Next Article in Journal
The Nutritional and Health Effects of the COVID-19 Pandemic on Patients with Diabetes Mellitus
Next Article in Special Issue
The Legacy Effect in the Prevention of Cardiovascular Disease
Previous Article in Journal
The Inhibitory Effect of Extra Virgin Olive Oil and Its Active Compound Oleocanthal on Prostaglandin-Induced Uterine Hypercontraction and Pain—Ex Vivo and In Vivo Study
Previous Article in Special Issue
Dietary Strategies for Metabolic Syndrome: A Comprehensive Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Very Long Chain Marine n-3 Polyunsaturated Fatty Acids in Atherothrombotic Heart Disease. A Brief Review, with a Focus on Metabolic Effects

by
Harald Arnesen
1,2,
Peder L. Myhre
2,3 and
Ingebjørg Seljeflot
1,2,*
1
Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Pb 4956 Nydalen, 0424 Oslo, Norway
2
Faculty of Medicine, University of Oslo, 0424 Oslo, Norway
3
Division of Medicine, Department of Cardiology, Akershus University Hospital, 1478 Lørenskog, Norway
*
Author to whom correspondence should be addressed.
Nutrients 2020, 12(10), 3014; https://doi.org/10.3390/nu12103014
Submission received: 28 August 2020 / Revised: 23 September 2020 / Accepted: 29 September 2020 / Published: 30 September 2020
(This article belongs to the Special Issue Dietary Intakes and Metabolic Disorders)

Abstract

:
The global burden of atherothrombotic heart disease should be considered as a life-style disorder where differences in dietary habits and related risk factors like limited physical activity and adiposity together play important roles. Related metabolic changes have been scientifically elucidated in recent decades, and the role of the very-long-chain marine fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been much focused on, especially their possible effects on processes like inflammation and thrombosis. In the present brief review of related metabolic mechanisms, the effects of these fatty acids in a clinical setting have been referred to, including some of the authors’ work on this topic. The main focus is the divergent results in the field and the important differences between the study population, the type of supplements and fresh marine sources, the proportion of EPA versus DHA dosages, and the duration of supplementation in clinical trials. We conclude that daily intake of at least 1 g of EPA + DHA may improve a dysmetabolic state in the population. The potential to reduce the risk and progression of atherothrombotic heart disease is still a matter of debate.

1. Background

Atherothrombotic heart disease is a dominating cause of morbidity and mortality worldwide [1,2]. Atherothrombotic heart disease comprises the continuous process of atherosclerosis, leading to narrowing of the coronary arteries and possibly the clinical manifestation angina pectoris, and the acute thrombotic occlusion of the artery as a basis for the life-threatening myocardial infarction [3,4]. Lifestyle factors like dietary habits have been highlighted as risk factors for atherothrombosis [5,6,7]. Preventive approaches like achievable dietary changes are obviously of major importance both for health and economic reasons. The importance of increased intake of very-long-chain n-3 polyunsaturated fatty acids (VLCM n-3 PUFAs), especially from marine sources, has been discussed over the last 50 years, back to the observations on Greenland Eskimos by the Danish scientists Dyerberg and Bang [8,9]. They found that the occurrence of coronary heart disease was very low among the Eskimos compared to the regular Danish population, and they attributed this observation to their special diet, which was composed mainly of seal and whale and was extremely rich in marine n-3 FAs. Later, a series of mechanisms for the potentially beneficial effects of such VLCM n-3 PUFA were published [10,11,12,13,14,15].
Relevant biologically active families of polyunsaturated FAs are the n-6 and n-3 FAs that contribute to several important metabolic pathways. However, as mammals lack enzymes to insert double bonds in the n-6 and n-3 position, linoleic acid (LA) (18:2 n-6) and alfa-linolenic acid (ALA) (18:3 n-3) are essential nutrients. In addition, the elongation and desaturation of these FAs to the metabolically important VLCM PUFAs are limited, and the n-3 and n-6 series are competing for the same enzymes, the elongases, and the desaturases (Figure 1). The metabolic key FAs in the two series are arachidonic acid (AA) (20:4 n-6) in the n-6 series and eicosapentaenoic acid (EPA) (20:5 n-3) and docosahexaenoic acid (DHA) (22:6 n-3) in the n-3 series [12,13,15].
In regular Western diet today, the content of LA (from green plants and red meat) is much higher than that of ALA. Thus, the competition for elongases and desaturases favors the synthesis of the n-6 series, i.e., mainly AA, which is metabolized into mainly pro-inflammatory and platelet activating derivatives. Therefore, the synthesis of the very-long-chain EPA and DHA are minimal, especially when the diet is relatively low in ALA [16,17]. Derivatives from EPA/DHA seem to be rather neutral with respect to inflammation and platelet activation, but contribute importantly to the resolution of inflammation by being so-called resolvins [13,18].
In recent decades, in addition to epidemiologic and registry studies, a series of observational and randomized clinical trials on the possible clinical effect of intake (dietary or as supplements) of VLCM n-3 PUFA have been conducted [13,14,15]. Additionally, the effects on in vivo VLCM n-3 PUFA in the circulation, adipose tissue, and erythrocyte membrane have been extensively studied [19,20,21]. The results are divergent, mainly depending on (i) the population studied, i.e., baseline health status and risk factors, primary versus secondary prevention, and background dietary intake of n-3 and n-6 FAs; (ii) the type of supplementation (fatty fish, cod liver oil, seal oil, or capsules); (iii) duration of supplementation; (iv) dosage of supplementation; (v) proportion of EPA and DHA; and (vi) the quality of capsule content.
As in all therapeutic trials, compliance with the investigational principle is relevant to assess the validity of the results. Dietary questionnaires are often used to characterize the diet, but are generally inadequate for scientific purposes, although validated scoring systems are available. In studies using supplementary capsules, “pill count” is often used as a measure of compliance. However, this approach has important scientific limitations relating to logistics and patient reliability. In addition, supplementary capsules are prone to peroxidation of the unsaturated FAs, although often add anti-oxidants. Thus, in vivo measurements of the relevant FAs in the circulation, erythrocyte membrane, and adipose tissue are considered to be the scientifically most reliable methods. This strategy also allows for the estimation of relevant indexes (EPA + DHA/total FAs % and n-6/n-3 ratios) [21,22,23].
It should be emphasized that the unsaturated FAs are prone to the peroxidation and saturation of their double bonds, potentially reducing their beneficial effects in atherosclerotic cardiovascular disease states [24,25]. This is often the case with commercial supplements of VLCM n-3 PUFAs if not enriched with antioxidants like vitamin E. Fresh marine fish products do not experience the same risk of peroxidation.

