds.
Advanced Research on Plant Lipids
. Dordrecht, The Netherlands:
Kluwer Academic Publishers
;
2003
:
49
52
.

25

Orikasa
Y
Yamada
A
Yu
R
, et al.
Characterisation of the eicosapentaenoic acid biosynthesis gene cluster from Shewanella sp. Strain SCRC-2738
.
Cell Mol Biol
.
2004
;
50
:
625
630
.

26

Morita
N
Nishida
T
Tanaka
N
Yano
Y
Okuyama
H
.
Enhancement of polyunsaturated fatty acid production by cerulenin treatment in polyunsaturated fatty acid-producing bacteria
.
Biotechnol Lett
.
2005
;
27
:
389
393
.

27

Amiri-Jami
M
Wang
H
Kakuda
Y
Griffiths
MW
.
Enhancement of polyunsaturated fatty acid production by Tn5 transposon in Shewanella baltica
.
Biotechnol Lett
.
2006
;
28
:
1187
1192
.

28

Orikasa
Y
Nishida
T
Yamada
A
, et al.
Recombinant production of docosahexaenoic acid in a polyketide biosynthesis mode in Escherichia coli
.
Biotechnol Lett
.
2006
;
28
:
1841
1847
.

29

Lee
SJ
Kim
CH
Seo
PS
Kwon
O
Hur
BK
Seo
JW
.
Enhancement of heterologous production of eicosapentaenoic acid in Escherichia coli by substitution of promoter sequences within the biosynthesis gene cluster
.
Biotechnol Lett
.
2008
;
30
:
2139
2142
.

30

Larsson
SC
Kumlin
M
Ingelman-Sundberg
M
Wolk
A
.
Dietary long-chain n-3 fatty acids for the prevention of cancer: a review of potential mechanisms
.
Am J Clin Nutr
.
2004
;
79
:
935
945
.

31

Brittish Nutrition Foundation
.
Briefing Paper: n-3 Fatty Acids and Health
. London:
British Nutrition Foundation
;
1999
.

32

Calder
PC
Burdge
GC
.
Fatty acids
. In:
Nicolaou
A
Kokotos
G
, eds.
Bioactive Lipids
. Bridgewater, UK:
The Oily Press
;
2004
:
1
36
.

33

Surette
ME
.
The science behind omega-3 fatty acids
.
Can Med Assoc J
.
2008
;
178
:
177
180
.

34

Calder
PC
Yaqoob
P
.
Omega-3 polyunsaturated fatty acids and human health outcomes
.
Biofactors
.
2009
;
35
:
266
272
.

35

Tilley
SL
Coffman
TM
Koller
BH
.
Mixed messages: modulation of inflammation and immune responses by prostaglandins and thromboxanes
.
J Clin Invest
.
2001
;
108
:
15
23
.

36

Kinsella
JE
Lokesh
B
Broughton
S
Whelan
J
.
Dietary polyunsaturated fatty acids and eicosanoids: potential effects on the modulation of inflammatory and immune cells: an overview
.
Nutrition
.
1990
;
6
:
24
44
.

37

Calder
PC
.
Polyunsaturated fatty acids, inflammatory processes and inflammatory bowel diseases
.
Mol Nutr Food Res
.
2008
;
52
:
885
897
.

38

Calder
PC
.
Polyunsaturated fatty acids, inflammation, and immunity
.
Lipids
.
2001
;
36
:
1007
1024
.

39

Simopoulos
AP
.
Omega-3 fatty acids in inflammation and autoimmune diseases
.
J Am Coll Nutr
.
2002
;
21
:
495
505
.

40

Bagga
D
Wang
L
Farias-Eisner
R
Glaspy
JA
Reddy
ST
.
Differential effects of prostaglandin derived from omega-6 and omega-3 polyunsaturated fatty acids on COX-2 expression and IL-6 secretion
.
Proc Natl Acad Sci USA
.
2003
;
100
:
1751
1756
.

