Hostname: page-component-8448b6f56d-m8qmq Total loading time: 0 Render date: 2024-04-23T07:36:18.432Z Has data issue: false hasContentIssue false

Very long-chain n-3 fatty acids and human health: fact, fiction and the future

Published online by Cambridge University Press:  17 October 2017

P. C. Calder*
Affiliation:
Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, MP887 Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
*
Corresponding author: P. C. Calder, email pcc@soton.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

EPA and DHA appear to be the most important n-3 fatty acids, but roles for n-3 docosapentaenoic acid are now also emerging. Intakes of EPA and DHA are usually low, typically below those recommended. Increased intakes result in higher concentrations of EPA and DHA in blood lipids, cells and tissues. Increased content of EPA and DHA modifies the structure of cell membranes and the function of membrane proteins. EPA and DHA modulate the production of lipid mediators and through effects on cell signalling can alter the patterns of gene expression. Through these mechanisms, EPA and DHA alter cell and tissue responsiveness in a way that often results in more optimal conditions for growth, development and maintenance of health. DHA has vital roles in brain and eye development and function. EPA and DHA have a wide range of physiological roles, which are linked to certain health or clinical benefits, particularly related to CVD, cancer, inflammation and neurocognitive function. The benefits of EPA and DHA are evident throughout the life course. Future research will include better identification of the determinants of variation of responses to increased intake of EPA and DHA; more in-depth dose–response studies of the effects of EPA and DHA; clearer identification of the specific roles of EPA, docosapentaenoic acid and DHA; testing strategies to enhance delivery of n-3 fatty acids to the bloodstream; and exploration of sustainable alternatives to fish-derived very long-chain n-3 fatty acids.

Type
Conference on ‘The future of animal products in the human diet: health and environmental concerns’
Copyright
Copyright © The Author 2017 

n-3 Fatty acids: structure, metabolic interrelationships, dietary sources and intakes

n-3 Fatty acids are a family of PUFA( Reference Burdge and Calder 1 ). They are defined by the position of the double bond closest to the methyl terminus of the hydrocarbon (acyl) chain. This is on carbon number three when counting the methyl carbon as number one. EPA (20:5n-3) and DHA (22:6n-3) appear to be the most important n-3 fatty acids( Reference Calder and Yaqoob 2 , Reference Calder 3 ), although roles for docosapentaenoic acid (DPA; 22:5n-3) are now also emerging( Reference Kaur, Cameron-Smith and Garg 4 , Reference Weylandt 5 ). Because of their long hydrocarbon chain, EPA, DPA and DHA are sometimes termed very long-chain n-3 fatty acids, in order to differentiate them from the 18-carbon plant-derived n-3 fatty acids like α-linolenic acid (ALA; 18:3n-3) and stearidonic acid (SDA; 18:4n-3). In the present paper, the term ‘very long-chain n-3 fatty acids’ will be used to individually and collectively describe EPA, DPA and DHA.

EPA, DPA and DHA are metabolically related to one another, and there is a pathway by which EPA can be synthesised from the simpler plant-derived n-3 fatty acids (Fig. 1). The conversion of ALA to EPA involves three steps catalysed, in turn, by delta-6 desaturase, elongase 5 and delta-5 desaturase (Fig. 1). Further conversion of EPA to DHA, via DPA, occurs by a complex pathway (Fig. 1) involving chain elongation catalysed by elongase 5, a second chain elongation catalysed by elongase 2 or 5, desaturation by delta 6-desaturase, and then removal of two carbon atoms by limited β-oxidation in peroxisomes. The enzymes of n-3 fatty acid interconversion are shared with the analogous n-6 fatty acid biosynthetic pathway of conversion of linoleic acid (18:2n-6) to arachidonic acid (ARA; 20:4n-6) and beyond. The high intake of linoleic acid relative to ALA in many Western diets( Reference Blasbalg, Hibbeln and Ramsden 6 ) favours linoleic acid conversion over that of ALA. This may be one explanation for the frequently reported low rate of conversion of ALA along this pathway( Reference Arterburn, Hall and Oken 7 , Reference Baker, Miles and Burdge 8 ), although this rate can be influenced by several other factors including age( Reference Bolton-Smith, Woodward and Tavendale 9 ), sex( Reference Childs, Romeu-Nadal and Burdge 10 Reference Burdge and Wootton 12 ), hormones( Reference Vessby, Gustafsson and Tengblad 13 ) and genetics( Reference Koletzko, Lattka and Zeilinger 14 ).

Fig. 1. The metabolic pathway of biosynthesis of EPA, docosapentaenoic acid and DHA.

EPA and DHA are found in fairly high amounts in most seafood, especially in fatty fish (sometimes called oily fish), in the blubber and tissues of sea mammals like whales and seals, in supplements like fish oils, cod liver oil and krill oil, in some algal oils, and in a limited number of pharmaceutical grade preparations( 15 , Reference Calder 16 ). There is at least a 10-fold range in content of these fatty acids per serving of seafood, with fatty fish (e.g. mackerel, salmon, trout, herring, tuna and sardines) being the richest source (Table 1). Hence, intake of very long-chain n-3 fatty acids is strongly influenced by fish consumption. In most Western countries only a relatively small proportion of the population regularly consume fatty fish. For example, in the UK it is estimated that only 25% of the adult population are regular fatty fish consumers( 17 ). Consequently intakes of very long-chain n-3 fatty acids are low in much of the population and mean intakes of EPA + DHA among adults in many Western populations are considered to be about 0·1–0·3 g/d( 17 ). However, it is difficult to be precise about this figure for several reasons, as discussed elsewhere( Reference Calder 3 ). A series of Australian studies in adults and children provide probably the most accurate data on intake of very long-chain n-3 fatty acids. Data from over 10 000 Australian adults identified mean daily intakes of EPA, DPA and DHA as 56, 26 and 106 mg, respectively, to give a total very long-chain n-3 fatty acid intake of 189 mg/d( Reference Meyer, Mann and Lewis 18 ), consistent with the oft-quoted average intake of these fatty acids among Western adults. However, median intakes of EPA, DPA and DHA were found to be only 8, 6 and 15 mg/d, respectively( Reference Meyer, Mann and Lewis 18 ). The large differences between mean and median intakes reflect the skewed distribution of the intake data. A more recent study using an updated nutrient composition database produced mean daily intake data for EPA, DPA and DHA of 75, 71 and 100 mg, respectively, in Australian adults, giving a mean total very long-chain n-3 fatty acid intake of 246 mg/d( Reference Howe, Meyer and Record 19 ). Again, median intakes were lower, being about 50% of the mean. The data suggest that 50% of Australian adults consume less than about 120 mg of very long-chain n-3 fatty acids daily. There is no reason to think that average intakes would be higher than this in other Western countries, and in many they may be lower. Australian children and adolescents aged 2–16 years consumed a mean of 79 mg/d EPA + DPA + DHA, with a median intake of 29 mg/d( Reference Rahmawaty, Charlton and Lyons-Wall 20 ). Intakes increased with age and were much higher in fish eaters than non-fish eaters( Reference Rahmawaty, Charlton and Lyons-Wall 20 ).

Table 1. Typical content of EPA, docosapentaenoic acid (DPA) and DHA (g/100 g food) in a selection of seafood and meat

Data are taken from ref. 15. Note that both very long-chain n-3 fatty acid content and portion size may vary. Modified from ref. Reference Calder3 with permission from John Wiley and Sons.

The oil used in many n-3 fatty acid supplements is sourced from fish; as such this may be referred to as fish oil. Other, non-fish, sources of oil such as krill and algae are also used in supplements. These different oils differ according to their very long-chain n-3 fatty acid content and the relative contributions of EPA and DHA (Table 2). Fig. 2 illustrates the estimated influence of daily consumption of a 1 g standard fish oil capsule, a 1 g ‘concentrated’ supplement, one teaspoon of cod liver oil, one meal of salmon and one or four capsules of a pharmaceutical grade preparation on daily intake of EPA + DHA. This shows that a person who does not consume fatty fish or n-3 fatty acid supplements can markedly increase their EPA + DHA intake through dietary change or through use of supplements.

Fig. 2. Typical intake of EPA + DHA from the background diet in an adult not regularly consuming fatty fish and what would be achieved by also consuming a 1 g standard fish oil (FO) capsule, a 1 g ‘concentrated’ supplement, one teaspoon of cod liver oil, one meal of salmon, or one or four capsules of the pharmaceutical grade preparation Omacor®. Reproduced from ref. Reference Calder3 with permission from John Wiley and Sons.

Table 2. Typical EPA and DHA contents of n-3 supplements

Reproduced from ref. Reference Calder16 with permission from John Wiley and Sons.

* Also known as Lovaza®.

Increased intake of EPA and DHA leads to increased content of EPA and DHA in blood, cells and tissues

As for all long-chain and very long-chain fatty acids, EPA and DHA are transported in the bloodstream esterified into TAG, phospholipids and cholesteryl esters as components of lipoproteins. They also occur in the bloodstream in the non-esterified form non-covalently bound to albumin. EPA and DHA are stored in adipose tissue esterified into TAG and they are found in all cell membranes esterified into phospholipids and related complex lipids. Cell membrane phospholipids and their fatty acid composition are important in determining the physical characteristics of cell membranes( Reference Stubbs and Smith 21 ), the manner in which membranes change in response to external stimuli( Reference Brenner 22 ) and the functional activities of membrane-bound proteins( Reference Murphy 23 ). EPA and DHA circulating in the bloodstream, stored in adipose tissue, and present in cell membranes may be regarded as transport, storage and functional pools, respectively. The contribution of EPA or DHA to the total fatty acids present within any of the transport, storage or functional pools differs according to the pool (Table 3)( Reference Browning, Walker and Mander 24 Reference Safarinejad 36 ). DHA is most often present in a greater concentration than EPA (Table 3). This is especially true in specific regions of the eye and brain where DHA makes a significant contribution to the fatty acid complement and EPA is virtually absent( Reference Skinner, Watt and Besson 27 , Reference Makrides, Neumann and Byard 28 , Reference Crawford, Casperd and Sinclair 37 ). Within cell membranes, EPA and DHA are distributed differently among the different phospholipid components and in the brain and eye specific phospholipids are especially rich in DHA( Reference Crawford, Casperd and Sinclair 37 , Reference Anderson 38 ).

Table 3. Typical EPA and DHA concentrations reported in different lipid pools in human subjects

Data are taken from the selected references and are expressed as % of total fatty acids. Reproduced from ref. Reference Calder16 with permission from John Wiley and Sons.

* Blood collected after an overnight fast.

Increased intakes of EPA and DHA from fish or from supplements are reflected in increased concentrations (and proportions) of both fatty acids in blood lipids, blood cells and many tissues. This has been reported many times for total plasma and serum lipids and for the complex lipid components of plasma and serum (i.e. TAG, phospholipids and cholesteryl esters)( Reference Browning, Walker and Mander 24 , Reference von Schacky, Fischer and Weber 39 Reference Popp-Snijders, Schouten and van Blitterswijk 47 ). It is also well described for erythrocytes( Reference Browning, Walker and Mander 24 , Reference Blonk, Bilo and Popp-Snijders 42 , Reference Katan, Deslypere and van Birgelen 43 , Reference Popp-Snijders, Schouten and van Blitterswijk 47 ), platelets ( Reference Browning, Walker and Mander 24 , Reference von Schacky, Fischer and Weber 39 , Reference Sanders and Roshanai 48 ) and leucocytes( Reference Browning, Walker and Mander 24 , Reference Healy, Wallace and Miles 26 , Reference Yaqoob, Pala and Cortina-Borja 44 Reference Faber, Berkhout and Vos 46 , Reference Sperling, Benincaso and Knoell 49 ). When their intake is increased, the content of EPA and DHA increases in several human tissues, including skeletal muscle( Reference McGlory, Galloway and Hamilton 31 ), heart( Reference Harris, Sands and Windsor 29 ), gut mucosa( Reference Hillier, Jewell and Dorrell 35 , Reference Sorensen, Rasmussen and Aardestrup 50 ) and adipose tissue( Reference Browning, Walker and Mander 24 , Reference Katan, Deslypere and van Birgelen 43 ). There is a dose- and time-dependent pattern of incorporation of both EPA and DHA( Reference Browning, Walker and Mander 24 , Reference Healy, Wallace and Miles 26 , Reference von Schacky, Fischer and Weber 39 , Reference Katan, Deslypere and van Birgelen 43 Reference Faber, Berkhout and Vos 46 ). Although EPA is incorporated more quickly than DHA( Reference Browning, Walker and Mander 24 , Reference Katan, Deslypere and van Birgelen 43 ), the precise pattern of EPA and DHA incorporation that occurs depends upon the specific nature of the lipid pool( Reference Browning, Walker and Mander 24 , Reference Katan, Deslypere and van Birgelen 43 ). Pools that are turning over rapidly show faster incorporation of EPA and DHA than slower turning over pools. For example, plasma lipids incorporate EPA and DHA more quickly than blood cells( Reference Browning, Walker and Mander 24 , Reference Katan, Deslypere and van Birgelen 43 ), and amongst blood cells, leucocytes have been usually shown to incorporate EPA and DHA more quickly than erythrocytes. However, dose- and time-dependence of EPA and DHA incorporation into tissues is not well investigated in human subjects, apart from for adipose tissue( Reference Browning, Walker and Mander 24 , Reference Katan, Deslypere and van Birgelen 43 ). Modification of human brain fatty acid composition is more difficult than for other tissues, especially beyond childhood.

The higher concentration of EPA and DHA achieved in various lipid pools through increased intake of EPA and DHA is maintained so long as the higher intake of EPA and DHA is maintained. If, after a period of increased intake of EPA and DHA, intake returns to the earlier lower level, then EPA and DHA concentrations decline, eventually returning to earlier levels. This is well described for blood lipids( Reference von Schacky, Fischer and Weber 39 , Reference Katan, Deslypere and van Birgelen 43 , Reference Yaqoob, Pala and Cortina-Borja 44 ), platelets( Reference von Schacky, Fischer and Weber 39 ), leucocytes( Reference Yaqoob, Pala and Cortina-Borja 44 ) and erythrocytes( Reference Katan, Deslypere and van Birgelen 43 , Reference Popp-Snijders, Schouten and van Blitterswijk 47 ). Just as the incorporation of EPA into different pools is faster than the incorporation of DHA, the loss of EPA is faster than the loss of DHA( Reference von Schacky, Fischer and Weber 39 , Reference Katan, Deslypere and van Birgelen 43 , Reference Yaqoob, Pala and Cortina-Borja 44 , Reference Popp-Snijders, Schouten and van Blitterswijk 47 ). The better retention of DHA than EPA may be because DHA is structurally and/or functionally preferred over EPA and that metabolic mechanisms have evolved to preserve it.