2. VLCM n-3 FA, Metabolic Disorders and Atherothrombotic Heart Disease

The metabolic syndrome is composed of the most important metabolic disorders related to atherothrombotic heart disease. It describes a cluster of risk factors for cardiovascular disease states (CVD) and diabetes type 2, including abdominal obesity (waist circumference ≥ 102 (men)/88 cm (women)), elevated fasting blood glucose (≥5.6 mmol/L), hypertension (blood pressure ≥ 130/85 or on treatment), elevated triglycerides (≥1.7 mmol/L), and low levels of HDL cholesterol (<1.0 (men)/1.3 (women) mmol/L), according to the scientific statement from the American Heart Association and National Heart, Lung, and Blood Institute [26].
Among these risk factors, triglycerides and hypertension have been given most attention as relevant targets for potential effects of the VLCM n-3 PUFA. Regarding triglycerides, fasting samples are most frequently used, although non-fasting values have lately been used in epidemiological studies [27]. Triglycerides have been repeatedly shown to be predictive of future CVD, and their importance in risk prediction has increased in parallel with the increased prevalence of the metabolic syndrome in the Western world. Already in 1992, this effect was demonstrated in two large population studies from Germany and Finland [28,29], and later was documented in several populations with long-term follow-up, like the PROCAM study [30]; the Oslo Study [31]; and, more recently, the Copenhagen Study [27].
Furthermore, the triglyceride-lowering effect of the VLCM n-3 PUFAs has been observed in numerous clinical trials and is considered a key pathway in preventing CVD. The mechanism by which these VLCM n-3 PUFAs reduce triglycerides is probably by reduced hepatic synthesis and secretion [32], and reduction of triglycerides has often been used as a measure of compliance in clinical trials. The dose-relationship is well known and is evident in the various clinical trials using a range of VLCM n-3 PUFA doses. In the GISSI-Prevenzione trial [33] using a low dose (0.85 g/day), the reduction in triglycerides was only 3.4% over 5.5 years, whereas in the CART study [34] with a very high dose of 5.1 g/day, the reduction over 1 year was 27%. In the SHOT [35] and DOIT [36] studies, relatively high doses of 3.4 g/day and 2.4 g/day, respectively, were used, resulting in 19 and 16% triglyceride reduction, respectively. In the recent REDUCE-IT study [37], the reduction with 4 g/day of icosapent ethyl (an ethyl-EPA that is metabolized to EPA after ingestion) over 4.9 years was 18%. In the latter study, the reduction of cardiovascular risk could not be explained solely by the reduction in triglycerides, suggesting other cardio-protective mechanisms from high-dose EPA [38]. In the 2019 European Society of Cardiology/European Atherosclerosis Society guidelines VLCM n-3 PUFA has a IIa recommendation for lowering triglycerides [39]. High doses of the VLCM n-3 PUFA are recommended for very high levels of triglycerides, mainly to avoid pancreatitis [40]. Whether even higher doses, e.g., 8–12 g/day would have even better longstanding effects is unclear, but at these doses bowel dysfunction may be a challenge.
In lipidology, there is a well-known inverse relationship between triglycerides and HDL-cholesterol, although this may differ between subtypes of HDL-cholesterol. Thus, with high levels of triglycerides low levels of HDL-cholesterol are regularly found. In studies on supplementation with VLCM n-3 PUFAs, the reduction in triglycerides is regularly accompanied by a moderate increase in HDL-cholesterol. A more specific effect on the synthesis or secretion of the latter has, however, not been clearly elucidated.
With respect to hypertension, the VLCM n-3 PUFAs have been shown to reduce both the systolic and diastolic blood pressure. Already in 1990, Bønaa et al. [41] could demonstrate in a randomized, control trial in 156 hypertensive men and women, a significant, although moderate, reduction of both SBP and DBP after six weeks supplementation with 5.1 g/day, and the effect was related to the measured increase in plasma n-3 FAs. Similar effects have later been demonstrated in larger meta-analyses [20,42].
In the metabolic syndrome, hyperglycemia and insulin resistance are linked to abdominal adiposity and a pro-inflammatory state in the adipose tissue (and in general). In murine and human studies investigating supplementation of VLCM n-3 PUFAs, a tendency to change from a pro-inflammatory state into an anti-inflammatory state of the adipose tissue has been shown [43,44]. Thus, a transition of macrophages from the pro-inflammatory M1-type to the anti- inflammatory M2-type in response to VLCM n-3 PUFAs has been described [45]. In vitro cellular studies have demonstrated that these metabolic effects are partly mediated by peroxisome–proliferator–activator receptor (PPAR) transcription factors, typically with two receptor subtypes, PPARα and PPARγ. Whereas PPARα is also thought to mediate the beneficial effect of VLCM n-3 PUFAs on hepatic insulin sensitivity, PPARγ mediates the effect on adiponectin upregulation [44]. Both subtypes are also known to counteract the activation of the key proinflammatory transcription factor NFkB [12,13,15,44,46] (Figure 2).
Another rapidly evolving area has been the role of micro-RNAs (miRNAs) in the inflammatory balance in white adipose tissue, and their possible anti-inflammatory role, also related to the presence of insulin resistance [43]. Protective effects of VLCM n-3 PUFAs has also been linked to certain miRNAs, and the importance of the ratio between n-6 and n-3 FAs in the diet, as well as in circulation and adipose tissue, has been emphasized [43].
Regarding adiposity per se, especially abdominal adiposity, the influence of VLCM n-3 PUFAs and the n-6/n-3 ratio has mainly been focused on the pro-inflammatory state and insulin resistance. It emerges from both animal and human studies that the modern Western diet with increased n-6/n-3 ratio is associated with increased incidence of adiposity, and increased pro-inflammatory activity in the adipose tissue, increased insulin resistance, and increased risk of type 2 diabetes. Results from studies with supplementation of VLCM n-3 PUFAs in the diet or as purified supplements indicate that reducing the n-6/n-3 ratio counteracts adiposity and normalizes the dysmetabolic state in the metabolic syndrome [43,46,47,48,49]. This effect may be linked to the anti-inflammatory properties of VLCM n-3 PUFAs by their role in resolution of inflammation (Figure 3).
Additionally, the possible importance of polymorphisms in the regulation of fatty acids desaturases (FADS) for the n-6/n-3 ratio has been focused on [50]. Specific clustering of these polymorphisms on chromosome 11 seems further to be related to sex and compliance with a Mediterranean diet.