41

Wallace
JL
.
Prostaglandin biology in inflammatory bowel disease
.
Gastroenterol Clin North Am
.
2001
;
30
:
971
980
.

42

Jupp
J
Hillier
K
Elliott
DH
, et al.
Colonic expression of leukotriene-pathway enzymes in inflammatory bowel diseases
.
Inflamm Bowel Dis
.
2007
;
13
:
537
546
.

43

Robinson
JG
Stone
NJ
.
Antiatherosclerotic and antithrombotic effects of omega-3 fatty acids
.
Am J Cardiol
.
2006
;
98
:
39
49
.

44

Goldman
DW
Pickett
WC
Goetzl
EJ
.
Human neutrophil chemotactic and degranulating activities of leukotriene B5 (LTB5) derived from eicosapentaenoic acid
.
Biochem Biophys Res Commun
.
1983
;
117
:
282
288
.

45

Lee
TH
Mencia-Huerta
JM
Shih
C
Corey
EJ
Lewis
RA
Austen
KF
.
Characterization and biologic properties of 5,12-dihydroxy derivatives of eicosapentaenoic acid, including leukotriene-B5 and the double lipoxygenase product
.
J Biol Chem
.
1984
;
259
:
2383
2389
.

46

Hong
S
Gronert
K
Devchand
PR
Moussignac
RL
Serhan
CN
.
Novel docosatrienes and 17S-resolvins generated from docosahexaenoic acid in murine brain, human blood, and glial cells. Autacoids in anti-inflammation
.
J Biol Chem
.
2003
;
278
:
14677
14687
.

47

Serhan
CN
Hong
S
Gronert
K
, et al.
Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals
.
J Exp Med
.
2002
;
196
:
1025
1037
.

48

Serhan
CN
Lu
Y
Hong
S
Yang
R
.
Mediator lipidomics: search algorithms for eicosanoids, resolvins, and protectins
.
Meth Enzymol
.
2007
;
432
:
275
317
.

49

Schwab
JM
Chiang
N
Arita
M
Serhan
CN
.
Resolvin E1 and protectin D1 activate inflammation-resolution programmes
.
Nature
.
2007
;
447
:
869
874
.

50

Serhan
CN
Clish
CB
Brannon
J
Colgan
SP
Chiang
N
Gronert
K
.
Novel functional sets of lipid-derived mediators with antiinflammatory actions generated from omega-3 fatty acids via cyclooxygenase 2-nonsteroidal anti-inflammatory drugs and transcellular processing
.
J Exp Med
.
2000
;
192
:
1197
1204
.

51

Weylandt
KH
Kang
JX
Wiedenmann
B
Baumgart
DC
.
Lipoxins and resolvins in inflammatory bowel disease
.
Inflamm Bowel Dis
.
2007
;
13
:
797
799
.

52

Endres
S
Ghorbani
R
Kelley
VE
, et al.
The effect of dietary supplementation with n-3 polyunsaturated fatty acids on the synthesis of interleukin-1 and tumor necrosis factor by mononuclear cells
.
N Engl J Med
.
1989
;
320
:
265
271
.

53

Yaqoob
P
Pala
HS
Cortina-Borja
M
Newsholme
EA
Calder
PC
.
Encapsulated fish oil enriched in a-tocopherol alters plasma phospholipid and mononuclear cell fatty acid compositions but not mononuclear cell functions
.
Eur J Clin Invest
.
2000
;
30
:
260
274
.

54

Healy
DA
Wallace
FA
Miles
EA
Calder
PC
Newsholm
P
.
Effect of low-to-moderate amounts of dietary fish oil on neutrophil lipid composition and function
.
Lipids
.
2000
;
35
:
763
768
.

55

Rees
D
Miles
EA
Banerjee
T
, et al.
Dose-related effects of eicosapentaenoic acid on innate immune function in healthy humans: a comparison of young and older men
.
Am J Clin Nutr
.
2006
;
83
:
331
342
.