Molecular and cellular effects of increased EPA and DHA content

Many, though not all, of the functional effects of EPA and DHA are considered to require their incorporation into cell membrane phospholipids( Reference Calder 51 Reference Calder 53 ) (Fig. 3). EPA and DHA are highly unsaturated and as a consequence they have been shown in some studies to decrease membrane order (i.e. increase membrane fluidity)( Reference Stubbs and Smith 21 , Reference Calder, Yaqoob and Harvey 54 ), although cells have mechanisms such as modifying membrane cholesterol content, to limit this effect. Through modulation of the physical properties of membranes, EPA and DHA provide a specific environment for membrane proteins like receptors, transporters, ion channels and signalling enzymes to function( Reference Brenner 22 , Reference Murphy 23 , Reference Miles and Calder 55 ). As a result, EPA and DHA can modulate cell responses that are dependent upon membrane protein function. This has been shown to be especially important in the eye where the presence of DHA enables optimal activity of the photoreceptor protein rhodopsin( Reference Niu, Mitchell and Lim 56 ). Cell membranes contain microdomains called rafts( Reference Pike 57 ). These have specific lipid and fatty acid compositions and act as platforms for receptor action and for the initiation of intracellular signalling pathways( Reference Pike 57 Reference Yaqoob 59 ). EPA and DHA can modify raft formation in a variety of cell types including neurons, immune cells and cancer cells( Reference Yaqoob 59 Reference Hou, McMurray and Chapkin 61 ), so affecting intracellular signalling pathways( Reference Yaqoob 59 Reference Calder 62 ). As a result of their effects on membrane-generated intracellular signals, EPA and DHA can modulate transcription factor activation and, subsequently, gene expression patterns in a variety of cell types( Reference Calder 51 , Reference Miles and Calder 55 , Reference Calder 63 ). NF-κB ( Reference Novak, Babcock and Jho 64 ), the peroxisome proliferator activated receptors( Reference Krey, Braissant and L'Horset 65 , Reference Marion-Letellier, Savoye and Ghosh 66 ), and the sterol regulatory element binding proteins( Reference Clarke 67 Reference Deckelbaum, Worgall and Seo 71 ) are amongst the transcription factors shown to be affected by EPA and DHA. The effects of EPA and DHA on transcription factor activation and modulation of gene expression are central to their role in controlling fatty acid and TAG metabolism, inflammation and adipocyte differentiation( Reference Calder 51 Reference Calder 53 , Reference Miles and Calder 55 , Reference Calder 63 ).

Fig. 3. General overview of the mechanisms by which very long-chain n-3 fatty acids can influence the function of cells. Modified from Calder( Reference Calder 72 ), Copyright (2011), with permission from Elsevier.

Increased abundance of EPA and DHA in cell membrane phospholipids is associated with decreased abundance of the n-6 PUFA ARA( Reference Healy, Wallace and Miles 26 , Reference von Schacky, Fischer and Weber 39 , Reference Yaqoob, Pala and Cortina-Borja 44 , Reference Rees, Miles and Banerjee 45 , Reference Calder 72 ). This alters the availability of substrates for synthesis of bioactive lipid mediators (Fig. 4). ARA is quantitatively the major substrate for the biosynthesis of various prostaglandins, thromboxanes and leukotrienes, together termed eicosanoids, which have well-established roles in regulation of inflammation, immunity, platelet aggregation, smooth muscle contraction and renal function. Eicosanoids are oxidised derivatives of 20-carbon PUFA and are produced via the cyclooxygenase (prostaglandins, thromboxanes), lipoxygenase (leukotrienes and other products) and cytochrome P450 pathways. Although they have a number of obvious important physiological roles, excess or inappropriate production of eicosanoids from ARA is associated with many disease processes( Reference Lewis, Austen and Soberman 73 , Reference Tilley, Coffman and Koller 74 ). Increasing the EPA and DHA content of cell membranes results in decreased production of eicosanoids from ARA( Reference von Schacky, Fischer and Weber 39 , Reference Rees, Miles and Banerjee 45 ), resulting in an impact of EPA and DHA on inflammation, immune function, blood clotting, vasoconstriction, and bone turnover amongst other processes. In addition to decreasing production of eicosanoids from ARA, EPA and DHA are themselves substrates for the synthesis of lipid mediators (Fig. 4). Some of these are simply analogues of those produced from ARA. For example, PG E3 produced from EPA is an analogue of PG E2 produced from ARA. Frequently, though not always, the EPA-derived mediator has weaker biological activity than the mediator derived from ARA( Reference Wada, DeLong and Hong 75 ). For example, thromboxane A3 produced from EPA is a much weaker platelet aggregator than thromboxane A2 produced from ARA( Reference Moncada and Vane 76 ). EPA and DHA are also substrates for more complex biosynthetic pathways that result in generation of mediators known as resolvins (E-series formed from EPA and D-series formed from DHA), protectins/neuroprotectins (formed from DHA) and maresins (formed from DHA)( Reference Bannenberg and Serhan 77 Reference Serhan and Chiang 79 ) (Fig. 4). It has recently been discovered that DPA gives rise to a similar family of mediators( Reference Weylandt 5 , Reference Dalli, Colas and Serhan 80 ). The major role of resolvins, protectins and maresins appears to be in the resolution of inflammation and modulation of immune function( Reference Bannenberg and Serhan 77 Reference Serhan and Chiang 79 ). It seems likely that many of the anti-inflammatory and immune modulating actions of EPA and DHA that are described in the literature( Reference Calder 52 , Reference Calder 53 , Reference Calder 72 , Reference Calder 81 ) are mediated through resolvins, protectins and maresins.

Fig. 4. Overview of the bioactive lipid mediators produced from arachidonic acid, EPA and DHA.

The above-mentioned mechanisms of action of EPA and DHA rely upon incorporation of those fatty acids into cell membrane phospholipids (Fig. 3). It is now recognised that EPA and DHA (in their non-esterified form) can also act directly via membrane G-protein coupled receptors that exhibit some specificity for very long-chain n-3 fatty acids over other fatty acids as ligands( Reference Oh, Talukdar and Bae 82 ). In particular, free fatty acid receptor 4 (also known as GPR120), which is highly expressed on inflammatory macrophages and on adipocytes, was shown in cell culture experiments to play a central role in mediating the anti-inflammatory effects of DHA on macrophages and the insulin-sensitising effects of DHA on adipocytes( Reference Oh, Talukdar and Bae 82 ).

Roles of EPA and DHA in supporting optimal cell and tissue function and promoting health

CVD

Native populations in Greenland, Northern Canada and Alaska consuming their traditional diet were found to have much lower rates of death from CVD than predicted, despite their high dietary fat intake( Reference Dyerberg, Bang and Stoffersen 83 Reference Newman, Middaugh and Propst 86 ). The protective component of the diet was suggested to be the very long-chain n-3 fatty acids consumed in very high amounts as a result of the regular intake of seal and whale meat, whale blubber and fatty fish( Reference Bang, Dyerberg and Hjorne 87 ). Low cardiovascular mortality is also seen in Japanese consuming a traditional diet( Reference Yano, MacLean and Reed 88 ) and this diet is rich in seafood including fatty fish and sometimes marine mammals, which contain significant amounts of EPA and DHA. Much evidence has now accumulated from prospective and case–control studies indicating that higher intake of EPA and DHA is associated with reduced risk of CVD outcomes in Western populations, although not all studies agree. These studies have been summarised and discussed in detail elsewhere( Reference Kris-Etherton, Harris and Appel 89 Reference Calder 93 ) and they have been subject to systematic review and meta-analysis. For example, Chowdhury et al. ( Reference Chowdhury, Warnakula and Kunutsor 94 ) brought together prospective studies examining the association of dietary or circulating fatty acids, including very long-chain n-3 fatty acids, with risk of coronary outcomes. Data from sixteen studies involving over 422 000 individuals showed a relative risk of 0·87 for those in the top third of dietary intake of very long-chain n-3 fatty acids compared with those in the lower third of intake. Data from thirteen studies involving over 20 000 individuals showed relative risks of 0·78, 0·79 and 0·75 for those in the top third of circulating EPA, DHA and EPA + DHA, respectively, compared with those in the lower third( Reference Chowdhury, Warnakula and Kunutsor 94 ). A smaller number of studies in fewer individuals gave a relative risk of 0·64 for circulating DPA( Reference Chowdhury, Warnakula and Kunutsor 94 ). A more recent analysis pooled data from nineteen studies that investigated the association between EPA or DHA concentration in a body compartment like plasma, serum, erythrocytes or adipose tissue and risk of future CHD in adults who were healthy at study entry( Reference Del Gobbo, Imamura and Aslibekyan 95 ). EPA and DHA were each associated with a lower risk of fatal CHD, with relative risks of 0·91 and 0·90, respectively( Reference Del Gobbo, Imamura and Aslibekyan 95 ). These analyses( Reference Chowdhury, Warnakula and Kunutsor 94 , Reference Del Gobbo, Imamura and Aslibekyan 95 ) support a clear role for EPA and DHA in primary prevention of CHD, and perhaps, more widely, of CVD, as discussed elsewhere( Reference Calder 96 ).

Beneficial modification of a broad range of risk factors probably explains the protective effect of very long-chain n-3 fatty acids towards CHD. These risk factors include plasma/serum TAG concentrations, blood pressure, thrombosis, cardiac function, vascular function and inflammation, which are all improved by very long-chain n-3 fatty acids( Reference Kris-Etherton, Harris and Appel 89 , Reference Calder 93 , Reference Harris 97 Reference Leslie, Cohen and Liddle 101 ). A recent meta-analysis of randomised controlled trials (RCT) evaluated the effects of EPA + DHA on a range of risk factors for CVD( Reference AbuMweis, Jew and Tayyem 102 ); the findings are summarised in Table 4. Significant effects of EPA + DHA were identified for plasma/serum TAG and HDL cholesterol concentrations, systolic and diastolic blood pressure, heart rate and C-reactive protein concentration, a marker of inflammation (Table 4). There was also a significant elevation of LDL concentration (Table 4), but this apparently deleterious effect may be offset by an increased size of LDL particles( Reference Mori, Burke and Puddey 103 ), rendering them less atherogenic. The improvement in the risk factor profile with very long-chain n-3 fatty acids would account for the lowered risk of coronary disease reported in many previous epidemiological studies and identified in the recent meta-analysis( Reference Chowdhury, Warnakula and Kunutsor 94 ) and pooled analysis( Reference Del Gobbo, Imamura and Aslibekyan 95 ) of cohort studies.

Table 4. Summary of the effects of very long-chain n-3 fatty acids on risk factors for CVD identified through the meta-analysis of AbuMweis et al. ( Reference AbuMweis, Jew and Tayyem 102 )

A role in prevention of CVD is an obvious important health benefit of EPA + DHA. There has also been great interest in the ability of very long-chain n-3 fatty acids to treat people with existing CVD. The outcome in studies in this area has most often been the occurrence of a major cardiovascular event (e.g. myocardial infarction (MI)), fatal MI, or death. Several large studies published between 1989 and 2008 reported lower rates of death in patients receiving very long-chain n-3 fatty acids( Reference Burr, Fehily and Gilbert 104 Reference Tavazzi, Maggioni and Marchioli 108 ). Doses of very long-chain n-3 fatty acids used in these studies were 500 mg to 1·6 g/d and durations of intervention were 1–5 years. As a result of these positive findings, meta-analyses published in the period 2002–2009 supported that very long-chain n-3 fatty acids lower mortality in patients with existing CVD( Reference Bucher, Hengstler and Schindler 109 Reference Leon, Shibata and Sivakumaran 111 ). For example, a meta-analysis including eleven studies involving almost 16 000 patients reported that, compared with control, very long-chain n-3 fatty acids lower the risk of fatal MI, sudden death and all-cause mortality (relative risks 0·7, 0·7 and 0·8, respectively)( Reference Bucher, Hengstler and Schindler 109 ). A second meta-analysis including fourteen studies involving over 20 000 patients reported that, compared with control, very long-chain n-3 fatty acids lower the risk of cardiac mortality and all-cause mortality (relative risks 0·68 and 0·77, respectively)( Reference Studer, Briel and Leimenstoll 110 ). It is likely that the mechanisms that reduce the likelihood of cardiovascular events and mortality in patients with established disease are different from the mechanisms that act to slow the development of atherosclerosis. Three key mechanisms have been suggested to contribute to this therapeutic effect of very long-chain n-3 fatty acids. The first is altered cardiac electrophysiology seen as lower heart rate( Reference Harris, Miller and Tighe 112 ), increased heart rate variability( Reference Xin, Wei and Li 113 ) and fewer arrhythmias( Reference Leaf and Xiao 114 ). These effects make the heart more able to respond robustly to stress. The second is an anti-thrombotic action resulting from the altered pattern of production of eicosanoid mediators from ARA and from EPA that control platelet aggregation( Reference von Schacky, Fischer and Weber 39 ). This effect would lower the likelihood of clot formation or would result in weaker clots less able to stop blood flow to affected organs. The third mechanism is the well-documented anti-inflammatory effect of very long-chain n-3 fatty acids, which would serve to stabilise atherosclerotic plaques preventing their rupture( Reference Thies, Garry and Yaqoob 115 , Reference Cawood, Ding and Napper 116 ). This effect would reduce the likelihood of a cardiovascular event (MI, stroke) from happening( Reference Calder and Yaqoob 117 ). Thus, there is a biological plausibility to EPA + DHA having a benefit in secondary prevention of cardiovascular events and mortality.

Despite the positive findings with very long-chain n-3 fatty acids, supported by meta-analyses and biologically plausible candidate mechanisms, more recent studies in patients with existing CVD have failed to replicate the earlier findings( Reference Galan, Kesse-Guyot and Czernichow 118 Reference Bosch, Gerstein and Dagenais 122 ). This has influenced some of the most recent meta-analyses, which have concluded that there is little protective effect of very long-chain n-3 fatty acids on cardiovascular mortality( Reference Kwak, Myung and Lee 123 Reference Rizos, Ntzani and Bika 125 ). Nevertheless, the meta-analysis by Rizos et al. ( Reference Rizos, Ntzani and Bika 125 ) which included the recent studies, but excluded the landmark GISSI Prevenzione trial( 105 ), did identify a reduction in cardiac death with very long-chain n-3 fatty acids (relative risk 0·91) and trends towards reductions in sudden death and MI (relative risks 0·91 and 0·89, respectively). It is important to recognise that the most recent studies of very long-chain n-3 fatty acids and cardiovascular mortality have been criticised for various reasons related to small sample size, the low dose of EPA + DHA used, the too short duration of follow up, and, in some studies, the low rate of events( Reference Calder and Yaqoob 126 ). Despite this, meta-analyses that include the GISSI-Prevenzione study and some of the more recent neutral studies, still report benefits from very long-chain n-3 fatty acids( Reference Casula, Soranna and Catapano 127 , Reference Wen, Dai and Gao 128 ). For example, Casula et al.( Reference Casula, Soranna and Catapano 127 ) identified reductions in cardiac death, sudden death and MI (relative risks 0·68, 0·67 and 0·75, respectively) with very long-chain n-3 fatty acids and a trend towards lower all-cause mortality (relative risk 0·89). Likewise, Wen et al.( Reference Wen, Dai and Gao 128 ) identified reductions in cardiac death, sudden death, MI and all-cause mortality (relative risks 0·88, 0·86, 0·86 and 0·92, respectively) with very long-chain n-3 fatty acids.

Thus, there is a significant literature gathered over more than 45 years from association studies, from RCT investigating the impact on risk factors, and from RCT investigating the effect on hard clinical outcomes like mortality that very long-chain n-3 fatty acids lower the risk of developing CVD, especially CHD and can be used to treat people with CVD. Although the most recent RCT in patients with CVD have produced findings that do not agree with the previously accumulated literature, it is too early to discard the earlier evidence. Instead reasons to explain the different findings need to be identified, in order to better understand the actions of EPA and DHA. The conclusion that very long-chain n-3 fatty acids have a role in reducing risk of CVD, especially CHD, remains well supported, for example by the American Heart Association( Reference Kris-Etherton, Harris and Appel 89 , Reference Siscovick, Barringer and Fretts 129 ).