3. VLCM n-3 PUFAs Role in Cardiovascular Risk Assessement

Since the Diet and Reinfarction Trial (DART) in 1989 found a 29% reduction in 2-year mortality associated with increased fish intake (fatty fish twice per week) in approximately 2000 post-MI patients [51], several randomized clinical trials have investigated the effect of VLCM n-3 PUFA in reducing cardiovascular risk. Most studies have been performed with use of supplements. The GISSI-Prevenzione trial, also a secondary prophylaxis study, demonstrated in 1999 a significant reduction in all-cause mortality (21%) and 45% reduction in sudden cardiac death in ~11,000 patients randomized to 0.85 g EPA + DHA/day versus placebo [33]. The first large RCT testing EPA alone was the Japan EPA Lipid Intervention Study (JELIS), randomizing more than 18,000 individuals already treated with statins, to 1.8 g EPA/day versus placebo for 5 years [52]. They found a 19% reduction in major coronary events, but no significant reduction in CV death. Interestingly, patients with hypertriglyceridemia were identified as super-responders, with a 53% reduction in major coronary events. In the last decade, several studies have investigated low-dose EPA + DHA in secondary prevention without any convincing cardiovascular benefit [53,54,55,56].
In 2018, three much anticipated RCTs on primary prevention were published. The Vitamin D and Omega-3 Trial (VITAL) randomized almost 26,000 healthy participants to 1 g EPA + DHA/day and followed them for 5 years [57]. They did not find any significant reduction in the composite endpoint of AMI, stroke, and CV death. The ASCEND (A Cardiovascular Study of Events in Diabetes) Study included more than 15,000 patients with diabetes mellitus and randomized them to ~1 g of EPA + DHA/day versus placebo [58]. Also in this study, there was no significant reduction in cardiovascular events over 7 years of follow-up. The third study was markedly different in design and treatment. The Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial (REDUCE-IT) tested 4 g/day of icosapent ethyl versus placebo in 8179 statin-treated adults with hypertriglyceridemia [37]. A striking 25% reduction in the primary composite endpoint of CV death, nonfatal MI, stroke, coronary revascularization, and unstable angina was observed after 5 years.
A separate mechanistic RCT (EVAPORATE) used serial coronary CT scans to evaluate changes in coronary plaque burden in REDUCE-IT-like patients with 4 g/day of icosapent ethyl [59]. This study showed significant reduction in the primary endpoint of changes in low-attenuating plaque volume (−17% vs. +109% in the placebo group), and a significant reduction also in fibro-fatty plaques (−34% vs. +32%) but not in calcified plaques (−1% vs. +15%) after 18 months. Based on these studies, and category A science Advisory from the American Heart Association [40], the Food and Drug Administration (FDA) in 2019 approved icosapent ethyl as an adjunctive therapy to reduce the risk of CV events in patients with hypertriglyceridemia and high risk of CVD.
During the last 30 years, we have studied a variety of effects of the VLCM n-3 PUFAs in randomized clinical trials as well as by use of extensive biobanks. Back in the early 1990s, the SHOT study [35] was performed to study whether supplementation with VLCM n-3 PUFAs (3.4 g/day of highly concentrated EPA + DHA) would positively influence the occlusion rate of coronary artery bypass grafts (CABG) compared to controls (n = 610). Vein graft occlusion rates after one year, assessed by angiography, were significantly lower (27%) in the VLCM n-3 PUFA group compared to controls (33%). Moreover, a significant trend to lower vein graft occlusion rate with increasing relative change in serum n-3 PUFA levels during the study period was observed. Simultaneously, serum levels of EPA increased from 38 to 92 (mg/L), and serum levels of DHA from 113 to 129 (mg/L), in the active treatment group. To the best of our knowledge, similar randomized trials have not been conducted.
In the CART study [34], performed later in the 1990s, supplementation with VLCM n-3 PUFAs was given daily for six months with the hypothesis to reduce the frequency of restenosis after percutaneous transluminal coronary angioplasty (PTCA). Patients were randomized to either 5.1 g/day of highly concentrated EPA + DHA or corn oil as placebo, starting two weeks prior to PTCA to allow for the FAs to be incorporated in the cell membranes before the procedure. Despite that, serum levels of EPA and DHA increased from 43 to 101 mg/L and 112 to 132 mg/L, respectively, in the VLCM n-3 PUFA group, no effects on the restenosis frequency were obtained (40.6% in the active treatment group vs. 35.4% in the placebo group). This is line with results from other PTCA studies conducted in the same time period using this very high dose of supplementation [60,61]. Sub-studies, however, revealed that whereas this high dose of VLCM n-3 PUFAs was followed by reduction in pro-thrombotic markers, markers of inflammation were increased, possibly related to lipid peroxidation visualized by reduction in vitamin E and increase in thiobarbituric acid-reactive substances [62].