56

Gibney
MJ
Hunter
B
.
The effects of short- and long-term supplementation with fish oil on the incorporation of n-3 polyunsaturated fatty acids into cells of the immune system in healthy volunteers
.
Eur J Clin Nutr
.
1993
;
47
:
255
259
.

57

Caughey
GE
Mantzioris
E
Gibson
RA
Cleland
LG
James
MJ
.
The effect on human tumor necrosis factor α and interleukin 1β production of diets enriched in n-3 fatty acids from vegetable oil or fish oil
.
Am J Clin Nutr
.
1996
;
63
:
116
122
.

58

Trebble
TM
Wootton
SA
Miles
EA
, et al.
Prostaglandin E2 production and T cell function after fish-oil supplementation: response to antioxidant cosupplementation
.
Am J Clin Nutr
.
2003
;
78
:
376
382
.

59

Kelley
DS
Taylor
PC
Nelson
GJ
, et al.
Docosahexaenoic acid ingestion inhibits natural killer cell activity and production of inflammatory mediators in young healthy men
.
Lipids
.
1999
;
34
:
317
324
.

60

Von Schacky
C
Kiefl
R
Jendraschak
E
Kaminski
WE
.
n-3 fatty acids and cysteinyl-leukotriene formation in humans in vitro, ex vivo, and in vivo
.
J Lab Clin Med
.
1993
;
121
:
302
309
.

61

Calder
PC
.
Dietary modification of inflammation with lipids
.
Proc Nutr Soc
.
2002
;
61
:
345
358
.

62

Ghosh
S
Karin
M
.
Missing pieces in the NF-kappaB puzzle
.
Cell
.
2002
;
109(Suppl)
:
S81
S96
.

63

Lo
CJ
Chiu
KC
Fu
M
Lo
R
Helton
S
.
Fish oil decreases macrophage tumor necrosis factor gene transcription by altering the NF kappa B activity
.
J Surg Res
.
1999
;
82
:
216
221
.

64

Novak
TE
Babcock
TA
Jho
DH
Helton
WS
Espat
NJ
.
NF-κB inhibition by ω-3 fatty acids modulates LPS-stimulated macrophage TNF-α transcription
.
Am J Physiol
.
2003
;
284
:
L84
L89
.

65

Zhao
Y
Joshi-Barve
S
Barve
S
Chen
LH
.
Eicosapentaenoic acid prevents LPS-induced TNF-α expression by preventing NF-κB activation
.
J Am Coll Nutr
.
2004
;
23
:
71
78
.

66

Hudert
CA
Weylandt
KH
Lu
Y
, et al.
Transgenic mice rich in endogenous omega-3 fatty acids are protected from colitis
.
Proc Natl Acad Sci USA
.
2006
;
103
:
11276
11281
.

67

Chawla
A
Repa
JJ
Evans
RM
Mangelsdorf
DJ
.
Nuclear receptors and lipid physiology: opening the X-files
.
Science
.
2001
;
294
:
1866
1870
.

68

Kota
BP
Huang
TH
Roufogalis
BD
.
An overview on biological mechanisms of PPARs
.
Pharmacol Res
.
2005
;
51
:
85
94
.

69

Poynter
ME
Daynes
RA
.
Peroxisome proliferator-activated receptor alpha activation modulates cellular redox status, represses nuclear factor-kappaB signaling, and reduces inflammatory cytokine production in aging
.
J Biol Chem
.
1998
;
273
:
32833
32841
.

70

Ricote
M
Huang
JT
Welch
JS
Glass
CK
.
The peroxisome proliferator-activated receptor (PPARgamma) as a regulator of monocyte/macrophage function
.
J Leukoc Biol
.
1999
;
66
:
733
739
.

71

Li
H
Ruan
XZ
Powis
SH
, et al.
EPA and DHA reduce LPS-induced inflammation responses in HK-2 cells: evidence for a PPAR-gamma-dependent mechanism
.
Kidney Int
.
2005
;
67
:
867
874
.