Cancer

EPA and DHA have a number of biological activities that may influence tumour cell viability and proliferation. For example, DHA can promote tumour cell apoptosis( Reference Gleissman, Johnsen and Kogner 130 , Reference Merendino, Costantini and Manzi 131 ), possibly through inducing oxidative stress. The replacement of ARA in cell membranes by EPA and DHA is also an important anti-cancer mechanism of action of n-3 PUFA, since ARA-derived eicosanoids like PG E2 and leukotriene B4 promote tumour cell proliferation, survival, migration and invasion( Reference Merendino, Costantini and Manzi 131 Reference Vaughan, Hassing and Lewandowski 133 ). Through these effects, EPA and DHA can directly influence cancer cells and the tumour environment, and they can also influence the host response to tumour bearing. Recent reviews provide a detailed description of the mechanisms by which very long-chain n-3 fatty acids affect tumour cell proliferation, survival, invasion and metastasis( Reference Gleissman, Johnsen and Kogner 130 , Reference Merendino, Costantini and Manzi 131 ); the ability of very long-chain n-3 fatty acids to enhance the effectiveness of anti-cancer treatments( Reference Merendino, Costantini and Manzi 131 , Reference Vaughan, Hassing and Lewandowski 133 ); and the current evidence of the efficacy of very long-chain n-3 fatty acids in human subjects in the context of cancer and its treatment( Reference Merendino, Costantini and Manzi 131 , Reference Vaughan, Hassing and Lewandowski 133 ).

Some prospective and case–control studies show that very long-chain n-3 fatty acids are associated with lower risk of colorectal and breast cancers, but there is inconsistency in the findings from such studies( Reference Murphy, Mourtzakis and Mazurak 134 ). Recent systematic reviews conclude that very long-chain n-3 fatty acids are protective against colorectal( Reference Yang, Wang and Ren 135 ) and breast( Reference Makarem, Chandran and Bandera 136 ) cancers, while a fairly recent prospective cohort study among post-menopausal women found that higher dietary intake of either EPA or DHA was associated with lower risk of developing breast cancer( Reference Sczaniecka, Brasky and Lampe 137 ). Whether very long-chain n-3 fatty acids increase or decrease risk of prostate cancer is currently under debate( Reference Brasky, Darke and Song 138 , Reference Crowe, Appleby and Travis 139 ).

In addition to effects that lower the risk of developing some forms of cancer, there seems to be a role for very long-chain n-3 fatty acids in patients who already have cancer. For example, quality of life and physical functioning can be improved in cancer patients with oral provision of very long-chain n-3 fatty acids. A systematic review( Reference Elia, Van Bokhorst-de van der Schueren and Garvey 140 ) concluded that lung cancer patients receiving supplements containing EPA and DHA had improved appetite, energy intake, body weight and quality of life. Breast cancer patients with a higher concentration of very long-chain n-3 fatty acids in their bloodstream had lower inflammation and less physical fatigue than seen in patients with a lower concentration of very long-chain n-3 fatty acids( Reference Alfano, Imayama and Neuhouser 141 ). Lung cancer patients given 1·8 g EPA + DHA daily had improved appetite and less fatigue than controls( Reference Cerchietti, Navigante and Castro 142 ). Van der Meij et al.( Reference van der Meij, Langius and Spreeuwenberg 143 ) reported improved quality of life, physical function, cognitive function and health status in patients with non-small cell lung cancer receiving 2·9 g EPA + DHA per day. The patients receiving very long-chain n-3 fatty acids also tended to have greater physical activity compared with the control group( Reference van der Meij, Langius and Spreeuwenberg 143 ).

EPA and DHA sensitise cultured tumour cells to chemotherapeutic agents, increasing the efficacy of those agents( Reference Sala-Vila, Folkes and Calder 144 ). The mechanism by which this occurs is not clear, but it might involve increased EPA and DHA content of tumour cell membranes resulting in increased lipid peroxidation in those membranes in the presence of the cancer therapeutic. If this occurred in vivo it would result in improved efficacy of the therapy and perhaps reduced side effects. Murphy et al. ( Reference Murphy, Mourtzakis and Chu 145 ) conducted a trial in patients with non-small cell lung cancer and showed that 2·5 g EPA + DHA daily caused a 2-fold increase in response rate to the chemotherapy being used and prolonged the period over which patients could receive the chemotherapy. They also reported a trend towards improved survival with very long-chain n-3 fatty acids. Bougnoux et al.( Reference Bougnoux, Hajjaji and Ferrasson 146 ) reported improved chemotherapy outcomes in breast cancer patients receiving 1·8 g DHA daily.

Cancer cachexia (loss of lean and fat tissue) is a complication that occurs in patients with advanced solid tumors and greatly increases risk of mortality. Weed et al. ( Reference Weed, Ferguson and Gaff 147 ) reported that patients with squamous cell cancer of the head and neck taking 3·08 g EPA + DHA daily had increased lean body mass. Murphy et al.( Reference Murphy, Mourtzakis and Chu 148 ) showed that 2·2 g EPA + DHA daily was able to maintain body weight and muscle mass during chemotherapy in patients with non-small cell lung cancer. In other studies in patients with cancer, very long-chain n-3 fatty acids increased body weight( Reference Fearon, Barber and Moses 149 , Reference Guarcello, Riso and Buosi 150 ).

Thus, there is increasing evidence from studies in human subjects that very long-chain n-3 fatty acids have a role in reducing risk of developing some cancers, particularly colorectal and breast. Furthermore, a number of intervention trials demonstrate that very long-chain n-3 fatty acids have a range of benefits in patients with various types of cancer. Most intervention studies have used approximately 2 g/d EPA + DHA. From their review of the literature Vaughan et al.( Reference Vaughan, Hassing and Lewandowski 133 ) concluded ‘There is now sufficient literature to suggest that the use of supplements containing EPA and DHA may have potential use as an effective adjuvant to chemotherapy treatment and may help ameliorate some of the secondary complications associated with cancer…… our investigations indicate that supplementation with fish oil or EPA/DHA (>1 g EPA and >0·8 g DHA daily) is associated with positive clinical outcomes.’

Inflammation

Inflammation is an essential component of normal host defence mechanisms, initiating the immune response and later playing a role in tissue repair. The inflammatory response is normally self-limiting (i.e. resolving) in order to protect the host from damage. However, the loss of the normal mechanisms inducing tolerance (e.g. to self, to endogenous commensal micro-organisms, or to environmental components such as foods) or loss of resolving factors can allow inflammation to become chronic and in this state the damage done to host tissues may become pathological( Reference Calder, Albers and Antoine 151 , Reference Calder, Ahluwalia and Albers 152 ). As such, inflammation is the central adverse response seen in a range of conditions including rheumatoid arthritis (RA), inflammatory bowel disease, multiple sclerosis, asthma, psoriasis, and atopic dermatitis. Furthermore, chronic low-grade inflammation is now recognised to be a contributor to CVD( Reference Ross 153 , Reference Hansson and Hermansson 154 ) and to play a role in cardiometabolic diseases like obesity, type-2 diabetes and non-alcoholic fatty liver disease and in cognitive decline( Reference Calder, Ahluwalia and Brouns 155 ). Cancer also has a permissive inflammatory component( Reference Colotta, Allavena and Sica 156 ).

Eicosanoids and related lipid mediators provide a direct link between fatty acids and inflammatory processes. As described earlier, the n-6 PUFA ARA is the precursor for the production of prostaglandins and leukotrienes that are directly involved in inflammatory processes( Reference Lewis, Austen and Soberman 73 , Reference Tilley, Coffman and Koller 74 ). In contrast to the effects of ARA, EPA and DHA give rise to mediators which are less pro-inflammatory, anti-inflammatory or inflammation resolving( Reference Calder 52 , Reference Calder 53 , Reference Calder 72 , Reference Bannenberg and Serhan 77 Reference Serhan and Chiang 79 , Reference Calder 81 ). In addition to their effects on lipid mediators (prostaglandins, leukotrienes, resolvins, protectins, maresins), EPA and DHA modulate many other aspects of inflammatory processes including leucocyte migration and production of inflammatory cytokines( Reference Calder 52 , Reference Calder 53 , Reference Calder 72 , Reference Calder 81 ). These effects on inflammation relate to the modulation of cell signalling, transcription factor activation and gene expression by EPA and DHA. There is good evidence that EPA and DHA given in combination at sufficiently high doses are anti-inflammatory and have a therapeutic role in inflammatory diseases. This has been most widely studied in RA( Reference Miles and Calder 157 ), inflammatory bowel disease( Reference Calder 158 ) and asthma( Reference Calder 159 ). Evidence of efficacy is strongest in RA, although high doses (several g/d EPA + DHA) are typically used( Reference Miles and Calder 157 ). Recent systematic reviews and meta-analyses have highlighted the benefits of EPA + DHA on arthritic pain( Reference Abdulrazaq, Innes and Calder 160 , Reference Senftleber, Nielsen and Andersen 161 ).

Allergic disease begins in infancy and the immune imbalances that predispose to allergy may be influenced by the relative exposure to n-6 and n-3 PUFA( Reference Calder, Kremmyda and Vlachava 162 ). Therefore, there has been significant interest in whether increased intake of very long-chain n-3 fatty acids by pregnant and breast-feeding women will reduce the risk of allergic disease in their babies. There is some evidence that increased intake of EPA and DHA during human pregnancy has an effect on the immune system of the baby( Reference Dunstan, Mori and Barden 163 Reference Noakes, Vlachava and Kremmyda 165 ) and that this is linked to reduced allergic symptoms later in childhood( Reference Dunstan, Mori and Barden 163 , Reference Furuhjelm, Warstedt and Larsson 166 , Reference Palmer, Sullivan and Gold 167 ). Best et al. ( Reference Best, Gold and Kennedy 168 ) reported a meta-analysis of offspring clinical outcomes from trials of increased maternal intake of very long-chain n-3 fatty acids in pregnancy. They identified lower risk of atopic eczema, and less likelihood of having a positive skin prick test to any allergen tested, to hens’ egg, or to any food extract, all in the first 12 months of life. A recent study reported that fish oil consumption by pregnant women decreased risk of persistent wheeze and asthma in the offspring at ages 3–5 years( Reference Bisgaard, Stokholm and Chawes 169 ). Supplementing the diets of very young infants has also now been shown to have immune effects consistent with reduced likelihood of allergy ( Reference D'Vaz, Meldrum and Dunstan 170 ). This area of research has been reviewed recently( Reference Willemsen 171 Reference Miles and Calder 173 ).

Thus, the anti-inflammatory actions of EPA and DHA are extensively demonstrated and the underlying mechanisms are increasingly understood. High doses of very long-chain n-3 fatty acids can be used to treat frank inflammatory conditions like RA, while lower doses likely have a role in protecting against development of childhood allergic disease and low-grade inflammatory conditions in adulthood.

Neurocognitive health

More than 50% of the dry weight of the brain is lipid, particularly structural and functional lipid (i.e. phospholipids). The human brain and retina contain an especially high proportion of DHA relative to other tissues but little EPA (Table 3). Grey matter phosphatidylethanolamine contains 24% of its fatty acids as DHA, whereas grey matter phosphatidylserine contains 37% of its fatty acids as DHA( Reference Lauritzen, Hansen and Jorgensen 174 ). DHA contributes 50–70% of the fatty acids present in the rod outer segments of the retina( Reference Anderson 38 ). These rod outer segments contain the eye's photoreceptors and DHA has been clearly shown to be essential for optimal visual functioning( Reference Niu, Mitchell and Lim 56 ). DHA is important for neurotransmission, neuronal membrane stability, neuroplasticity and signal transduction( Reference Lauritzen, Hansen and Jorgensen 174 Reference Parletta, Milte and Meyer 176 ). An adequate supply of very long-chain n-3 fatty acids, especially DHA, seems essential for optimal visual, neural and behavioural development of the infant/child. The need for DHA so early in life was demonstrated in studies conducted in the 1980s and 1990s with pre-term infants, where formulas that included DHA (and often also ARA) were shown to improve visual development( Reference Uauy, Birch and Birch 177 Reference Carlson, Werkman and Tolley 182 ). However, a recent meta-analysis of seventeen trials of inclusion of DHA (and ARA) in infant feeds involving 2260 preterm infants found little evidence to support improved visual acuity or neurodevelopment, although three out of seven studies of neurodevelopment did report some benefit of the PUFA( Reference Moon, Rao and Schulzke 183 ). Studies of DHA in preterm infants are discussed in detail elsewhere( Reference Molloy, Doyle and Makrides 184 ). The literature on the effect of DHA on visual and cognitive outcomes in term infants is mixed with some studies reporting benefits( Reference Agostoni, Trojan and Bellu 185 , Reference Willatts, Forsyth and DiModugno 186 ) and others not( Reference Scott, Janowsky and Carroll 187 Reference Auested, Scott and Janowsky 190 ). One reason for this might be that an early beneficial effect of DHA is lost with time so that early assessments show benefit and later assessments do not; at least one study has shown this( Reference Agostoni, Trojan and Bellu 185 , Reference Agostoni, Trojan and Bellu 191 ). A recent meta-analysis of fifteen trials of inclusion of DHA (and ARA) in infant feeds involving 1889 term infants found little evidence to support improved visual or neurodevelopment, although four out of nine studies of visual acuity and two out of eleven studies of cognitive development reported benefit( Reference Jasani, Simmer and Patole 192 ). Despite the inconsistencies in the literature, it still seems important that pregnant and breastfeeding women and infants consuming formula instead of breast milk have adequate intakes of very long-chain n-3 fatty acids, especially DHA. A systematic review and meta-analysis of eleven RCT of very long-chain n-3 fatty acids in pregnancy involving over 5000 women could not conclusively support or refute that very long-chain n-3 fatty acids in pregnancy improve infant visual or cognitive development( Reference Gould, Smithers and Makrides 193 ).

Very long-chain n-3 fatty acids are likely to have important roles in the brain beyond infancy and are probably important for brain function throughout the life course. A number of studies have reported lower levels of EPA and DHA in the bloodstream of children with attention deficit hyperactivity disorder (ADHD) or autistic spectrum disorders than in control children( Reference Richardson 194 ). It is possible that these and other developmental disorders might be related to some sort of fatty acid deficiency state. If this is the case, then normalisation of fatty acid levels should lead to clinical benefit in these conditions. This has been examined in a number of trials in children and adolescents with attention, learning, or behavioural disorders, some showing some improvements( Reference Stevens, Zhang and Peck 195 Reference Sheppard, Boone and Gracious 205 ) and others finding no effect( Reference Voigt, Llorente and Jensen 206 Reference Mankad, Dupuis and Smile 214 ). These trials have been reviewed many times, with differing conclusions. The different findings may relate to the dose of n-3 fatty acids used, the duration of supplementation, the precise outcome(s) measured and differences in the children studied. Indeed, one review concluded that studies using higher doses of very long-chain n-3 fatty acids or of longer duration or in children/adolescents with low socioeconomic status were more likely to find effects( Reference Frensham, Bryan and Parletta 215 ). A recent meta-analysis of five RCT involving 189 children with autism spectrum disorder identified benefit of very long-chain n-3 fatty acids on some outcomes, but concluded that the limited number of studies and small sample sizes restrict the ability to make firm conclusions( Reference Horvath, Łukasik and Szajewska 216 ). Another recent meta-analysis used data from fifteen case–control studies involving 1193 individuals with autism to show lower blood EPA, DHA and ARA and higher ratio of n-6 to n-3 PUFA in autism( Reference Mazahery, Stonehouse and Delshad 217 ). Data from four RCT involving 107 individuals with autism showed improvements in some outcomes with very long-chain n-3 fatty acids, but the authors suggested the need for larger and longer studies in order to be clear about the effect( Reference Mazahery, Stonehouse and Delshad 217 ). A meta-analysis of ten RCT of very long-chain n-3 fatty acids in children with ADHD showed no improvements in measures of emotional lability, oppositional behaviour, conduct problems or aggression( Reference Cooper, Tye and Kuntsi 218 ). However, subgroup analyses of higher quality studies and those meeting strict inclusion criteria found a significant reduction in emotional lability and oppositional behaviour( Reference Cooper, Tye and Kuntsi 218 ). The authors concluded that a number of treatment effects may have failed to reach statistical significance due to small sample sizes and within and between study heterogeneity in terms of design and study participants( Reference Cooper, Tye and Kuntsi 218 ). In seven RCT involving 534 adolescents with ADHD, very long-chain n-3 fatty acids improved ADHD clinical symptom scores, while in three RCT involving 214 adolescents, very long-chain n-3 fatty acids improved cognitive measures associated with attention( Reference Chang, Su and Mondelli 219 ). A very recent systematic review was supportive of use of n-3 fatty acid supplements in children with ADHD( Reference Derbyshire 220 ).