The possible pro-oxidative properties of high doses of VLCM n-3 PUFAs have been discussed with conflicting conclusions [24,25,63,64]. This topic has mainly been explored in experimental studies, and limited in the context of patients with cardiovascular disease. In the OVITES study, 6 weeks’ worth of supplementation with either low (0.85 g/day) or high (5.1 g/day) dose of VLCM n-3 PUFA, or the latter amount + vitamin E 400 mg/day, was explored in high risk individuals. In the low FA group, no changes in inflammatory or peroxidation markers were observed, whereas in the high FA group alone significantly increased inflammation and peroxidation were noted, without any change when vitamin E was added [65]. Thus, the high dose of VLCM n-3 PUFA may induce lipid peroxidation in individuals at risk of atherosclerosis with a subsequent proinflammatory state, not counteracted by vitamin E as an antioxidant, probably because peroxidation mainly occurs at the cellular level. Nevertheless, whether higher doses have a better longstanding effect is still unclear, and is probably dependent on the population. It should be noted that daily dietary amounts in Greenland Eskimos were described as being about 10 g and even more [66]; however, this population at that time is not comparable with the general Western world population today.
In the DOIT study [36,67], the possible modulating effect of 3-year diet counselling was compared to that of supplementation with VLCM n-3 PUFAs given as capsules, on the progression of atherosclerosis assessed as structural measures of carotid IMT (cIMT), plaques score, and vascular function, as well as on clinical end-points. Such comparisons in clinical studies on atherosclerosis had hitherto not been extensively studied. Men at high risk for CVD were randomized in a factorial design to dietary counselling and/or VLCM n-3 PUFA capsules (2.4 g VLCM n-3 FA/day) in a placebo-controlled fashion. The dietary advices were to increase the use of vegetable oils, vegetables, fruits, and fish, and to decrease the use of meat and fat from animal sources. Reduced progression in cIMT was observed in groups with dietary counselling, whereas vascular function was significantly improved in the VLCM n-3 PUFA groups and significantly correlated to changes in serum EPA concentrations. When applying the so-called Plasma Index (PI) (EPA+DHA/total FAs) [22] to the n-3 PUFA groups, baseline levels of total FA were 5.5 g/L, PI 2.2% with EPA and 3.3% with DHA, and after 36 months total FA was 5.0 g/L. PI with EPA was increased to 4.5% and with DHA to 4.3%. Although the study was not geared toward clinical endpoints, a 50% reduction of all-cause mortality in patients randomized to VLCM n-3 PUFA supplementation, compared with placebo, was obtained [67].
Vegetable oils are increasingly used in fish farming industry due to shortness of marine resources; however, the effects on human health of this feeding change are largely unknown. We studied the effects of dietary intake of Atlantic salmon, fed on different fatty acid sources, on the lipid profile, vascular inflammation, and peroxidation in patients with stable coronary heart disease, in a double-blind, randomized dietary intervention study (“Fiord-to-Table”) [68,69]. Patients were randomized into three groups consuming approximately 700 g per week for 6 weeks of differently fed Atlantic salmon (fed (i) 100% South American fish oil, (ii) 50% South American fish oil/50% rapeseed oil, and (iii) 100% rapeseed oil, respectively). Significant differences between the groups in serum levels of total VLCM n-3 PUFA and the n-3/n-6 FA ratio were demonstrated, and interestingly, the fatty acid composition of the fish feed/pellets and fish fillets was highly mirrored in the serum FA profile of the patients after the intervention: “The patient’s serum fatty acid profile became what the salmon was fed” (Figure 4). Additionally, significant reduction in serum triglycerides and inflammatory markers were obtained in patients receiving the 100% South American fish oil.
Possible changes in dietary habits during the study period when ingestion of 700 g salmon per week was also examined [69]. In the total population, significant changes were observed for fatty fish (10.2–75.9 g/day), lean fish (18.6–0.0 g/day), processed fish (16.4–0.8 g/day), processed meat (23.1–15.8 g/day), pork (11.1–7.8 g/day), and meat stew (30.2–22.7 g/day). Thus, a healthier diet was obtained, and the beneficial results of vascular inflammation with 100% South American fish oil were conferred by the difference in feed of the farmed salmon. To the best of our knowledge, similar studies have not been conducted since.
In this context, the quality of farmed salmon on the market can be discussed, as can the toxicity of farmed fish. In this “Fiord-to-Table”study, the latter was explored, showing levels of inorganic contaminants in plasma to be only slightly changed over the study period [70].
The ongoing OMEMI Trial (NCT01841944NC) [71] is a prospective, randomized, placebo-controlled, double-blind multicenter trial, aimed to investigate the effects of supplementation with 1.8 g/day of VLCM n-3 PUFAs (Pikasol®) on cardiovascular morbidity and mortality during a follow-up period of 2 years in an elderly population (70–82 years) who have experienced an AMI. Compliance is secured by measurement of FAs in serum phospholipids. Our hypothesis is that supplementation of VLCM n-3 PUFAs will reduce the risk for cardiovascular events. All patients are included and follow-up is finalized. The results will be presented at the end of 2020.