72

Calder
PC
.
Session 3: Joint Nutrition Society and Irish Nutrition and Dietetic Institute Symposium on “Nutrition and autoimmune disease” PUFA, inflammatory processes and rheumatoid arthritis
.
Proc Nutr Soc
.
2008
;
67
:
409
418
.

73

Sijben
JWC
Calder
PC
.
Differential immunomodulation with long-chain n-3 PUFA in health and chronic disease
.
Proc Nutr Soc
.
2007
;
66
:
237
259
.

74

Shoda
R
Matsueda
K
Yamato
S
Umeda
N
.
Epidemiologic analysis of Crohn disease in Japan: increased dietary intake of n-6 polyunsaturated fatty acids and animal protein relates to the increased incidence of Crohn disease in Japan
.
Am J Clin Nutr
.
1996
;
63
:
741
745
.

75

Sakamoto
N
Kono
S
Wakai
K
, et al.
Dietary risk factors for inflammatory bowel disease: a multicenter case-control study in Japan
.
Inflamm Bowel Dis
.
2005
;
11
:
154
163
.

76

Shimizu
T
Fujii
T
Suzuki
R
, et al.
Effects of highly purified eicosapentaenoic acid on erythrocyte fatty acid composition and leukocyte and colonic mucosa leukotriene B4 production in children with ulcerative colitis
.
J Pediatr Gastroenterol Nutr
.
2003
;
37
:
581
585
.

77

Hawthorne
AB
Daneshmend
TK
Hawkey
CJ
.
Treatment of ulcerative colitis with fish oil supplementation: a prospective 12 month randomised controlled trial
.
Gut
.
1992
;
33
:
922
928
.

78

Trebble
TM
Arden
NK
Wootton
SA
, et al.
Fish oil and antioxidants alter the composition and function of circulating mononuclear cells in Crohn disease
.
Am J Clin Nutr
.
2004
;
80
:
1137
1144
.

79

Chapkin
RS
Davidson
LA
Ly
L
Weeks
BR
Lupton
JR
McMurray
DN
.
Immunomodulatory effects of (n-3) fatty acids: putative link to inflammation and colon cancer
.
J Nutr
.
2007
;
137
:
200
204
.

80

Belluzzi
A
Brignola
C
Campieri
M
Pera
A
Boschi
S
Miglioli
M
.
Effect of an enteric-coated fish-oil preparation on relapses in Crohn's disease
.
N Engl J Med
.
1996
;
334
:
1557
1560
.

81

Ferrucci
L
Cherubini
A
Bandinelli
S
, et al.
Relationship of plasma polyunsaturated fatty acids to circulating inflammatory markers
.
J Clin Endocrinol Metab
.
2006
;
91
:
439
446
.

82

Serhan
CN
Savill
J
.
Resolution of inflammation: the beginning programs the end
.
Nat Immunol
.
2005
;
6
:
1191
1197
.

83

Ross
R
.
Atherosclerosis – an inflammatory disease
.
N Engl J Med
.
1999
;
340
:
115
126
.

84

Bang
HO
Dyerberg
J
.
Lipid metabolism and ischemic heart disease in Greenland Eskimos
.
Acta Med Scand
.
1976
;
200
:
69
73
.

85

Menotti
A
Kromhout
D
Blackburn
H
Fidanza
F
Buzina
R
Nissinen
A
.
Food intake patterns and 25-year mortality from coronary heart disease: cross-cultural correlations in the Seven Countries Study. The Seven Countries Study Research Group
.
Eur J Epidemiol
.
1999
;
15
:
507
515
.

86

Holub
BJ
.
Clinical nutrition: 4. Omega-3 fatty acids in cardiovascular care
.
CMAJ
.
2002
;
166
:
608
615
.

87

Lemaitre
RN
King
IB
Mozaffarian
D
Kuller
LH
Tracy
RP
Siscovick
DS
.
n-3 polyunsaturated fatty acids, fatal ischemic heart disease, and nonfatal myocardial infarction in older adults: the Cardiovascular Health Study
.
Am J Clin Nutr
.
2003
;
77
:
279
280
.