Over 35 years ago Rudin suggested that mental disorders might result from a deficiency in very long-chain n-3 fatty acids and might respond to provision of these fatty acids( Reference Rudin 221 ). Data from nine countries demonstrated a significant correlation between high annual fish consumption and lower prevalence of major depression( Reference Hibbeln 222 ), an observation that is compatible with a proposed protective effect of very long-chain n-3 fatty acids. A reduction in depressive symptoms was reported in a small study using a very high dose of EPA + DHA (9·6 g/d)( Reference Su, Huang and Chiu 223 ), while this effect was not seen in a study using a lower dose of DHA alone (2 g/d)( Reference Marangell, Martinez and Zboyan 224 ). Intervention with 6·2 g/d EPA + DHA in patients with bipolar manic depression resulted in significant improvements in nearly all outcomes, especially with respect to depressive symptoms, after 4 months( Reference Stoll, Severus and Freeman 225 ). Likewise, 2 g/d EPA improved symptoms in patients with unipolar depressive disorder after 4 weeks( Reference Nemets, Stahl and Belmaker 226 ). A meta-analysis of thirteen RCT involving 1233 participants with major depressive disorder showed an overall beneficial effect of very long-chain n-3 fatty acids on depressive symptoms( Reference Mocking, Harmsen and Assies 227 ); interestingly higher EPA dose was one factor associated with better outcome for n-3 PUFA supplementation. This finding fits with an earlier analysis of fifteen RCT investigating the effects of EPA, which concluded that supplements containing ≥60% EPA in doses ranging from 0·2 to 2·2 g/d EPA were effective against primary depression( Reference Sublette, Ellis and Geant 228 ). There is also evidence from meta-analysis that depressive symptoms seen in bipolar disorder may be improved by the adjunctive use of very long-chain n-3 fatty acids( Reference Sarris, Mischoulon and Schweitzer 229 ).Another meta-analysis that included twenty-five studies involving 1373 participants identified a ‘small-to-modest benefit’ for depressive symptomology with very long-chain n-3 fatty acids( Reference Appleton, Sallis and Perry 230 ) but the authors expressed doubts about the robustness of their finding.

Schizophrenic patients have lower levels of EPA and DHA in their erythrocytes than do controls( Reference Peet, Laugharne and Rangarajan 231 Reference Hoen, Lijmer and Duran 235 ). The first trial of very long-chain n-3 fatty acids in schizophrenia identified clinical improvement with EPA (2 g/d), but not with DHA( Reference Peet, Brind and Ramchand 236 ), while subsequent trials also showed benefit with EPA( Reference Peet and Horrobin 237 , Reference Fenton, Dickerson and Boronow 238 ), but not all studies have seen this( Reference Emsley, Myburgh and Oosthuizen 239 ). A recent study reported significant benefits of very long-chain n-3 fatty acids given for 26 weeks to patients with schizophrenia( Reference Pawełczyk, Grancow-Grabka and Kotlicka-Antczak 240 ). Although these findings are encouraging, a Cochrane review concluded that very long-chain n-3 fatty acids should be regarded only as an experimental treatment for schizophrenia( Reference Joy, Mumby-Croft and Joy 241 ). A study reported significant benefit from 1 g/d EPA in borderline personality disorder( Reference Zanarini and Frankenburg 242 ), while a small number of studies report anti-aggressive effects of DHA( Reference Hamazaki, Sawazaki and Itomura 243 , Reference Hamazaki, Thienprasert and Kheovichai 244 ).

Post-mortem studies showed that the brains of people with Alzheimer's disease contain less DHA than those without the disease( Reference Prasad, Lovell and Yatin 245 Reference Soderberg, Edlund and Kristensson 248 ) and some studies have linked low blood levels of very long-chain n-3 fatty acids to dementia( Reference Conquer, Tierney and Zecevic 249 ). Sinn et al. reported that 1·8 g EPA + DHA daily for 6 months reduced depressive symptoms and improved cognition in adults with mild cognitive impairment( Reference Sinn, Milte and Street 250 ). Improved memory performance in subjects with mild Alzheimer's disease was reported with 1·5 g EPA + DHA daily for 6 months( Reference Scheltens, Twisk and Blesa 251 ). However, a number of other studies using various doses and ratios of EPA and DHA reported no effect on cognitive performance in people with Alzheimer's disease( Reference Quinn, Raman and Thomas 252 Reference Phillips, Childs and Calder 257 ). Meta-analyses provide a mixed view of the findings for n-3 fatty acid treatment in the area of cognitive impairment, reflecting the mixed findings from individual trials. A Cochrane review of RCT studying the role of very long-chain n-3 fatty acids in preventing cognitive decline in healthy older people showed no benefits( Reference Sydenham, Dangour and Lim 258 ), while two more recent meta-analyses differ in their findings. One meta-analysis of six RCT of duration 3–40 months and using 0·l4–1·8 g EPA + DHA daily identified a slower rate of cognitive decline in those receiving very long-chain n-3 fatty acids( Reference Zhang, Hou and Li 259 ). In contrast, a meta-analysis of three RCT involving 632 participants with mild to moderate Alzheimer's disease and followed for 6, 12 or 18 months found no evidence of a benefit from very long-chain n-3 fatty acids on any outcome that was assessed( Reference Burckhardt, Herke and Wustmann 260 ). A recent large trial found no effects of very long-chain n-3 fatty acids (0·225 g EPA plus 0·8 g DHA daily) over 3 years on cognitive decline( Reference Andrieu, Guyonnet and Coley 261 ).

Thus, DHA is a key structural component of the brain and retina, where it plays particular, unique functional roles. A supply of DHA is very important early in life, especially during the fetal and early infant periods when the eye and central nervous system are developing. Since the supply must come from maternal sources (via the placenta and breast milk), maternal DHA status is likely to be important in determining eye and brain development early in life. Newly emerging areas of interest relate to the influence of very long-chain n-3 fatty acids on childhood developmental disorders, adult psychiatric and psychological disorders, and neurodegenerative diseases of ageing. These conditions appear to be associated with a lower status of very long-chain n-3 fatty acids. Additionally, there is some epidemiological evidence for a lowered risk of psychiatric, psychological disorders and neurodegenerative disorders with increased dietary intake of very long-chain n-3 fatty acids. Intervention studies indicate that there may be some benefit from very long-chain n-3 fatty acids in childhood developmental and adult psychiatric and psychological disorders, particularly in depression. Interestingly many of these studies are indicative that EPA is more important than DHA, which contrasts with the relative roles of these two fatty acids in very early eye and brain development. Although there may be a role for very long-chain n-3 fatty acids in slowing cognitive decline( Reference Zhang, Hou and Li 259 ), this has not been well demonstrated( Reference Sydenham, Dangour and Lim 258 , Reference Burckhardt, Herke and Wustmann 260 ). There is a clear need for larger, longer and higher quality human trials in this area of research.

Recommendations for intake of EPA and DHA acid

The demonstration of physiological actions of EPA and DHA that result in improved health outcomes and reduced risk of disease, along with the increased understanding of the molecular and cellular mechanisms of action involved, indicates a need to set recommendations for the intake of these important fatty acids. However, the exact requirement for very long-chain n-3 fatty acids in order to maintain health is not known. Furthermore, there has been a lack of clarity about the extent to which EPA and DHA can be synthesised in human subjects so long as there is sufficient intake of the precursor ALA( Reference Baker, Miles and Burdge 8 ). Nevertheless, the recognition of the health benefits of EPA and DHA has resulted in several recommendations to increase the intake of fish and, more specifically, of EPA and DHA by various governmental, non-governmental and professional agencies( 17 , Reference Kris-Etherton, Harris and Appel 89 , Reference Siscovick, Barringer and Fretts 129 , Reference Simopolous, Leaf and Salem 262 267 ). These recommendations are summarised in Table 5.

Table 5. Some of the current recommendations for intake of very long-chain n-3 fatty acids

The future of research in n-3 PUFA

Very long-chain n-3 fatty acids have been a focus of research since the late 1980s, although important pioneering work in the field was performed before then. A PubMed search conducted in late August 2017 identified over 25 500 publications using the search term ‘omega-3’ and 28 500 publications using the search term ‘fish oil’. The earliest publications identified using those terms were from 1922 and 1945, respectively, although the earliest of the almost 1000 publications identified with the search term ‘cod liver oil’ was from 1845. By 1990, there was a publication on the topic of n-3 fatty acids on average every day. By the mid-2010s this had risen to almost five publications every day. Thus, a tremendous effort has been, and is continuing to be, expended on research in the area of n-3 fatty acids. This research effort obviously falls into many categories, not all related to human health and well-being, although much of it does. Research aimed at better defining the roles that very long-chain n-3 fatty acids play in human growth, development, function, health, well-being and disease risk and better understanding the molecular and cellular mechanisms involved will continue for the foreseeable future. Despite the vast amount of knowledge that is currently available about n-3 PUFA, there remain significant gaps and inconsistencies in the literature that impair the ability to make robust recommendations for both the healthy population and specific patient groups. One reason for this is that it is not clear what the extent of the variation in response to very long-chain n-3 fatty acids is within the population or amongst particular subgroups, or what the determinants of such variation might be, although these are likely to include other dietary components, sex, body composition, genetics, stage in the life course, physiological state and the presence of disease. These determinants are likely to be more thoroughly explored in the coming years, with researchers taking advantage of new analytical and data handling technologies to study larger numbers of samples. Surprisingly, there is insufficient information currently available on dose responses of many physiological and pathological outcomes to very long-chain n-3 fatty acid intervention. This hampers the ability to use the full potential of very long-chain n-3 fatty acids in both public health and clinical settings. Most research on very long-chain n-3 fatty acids to date has focused on EPA and DHA, usually studied in combination. This has resulted in many researchers thinking that EPA and DHA have the same biological effects. This seems not to be the case( Reference Mori and Woodman 268 , Reference Wei and Jacobson 269 ), but again this is an area of research that is likely to become better explored in the next period. Related to this, biological actions of DPA have emerged( Reference Kaur, Cameron-Smith and Garg 4 , Reference Weylandt 5 ) but the effects of DPA and its mechanisms of action are poorly described at the moment. It seems likely that DPA will become a greater focus of research in this area. It is also likely that research into the molecular mechanisms of the action of each individual very long-chain n-3 fatty acid will continue, with membrane structure–function relationships, early intracellular signalling pathways, transcription factors, novel membrane receptors and novel lipid mediators all being important candidates for future study. Whatever the mechanism involved, it is evident that EPA, DPA and DHA consumed orally need to be found at the site of their action in the body. As described earlier, it is also clear that the effects of these fatty acids on cell and tissue function are dose dependent( Reference von Schacky, Fischer and Weber 39 , Reference Harris, Windsor and Dujovne 40 , Reference Blonk, Bilo and Popp-Snijders 42 , Reference Rees, Miles and Banerjee 45 ). Thus, it seems likely that strategies for more effective delivery of these bioactive fatty acids will be explored. These might include different chemical( Reference Schuchardt and Hahn 270 , Reference Ghasemifard, Turchini and Sinclair 271 ) or physical( Reference Raatz, Johnson and Bukowski 272 ) formulations.

The richest dietary source of very long-chain n-3 fatty acids is seafood, especially fatty fish (Table 1). Despite recommendations by many authorities to increase consumption of fish, the majority of the Western population are low consumers. An alternative to eating fish is to use n-3 supplements (fish oil) which give an assured intake of EPA and DHA. A concern is that the supply of fish for human consumption and for the production of oil for use in n-3 supplements is not sustainable. Thus, alternative sources need to be sought and then proven to be beneficial to human health. Such alternatives include algal oils, some of which are already widely used in the infant formula industry and seed oils from plants GM to produce EPA and DHA( Reference Ruiz-Lopez, Usher and Sayanova 273 ). Finally, although the focus of the present paper is the very long-chain n-3 fatty acids EPA, DPA and DHA, plant n-3 fatty acids are widely available and are sustainable. Most research on plant n-3 fatty acids conducted to date has been on ALA as an alternative to very long-chain n-3 fatty acids( Reference Baker, Miles and Burdge 8 ), but there has been some research on SDA( Reference Baker, Miles and Burdge 8 , Reference Walker, Jebb and Calder 274 ). From the research conducted to date, ALA and SDA need to be converted to very long-chain n-3 fatty acids by the pathway shown in Fig. 1 to be biologically effective( Reference Baker, Miles and Burdge 8 ). However, the studies done so far show very poor conversion of ALA and SDA to DHA in human subjects, although SDA is a better precursor for EPA synthesis than ALA( Reference James, Ursin and Cleland 275 ). Future research will attempt to identify strategies to enhance endogenous biosynthesis of very long-chain n-3 fatty acids from their simpler plant precursors. These strategies might include modifying other aspects of the diet or taking advantage of genetic polymorphisms that favour conversion. Thus, there are many important questions yet to be answered and this assures continued research in the field of n-3 fatty acids and human health.

Summary and conclusions

n-3 Fatty acids are a family of PUFA that contribute to human development, health and well-being. Functionally the most important n-3 fatty acids are the very long-chain EPA and DHA found in fatty fish and in supplements; roles for DPA are now emerging. Intakes of EPA and DHA are typically low and much below those that are recommended. Increased intakes of EPA and DHA, either from fish or from supplements, are reflected in greater incorporation into blood lipid, cell and tissue pools. Increased content of EPA and DHA can modify the structure of cell membranes and also the function of membrane proteins involved as receptors, signalling proteins, transporters and enzymes. EPA and DHA also modify the production of lipid mediators and through effects on cell signalling can alter patterns of gene expression. Through these actions EPA and DHA act to alter cellular responsiveness in a manner that seems to result in more optimal conditions for growth, development and maintenance of health. The effects of very long-chain n-3 fatty acids are evident right through the life course meaning that there is a need for all sectors of the population to increase the intake of these important nutrients. EPA and DHA have a wide range of physiological roles, which are linked to certain health or clinical benefits. A number of risk factors for CVD are modified in a beneficial way by increased intake of EPA and DHA; these include blood pressure, platelet reactivity and thrombosis, plasma TAG concentrations, vascular function, cardiac arrhythmias, heart rate variability and inflammation. Thus, there is a key role for these fatty acids in prevention and slowing progression of CVD. Furthermore, some supplementation studies with EPA and DHA have demonstrated reduced mortality in at risk patients, such as post-MI, indicating a therapeutic role, although this is currently disputed by some. A number of other, non-cardiovascular, actions of EPA and DHA have also been documented, suggesting that increased intake of these fatty acids could be of benefit in reducing the risk of (i.e. protecting from) or treating many conditions. For example, they have been used successfully in RA and, in some studies, in inflammatory bowel disease, and may be useful in other inflammatory conditions like asthma and psoriasis. EPA and DHA may also have a role as part of cancer therapy; some recent studies show that they improve the effectiveness of chemotherapeutic agents. DHA has an important structural role in the eye and brain, and its supply early in life when these tissues are developing is known to be of importance in terms of optimising visual and neurological development. For this reason it is very important that pregnant and breast-feeding women have adequate DHA intake. Recent studies have highlighted the potential for EPA and DHA to contribute to enhanced mental development and improved childhood learning and behaviour and to reduce the burden of psychiatric and depressive illnesses in adults, although these areas of possible action require more robust scientific support. There may also be a role for EPA and DHA in preventing neurodegenerative disease of ageing, but evidence for this is currently weak. The effects of EPA and DHA on health outcomes are likely to be dose-dependent, but clear dose–response data have not been identified in most cases. Also in many cases it is not clear whether both EPA and DHA have the same effect or potency and therefore which one will be the most important for a particular indication. Thus, despite several decades of productive research on the health effects of very long-chain n-3 fatty acids and the mechanisms involved, important questions are currently unanswered and many areas remain to be explored in future research.