4. Summary

The global burden of atherothrombotic heart disease should be considered a life-style disorder where differences in dietary habits and related risk factors like limited physical activity and adiposity together play important roles. Related metabolic changes have been scientifically elucidated in recent decades, and the role of VLCM FAs EPA and DHA has gained increased interest. Their possible effects on pathophysiological processes like inflammation and thrombosis have been highlighted.
In the present brief review, we have mainly focused on metabolic mechanisms in atherothrombotic heart disease, and the effects of these FAs in the clinical setting. The main focus has been on the divergent results in the field and the important differences in study population, supplements versus fresh marine sources, the proportion of EPA versus DHA, and the dosages and duration of supplementation.
We conclude that daily intake of at least 1 g of VLCM FAs may improve the dysmetabolic state. The potential to reduce the risk and progression of atherothrombotic heart disease is, however, still a matter of debate.

Author Contributions

H.A. drafted the manuscript, P.L.M. mainly responsible for implementing the late clinical trials, I.S. contributed to finalizing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Cardiovascular Diseases Fact Sheet: World Health Organization. 2017. Available online: http://www.who.int/mediacentre/factsheets/fs317/en/ (accessed on 17 May 2017).
  2. Timmis, A.; Townsend, N.; Gale, C.; Grobbee, R.; Maniadakis, N.; Flather, M.; Wilkins, E.; Wright, L.; Vos, R.; Blum, M.; et al. European Society of Cardiology: Cardiovascular Disease Statistics 2017. Eur. Heart J. 2017, 39, 508–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Libby, P.; Ridker, P.M.; Hansson, G.K. Progress and challenges in translating the biology of atherosclerosis. Nature 2011, 473, 317–325. [Google Scholar] [CrossRef]
  4. Badimon, L.; Padró, T.; Vilahur, G. Atherosclerosis, platelets and thrombosis in acute ischaemic heart disease. Eur. Heart J. Acute Cardiovasc. Care 2012, 1, 60–74. [Google Scholar] [CrossRef] [PubMed]
  5. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N. Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef] [PubMed]
  6. Badimon, L.; Chagas, P.; Chiva-Blanch, G. Diet and Cardiovascular Disease: Effects of Foods and Nutrients in Classical and Emerging Cardiovascular Risk Factors. Curr. Med. Chem. 2019, 26, 3639–3651. [Google Scholar] [CrossRef] [PubMed]
  7. De Lorgeril, M.; Salen, P.; Martin, J.L.; Monjaud, I.; Delaye, J.; Mamelle, N. Mediterranean Diet, Traditional Risk Factors, and the Rate of Cardiovascular Complications after Myocardial Infarction. Final report of the Lyon Diet Heart Study. Circulation 1999, 9, 324. [Google Scholar] [CrossRef] [Green Version]
  8. Bang, H.; Dyerberg, J.; Nielsen, A.B.; Nielsen, A. Plasma lipid and lipoprotein pattern in greenlandic west-coast eskimos. Lancet 1971, 297, 1143–1146. [Google Scholar] [CrossRef]
  9. Dyerberg, J.; Bang, H.; Stoffersen, E.; Moncada, S.; Vane, J. Eicosapentaenoic acid and prevention of thrombosis and atherosclerosis? Lancet 1978, 312, 117–119. [Google Scholar] [CrossRef]
  10. Siddiqui, R.A.; Harvey, K.A.; Zaloga, G.P. Modulation of enzymatic activities by n-3 polyunsaturated fatty acids to support cardiovascular health. J. Nutr. Biochem. 2008, 19, 417–437. [Google Scholar] [CrossRef]
  11. Schmidt, E.B.; Arnesen, H.; de Caterina, R.; Rasmussen, L.H.; Kristensen, S.D. Marine n-3 polyunsaturated fatty acids and coronary heart disease. Part I. Background, epidemiology, animal data, effects on risk factors and safety. Thromb. Res. 2005, 115, 163–170. [Google Scholar] [CrossRef]
  12. De Caterina, R. n–3 Fatty Acids in Cardiovascular Disease. N. Engl. J. Med. 2011, 364, 2439–2450. [Google Scholar] [CrossRef] [PubMed]
  13. Innes, J.K.; Calder, P.C. Marine Omega-3 (N-3) Fatty Acids for Cardiovascular Health: An Update for 2020. Int. J. Mol. Sci. 2020, 21, 1362. [Google Scholar] [CrossRef] [Green Version]
  14. Delgado-Lista, J.; Pérez-Martínez, P.; Miranda, J.L.; Pérez-Jiménez, F. Long chain omega-3 fatty acids and cardiovascular disease: A systematic review. Br. J. Nutr. 2012, 107 (Suppl. 2), S201–S213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Watanabe, Y.; Tatsuno, I. Prevention of Cardiovascular Events with Omega-3 Polyunsaturated Fatty Acids and the Mechanism Involved. J. Atheroscler. Thromb. 2020, 27, 183–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Burdge, G.C.; Calder, P.C. Conversion of α-linolenic acid to longer-chain polyunsaturated fatty acids in human adults. Reprod. Nutr. Dev. 2005, 45, 581–597. [Google Scholar] [CrossRef] [PubMed]
  17. Bork, C.S. Alpha-Linolenic Acid and the Risk of Atherosclerotic Cardiovascular Disease. Ph.D. Thesis, Faculty of Medicine Aalborg University, Aalborg, Denmark, 2020. [Google Scholar]
  18. Serhan, C.N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 2014, 510, 92–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Nordøy, A.; Marchioli, R.; Arnesen, H.; Videbaek, J. n-3 polyunsaturated fatty acids and cardiovascular diseases: To whom, how much, preparations. Lipids 2001, 36, S127–S129. [Google Scholar]
  20. AbuMweis, S.S.; Jew, S.; Tayyem, R.; Agraib, L. 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. 2017, 31, 67–84. [Google Scholar] [CrossRef]
  21. Harris, W.S.; Tintle, N.L.; Etherton, M.R.; Vasan, R.S. Erythrocyte long-chain omega-3 fatty acid levels are inversely associated with mortality and with incident cardiovascular disease: The Framingham Heart Study. J. Clin. Lipidol. 2018, 12, 718–727.E6. [Google Scholar] [CrossRef] [Green Version]