88

He
K
Song
Y
Daviglus
ML
, et al.
Accumulated evidence on fish consumption and coronary heart disease mortality: a meta-analysis of cohort studies
.
Circulation
.
2004
;
109
:
2705
2711
.

89

Mozaffarian
D
Geelen
A
Brouwer
IA
Geleijnse
JM
Zock
PL
Katan
MB
.
Effect of fish oil on heart rate in humans: a meta-analysis of randomized controlled trials
.
Circulation
.
2005
;
112
:
1945
1952
.

90

Yokoyama
M
Origasa
H
Matsuzaki
M
, et al.
Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis
.
Lancet
.
2007
;
369
:
1090
1098
.

91

Gissi
HF
Tavazzi
L
Maggioni
A
, et al.
Effect on n-3 polyunsaturated fatty acids in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial
.
Lancet
.
2008
;
372
:
1223
1230
.

92

Folsom
AR
Demissie
Z
.
Fish intake, marine omega-3 fatty acids, and mortality in a cohort of postmenopausal women
.
Am J Epidemiol
.
2004
;
160
:
1005
1010
.

93

Streppel
MT
Ocké
MC
Boshuizen
HC
Kok
FJ
Kromhout
D
.
Long-term fish consumption and n-3 fatty acid intake in relation to (sudden) coronary heart disease death: the Zutphen study
.
Eur Heart J
.
2008
;
29
:
2024
2030
.

94

Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto Miocardio (GISSI) (Italian Group for the Study of the Survival of Myocardial Infarction)
.
Dietary supplementation with n-3 polyunsaturated fatty acids and vitamin E after myocardial infarction: results of the GISSI-Prevenzione trial. Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto miocardico
.
Lancet
.
1999
;
354
:
447
455
.

95

Matsuzaki
M
Yokoyama
M
Saito
Y
, et al.
Incremental effects of eicosapentaenoic acid on cardiovascular events in statin-treated patients with coronary artery disease
.
Circ J
.
2009
;
73
:
1283
1290
.

96

He
K
.
Fish, long-chain omega-3 polyunsaturated fatty acids and prevention of cardiovascular disease – eat fish or take fish oil supplements?
Prog Cardiovasc Dis
.
2009
;
52
:
95
114
.

97

Van de Werf
F
Bax
J
Betriu
A
, et al.
Management of acute myocardial infarction in patients presenting with persistent ST-segment elevation: the Task Force on the Management of ST-Segment Elevation Acute Myocardial Infarction of the European Society of Cardiology
.
Eur Heart J
.
2008
;
29
:
2909
2945
.

98

British Cardiac Society, British Hypertension Society, Diabetes UK, and HEART UK. Primary Care Cardiovascular Society, and Stroke Association
.
JBS 2: Joint British Societies' guidelines on prevention of cardiovascular disease in clinical practice
.
Heart
.
2005
;
91
:
v1
v52
.

99

Julien
C
Berthiaume
L
Hadj-Tahar
A
, et al.
Postmortem brain fatty acid profile of levodopa-treated Parkinson disease patients and Parkinsonian monkeys
.
Neurochem Int
.
2006
;
48
:
404
414
.

100

Salem
N
Wegher
B
Mena
P
Uauy
R
.
Arachidonic and docosahexaenoic acids are biosynthesized from their 18-carbon precursors in human infants
.
Proc Natl Acad Sci USA
.
1996
;
93
:
49
54
.

101

Fedorova
I
Salem
N
Jr
.
Omega-3 fatty acids and rodent behaviour
.
Prostaglandins Leukot Essent Fatty Acids
.
2006
;
75
:
271
289
.

102

Pawlosky
RJ
Bacher
J
Salemn
N
Jr
.
Ethanol consumption alters electroretinograms and depletes neural tissues of docosahexaenoic acid in rhesus monkeys: nutritional consequences of a low n-3 fatty acid diet
.
Alcohol Clin Exp Res
.
2001
;
25
:
1758
1765
.