Conflict of interest

The author serves on the Scientific Advisory Boards of Pronova BioPharma (part of BASF), Smartfish, DSM, FrieslandCampina and Danone Nutricia Research.

Financial Support

None.

Authorship

The author had sole responsibility for all aspects of preparation of this paper.

References

1. Burdge, GC & Calder, PC (2015) Introduction to fatty acids and lipids. World Rev Nutr Diet 112, 116.Google Scholar
2. Calder, PC & Yaqoob, P (2009) Understanding omega-3 polyunsaturated fatty acids. Postgrad Med 121, 148157.Google Scholar
3. Calder, PC (2014) Very long chain omega-3 (n-3) fatty acids and human health. Eur J Lipid Sci Technol 116, 12801300.Google Scholar
4. Kaur, G, Cameron-Smith, D, Garg, M et al. (2011) Docosapentaenoic acid (22:5n-3): a review of its biological effects. Prog. Lipid Res 50, 2834.Google Scholar
5. Weylandt, KH (2016) Docosapentaenoic acid derived metabolites and mediators – the new world of lipid mediator medicine in a nutshell. Eur J Pharmacol 785, 108115.CrossRefGoogle Scholar
6. Blasbalg, TL, Hibbeln, JR, Ramsden, CE et al. (2011) Changes in consumption of omega-3 and omega-6 fatty acids in the United States during the 20th century. Am J Clin Nutr 93, 950962.CrossRefGoogle Scholar
7. Arterburn, LM, Hall, EB & Oken, H (2006) Distribution, interconversion, and dose response of n-3 fatty acids in humans. Am J Clin Nutr 83, 1467S1476S.Google Scholar
8. Baker, EJ, Miles, EA, Burdge, GC et al. (2016) Metabolism and functional effects of plant-derived omega-3 fatty acids in humans. Prog Lipid Res 64, 3056.CrossRefGoogle ScholarPubMed
9. Bolton-Smith, C, Woodward, M & Tavendale, R (1997) Evidence for age-related differences in the fatty acid composition of human adipose tissue, independent of diet. Eur J Clin Nutr 51, 619624.Google Scholar
10. Childs, CE, Romeu-Nadal, M, Burdge, GC et al. (2008) Gender differences in the n-3 fatty acid content of tissues. Proc Nutr Soc 67, 1927.Google Scholar
11. Burdge, GC, Jones, AE & Wootton, SA (2002) Eicosapentaenoic and docosapentaenoic acids are the principal products of alpha-linolenic acid metabolism in young men. Brit J Nutr 88, 355363.CrossRefGoogle ScholarPubMed
12. Burdge, GC & Wootton, SA (2002) Conversion of alpha-linolenic acid to eicosapentaenoic, docosapentaenoic and docosahexaenoic acids in young women. Brit J Nutr 88, 411420.Google Scholar
13. Vessby, B, Gustafsson, IB, Tengblad, S et al. (2002) Desaturation and elongation of fatty acids and insulin action. Ann NY Acad Sci 967, 183195.Google Scholar
14. Koletzko, B, Lattka, E, Zeilinger, S et al. (2011) Genetic variants of the fatty acid desaturase gene cluster predict amounts of red blood cell docosahexaenoic acid and other polyunsaturated fatty acids in pregnant women: findings from the Avon Longitudinal Study of Parents and Children. Am J Clin Nutr 93, 211219.CrossRefGoogle ScholarPubMed
15. British Nutrition Foundation (1999) N-3 Fatty Acids and Health. London: British Nutrition Foundation.Google Scholar
16. Calder, PC (2017) Omega-3: the good oil. Nutr Bull 42, 132140.Google Scholar
17. Scientific Advisory Committee on Nutrition/Committee on Toxicity (2004) Advice on Fish Consumption: Benefits and Risks. London: TSO.Google Scholar
18. Meyer, BJ, Mann, NJ, Lewis, JL et al. (2003) Dietary intakes and food sources of omega-6 and omega-3 polyunsaturated fatty acids. Lipids 38, 391398.CrossRefGoogle ScholarPubMed
19. Howe, P, Meyer, B, Record, S et al. (2006) Dietary intake of long-chain omega-3 polyunsaturated fatty acids: contribution of meat sources. Nutrition 22, 4753.Google Scholar
20. Rahmawaty, S, Charlton, K, Lyons-Wall, P et al. (2013) Dietary intake and food sources of EPA, DPA and DHA in Australian children. Lipids 48, 869877.Google Scholar
21. Stubbs, CD & Smith, AD (1984) The modification of mammalian membrane polyunsaturated fatty acid composition in relation to membrane fluidity and function. Biochim Biophys Acta 779, 89137.CrossRefGoogle ScholarPubMed
22. Brenner, RR (1984) Effect of unsaturated fatty acids on membrane structure and enzyme kinetics. Prog Lipid Res 23, 6996.Google Scholar
23. Murphy, MG (1990) Dietary fatty acids and membrane protein function. J Nutr Biochem 1, 6879.Google Scholar
24. Browning, LM, Walker, CG, Mander, AP et al. (2012) Incorporation of eicosapentaenoic and docosahexaenoic acids into lipid pools when given as supplements providing doses equivalent to typical intakes of oily fish. Am J Clin Nutr 96, 748758.CrossRefGoogle ScholarPubMed
25. Miles, EA, Noakes, PS, Kremmyda, LS et al. (2011) The Salmon in Pregnancy Study: study design, subject characteristics, maternal fish and marine n-3 fatty acid intake, and marine n-3 fatty acid status in maternal and umbilical cord blood. Am J Clin Nutr 94, 1986S1992S.Google Scholar
26. Healy, DA, Wallace, FA, Miles, EA et al. (2000) The effect of low to moderate amounts of dietary fish oil on neutrophil lipid composition and function. Lipids 35, 763768.Google Scholar
27. Skinner, ER, Watt, C, Besson, JA et al. (1993) Differences in the fatty acid composition of the grey and white matter of different regions of the brains of patients with Alzheimer's disease and control subjects. Brain 116, 717725.Google Scholar
28. Makrides, M, Neumann, MA, Byard, RW et al. (1994) Fatty acid composition of brain, retina, and erythrocytes in breast- and formula-fed infants. Am J Clin Nutr 60, 189194.Google Scholar
29. Harris, WS, Sands, SA, Windsor, SL et al. (2004) Omega-3 fatty acids in cardiac biopsies from heart transplantation patients: correlation with erythrocyes and response to supplementation. Circulation 110, 16451649.Google Scholar
30. Metcalf, RG, James, MJ, Gibson, RA et al. (2007) Effects of fish-oil supplementation on myocardial fatty acids in humans. Am J Clin Nutr 85, 12221228.Google Scholar
31. McGlory, C, Galloway, SD, Hamilton, DL et al. (2014) Temporal changes in human skeletal muscle and blood lipid composition with fish oil supplementation. Prostagland Leukotr Essent Fatty Acids 90, 199206.Google Scholar
32. Smith, GI, Atherton, P, Reeds, DN et al. (2011) Omega-3 polyunsaturated fatty acids augment the muscle protein anabolic response to hyperinsulinaemia-hyperaminoacidaemia in healthy young and middle-aged men and women. Clin Sci 121, 267278.Google Scholar
33. Araya, J, Rodrigo, R, Videla, LA et al. (2004) Increase in long-chain polyunsaturated fatty acid n-6/n-3 ratio in relation to hepatic steatosis in patients with non-alcoholic fatty liver disease. Clin Sci 106, 635643.CrossRefGoogle ScholarPubMed
34. Elizondo, A, Araya, J, Rodrigo, R et al. (2007) Polyunsaturated fatty acid pattern in liver and erythrocyte phospholipids from obese patients. Obesity 15, 2431.Google Scholar
35. Hillier, K, Jewell, R, Dorrell, L et al. (1991) Incorporation of fatty acids from fish oil and olive oil into colonic mucosal lipids and effects upon eicosanoid synthesis in inflammatory bowel disease. Gut 32, 11511155.Google Scholar
36. Safarinejad, MR (2011) Effect of omega-3 polyunsaturated fatty acid supplementation on semen profile and enzymatic anti-oxidant capacity of seminal plasma in infertile men with idiopathic oligoasthenoteratospermia: a double-blind, placebo-controlled, randomised study. Andrologia 43, 3847.Google Scholar
37. Crawford, MA, Casperd, NM & Sinclair, AJ (1976) The long chain metabolites of linoleic and linolenic acids in liver and brain in herbivores and carnivores. Comp Biochem Physiol 54B, 395401.Google Scholar
38. Anderson, RE (1970) Lipids of ocular tissues. IV. A comparison of the phospholipids from the retina of six mammalian species. Exp Eye Res 10, 339344.Google Scholar
39. von Schacky, C, Fischer, S & Weber, PC (1985) Long term effects of dietary marine ω−3 fatty acids upon plasma and cellular lipids, platelet function, and eicosanoid formation in humans. J Clin Invest 76, 16261631.Google Scholar
40. Harris, WS, Windsor, SL & Dujovne, CA (1991) Effects of four doses of n-3 fatty acids given to hyperlipidemic patients for six months. J Am Coll Nutr 10, 220227.Google Scholar
41. Marsen, TA, Pollok, M, Oette, K et al. (1992) Pharmacokinetics of omega-3 fatty acids during ingestion of fish oil preparations. Prostagland Leukotr Essent Fatty Acids 46, 191196.CrossRefGoogle ScholarPubMed
42. Blonk, MC, Bilo, HJ, Popp-Snijders, C et al. (1990) Dose-response effects of fish oil supplementation in healthy volunteers. Am J Clin Nutr 52, 120127.Google Scholar
43. Katan, MB, Deslypere, JP, van Birgelen, APJM et al. (1997) Kinetics of the incorporation of dietary fatty acids into serum cholesteryl esters, erythrocyte membranes and adipose tissue: an 18 month controlled study. J Lipid Res 38, 20122022.CrossRefGoogle ScholarPubMed
44. Yaqoob, P, Pala, HS, Cortina-Borja, M et al. (2000) Encapsulated fish oil enriched in α-tocopherol alters plasma phospholipid and mononuclear cell fatty acid compositions but not mononuclear cell functions. Eur J Clin Invest 30, 260274.Google Scholar
45. Rees, D, Miles, EA, Banerjee, T et al. (2006) Dose-related effects of eicosapentaenoic acid on innate immune function in healthy humans: a comparison of young and older men. Am J Clin Nutr 83, 331342.CrossRefGoogle Scholar
46. Faber, J, Berkhout, M, Vos, AP et al. (2011) Supplementation with a fish oil-enriched, high-protein medical food leads to rapid incorporation of EPA into white blood cells and modulates immune responses within one week in healthy men and women. J Nutr 141, 964970.CrossRefGoogle ScholarPubMed
47. Popp-Snijders, C, Schouten, JA, van Blitterswijk, WJ et al. (1986) Changes in membrane lipid composition of human erythrocytes after dietary supplementation of (n-3) fatty acids: maintenance of membrane fluidity. Biochim Biophys Acta 854, 3137.Google Scholar
48. Sanders, TAB & Roshanai, F (1983) The influence of different types of ω3 polyunsaturated fatty acids on blood lipids and platelet function in healthy volunteers. Clin Sci 64, 9199.Google Scholar
49. Sperling, RI, Benincaso, AI, Knoell, CT et al. (1993) Dietary ω−3 polyunsaturated fatty acids inhibit phosphoinositide formation, and chemotaxis in neutrophils. J Clin Invest 91, 651660.Google Scholar
50. Sorensen, LS, Rasmussen, HH, Aardestrup, IV et al. (2014) Rapid incorporation of ω−3 fatty acids into colonic tissue after oral supplementation in patients with colorectal cancer: a randomized, placebo-controlled intervention trial. J Parent Ent Nutr 38, 617624.Google Scholar
51. Calder, PC (2012) Mechanisms of action of (n-3) fatty acids. J Nutr 142, 592S599S.Google Scholar
52. Calder, PC (2013) Omega-3 polyunsaturated fatty acids and inflammatory processes: nutrition or pharmacology? Brit J Clin Pharmacol 75, 645662.Google Scholar
53. Calder, PC (2013) n-3 Fatty acids, inflammation and immunity: new mechanisms to explain old actions. Proc Nutr Soc 72, 326336.CrossRefGoogle ScholarPubMed
54. Calder, PC, Yaqoob, P, Harvey, DJ et al. (1994) The incorporation of fatty acids by lymphocytes and the effect on fatty acid composition and membrane fluidity. Biochem J 300, 509518.Google Scholar
55. Miles, EA & Calder, PC (1998) Modulation of immune function by dietary fatty acids. Proc Nutr Soc 57, 277292.CrossRefGoogle ScholarPubMed
56. Niu, SL, Mitchell, DC, Lim, SY et al. (2004) Reduced G protein-coupled signaling efficiency in retinal rod outer segments in response to n-3 fatty acid deficiency. J Biol Chem 279, 3109831104.Google Scholar
57. Pike, LJ (2003) Lipid rafts: bringing order to chaos. J Lipid Res 44, 655667.Google Scholar
58. Simons, K & Gerl, MJ (2010) Revitalizing membrane rafts: new tools and insights. Nat Rev Mol Cell Biol 11, 688699.Google Scholar
59. Yaqoob, P (2009) The nutritional significance of lipid rafts. Annu Rev Nutr 29, 257282.CrossRefGoogle ScholarPubMed
60. Calder, PC & Yaqoob, P (2007) Lipid rafts – Composition, characterization and controversies. J Nutr 137, 545547.Google Scholar
61. Hou, TY, McMurray, DN & Chapkin, RS (2016) Omega-3 fatty acids, lipid rafts, and T cell signaling. Eur J Pharmacol 785, 29.Google Scholar
62. Calder, PC (2013) Fat chance to enhance B cell function. J Leuk Biol 93, 457459.Google Scholar
63. Calder, PC (2013) Long chain fatty acids and gene expression in inflammation and immunity. Curr Opin Clin Nutr Metab Care 16, 425433.Google Scholar
64. Novak, TE, Babcock, TA, Jho, DH et al. (2003) NF-kappa B inhibition by omega-3 fatty acids modulates LPS-stimulated macrophage TNF-alpha transcription. Am J Physiol 284, L84L89.Google Scholar
65. Krey, G, Braissant, O, L'Horset, F et al. (1997) Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor ligand assay. Mol Endocrinol 11, 779791.Google Scholar
66. Marion-Letellier, R, Savoye, G & Ghosh, S (2016) Fatty acids, eicosanoids and PPAR gamma. Eur J Pharmacol 785, 4449.Google Scholar
67. Clarke, SD (2004) The multi-dimensional regulation of gene expression by fatty acids: polyunsaturated fats as nutrient sensors. Curr Opin Lipidol 15, 1318.CrossRefGoogle ScholarPubMed
68. Lapillonne, A, Clarke, SD & Heird, WC (2004) Polyunsaturated fatty acids and gene expression. Curr Opin Clin Nutr Metab Care 7, 151156.CrossRefGoogle ScholarPubMed
69. Jump, DB (2002) Dietary polyunsaturated fatty acids and regulation of gene transcription. Curr Opin Lipidol 13, 1551564.CrossRefGoogle ScholarPubMed
70. Jump, DB (2008) N-3 polyunsaturated fatty acid regulation of hepatic gene transcription. Curr Opin Lipidol 19, 242247.Google Scholar
71. Deckelbaum, RJ, Worgall, TS & Seo, T (2006) N-3 fatty acids and gene expression. Am J Clin Nutr 83, 1520S1525S.Google Scholar
72. Calder, PC (2011) Fatty acids and inflammation: the cutting edge between food and pharma. Eur J Pharmacol 668, S50S58.CrossRefGoogle ScholarPubMed
73. Lewis, RA, Austen, KF & Soberman, RJ (1990) Leukotrienes and other products of the 5-lipoxygenase pathway: biochemistry and relation to pathobiology in human diseases. N Engl J Med 323, 645655.Google ScholarPubMed
74. Tilley, SL, Coffman, TM & Koller, BH (2001) Mixed messages: modulation of inflammation and immune responses by prostaglandins and thromboxanes. J Clin Invest 108, 1523.CrossRefGoogle Scholar
75. Wada, M, DeLong, CJ, Hong, YH et al. (2007) Enzymes and receptors of prostaglandin pathways with arachidonic acid-derived versus eicosapentaenoic acid-derived substrates and products. J Biol Chem 282, 2225422266.Google Scholar
76. Moncada, S & Vane, JR (1979) The role of prostacyclin in vascular tissue. Fed Proc 38, 6871.Google Scholar
77. Bannenberg, G & Serhan, CN (2010) Specialized pro-resolving lipid mediators in the inflammatory response: an update. Biochim Biophys Acta Mol Cell Biol Lipids 1801, 12601273.Google Scholar
78. Serhan, CN, Chiang, N & van Dyke, TE (2008) Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol 8, 349361.CrossRefGoogle ScholarPubMed
79. Serhan, CN & Chiang, N (2013) Resolution phase lipid mediators of inflammation: agonists of resolution. Curr Opin Pharmacol 13, 632640.Google Scholar
80. Dalli, J, Colas, RA & Serhan, CN (2013) Novel n-3 immunoresolvents: structures and roles. Sci Rep 3, 1940.Google Scholar
81. Calder, PC (2015) Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochim Biophys Acta 1851, 469484.Google Scholar
82. Oh, DY, Talukdar, S, Bae, EJ et al. (2010) GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 142, 687698.Google Scholar
83. Dyerberg, J, Bang, HO, Stoffersen, E et al. (1978) Eicosapentaenoic acid and prevention of thrombosis and atherosclerosis. Lancet ii, 117119.Google Scholar
84. Kromann, N & Green, A (1980) Epidemiological studies in the Upernavik District, Greenland. Acta Med Scand 208, 401406.Google Scholar
85. Bjerregaard, P & Dyerberg, J (1988) Mortality from ischemic heart disease and cerebrovascular disease in Greenland. Int J Epidemiol 17, 514519.Google Scholar
86. Newman, WP, Middaugh, JP, Propst, MT et al. (1993) Atherosclerosis in Alaska Natives and non-natives. Lancet 341, 10561057.Google Scholar
87. Bang, HO, Dyerberg, J & Hjorne, N (1976) The composition of foods consumed by Greenland Eskimos. Acta Med Scand 200, 6973.Google Scholar
88. Yano, K, MacLean, CJ, Reed, DM et al. (1988) A comparison of the 12-year mortality and predictive factors of coronary heart disease among Japanese men in Japan and Hawaii. Am J Epidemiol 127, 476487.Google Scholar
89. Kris-Etherton, PM, Harris, WS & Appel, LJ (2002) Fish consumption, fish oil, omega-3 fatty acids, and cardiovascular disease. American Heart Association Nutrition Committee. Circulation 106, 27472757.Google Scholar
90. von Schacky, C (2004) Omega-3 fatty acids and cardiovascular disease. Curr Opin Clin Nutr Metab Care 7, 131136.Google Scholar
91. London, B, Albert, C, Anderson, M et al. (2007) Omega-3 fatty acids and cardiac arrhythmias: prior studies and recommendations for future research: a report from the National Heart, Lung, and Blood Institute and Office Of Dietary Supplements Omega-3 Fatty Acids and their Role in Cardiac Arrhythmogenesis Workshop. Circulation 116, e320e335.Google Scholar
92. Wang, C, Harris, WS, Chung, M et al. (2006) n-3 Fatty acids from fish or fish-oil supplements, but not alpha-linolenic acid, benefit cardiovascular disease outcomes in primary- and secondary-prevention studies: a systematic review. Am J Clin Nutr 84, 517.Google Scholar
93. Calder, PC (2004) n-3 Fatty acids and cardiovascular disease: evidence explained and mechanisms explored. Clin Sci 107, 111.Google Scholar
94. Chowdhury, R, Warnakula, S, Kunutsor, S et al. (2014) Association of dietary, circulating, and supplement fatty acids with coronary risk: a systematic review and meta-analysis. Ann Intern Med 160, 398406.Google Scholar
95. Del Gobbo, LC, Imamura, F, Aslibekyan, S et al. (2016) ω−3 Polyunsaturated fatty acid biomarkers and coronary heart disease: pooling project of 19 cohort studies. JAMA Intern Med 176, 11551166.Google Scholar
96. Calder, PC (2017) New evidence that omega-3 fatty acids have a role in primary prevention of coronary heart disease. J Pub Health Emerg 1, 35.Google Scholar
97. Harris, WS (1996) N-3 fatty acids and lipoproteins: comparison of results from human and animal studies. Lipids 31, 243252.Google Scholar
98. Saravanan, P, Davidson, NC, Schmidt, EB et al. (2010) Cardiovascular effects of marine omega-3 fatty acids. Lancet 376, 540550.Google Scholar
99. De Caterina, R (2011) n-3 fatty acids in cardiovascular disease. N Engl J Med 364, 24392450.CrossRefGoogle ScholarPubMed
100. Balk, EM, Lichtenstein, AH, Chung, M et al. (2006) Effects of omega-3 fatty acids on serum markers of cardiovascular disease risk: a systematic review. Atherosclerosis 189, 1930.Google Scholar
101. Leslie, MA, Cohen, DJ, Liddle, DM et al. (2015) A review of the effect of omega-3 polyunsaturated fatty acids on blood triacylglycerol levels in normolipidemic and borderline hyperlipidemic individuals. Lipids Health Dis 14, 53.Google Scholar
102. AbuMweis, S, Jew, S, Tayyem, R et al. (2017) Eicosapentaenoic acid and docosahexaenoic acid containing supplements modulate risk factors for cardiovascular disease: a meta-analysis of randomised placebo-control human clinical trials. J Hum Nutr Diet (In the Press).Google Scholar
103. Mori, TA, Burke, V, Puddey, IB et al. (2000) Purified eicosapentaenoic and docosahexaenoic acids have differential effects on serum lipids and lipoproteins, LDL particle size, glucose, and insulin in mildly hyperlipidemic men. Am J Clin Nutr 71, 10851094.CrossRefGoogle ScholarPubMed
104. Burr, ML, Fehily, AM, Gilbert, JF et al. (1989) Effects of changes in fat, fish and fibre intake on death and myocardial reinfarction: diet and reinfarction trial (DART). Lancet ii, 757761.Google Scholar
105. Anonymous (1999) Dietary supplementation with n-3 polyunsaturated fatty acids and vitamin E after myocardial infarction: results of the GISSI-Prevenzione trial. Lancet 354, 447455.Google Scholar
106. Marchioli, R, Barzi, F, Bomba, E et al. (2002) Early protection against sudden death by n-3 polyunsaturated fatty acids after myocardial infarction - Time-course analysis of the results of the Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto Miocardico (GISSI)-Prevenzione. Circulation 105, 18971903.Google Scholar
107. Yokoyama, M, Origasa, H, Matsuzaki, M et al. (2007) Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet 369, 10901098.Google Scholar
108. Gissi-HF Investigators, Tavazzi, L, Maggioni, AP, Marchioli, R et al. (2008). Effect of n-3 polyunsaturated fatty acids in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial. Lancet 372, 12231230.Google Scholar
109. Bucher, HC, Hengstler, P, Schindler, C et al. (2002) N-3 polyunsaturated fatty acids in coronary heart disease: a meta-analysis of randomized controlled trials. Am J Med 112, 298304.Google Scholar
110. Studer, M, Briel, M, Leimenstoll, B et al. (2005) Effect of different antilipidemic agents and diets on mortality: a systematic review. Arch Intern Med 165, 725730.Google Scholar
111. Leon, H, Shibata, MC, Sivakumaran, S et al. (2009) Effect of fish oil on arrhythmias and mortality: systematic review. Brit Med J 338, a2931.Google Scholar
112. Harris, WS, Miller, M, Tighe, AP et al. (2008) Omega-3 fatty acids and coronary heart disease risk: clinical and mechanistic perspectives. Atherosclerosis 197, 1224.Google Scholar
113. Xin, W, Wei, W & Li, XY (2013) Short-term effects of fish-oil supplementation on heart rate variability in humans: a meta-analysis of randomized controlled trials. Am J Clin Nutr 97, 926935.Google Scholar
114. Leaf, A & Xiao, YF (2001) The modulation of ionic currents in excitable tissues by n-3 polyunsaturated fatty acids. J Memb Biol 184, 263271.Google Scholar
115. Thies, F, Garry, JMC, Yaqoob, P et al. (2003) Association of n-3 polyunsaturated fatty acids with stability of atherosclerotic plaques: a randomised controlled trial. Lancet 361, 477485.Google Scholar
116. Cawood, AL, Ding, R, Napper, FL et al. (2010) Eicosapentaenoic acid (EPA) from highly concentrated n-3 fatty acid ethyl esters is incorporated into advanced atherosclerotic plaques and higher plaque EPA is associated with decreased plaque inflammation and increased stability. Atherosclerosis 212, 252259.CrossRefGoogle ScholarPubMed
117. Calder, PC & Yaqoob, P (2010) Omega-3 (n-3) fatty acids, cardiovascular disease and stability of atherosclerotic plaques. Cell Mol Biol 56, 2837.Google Scholar
118. Galan, P, Kesse-Guyot, E, Czernichow, S et al. (2010) Effects of B vitamins and omega 3 fatty acids on cardiovascular diseases: a randomised placebo controlled trial. BMJ 341, c6273.Google Scholar
119. Kromhout, D, Giltay, EJ, Geleijnse, JM et al. (2010) n-3 fatty acids and cardiovascular events after myocardial infarction. N Engl J Med 363, 20152026.Google Scholar
120. Rauch, B, Schiele, R, Schneider, S et al. (2010) OMEGA, a randomized, placebo-controlled trial to test the effect of highly purified omega-3 fatty acids on top of modern guideline-adjusted therapy after myocardial infarction. Circulation 122, 21522159.Google Scholar
121. Risk and Prevention Study Collaborative Group, Roncaglioni, MC, Tombesi, M, Avanzini, F et al. (2013) n-3 fatty acids in patients with multiple cardiovascular risk factors. N Engl J Med 368, 18001808.Google ScholarPubMed
122. Bosch, J, Gerstein, HC, Dagenais, GR et al. (2012) n-3 fatty acids and cardiovascular outcomes in patients with dysglycemia. N Engl J Med 367, 309318.Google Scholar
123. Kwak, SM, Myung, SK, Lee, YJ et al. (2012) Efficacy of omega-3 fatty acid supplements (eicosapentaenoic acid and docosahexaenoic acid) in the secondary prevention of cardiovascular disease: a meta-analysis of randomized, double-blind, placebo-controlled trials. Arch Intern Med 172, 686694.Google Scholar
124. Kotwal, S, Jun, M, Sullivan, D et al. (2012) Omega 3 Fatty acids and cardiovascular outcomes: systematic review and meta-analysis. Circ Cardiovasc Qual Outcomes 5, 808818.Google Scholar
125. Rizos, EC, Ntzani, EE, Bika, E et al. (2012) Association between omega-3 fatty acid supplementation and risk of major cardiovascular disease events: a systematic review and meta-analysis. J Am Med Assoc 308, 10241033.Google Scholar
126. Calder, PC & Yaqoob, P (2012) Marine omega-3 fatty acids and coronary heart disease. Curr Opin Cardiol 27, 412419.Google Scholar
127. Casula, M, Soranna, D, Catapano, AL et al. (2013) Long-term effect of high dose omega-3 fatty acidsupplementation for secondary prevention of cardiovascular outcomes: a meta-analysis of randomized, double blind, placebo controlled trials. Atheroscler Suppl 14, 243251.Google Scholar
128. Wen, YT, Dai, JH & Gao, Q (2014) Effects of omega-3 fatty acid on major cardiovascular events and mortality in patients with coronary heart disease: a meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis 24, 470475.CrossRefGoogle ScholarPubMed
129. Siscovick, DS, Barringer, TA, Fretts, AM et al. (2017) Omega-3 polyunsaturated fatty acid (fish oil) supplementation and the prevention of clinical cardiovascular disease: a science advisory from the American Heart Association. Circulation 135, e867e884.Google Scholar
130. Gleissman, H, Johnsen, JI & Kogner, P (2010) Omega-3 fatty acids in cancer, the protectors of good and the killers of evil? Exp Cell Res 316, 13651373.Google Scholar
131. Merendino, N, Costantini, L, Manzi, L et al. (2013) Dietary ω−3 polyunsaturated fatty acids DHA: a potential adjuvant in the treatment of cancer. BioMed Res Int 2013; 310186.Google Scholar
132. Wang, D & Dubois, RN (2010) Eicosanoids and cancer. Nat Rev Cancer 10, 181193.Google Scholar
133. Vaughan, VC, Hassing, MR & Lewandowski, PA (2013) Marine polyunsaturated fatty acids and cancer therapy. Brit J Cancer 108, 486492.Google Scholar
134. Murphy, RA, Mourtzakis, M & Mazurak, VC (2012) n-3 polyunsaturated fatty acids: the potential role for supplementation in cancer. Curr Opin Clin Nutr Metab Care 15, 246251.Google Scholar
135. Yang, B, Wang, FL, Ren, XL et al. (2014) Biospecimen long-chain n-3 PUFA and risk of colorectal cancer: a meta-analysis of data from 60,627 individuals. PLoS ONE 9, e110574.Google Scholar
136. Makarem, N, Chandran, U, Bandera, EV et al. (2013) Dietary fat in breast cancer survival. Annu Rev Nutr 33, 319348.Google Scholar
137. Sczaniecka, AK, Brasky, TM, Lampe, JW et al. (2012) Dietary intake of specific fatty acids and breast cancer risk among postmenopausal women in the VITAL cohort. Nutr Cancer 64, 11311142.Google Scholar
138. Brasky, TM, Darke, AK, Song, X et al. (2013) Plasma phospholipid fatty acids and prostate cancer risk in the SELECT trial. J Natl Cancer Inst 105, 11321141.Google Scholar
139. Crowe, FL, Appleby, PN, Travis, RC et al. (2014) Circulating fatty acids and prostate cancer risk: individual participant meta-analysis of prospective studies. J Natl Cancer Inst 106, dju240.Google Scholar
140. Elia, M, Van Bokhorst-de van der Schueren, MA, Garvey, J et al. (2006) Enteral (oral or tube administration) nutritional support and eicosapentaenoic acid in patients with cancer: a systematic review. Int J Oncol 28, 523.Google Scholar
141. Alfano, CM, Imayama, I, Neuhouser, ML et al. (2012) Fatigue, inflammation, and ω−3 and ω−6 fatty acid intake among breast cancer survivors. J Clin Oncol 30, 12801287.Google Scholar
142. Cerchietti, LC, Navigante, AH & Castro, MA (2007) Effects of eicosapentaenoic and docosahexaenoic n-3 fatty acids from fish oil and preferential Cox-2 inhibition on systemic syndromes in patients with advanced lung cancer. Nutr Cancer 59, 1420.Google Scholar
143. van der Meij, BS, Langius, JA, Spreeuwenberg, MD et al. (2012) Oral nutritional supplements containing n-3 polyunsaturated fatty acids affect quality of life and functional status in lung cancer patients during multimodality treatment: an RCT. Eur J Clin Nutr 66, 399404.Google Scholar
144. Sala-Vila, A, Folkes, J & Calder, PC (2010) The effect of three lipid emulsions differing in fatty acid composition on growth, apoptosis and cell cycle arrest in the HT-29 colorectal cancer cell line. Clin Nutr 29, 519524.Google Scholar
145. Murphy, RA, Mourtzakis, M, Chu, QS et al. (2011) Supplementation with fish oil increases first-line chemotherapy efficacy in patients with advanced nonsmall cell lung cancer. Cancer 117, 37743780.Google Scholar
146. Bougnoux, P, Hajjaji, N, Ferrasson, MN et al. (2009) Improving outcome of chemotherapy of metastatic breast cancer by docosahexaenoic acid: a phase II trial. Brit J Cancer 101, 19781985.Google Scholar
147. Weed, HG, Ferguson, ML, Gaff, RL et al. (2011) Lean body mass gain in patients with head and neck squamous cell cancer treated perioperatively with a protein- and energy-dense nutritional supplement containing eicosapentaenoic acid. Head Neck 33, 10271033.Google Scholar
148. Murphy, RA, Mourtzakis, M, Chu, QS et al. (2011) Nutritional intervention with fish oil provides a benefit over standard of care for weight and skeletal muscle mass in patients with nonsmall cell lung cancer receiving chemotherapy. Cancer 117, 17751782.Google Scholar
149. Fearon, KC, Barber, MD, Moses, AG et al. (2006) Double-blind, placebo-controlled, randomized study of eicosapentaenoic acid diester in patients with cancer cachexia. J Clin Oncol 24, 34013407.Google Scholar
150. Guarcello, M, Riso, S, Buosi, R et al. (2007) EPA-enriched oral nutritional support in patients with lung cancer: effects on nutritional status and quality of life. Nutr Therap Metab 25, 2530.Google Scholar
151. Calder, PC, Albers, R, Antoine, JM et al. (2009) Inflammatory disease processes and interactions with nutrition. Brit J Nutr 101, Suppl. 1, S145.Google Scholar
152. Calder, PC, Ahluwalia, N, Albers, R et al. (2013) A consideration of biomarkers to be used for evaluation of inflammation in human nutritional studies. Brit J Nutr 109, Suppl. 1, S1S34.Google Scholar
153. Ross, R (1999) Atherosclerosis--an inflammatory disease. N Engl J Med 340, 115126.Google Scholar
154. Hansson, GK & Hermansson, A (2011) The immune system in atherosclerosis. Nat Immunol 12, 204212.Google Scholar
155. Calder, PC, Ahluwalia, N, Brouns, F et al. (2011) Dietary factors and low-grade inflammation in relation to overweight and obesity. Brit J Nutr 106, Suppl. 3, S5S78.Google Scholar
156. Colotta, F, Allavena, P, Sica, A et al. (2009) Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 30, 10731081.Google Scholar
157. Miles, EA & Calder, PC (2012) Influence of marine n-3 polyunsaturated fatty acids on immune function and a systematic review of their effects on clinical outcomes in rheumatoid arthritis. Brit J Nutr 107, Suppl. 2, S171S184.Google Scholar
158. Calder, PC (2009) Fatty acids and immune function: relevance to inflammatory bowel diseases. Int Rev Immunol 28, 506534.CrossRefGoogle ScholarPubMed
159. Calder, PC (2006) n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr 83, 1505S1519S.Google Scholar
160. Abdulrazaq, M, Innes, JK & Calder, PC (2017) Effect of ω−3 polyunsaturated fatty acids on arthritic pain: a systematic review. Nutrition 39–40, 5766.Google Scholar
161. Senftleber, NK, Nielsen, SM, Andersen, JR et al. (2017) Marine oil supplements for arthritis pain: a systematic review and meta-analysis of randomized trials. Nutrients 9, 42.Google Scholar
162. Calder, PC, Kremmyda, LS, Vlachava, M et al. (2010) Is there a role for fatty acids in early life programming of the immune system? Proc Nutr Soc 69, 373380.Google Scholar
163. Dunstan, JA, Mori, TA, Barden, A et al. (2003) Maternal fish oil supplementation in pregnancy reduces interleukin-13 levels in cord blood of infants at high risk of atopy. Clin Exp Allergy 33, 442448.Google Scholar
164. Dunstan, JA, Mori, TA, Barden, A et al. (2003) Fish oil supplementation in pregnancy modifies neonatal allergen-specific immune responses and clinical outcomes in infants at high risk of atopy: a randomized, controlled trial. J Allergy Clin Immunol 112, 11781184.Google Scholar
165. Noakes, PS, Vlachava, M, Kremmyda, LS et al. (2012) Increased intake of oily fish in pregnancy: effects on neonatal immune responses and on clinical outcomes in infants at 6 mo. Am J Clin Nutr 95, 395404.Google Scholar
166. Furuhjelm, C, Warstedt, K, Larsson, J et al. (2009) Fish oil supplementation in pregnancy and lactation may decrease the risk of infant allergy. Acta Paediatr 98, 14611467.CrossRefGoogle ScholarPubMed
167. Palmer, DJ, Sullivan, T, Gold, MS et al. (2012) Effect of n-3 long chain polyunsaturated fatty acid supplementation in pregnancy on infants’ allergies in first year of life: randomised controlled trial. Brit Med J 344, e184.Google Scholar
168. Best, KP, Gold, M, Kennedy, D et al. (2016) Omega-3 long-chain PUFA intake during pregnancy and allergic disease outcomes in the offspring: asystematic review and meta-analysis of observational studies and randomized controlled trials. Am J Clin Nutr 103, 128143.Google Scholar
169. Bisgaard, H, Stokholm, J, Chawes, BL et al. (2016) Fish oil-derived fatty acids in pregnancy and wheeze and asthma in offspring. N Engl J Med 375, 25302539.Google Scholar
170. D'Vaz, N, Meldrum, SJ, Dunstan, JA et al. (2012) Fish oil supplementation in early infancy modulates developing infant immune responses. Clin Exp Allergy 42, 12061216.Google Scholar
171. Willemsen, LEM (2016) Dietary n-3 long chain polyunsaturated fatty acids in allergy prevention and asthma treatment. Eur J Pharmacol 785, 174186.Google Scholar
172. Klemens, CM, Berman, DR & Mozurkewich, EL (2011) The effect of perinatal omega-3 fatty acid supplementation on inflammatory markers and allergic disease: a systematic review. Brit J Obstet Gynaecol 118, 916925.Google Scholar
173. Miles, EA & Calder, PC (2017) Can early omega-3 fatty acid exposure reduce risk of childhood allergic disease? Nutrients 9, 784.Google Scholar
174. Lauritzen, L, Hansen, HS, Jorgensen, MH et al. (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, 194.Google Scholar
175. Salem, N, Litman, B, Kim, HY et al. (2001) Mechanisms of action of docosahexaenoic acid in the nervous system. Lipids 36, 945959.Google Scholar
176. Parletta, N, Milte, CM & Meyer, BJ (2013) Nutritional modulation of cognitive function and mental health. J Nutr Biochem 24, 725743.Google Scholar
177. Uauy, RD, Birch, DG, Birch, EE et al. (1990) Effect of dietary omega-3 fatty acids on retinal function of very-low-birth-weight neonates. Pediatr Res 28, 485492.Google Scholar
178. Birch, DG, Birch, EE, Hoffman, DR et al. (1992) Retinal development in very-low-birth-weight infants fed diets differing in omega-3 fatty acids. Invest Opthalmol Vis Sci 33, 23652376.Google Scholar
179. Birch, EE, Birch, DG, Hoffman, DR et al. (1992) Dietary essential fatty acid supply and visual acuity development. Invest Opthalmol Vis Sci 33, 32423253.Google Scholar
180. Carlson, SE, Werkman, SH, Rhodes, PG et al. (1993) Visual acuity development in healthy preterm infants: effect of marine oil supplementation. Am J Clin Nutr 58, 3542.Google Scholar
181. Carlson, SE, Ford, AJ, Werkman, SH et al. (1996) Visual acuity and fatty acid status of term infants fed human milk and formulas with and without docosahexaenoate and arachidonate from egg yolk lecithin. Pediatr Res 39, 882888.Google Scholar
182. Carlson, SE, Werkman, SH & Tolley, EA (0000) Effect of long-chain n-3 fatty acid supplementation on visual acuity and growth of preterm infants with and without bronchopulmonary dysplasia. Am J Clin Nutr 63, 687697.Google Scholar
183. Moon, K, Rao, SC, Schulzke, SM et al. (2016) Long-chain polyunsaturated fatty acid supplementation in preterm infants. Cochrane Database Syst Rev 12, CD000375.Google Scholar
184. Molloy, C, Doyle, LW, Makrides, M et al. (2012) Docosahexaenoic acid and visual functioning in preterm infants: a review. Neuropsychol Rev 22, 425437.Google Scholar
185. Agostoni, C, Trojan, S, Bellu, R et al. (1995) Neurodevelopmental quotient of healthy term infants at 4 months and feeding practice: the role of long-chain polyunsaturated fatty acids. Pediatr Res 38, 262266.Google Scholar
186. Willatts, P, Forsyth, JS, DiModugno, MK et al. (1998) Effect of long-chain polyunsaturated fatty acids in infant formula on problem solving at 10 months of age. Lancet 352, 688691.Google Scholar
187. Scott, DT, Janowsky, JS, Carroll, RE et al. (1998) Formula supplementation with long-chain polyunsaturatedfatty acids: are there developmental benefits? Pediatrics 102, E59.Google Scholar
188. Makrides, M, Neumann, MA, Simmer, K et al. (2000) A critical appraisal of the role of dietary long-chain polyunsaturated fatty acids on neural indices of term infants: a randomized, controlled trial. Pediatrics 105, 3238.Google Scholar
189. Auested, N, Halter, R, Hall, RT et al. (2001) Growth and development of term infants fed long-chain polyunsaturated fatty acids: a double-masked, randomized, parallel, prospective, multivariate study. Pediatrics 108, 272381.Google Scholar