  22. Alfaddagh, A.; Elajami, T.K.; Saleh, M.; Mohebali, D.; Bistrian, B.R.; Welty, F.K. An omega-3 fatty acid plasma index ≥4% prevents progression of coronary artery plaque in patients with coronary artery disease on statin treatment. Atherosclerosis 2019, 285, 153–162. [Google Scholar] [CrossRef]
  23. Von Schacky, C. Omega-3 fatty Acids in cardiovascular disease—An uphill battle. Prostaglandins Leukot. Essent. Fat. Acids 2015, 92, 41–47. [Google Scholar] [CrossRef] [PubMed]
  24. Albert, B.B.; Cameron-Smith, D.; Hofman, P.L.; Cutfield, W.S. Oxidation of Marine Omega-3 Supplements and Human Health. BioMed Res. Int. 2013, 2013, 464921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Nenseter, M.S.; Drevon, C.A. Dietary polyunsaturates and peroxidation of low density lipoprotein. Curr. Opin. Lipidol. 1996, 7, 8–13. [Google Scholar] [CrossRef] [PubMed]
  26. Grundy, S.M.; Cleeman, J.I.; Daniels, S.R.; Donato, K.A.; Eckel, R.; Franklin, B.A.; Gordon, D.J.; Krauss, R.M.; Savage, P.J.; Smith, S.C.; et al. Diagnosis and Management of the Metabolic Syndrome: An American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation 2005, 112, 2735–2752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Langsted, A.; Freiberg, J.J.; Tybjaerg-Hansen, A.; Schnohr, P.; Jensen, G.B.; Nordestgaard, B.G.; Tybjærg-Hansen, A. Nonfasting cholesterol and triglycerides and association with risk of myocardial infarction and total mortality: The Copenhagen City Heart Study with 31 years of follow-up. J. Intern. Med. 2010, 270, 65–75. [Google Scholar] [CrossRef]
  28. Assmann, G.; Schulte, H. Role of triglycerides in coronary artery disease: Lessons from the prospective cardiovascular Münster study. Am. J. Cardiol. 1992, 70, H10–H13. [Google Scholar] [CrossRef]
  29. Manninen, V.; Tenkanen, L.; Koskinen, P.; Huttunen, J.K.; Mänttäri, M.; Heinonen, O.P.; Frick, M.H. Joint effects of serum triglyceride and LDL cholesterol and HDL cholesterol concentrations on coronary heart disease risk in the Helsinki Heart Study. Implications for treatment. Circulation 1992, 85, 37–45. [Google Scholar] [CrossRef] [Green Version]
  30. Assmann, G.; Schulte, H.; Funke, H.; Von Eckardstein, A. The emergence of triglycerides as a significant independent risk factor in coronary artery disease. Eur. Heart J. 1998, 19 (Suppl. M), M8–M14. [Google Scholar]
  31. Hjerkinn, E.M.; Sandvik, L.; Hjermann, I.; Arnesen, H. Fasting triglycerides as a predictor of long-term mortality in middle-aged men with combined hyperlipidaemia. Scand. J. Clin. Lab. Investig. 2003, 63, 273–278. [Google Scholar] [CrossRef]
  32. Harris, W.S.; Bulchandani, D. Why do omega-3 fatty acids lower serum triglycerides? Curr. Opin. Lipidol. 2006, 17, 387–393. [Google Scholar] [CrossRef]
  33. GISSI-Prevenzione Investigators (Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto miocardico). Dietary supplementation with n-3 polyunsaturated fatty acids and vitamin E after myocardial infarction: Results of the GISSI-Prevenzione trial. Lancet 1999, 354, 447–455. [Google Scholar]
  34. Johansen, O.; Brekke, M.; Seljeflot, I.; Abdelnoor, M.; Arnesen, H. n-3 fatty acids do not prevent restenosis after coronary angioplasty: Results from the CART study. Coronary Angioplasty Restenosis Trial. J. Am. Coll. Cardiol. 1999, 33, 1619–1626. [Google Scholar] [CrossRef] [Green Version]
  35. Eritsland, J.; Arnesen, H.; Grønseth, K.; Fjeld, N.B.; Abdelnoor, M. Effect of dietary supplementation with n-3 fatty acids on coronary artery bypass graft patency. Am. J. Cardiol. 1996, 77, 31–36. [Google Scholar] [CrossRef]
  36. Hjerkinn, E.M.; Abdelnoor, M.; Breivik, L.; Bergengen, L.; Ellingsen, I.; Seljeflot, I.; Aase, O.; Klemsdal, T.O.; Hjermann, I.; Arnesen, H. Effect of diet or very long chain ω-3 fatty acids on progression of atherosclerosis, evaluated by carotid plaques, intima-media thickness and by pulse wave propagation in elderly men with hypercholesterolaemia. Eur. J. Cardiovasc. Prev. Rehabil. 2006, 13, 325–333. [Google Scholar] [CrossRef]
  37. Bhatt, D.L.; Steg, P.G.; Miller, M.; Brinton, E.A.; Jacobson, T.A.; Ketchum, S.B.; Doyle, R.T.; Juliano, R.A.; Jiao, L.; Granowitz, C.; et al. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N. Engl. J. Med. 2019, 380, 11–22. [Google Scholar] [CrossRef]
  38. Sheikh, O.; Hei, A.G.V.; Battisha, A.; Hammad, T.; Pham, S.; Chilton, R. Cardiovascular, electrophysiologic, and hematologic effects of omega-3 fatty acids beyond reducing hypertriglyceridemia: As it pertains to the recently published REDUCE-IT trial. Cardiovasc. Diabetol. 2019, 18, 84. [Google Scholar] [CrossRef] [Green Version]
  39. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A.; et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk: The Task Force for the management of dyslipidaemias of the European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS). Eur. Heart J. 2019, 41, 111–188. [Google Scholar] [CrossRef]
  40. Skulas-Ray, A.C.; Wilson, P.W.; Harris, W.S.; Brinton, E.A.; Kris-Etherton, P.M.; Richter, C.K.; Jacobson, T.A.; Engler, M.B.; Miller, M.; Robinson, J.G.; et al. Omega-3 Fatty Acids for the Management of Hypertriglyceridemia: A Science Advisory From the American Heart Association. Circulation 2019, 140, e673–e691. [Google Scholar] [CrossRef] [Green Version]
  41. Bønaa, K.H.; Bjerve, K.S.; Straume, B.; Gram, I.T.; Thelle, D. Effect of Eicosapentaenoic and Docosahexaenoic Acids on Blood Pressure in Hypertension. N. Engl. J. Med. 1990, 322, 795–801. [Google Scholar] [CrossRef]