103

Murthy
M
Hamilton
J
Greiner
RS
Moriguchi
T
Salem
N
Jr
Kim
HY
.
Differential effects of n-3 fatty acid deficiency on phospholipid molecular species composition in the rat hippocampus
.
J Lipid Res
.
2002
;
43
:
611
617
.

104

Levant
B
Ozias
MK
Carlson
SE
.
Specific brain regions of female rats are differentially depleted of docosahexaenoic acid by reproductive activity and an (n-3) fatty acid-deficient diet
.
J Nutr
.
2007
;
137
:
130
134
.

105

Heude
B
Ducimetière
P
Berr
C
.
Cognitive decline and fatty acid composition of erythrocyte membranes – The EVA Study
.
Am J Clin Nutr
.
2003
;
77
:
803
808
.

106

Schaefer
EJ
Bongard
V
Beiser
AS
, et al.
Plasma phosphatidylcholine docosahexaenoic acid content and risk of dementia and Alzheimer disease: the Framingham Heart Study
.
Arch Neurol
.
2006
;
63
:
1545
1550
.

107

Hashimoto
M
Hossain
S
Shimada
T
, et al.
Docosahexaenoic acid provides protection from impairment of learning ability in Alzheimer's disease model rats
.
J Neurochem
.
2002
;
81
:
1084
1091
.

108

Lim
GP
Calon
F
Morihara
T
, et al.
A diet enriched with the omega-3 fatty acid docosahexaenoic acid reduces amyloid burden in an aged Alzheimer mouse model
.
J Neurosci
.
2005
;
25
:
3032
3040
.

109

Hashimoto
M
Hossain
S
Agdul
H
Shido
O
.
Docosahexaenoic acid-induced amelioration on impairment of memory learning in amyloid beta-infused rats relates to the decreases of amyloid beta and cholesterol levels in detergent-insoluble membrane fractions
.
Biochim Biophys Acta
.
2005
;
1738
:
91
98
.

110

Barberger-Gateau
P
Letenneur
L
Deschamps
V
Pérès
K
Dartigues
JF
Renaud
S
.
Fish, meat, and risk of dementia: cohort study
.
BMJ
.
2002
;
325
:
932
933
.

111

Morris
MC
Evans
DA
Bienias
JL
, et al.
Consumption of fish and n-3 fatty acids and risk of incident Alzheimer disease
.
Arch Neurol
.
2003
;
60
:
940
946
.

112

Kalmijn
S
Van Boxtel
MP
Ocke
M
, et al.
Dietary intake of fatty acids and fish in relation to cognitive performance at middle age
.
Neurology
.
2004
;
62
:
275
280
.

113

Van Gelder
BM
Tijhuis
M
Kalmijn
S
Kromhout
D
.
Fish consumption, n-3 fatty acids, and subsequent 5-y cognitive decline in elderly men: the Zutphen Elderly Study
.
Am J Clin Nutr
.
2007
;
85
:
1142
1147
.

114

Waitzberg
DL
Torrinhas
RS
.
Fish oil lipid emulsions and immune response: what clinicians need to know
.
Nutr Clin Pract
.
2009
;
24
:
487
499
.

115

Grimm
H
Mertes
N
Goeters
C
, et al.
Improved fatty acid and leukotriene pattern with a novel lipid emulsion in surgical patients
.
Eur J Nutr
.
2006
;
45
:
55
60
.

116

Liang
B
Wang
S
Ye
YJ
, et al.
Impact of postoperative omega-3 fatty acid-supplemented parenteral nutrition on clinical outcomes and immunomodulations in colorectal cancer patients
.
World J Gastroenterol
.
2008
;
14
:
2434
2439
.

117

Kris-Etherton
PM
.
Fish consumption, fish oil, omega-3 fatty acids, and cardiovascular disease
.
Circulation
.
2002
;
106
:
2747
2757
.