190. Auested, N, Scott, DT, Janowsky, JS et al. (2003) Visual, cognitive, and language assessments at 39 months: a follow-up study of children fed formulas containing long-chain polyunsaturated fatty acids to 1 year of age. Pediatrics 112, e177e183.Google Scholar
191. Agostoni, C, Trojan, S, Bellu, R et al. (1997) Developmental quotient at 24 months and fatty acid composition of diet in early infancy: a follow up study. Arch Dis Child 1997, 76, 421424.Google Scholar
192. Jasani, B, Simmer, K, Patole, SK et al. (2017) Long chain polyunsaturated fatty acid supplementation in infants born at term. Cochrane Database Syst Rev 3, CD000376.Google Scholar
193. Gould, JF, Smithers, LG & Makrides, M (2013) The effect of maternal omega-3 (n-3) LCPUFA supplementation during pregnancy on early childhood cognitive and visual development; a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 97, 531544.CrossRefGoogle ScholarPubMed
194. Richardson, AJ (2004) Clinical trials of fatty acid treatment in ADHD, dyslexia, dyspraxia and the autistic spectrum. Prostagland Leuk Essent Fatty Acids 70, 383390.Google Scholar
195. Stevens, L, Zhang, W, Peck, L et al. (2003) EFA supplementation in children with inattention, hyperactivity, and other disruptive behaviors. Lipids 38, 10071021.CrossRefGoogle ScholarPubMed
196. Richardson, AJ & Puri, BK (2002) A randomized double-blind, placebo-controlled study of the effects of supplementation with highly unsaturated fatty acids on ADHD-related symptoms in children with specific learning difficulties. Prog Neuropsychopharmacol Biol Psych 26, 233239.Google Scholar
197. Yui, K, Koshiba, M, Nakamura, S et al. (2012) Effects of large doses of arachidonic acid added to docosahexaenoic acid on social impairment in individuals with autism spectrum disorders: a double-blind, placebo-controlled, randomized trial. J Clin Psychopharmacol 32, 200206.Google Scholar
198. Meguid, NA, Atta, HM, Gouda, AS et al. (2008) Role of polyunsaturated fatty acids in the management of Egyptian children with autism. Clin Biochem 41, 10441048.Google Scholar
199. Perera, H, Jeewandara, KC, Seneviratne, S et al. (2012) Combined ω3 and ω6 supplementation in children with attention-deficit hyperactivity disorder (ADHD) refractory to methylphenidate treatment: a double-blind, placebo-controlled study. J Child Neurol 27, 747753.Google Scholar
200. Milte, CM, Parletta, N, Buckley, JD et al. (2012) Eicosapentaenoic and docosahexaenoic acids, cognition, and behavior in children with attention-deficit/hyperactivity disorder: a randomized controlled trial. Nutrition 28, 670677.Google Scholar
201. Gustafsson, PA, Birberg-Thornberg, U, Duchén, K et al. (2010) EPA supplementation improves teacher-rated behaviour and oppositional symptoms in children with ADHD. Acta Paediatr 99, 15401549.Google Scholar
202. Bélanger, SA, Vanasse, M, Spahis, S et al. (2009) Omega-3 fatty acid treatment of children with attention-deficit hyperactivity disorder: a randomized, double-blind, placebo-controlled study. Paediatr Child Health 14, 8998.CrossRefGoogle ScholarPubMed
203. Sorgi, PJ, Hallowell, EM, Hutchins, HL et al. (2007) Effects of an open-label pilot study with high-dose EPA/DHA concentrates on plasma phospholipids and behavior in children with attention deficit hyperactivity disorder. Nutr J 6, 16.Google Scholar
204. Bos, DJ, Oranje, B, Veerhoek, ES et al. (2015) Reduced symptoms of inattention after dietary omega-3 fatty acid supplementation in boys with and without attention deficit/hyperactivity disorder. Neuropsychopharmacol 40, 22982306.Google Scholar
205. Sheppard, KW, Boone, KM, Gracious, B et al. (2017) Effect of omega-3 and -6 supplementation on language in preterm toddlers exhibiting autism spectrum disorder symptoms. J Autism Dev Disord (In the Press).Google Scholar
206. Voigt, RG, Llorente, AM, Jensen, CL et al. (2001) A randomized, double-blind, placebo-controlled trial of docosahexaenoic acid supplementation in children with attention-deficit/hyperactivity disorder. J Pediatr 139, 189196.Google Scholar
207. Hirayama, S, Hamazaki, T & Terasawa, K (2004) Effect of docosahexaenoic acid-containing food administration on symptoms of attention-deficit/hyperactivity disorder - a placebo-controlled double-blind study. Eur J Clin Nutr 58, 467473.Google Scholar
208. Bent, S, Bertoglio, K, Ashwood, P et al. (2011) A pilot randomized controlled trial of omega-3 fatty acids for autism spectrum disorder. J Autism Dev Disord 41, 545554.Google Scholar
209. Politi, P, Cena, H, Comelli, M et al. (2008) Behavioral effects of omega-3 fatty acid supplementation in young adults with severe autism: an open label study. Arch Med Res 39, 682685.Google Scholar
210. Amminger, GP, Berger, GE, Schäfer, MR et al. (2007) Omega-3 fatty acids supplementation in children with autism: a double-blind randomized, placebo-controlled pilot study. Biol Psychiatry 61, 551553.Google Scholar
211. Johnson, M, Ostlund, S, Fransson, G et al. (2009) Omega-3/omega-6 fatty acids for attention deficit hyperactivity disorder: a randomized placebo-controlled trial in children and adolescents. J Atten Disord 12, 394401.Google Scholar
212. Raz, R, Carasso, RL & Yehuda, S (2009) The influence of short-chain essential fatty acids on children with attention-deficit/hyperactivity disorder: a double-blind placebo-controlled study. J Child Adolesc Psychopharmacol 19, 167177.Google Scholar
213. Voigt, RG, Mellon, MW, Katusic, SK et al. (2014) Dietary docosahexaenoic acid supplementation in children with autism. J Pediatr Gastroenterol Nutr 58, 715722.Google Scholar
214. Mankad, D, Dupuis, A, Smile, S et al. (2015) A randomized, placebo controlled trial of omega-3 fatty acids in the treatment of young children with autism. Mol Autism 6, 18.Google Scholar
215. Frensham, LJ, Bryan, J & Parletta, N (2012) Influences of micronutrient and omega-3 fatty acid supplementation on cognition, learning, and behaviour: methodological considerations and implications for children and adolescents in developed societies. Nutr Rev 70, 594610.Google Scholar
216. Horvath, A, Łukasik, J & Szajewska, H (2017) ω−3 Fatty acid supplementation does not affect autism spectrum disorder in children: a systematic review and meta-analysis. J Nutr 147, 367376.Google Scholar
217. Mazahery, H, Stonehouse, W, Delshad, M et al. (2017) Relationship between long chain n-3 polyunsaturated fatty acids and autism spectrum disorder: systematic review and meta-analysis of case-control and randomised controlled trials. Nutrients 9, 155.Google Scholar
218. Cooper, RE, Tye, C, Kuntsi, J et al. (2016) The effect of omega-3 polyunsaturated fatty acid supplementation on emotional dysregulation, oppositional behaviour and conduct problems in ADHD: a systematic review and meta-analysis. J Affect Disord 190, 474482.Google Scholar
219. Chang, JC, Su, KP, Mondelli, V et al. (2017) Omega-3 polyunsaturated fatty acids in youths with attention deficit hyperactivity disorder (ADHD): A systematic review and meta-analysis of clinical trials and biological studies. Neuropsychopharmacol (In the Press).Google Scholar
220. Derbyshire, E (2017) Do omega-3/6 fatty acids have a therapeutic role in children and young people with ADHD? J Lipids (In the Press).Google Scholar
221. Rudin, DO (1981) The major psychoses and neuroses as omega-3 essential fatty acid deficiency syndrome: substrate pellagra. Biol Psychiatry 16, 837850.Google Scholar
222. Hibbeln, JR (1998) Fish consumption and major depression. Lancet 351, 1213.Google Scholar
223. Su, KP, Huang, SY, Chiu, CC et al. (2003) Omega-3 fatty acids in major depressive disorder. A preliminary double-blind, placebo-controlled trial. Eur Neuropsychopharmacol 13, 267271.Google Scholar
224. Marangell, LB, Martinez, JM, Zboyan, HA et al. (2003) A double-blind, placebo-controlled study of the omega-3 fatty acid docosahexaenoic acid in the treatment of major depression. Am J Psychiatry 160, 996998.Google Scholar
225. Stoll, AL, Severus, WE, Freeman, MP et al. (1999) Omega 3 fatty acids in bipolar disorder: a preliminary double-blind, placebo-controlled trial. Arch Gen Psychiatry 56, 407412.Google Scholar
226. Nemets, B, Stahl, Z & Belmaker, RH (2002) Addition of omega-3 fatty acid to maintenance medication treatment for recurrent unipolar depressive disorder. Am J Psychiatry 159, 477479.Google Scholar
227. Mocking, RJ, Harmsen, I, Assies, J et al. (2016) Meta-analysis and meta-regression of omega-3 polyunsaturated fatty acid supplementation for major depressive disorder. Transl Psychiatry 6, e756.Google Scholar
228. Sublette, ME, Ellis, SP, Geant, AL et al. (2012) Meta-analysis of the effects of eicosapentaenoic acid (EPA) in clinical trials in depression. J Clin Psychiatry 72, 15771584.Google Scholar
229. Sarris, J, Mischoulon, D & Schweitzer, I (2012) Omega-3 for bipolar disorder: meta-analyses of use in mania and bipolar depression. J Clin Psychiatry 73, 8186.Google Scholar
230. Appleton, KM, Sallis, HM, Perry, R et al. (2016) ω−3 Fatty acids for major depressive disorder in adults: an abridged Cochrane review. BMJ Open 6, e010172.Google Scholar
231. Peet, M, Laugharne, J, Rangarajan, N et al. (1995) Depleted red cell membrane essential fatty acids in drug-treated schizophrenic patients. J Psychiatr Res 29, 227232.Google Scholar
232. Yao, JK, van Kammen, DP & Welker, JA (1994) Red blood cell membrane dynamics in schizophrenia. II. Fatty acid composition. Schizophr Res 13, 217226.Google Scholar
233. Glen, AI, Glen, EM, Horrobin, DF et al. (1994) A red cell membrane abnormality in a subgroup of schizophrenic patients: evidence for two diseases. Schizophr Res 12, 5361.Google Scholar
234. Edwards, R, Peet, M, Shay, J et al. (1998) Omega-3 polyunsaturated fatty acid levels in the diet and in red blood cell membranes of depressed patients. J Affect Disord 48, 149155.Google Scholar
235. Hoen, WP, Lijmer, JG, Duran, M et al. (2013) Red blood cell polyunsaturated fatty acids measured in red blood cells and schizophrenia: a meta-analysis. Psychiatry Res 207, 112.Google Scholar
236. Peet, M, Brind, J, Ramchand, CN et al. (2001) Two double-blind placebo-controlled pilot studies of eicosapentaenoic acid in the treatment of schizophrenia. Schizophr Res 49, 243251.Google Scholar
237. Peet, M & Horrobin, DF (2002) A dose-ranging study of the effects of ethyl-eicosapentaenoate in patients with ongoing depression despite apparently adequate treatment with standard drugs. Arch Gen Psychiatry 59, 913919.Google Scholar
238. Fenton, WS, Dickerson, F, Boronow, J et al. (2001) A placebo-controlled trial of omega-3 fatty acid (ethyl eicosapentaenoic acid) supplementation for residual symptoms and cognitive impairment in schizophrenia. Am J Psychiatry 158, 20712074.Google Scholar
239. Emsley, R, Myburgh, C, Oosthuizen, P et al. (2002) Randomized, placebo-controlled study of ethyl-eicosapentaenoic acid as supplemental treatment in schizophrenia. Am J Psychiatry 159, 15961598.Google Scholar
240. Pawełczyk, T, Grancow-Grabka, M, Kotlicka-Antczak, M et al. (2016) A randomized controlled study of the efficacy of six-month supplementation with concentrated fish oil rich in omega-3 polyunsaturated fatty acids in first episode schizophrenia. J Psychiatr Res 73, 3444.Google Scholar
241. Joy, CB, Mumby-Croft, R & Joy, LA (2006) Polyunsaturated fatty acid supplementation for schizophrenia. Cochrane Database Syst Rev 3, CD001257.Google Scholar
242. Zanarini, MC & Frankenburg, FR (2003) N-3 fatty acid treatment of women with borderline personality disorder: a double-blind, placebo-controlled pilot study. Am J Psychiatry 160, 167169.Google Scholar
243. Hamazaki, T, Sawazaki, S, Itomura, M et al. (1996) The effect of docosahexaenoic acid on aggression in young adults. A placebo-controlled double-blind study. J Clin Invest 97, 11291133.Google Scholar
244. Hamazaki, T, Thienprasert, A, Kheovichai, K et al. (2002) The effect of docosahexaenoic acid on aggression in elderly Thai subjects--a placebo-controlled double-blind study. Nutr Neurosci 5, 3741.Google Scholar
245. Prasad, MR, Lovell, MA, Yatin, M et al. (1998) Regional membrane phospholipid alterations in Alzheimer's disease. Neurochem Res 23, 8188.Google Scholar
246. Tully, AM, Roche, HM, Doyle, R et al. (2003) Low serum cholesteryl ester-docosahexaenoic acid levels in Alzheimer's disease: a case-control study. Brit J Nutr 89, 483489.Google Scholar
247. Cunnane, SC, Schneider, JA, Tangney, C et al. (2012) Plasma and brain fatty acid profile in mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis 29, 691697.Google Scholar
248. Soderberg, M, Edlund, C, Kristensson, K et al. (1991) Fatty acid composition of brain phospholipids in aging and in Alzheimer's disease. Lipids 26, 421425.Google Scholar
249. Conquer, JA, Tierney, MC, Zecevic, J et al. (2000) Fatty acid analysis of blood plasma of patients with Alzheimer's disease, other types of dementia, and cognitive impairment. Lipids 35, 13051312.Google Scholar
250. Sinn, N, Milte, CM, Street, SJ et al. (2012) Effects of n-3 fatty acids, EPA v. DHA, on depressive symptoms, quality of life, memory and executive function in older adults with mild cognitive impairment: a 6-month randomised controlled trial. Brit J Nutr 107, 16821693.Google Scholar
251. Scheltens, P, Twisk, JW, Blesa, R et al. (2012) Efficacy of Souvenaid in mild Alzheimer's disease: results from a randomized, controlled trial. J Alzheimers Dis 31, 225236.Google Scholar
252. Quinn, JF, Raman, R, Thomas, RG et al. (2010) Docosahexaenoic acid supplementation and cognitive decline in Alzheimer disease: a randomized trial. J Am Med Assoc 304, 19031911.Google Scholar
253. Freund-Levi, Y, Eriksdotter-Jönhagen, M, Cederholm, T et al. (2006) Omega-3 fatty acid treatment in 174 patients with mild to moderate Alzheimer disease: OmegAD study: a randomized double-blind trial. Arch Neurol 63, 14021408.Google Scholar
254. Freund-Levi, Y, Basun, H, Cederholm, T et al. (2008) Omega-3 supplementation in mild to moderate Alzheimer's disease: effects on neuropsychiatric symptoms. Int J Geriatr Psychiatry 23, 161169.Google Scholar
255. Kotani, S, Sakaguchi, E, Warashina, S et al. (2006) Dietary supplementation of arachidonic and docosahexaenoic acids improves cognitive dysfunction. Neurosci Res 56, 159164.Google Scholar
256. Boston, PF, Bennett, A, Horrobin, DF et al. (2204) Ethyl-EPA in Alzheimer's disease - a pilot study. Prostagland Leukot Essent Fatty Acids 71, 341346.Google Scholar
257. Phillips, MA, Childs, CE, Calder, PC et al. (2015) No effect of omega-3 fatty acid supplementation on cognition and mood in individuals with cognitive impairment and probable Alzheimer's disease: a randomised controlled trial. Int J Mol Sci 16, 2460024613.Google Scholar
258. Sydenham, E, Dangour, AD & Lim, WS (2012) Omega 3 fatty acid for the prevention of cognitive decline and dementia. Cochrane Database Syst Rev 6, CD005379.Google Scholar
259. Zhang, XW, Hou, WS, Li, M et al. (2016) Omega-3 fatty acids and risk of cognitive decline in the elderly: a meta-analysis of randomized controlled trials. Aging Clin Exp Res 28, 165166.CrossRefGoogle ScholarPubMed
260. Burckhardt, M, Herke, M, Wustmann, T et al. (2016) Omega-3 fatty acids for the treatment of dementia. Cochrane Database Syst Rev 4, CD009002.Google Scholar
261. Andrieu, S, Guyonnet, S, Coley, N et al. (2017) 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 16, 377389.Google Scholar
262. Simopolous, AP, Leaf, A & Salem, N (1999) Essentiality and recommended dietary intakes for omega-6 and omega-3 fatty acids. Ann Nutr Metab 43, 127130.Google Scholar
263. International Society for the Study of Fatty Acids and Lipids (2004) Recommendations for the intake of polyunsaturated fatty acids in healthy adults (ISSFAL Policy Statement 3). http://www.issfal.org/news-links/resources/publications/PUFAIntakeReccomdFinalReport.pdf.Google Scholar
264. French Agency for Food, Environmental and Occupational Health Safety (2003) The omega-3 fatty acids and the cardiovascular system: nutritional benefits and claims. http://www.afssa.fr?Documents?NUT-Ra-omega3EN.pdf.Google Scholar
265. National Health and Medical Research Council (2006) Nutrient reference values for Australia and New Zealand including recommended dietary intakes. http://www.nhmrc.gov.au/publications/synopses/n35syn.htm.Google Scholar
266. Food and Agricultural Organisation of the United Nations (2010) Fat and Fatty Acids in Human Nutrition: report of an Expert Consultation. Food and Agricultural Organisation of the United Nations, Rome.Google Scholar
267. European Food Safety Authority (2010) Scientific opinion on dietary reference values for fats, including saturated fatty acids, polyunsaturated fatty acids, monounsaturated fatty acids, trans fatty acids and cholesterol. EFSA J 8, 1461.Google Scholar
268. Mori, TA & Woodman, RJ (2006) The independent effects of eicosapentaenoic acid and docosahexaenoic acid on cardiovascular risk factors in humans. Curr Opin Clin Nutr Metab Care 9, 95104.Google Scholar
269. Wei, MY & Jacobson, TA (2011) Effects of eicosapentaenoic acid versus docosahexaenoic acid on serum lipids: a systematic review and meta-analysis. Curr Atheroscler Rep 13, 474483.Google Scholar
270. Schuchardt, JP & Hahn, A (2013) Bioavailability of long-chain omega-3 fatty acids. Prostaglandins Leukot Essent Fatty Acids 89, 18.Google Scholar
271. Ghasemifard, S, Turchini, GM & Sinclair, AJ (2014) Omega-3 long chain fatty acid ‘bioavailability’: a review of evidence and methodological considerations. Prog Lipid Res 56, 92108.Google Scholar
272. Raatz, SK, Johnson, LK & Bukowski, MR (2016) Enhanced bioavailability of EPA from emulsified fish oil preparations versus capsular triacylglycerol. Lipids 51, 643651.Google Scholar
273. Ruiz-Lopez, N, Usher, S, Sayanova, OV et al. (2015) Modifying the lipid content and composition of plant seeds: engineering the production of LC-PUFA. Appl Microbiol Biotechnol 99, 143154.Google Scholar
274. Walker, CG, Jebb, SA & Calder, PC (2013) Stearidonic acid as a supplemental source of ω−3 polyunsaturated fatty acids to enhance status for improved human health. Nutrition 29, 363369.Google Scholar
275. James, MJ, Ursin, VM & Cleland, LG (2003) Metabolism of stearidonic acid in human subjects: comparison with the metabolism of other n-3 fatty acids. Am J Clin Nutr 77, 11401145.Google Scholar
Figure 0