  42. Miller, P.E.; Van Elswyk, M.; Alexander, D.D. Long-chain omega-3 fatty acids eicosapentaenoic acid and docosahexaenoic acid and blood pressure: A meta-analysis of randomized controlled trials. Am. J. Hypertens. 2014, 27, 885–896. [Google Scholar] [CrossRef]
  43. Pahlavani, M.; Ramalho, T.; Koboziev, I.; Lemieux, M.J.; Jayarathne, S.; Ramalingam, L.; Filgueiras, L.R.; Moustaid-Moussa, N. Adipose tissue inflammation in insulin resistance: Review of mechanisms mediating anti-inflammatory effects of omega-3 polyunsaturated fatty acids. J. Investig. Med. 2017, 65, 1021–1027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Kalupahana, N.S.; Goonapienuwala, B.L.; Moustaid-Moussa, N. Omega-3 Fatty Acids and Adipose Tissue: Inflammation and Browning. Annu. Rev. Nutr. 2020, 40. [Google Scholar] [CrossRef] [PubMed]
  45. Poledne, R.; Malinska, H.; Kubatova, H.; Fronek, J.; Thieme, F.; Kauerova, S.; Lesná, I.K. Polarization of Macrophages in Human Adipose Tissue is Related to the Fatty Acid Spectrum in Membrane Phospholipids. Nutrients 2019, 12, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Simopoulos, A.P. An Increase in the Omega-6/Omega-3 Fatty Acid Ratio Increases the Risk for Obesity. Nutrients 2016, 8, 128. [Google Scholar] [CrossRef] [Green Version]
  47. Lorente-Cebrián, S.; Costa, A.G.V.; Navas-Carretero, S.; Zabala, M.; Martínez, J.A.; Moreno-Aliaga, M.J. Role of omega-3 fatty acids in obesity, metabolic syndrome, and cardiovascular diseases: A review of the evidence. J. Physiol. Biochem. 2013, 69, 633–651. [Google Scholar] [CrossRef]
  48. Martínez-Fernández, L.; Laiglesia, L.; Huerta, A.E.; Martínez, J.A.; Moreno-Aliaga, M.J. Omega-3 fatty acids and adipose tissue function in obesity and metabolic syndrome. Prostaglandins Other Lipid Mediat. 2015, 121, 24–41. [Google Scholar] [CrossRef]
  49. Albracht-Schulte, K.; Kalupahana, N.S.; Ramalingam, L.; Wang, S.; Rahman, S.M.; Robert-McComb, J.; Moustaid-Moussa, N. Omega-3 fatty acids in obesity and metabolic syndrome: A mechanistic update. J. Nutr. Biochem. 2018, 58, 1–16. [Google Scholar] [CrossRef]
  50. Coltell, O.; Sorli, J.V.; Asensio, E.M.; Barragán, R.; González, J.I.; Gimenez-Alba, I.M.; Zanon-Moreno, V.C.; Estruch, R.; Sabio, J.B.R.; Pascual, E.C.; et al. Genome-Wide Association Study for Serum Omega-3 and Omega-6 Polyunsaturated Fatty Acids: Exploratory Analysis of the Sex-Specific Effects and Dietary Modulation in Mediterranean Subjects with Metabolic Syndrome. Nutrients 2020, 12, 310. [Google Scholar] [CrossRef] [Green Version]
  51. Burr, M.; Gilbert, J.; Holliday, R.; Elwood, P.; Fehily, A.; Rogers, S.; Sweetnam, P.; Deadman, N. Effects of changes in fat, fish, and fibre intakes on death and myocardial reinfarction: Diet and reinfarction trial (dart). Lancet 1989, 334, 757–761. [Google Scholar] [CrossRef]
  52. Yokoyama, M.; Origasa, H.; Matsuzaki, M.; Matsuzawa, Y.; Saito, Y.; Ishikawa, Y.; Oikawa, S.; Sasaki, J.; Hishida, H.; Itakura, H.; et al. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): A randomised open-label, blinded endpoint analysis. Lancet 2007, 369, 1090–1098. [Google Scholar] [CrossRef]
  53. Kromhout, D.; Giltay, E.J.; Geleijnse, J.M. n–3 Fatty Acids and Cardiovascular Events after Myocardial Infarction. N. Engl. J. Med. 2010, 363, 2015–2026. [Google Scholar] [CrossRef] [PubMed]
  54. Rauch, B.; Schiele, R.; Schneider, S.; Diller, F.; Victor, N.; Gohlke, H.; Gottwik, M.; Steinbeck, G.; Del Castillo, U.; Sack, R.; et al. 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 2010, 122, 2152–2159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Galan, P.; Kesse-Guyot, E.; Czernichow, S.; Briançon, S.; Blacher, J.; Hercberg, S. Effects of B vitamins and omega 3 fatty acids on cardiovascular diseases: A randomised placebo controlled trial. BMJ 2010, 341, c6273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Bosch, J.; Gerstein, H.C.; Dagenais, G.R.; Díaz, R.; Dyal, L.; Jung, H.; Maggiono, A.P.; Probstfield, J.; Ramachandran, A.; Riddle, M.C.; et al. n-3 fatty acids and cardiovascular outcomes in patients with dysglycemia. N. Engl. J. Med. 2012, 367, 309–318. [Google Scholar]
  57. Manson, J.E.; Cook, N.R.; Lee, I.M.; Christen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Albert, C.M.; Gordon, D.; Copeland, T.; et al. Marine n-3 fatty acids and prevention of cardiovascular disease and cancer. Yearb. Paediatr. Endocrinol. 2019, 380, 23–32. [Google Scholar] [CrossRef]
  58. Bowman, L.; Mafham, M.; Wallendszus, K.; Stevens, W.; Buck, G.; Barton, J.; Murphy, K.; Aung, T.; Haynes, R.; Cox, J.; et al. Effects of n-3 Fatty Acid Supplements in Diabetes Mellitus. N. Engl. J. Med. 2018, 379, 1540–1550. [Google Scholar]
  59. Budoff, M.J.; Bhatt, D.L.; Kinninger, A.; Laksmanan, S.; Muhlestein, J.B.; Le, V.T.; May, H.T.; Shaikh, K.; Shekar, C.; Roy, S.K.; et al. Effect of icosapent ethyl on progression of coronary atherosclerosis in patients with elevated triglycerides on statin therapy: Final results of the EVAPORATE trial. Eur. Heart J. 2020. [Google Scholar] [CrossRef]
  60. Leaf, A.; Jorgensen, M.B.; Jacobs, A.K.; Cote, G.; Schoenfeld, D.A.; Scheer, J.; Weiner, B.H.; Slack, J.D.; Kellett, M.A.; Raizner, A.E. Do fish oils prevent restenosis after coronary angioplasty? Circulation 1994, 90, 2248–2257. [Google Scholar] [CrossRef] [Green Version]