Fig. 1. The metabolic pathway of biosynthesis of EPA, docosapentaenoic acid and DHA.

Figure 1

Table 1. Typical content of EPA, docosapentaenoic acid (DPA) and DHA (g/100 g food) in a selection of seafood and meat

Figure 2

Fig. 2. Typical intake of EPA + DHA from the background diet in an adult not regularly consuming fatty fish and what would be achieved by also consuming a 1 g standard fish oil (FO) capsule, a 1 g ‘concentrated’ supplement, one teaspoon of cod liver oil, one meal of salmon, or one or four capsules of the pharmaceutical grade preparation Omacor®. Reproduced from ref. 3 with permission from John Wiley and Sons.

Figure 3

Table 2. Typical EPA and DHA contents of n-3 supplements

Figure 4

Table 3. Typical EPA and DHA concentrations reported in different lipid pools in human subjects

Figure 5

Fig. 3. General overview of the mechanisms by which very long-chain n-3 fatty acids can influence the function of cells. Modified from Calder(72), Copyright (2011), with permission from Elsevier.

Figure 6

Fig. 4. Overview of the bioactive lipid mediators produced from arachidonic acid, EPA and DHA.

Figure 7

Table 4. Summary of the effects of very long-chain n-3 fatty acids on risk factors for CVD identified through the meta-analysis of AbuMweis et al.(102)

Figure 8

Table 5. Some of the current recommendations for intake of very long-chain n-3 fatty acids