  61. Cairns, J.A.; Gill, J.; Morton, B.; Roberts, R.; Gent, M.; Hirsh, J.; Holder, D.; Finnie, K.; Marquis, J.F.; Naqvi, S.; et al. Fish Oils and Low-Molecular-Weight Heparin for the Reduction of Restenosis after Percutaneous Transluminal Coronary Angioplasty. Circulation 1996, 94, 1553–1560. [Google Scholar] [CrossRef]
  62. Johansen, O.; Seljeflot, I.; Høstmark, A.T.; Arnesen, H. The effect of supplementation with omega-3 fatty acids on soluble markers of endothelial function in patients with coronary heart disease. Arter. Thromb. Vasc. Biol. 1999, 19, 1681–1686. [Google Scholar] [CrossRef] [Green Version]
  63. Guichardant, M.; Calzada, C.; Bernoud-Hubac, N.; Lagarde, M.; Véricel, E. Omega-3 polyunsaturated fatty acids and oxygenated metabolism in atherothrombosis. Biochim. Biophys. Acta 2015, 1851, 485–495. [Google Scholar] [CrossRef] [PubMed]
  64. Mæhre, H.K.; Jensen, I.-J.; Elvevoll, E.O.; Eilertsen, K.-E. ω-3 Fatty Acids and Cardiovascular Diseases: Effects, Mechanisms and Dietary Relevance. Int. J. Mol. Sci. 2015, 16, 22636–22661. [Google Scholar] [CrossRef] [PubMed]
  65. Arnesen, H.; Seljeflot, I. Studies on very long chain marine n-3 fatty acids in patients with atherosclerotic heart disease with special focus on mechanisms, dosage and formulas of supplementation. Cell. Mol. Biol. 2010, 56, 18–27. [Google Scholar] [PubMed]
  66. Bang, H.O.; Dyerberg, J.; Sinclair, H.M. The composition of the Eskimo food in north western Greenland. Am. J. Clin. Nutr. 1980, 33, 2657–2661. [Google Scholar] [CrossRef] [Green Version]
  67. Einvik, G.; Klemsdal, T.O.; Sandvik, L.; Hjerkinn, E.M. A randomized clinical trial on n-3 polyunsaturated fatty acids supplementation and all-cause mortality in elderly men at high cardiovascular risk. Eur. J. Cardiovasc. Prev. Rehabil. 2010, 17, 588–592. [Google Scholar] [CrossRef] [PubMed]
  68. Seierstad, S.L.; Seljeflot, I.; Johansen, O.; Hansen, R.; Haugen, M.; Rosenlund, G.; Frøyland, L.; Arnesen, H. Dietary intake of differently fed salmon; the influence on markers of human atherosclerosis. Eur. J. Clin. Investig. 2005, 35, 52–59. [Google Scholar] [CrossRef]
  69. Gharagozlian, S.; Hansen, R.; Haugen, M.; Johansen, O.; Seierstad, S.L.; Seljeflot, I.; Arnesen, H. Changes in dietary pattern when including 700 g of salmon per week to patients with atherosclerotic heart disease. Clin. Nutr. ESPEN 2017, 19, 38–44. [Google Scholar] [CrossRef]
  70. Bethune, C.; Seierstad, S.L.; Seljeflot, I.; Johansen, O.; Arnesen, H.; Meltzer, H.M.; Rosenlund, G.; Frøyland, L.; Lundebye, A.-K. Dietary intake of differently fed salmon: A preliminary study on contaminants. Eur. J. Clin. Investig. 2006, 36, 193–201. [Google Scholar] [CrossRef]
  71. Laake, K.; Myhre, P.; Nordby, L.M.; Seljeflot, I.; Abdelnoor, M.; Smith, P.; Tveit, A.; Arnesen, H.; Solheim, S. Effects of omega 3 supplementation in elderly patients with acute myocardial infarction: Design of a prospective randomized placebo controlled study. BMC Geriatr. 2014, 14, 74. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Metabolic pathway of omega-6 and omega-3 fatty acid synthesis. Copied from Siddiqui RA, Harvey KA, Zaloga GP. J. Nutr. Biochem. 2008, 19, 417–437 [10], with permission.
Figure 1. Metabolic pathway of omega-6 and omega-3 fatty acid synthesis. Copied from Siddiqui RA, Harvey KA, Zaloga GP. J. Nutr. Biochem. 2008, 19, 417–437 [10], with permission.
Nutrients 12 03014 g001
Figure 2. Modulation of white adipose tissue function by omega-3 polyunsaturated fatty acids (ω-3 PUFAs). Copied from Kalupahana NS et al., with permission. Annu. Rev. Nutr. 2020 Jun 16. doi:10.1146/annurev-nutr-122319-034142 [44].
Figure 2. Modulation of white adipose tissue function by omega-3 polyunsaturated fatty acids (ω-3 PUFAs). Copied from Kalupahana NS et al., with permission. Annu. Rev. Nutr. 2020 Jun 16. doi:10.1146/annurev-nutr-122319-034142 [44].
Nutrients 12 03014 g002
Figure 3. Lipid mediators in the acute inflammatory response, resolution, and other outcomes. Copied from Serhan CN, Nature 2014, 510, 92–101 [18], with permission.
Figure 3. Lipid mediators in the acute inflammatory response, resolution, and other outcomes. Copied from Serhan CN, Nature 2014, 510, 92–101 [18], with permission.
Nutrients 12 03014 g003
Figure 4. Patterns of the sum of n-3 FA and the ratio n-3/n-6 FA in (a) the three different feeds, (b) corresponding salmon fillets, and (c) serum fatty acids in patients, after the 6-week dietary intervention. FO: fish oil; RO: rapeseed oil (Seierstad SL et al. [67]).
Figure 4. Patterns of the sum of n-3 FA and the ratio n-3/n-6 FA in (a) the three different feeds, (b) corresponding salmon fillets, and (c) serum fatty acids in patients, after the 6-week dietary intervention. FO: fish oil; RO: rapeseed oil (Seierstad SL et al. [67]).
Nutrients 12 03014 g004

Share and Cite

MDPI and ACS Style

Arnesen, H.; Myhre, P.L.; Seljeflot, I. Very Long Chain Marine n-3 Polyunsaturated Fatty Acids in Atherothrombotic Heart Disease. A Brief Review, with a Focus on Metabolic Effects. Nutrients 2020, 12, 3014. https://doi.org/10.3390/nu12103014

AMA Style

Arnesen H, Myhre PL, Seljeflot I. Very Long Chain Marine n-3 Polyunsaturated Fatty Acids in Atherothrombotic Heart Disease. A Brief Review, with a Focus on Metabolic Effects. Nutrients. 2020; 12(10):3014. https://doi.org/10.3390/nu12103014

Chicago/Turabian Style

Arnesen, Harald, Peder L. Myhre, and Ingebjørg Seljeflot. 2020. "Very Long Chain Marine n-3 Polyunsaturated Fatty Acids in Atherothrombotic Heart Disease. A Brief Review, with a Focus on Metabolic Effects" Nutrients 12, no. 10: 3014. https://doi.org/10.3390/nu12103014

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop