Hostname: page-component-8448b6f56d-wq2xx Total loading time: 0 Render date: 2024-04-25T05:18:45.372Z Has data issue: false hasContentIssue false

The impact of fatty acid desaturase genotype on fatty acid status and cardiovascular health in adults

Published online by Cambridge University Press:  16 August 2016

Colette M. O'Neill*
Affiliation:
Department of Nutrition and Preventive Medicine, Norwich Medical School, BCRE, University of East Anglia, James Watson Road, Norwich NR4 7UQ, UK Cork Centre for Vitamin D and Nutrition Research, School of Food and Nutritional Sciences, University College Cork, Ireland
Anne-Marie Minihane
Affiliation:
Department of Nutrition and Preventive Medicine, Norwich Medical School, BCRE, University of East Anglia, James Watson Road, Norwich NR4 7UQ, UK
*
*Corresponding author: C. M. O'Neill, email colette.oneill@ucc.ie
Rights & Permissions [Opens in a new window]

Abstract

The aim of this review was to determine the impact of the fatty acid desaturase (FADS) genotype on plasma and tissue concentrations of the long-chain (LC) n-3 PUFA, including EPA and DHA, which are associated with the risk of several diet-related chronic diseases, including CVD. In addition to dietary intakes, which are low for many individuals, tissue EPA and DHA are also influenced by the rate of bioconversion from α-linolenic acid (αLNA). Δ-5 and Δ-6 desaturase enzymes, encoded for by FADS1 and FADS2 genes, are key desaturation enzymes involved in the bioconversion of essential fatty acids (αLNA and linoleic acid (LA)) to longer chained PUFA. In general, carriers of FADS minor alleles tend to have higher habitual plasma and tissue levels of LA and αLNA, and lower levels of arachidonic acid, EPA and also to a lesser extent DHA. In conclusion, available research findings suggest that FADS minor alleles are also associated with reduced inflammation and CVD risk, and that dietary total fat and fatty acid intake have the potential to modify relationships between FADS gene variants and circulating fatty acid levels. However to date, neither the size-effects of FADS variants on fatty acid status, nor the functional SNP in FADS1 and 2 have been identified. Such information could contribute to the refinement and targeting of EPA and DHA recommendations, whereby additional LC n-3 PUFA intakes could be recommended for those carrying FADS minor alleles.

Type
Irish Section Postgraduate Meeting
Copyright
Copyright © The Authors 2016 

Plasma and tissue long-chain (LC) PUFA concentrations are associated with the risk of several diet-related chronic diseases, including CVD( Reference Bucher, Hengstler and Schindler 1 Reference Dacks, Shineman and Fillit 5 ). Therefore it is important that the determinants of LC-PUFA metabolism, and concentrations in the circulation and in target tissues are fully understood. n-3 Fatty acids are PUFA, which contain the first double bond at the third carbon atom from the methyl end of the fatty acid. There are three major LC n-3 PUFA in the human diet and mammalian tissues, namely α-linolenic acid (αLNA), EPA and DHA. Although the most effective means to increase EPA and DHA status is through increased consumption of fish, bioconversion from the essential fatty acid, αLNA, represents a significant source and in particular in non-fish/EPA plus DHA supplement consumers who have 57–80 % lower intakes than fish eaters, with EPA and DHA derived from the sequential desaturation and elongation from αLNA( Reference Welch, Shakya-Shrestha and Lentjes 6 ).

The potential health benefits associated with consumption of EPA and DHA are numerous, with the most studied and accepted being a reduction in CVD risk. As summarised in several systematic reviews and meta-analysis of prospective epidemiological studies and randomised controlled trials, the ability of LC n-3 PUFA to reduce all-cause mortality and cardiovascular mortality has been widely described( Reference Bucher, Hengstler and Schindler 1 , Reference Mente, de Koning and Shannon 2 , Reference Wang, Harris and Chung 4 , Reference León, Shibata and Sivakumaran 7 , Reference Studer, Briel and Leimenstoll 8 ). However, it should be noted that this is not a fully consistent finding, with the heterogeneity in responsiveness as yet not fully understood( Reference Rizos, Ntzani and Bika 9 , Reference Kwak, Myung and Lee 10 ). Consumption of EPA and DHA has also been shown to be associated with many other diseases, for example, autoimmune diseases such as rheumatoid arthritis, cancer, diabetes, respiratory diseases, gastrointestinal diseases, Alzheimer's disease, depression, as well as psychotic disorders, for example schizophrenia( Reference Fetterman and Zdanowicz 11 Reference Jump, Depner and Tripathy 14 ).

The current recommended intakes for EPA plus DHA in the UK are ⩾450 mg/d( 15 ). This recommendation is based largely on the cardiovascular benefits of these fatty acids and can be achieved by consuming two portions of fish per week, one of which should be oily( 15 ). However, the estimated EPA and DHA consumption in adults in the UK is approximately 270 mg/d for men and 220 mg/d for women, which is far below the recommended minimal intake( Reference Welch, Shakya-Shrestha and Lentjes 6 ). Furthermore, mean population intakes are known to be highly skewed, with a large proportion of the population who do not consume fish or an EPA/DHA-containing supplement having a typical EPA plus DHA intake of <50 mg/d( Reference Welch, Shakya-Shrestha and Lentjes 6 , Reference Minihane 16 ).

n-6 PUFA, including linoleic acid (LA) and arachidonic acid (AA), contain the first double bond at the sixth carbon atom from the methyl end of the fatty acid. LA is an essential fatty acid that is found in vegetable oils and is the most abundant PUFA in the modern Western diet( Reference Chilton, Murphy and Wilson 17 ). LA can be metabolised to AA, which in turn, is a precursor of eicosanoids, such as PG, thromboxanes and leukotrienes. These eicoisanoids tend to be pro-inflammatory and therefore may negatively impact on the development of CVD( Reference Martinelli, Girelli and Malerba 18 ).

There is now a large published literature reporting on the impact of individual gene variants on LC-PUFA metabolism and CVD incidence and biomarker profiles. This review will focus on the fatty acid desaturase (FADS) genotypes, which are emerging as the most significant common genetic determinants identified to date. Accumulating evidence suggests that the locus may, in the future, be useful in stratification and targeting of LC-PUFA recommendations towards individuals likely to be deficient and responsive.

PUFA bioconversion and the fatty acid desaturase genotype

In addition to dietary intake, tissue EPA and DHA is influenced by the rate of bioconversion from αLNA, which involves multiple desaturation and elongation steps (Fig. 1). The Δ-5 and Δ-6 desaturase enzymes are the key rate-limiting enzymes in this pathway( Reference Sprecher 19 ). The human desaturase complementary DNA were first cloned in 1999 by Cho et al.( Reference Cho, Nakamura and Clarke 20 , Reference Cho, Nakamura and Clarke 21 ) and were later identified as FADS1 and FADS2 in the human genome( Reference Marquardt, Stohr and White 22 ), located in a cluster on chromosome 11 (11q12–13.1). Δ-5 desaturase and Δ-6 desaturase are found in many human tissues, but the liver is the site at which they are most highly expressed( Reference Cho, Nakamura and Clarke 20 , Reference Cho, Nakamura and Clarke 21 ). LA and αLNA are metabolised by the same series of enzymes. EPA and DHA are produced at limited conversion rates of 0·2–6 % for EPA and <0·1 % for DHA in human males and post-menopausal females, with higher rates evident in pre-menopausal females( Reference Burdge 23 ). The more efficient EPA and DHA synthesis in pre-menopausal women is thought to be an evolutionary adaptation, so that younger females have sufficient LC-PUFA to meet the demands of pregnancy and the developing fetus. As will be described, variation across the FADS gene region appears to be important in modulating LC-PUFA status. The functional SNP in FADS1 and 2 have not yet been identified.

Fig. 1. Synthesis of long-chain PUFA from linoleic acid (LA) and alpha-linolenic acid (αLNA). Both LA (n-6) and αLNA (n-3) are elongated, desaturated and β-oxidised using the same enzyme system. AA, arachidonic acid.

Impact of fatty acid desaturase genotype on PUFA status

Using both a candidate gene (Table 1) and a genome wide association study (Table 2) approach, numerous studies have reported associations between variations in the FADS locus and desaturase activity and fatty acid status in human subjects. Desaturase activity can be approximated by calculating the product-to-precursor ratio of fatty acids. In 2006, Schaeffer et al.( Reference Schaeffer, Gohlke and Muller 24 ) analysed eighteen SNP and reconstructed haplotypes in the FADS1–2 cluster in 727 adults. A five-locus FADS haplotype accounted for 27·7, 5·2 and 1·4 % of the variation in AA, EPA and DHA in serum phospholipids, respectively. The minor alleles were associated with higher αLNA and LA and lower γ-linolenic acid, AA, EPA and n-3 docosapentaenoic acid concentrations, with no significant impact on DHA( Reference Schaeffer, Gohlke and Muller 24 ). More recently, Ameur et al. performed genome wide genotyping in 5652 individuals, and targeted resequencing (n 960) of the FADS region, across five European population cohorts and reported that present-day human subjects have two common FADS haplotypes, which are defined by twenty-eight closely linked SNP, one of which was considered to be more efficient in relation to the biosynthesis of LC-PUFA( Reference Ameur, Enroth and Johansson 25 ). This FADS haplotype was associated with lower levels of LA (borderline significant) and αLNA and higher levels of EPA, γ-linolenic acid, DHA and AA. Over the last decade, a number of other candidate gene approach studies, as well as genome wide association studies, have been conducted and the association between FADS SNP/haplotypes and PUFA status, as well as desaturase activities, in plasma have been confirmed and extended to tissue fatty acid composition (Tables 1 and 2). However, information on how factors, including n-3 PUFA intakes, health status and ethnicity, may influence the penetrance of the FADS genotype, and in turn the effect size, is relatively unknown. Further research, expanding on the recent research by Wang et al.( Reference Wang, Athinarayanan and Jiang 26 ), is also required to determine the functional SNP, as well the molecular mechanism(s) responsible for the effect of the FADS genotype on EPA and DHA status. Wang et al. examined the association between six FADS SNP and the lipidomic profile and FADS13 expression in liver samples (n 154) and reported all six alleles to be associated with FADS1 (but not FADS2 and 3) gene expression and protein levels, suggesting that the causal variant(s) may be located at FADS1 ( Reference Wang, Athinarayanan and Jiang 26 ). In addition, twenty out of forty-two highly linked SNP were located in the transcription factor-binding sites of the locus. Although it is unclear exactly which SNP is causal and exactly how the SNP influences transcription factor binding and activation of FADS1, the findings add considerable credibility to the observations that FADS genotypes influence EPA and DHA status.

Table 1. Candidate gene studies: associations between fatty acid desaturase SNP and fatty acid status and cardiovascular health

AA, arachidonic acid; LA, linoleic acid; αLNA, alpha-linolenic acid, MI, myocardial infarction, CAD, coronary artery disease; hs-CRP, high sensitivity C-reactive protein; EDA, eicosadienoic acid; LD, linkage disequilibrium; D6D, Δ-6-desaturase; D5D, Δ-5-desaturase; DGLA, dihomo-γ-linolenic acid; HR, hazard ratio; ACS, acute coronary syndrome; VLCFA, very long-chained fatty acids; T2D, type 2 diabetes.

Table 2. Genome wide association studies: associations between fatty acid desaturase SNP and fatty acid status and cardiovascular health

PC, phosphatidylcholine; D5D, Δ-5-desaturase; AA, arachidonic acid; TC, total cholesterol; LA, linoleic acid; αLNA, alpha-linolenic acid; GLA, γ-linolenic acid; DGLA, dihomo-γ-linolenic acid; TFA, trans-fatty acid.

Impact of fatty acid desaturase genotype on cardiovascular health

The majority of studies to date suggest that FADS minor alleles (associated with decreased desaturase activity) are associated with reduced inflammation, total cholesterol, LDL-cholesterol and coronary artery disease risk (Tables 1 and 2)( Reference Martinelli, Girelli and Malerba 18 , Reference Kwak, Paik and Kim 27 Reference Sabatti, Service and Hartikainen 31 ). In the Verona Heart Study (2008), a coronary artery disease incidence of 84 v. 66 % was evident in individuals with six to seven v. two to three risk alleles and a higher AA:LA ratio was an independent risk factor for coronary artery disease ( Reference Martinelli, Girelli and Malerba 18 ). A potential reason for these findings could involve the high LA intakes in the Western diet, resulting in reduced synthesis of LC n-3 PUFA from αLNA( Reference Blasbalg, Hibbeln and Ramsden 32 ). The higher n-6 conversion also leads to increased levels of AA, which is a direct precursor of many pro-inflammatory eicosanoids( Reference Hester, Murphy and Uhlson 33 , Reference Mathias, Pani and Chilton 34 ). Hester et al.( Reference Hester, Murphy and Uhlson 33 ) recently showed that subjects with the major allele for FADS SNP rs174537 had significantly higher levels of pro-inflammatory eicosanoids, LTB4 and 5-HETE, compared with minor allele carriers( Reference Hester, Murphy and Uhlson 33 ). However, a few studies have reported contradictory results( Reference Qin, Sun and Ye 35 Reference Song, Cao and Qin 37 ) which could be due to the ethnicity of the participants or differences in the n-6 : n-3 PUFA content of the habitual diet. For example, two studies carried out in a Chinese-Han population reported the frequency of the rs174556 minor allele to be significantly higher in cases of both coronary artery disease and acute coronary syndrome compared with control groups( Reference Qin, Sun and Ye 35 , Reference Song, Cao and Qin 37 ).

Impact of diet composition on the relationship between the fatty acid desaturase genotype and PUFA and cardiovascular health status

There have been a number of studies that show that diet composition can influence the relationship between FADS genotype and plasma fatty acid and lipid status (Table 3). In 2012, Hellstrand et al. reported that the FADS rs174547 minor allele was associated with lower LDL-cholesterol among individuals in the lowest tertile of LC n-3 PUFA intakes( Reference Hellstrand, Sonestedt and Ericson 38 ). A significant interaction between rs174547 and the ratio of αLNA and LA intakes on HDL-C was also observed( Reference Hellstrand, Sonestedt and Ericson 38 ). More recently, a 14-year follow-up in 24 032 participants reported that the αLNA:LA intake ratio was inversely associated with CVD risk only among participants homozygous for the rs174547 minor allele( Reference Hellstrand, Ericson and Gullberg 39 ). αLNA intakes were also inversely associated with ischaemic stroke in this genotype group. In addition to observational analysis, the impact of FADS variants on response to LC-PUFA supplementation has also been examined. Gillingham et al. carried out a randomised crossover trial in thirty-six hyperlipidemic subjects in which three diets (enriched with flaxseed oil or high-oleic acid canola oil compared with a typical Western diet) were consumed for 4 weeks and five FADS SNP were analysed( Reference Gillingham, Harding and Rideout 40 ). Subjects with minor allele variants (rs174545, rs174583, rs174561, rs174537) had decreased desaturase activity, but an increase in αLNA intakes resulted in greater increases in plasma EPA than in major allele homozygotes consuming αLNA intakes typical of a Western diet( Reference Gillingham, Harding and Rideout 40 ). Cormier et al. conducted a study in 208 subjects examining the impact of fish-oil supplementation (1·9–2·2 g/d EPA and 1·1 g/d DHA) for 6 weeks and nineteen FADS SNP on plasma TAG and reported that rs174546 was associated with TAG, but no significant genotype by supplementation interaction was observed( Reference Cormier, Rudkowska and Paradis 41 ). In terms of whole-diet interventions, one study to date has examined the interaction of FADS genotype and the Mediterranean diet on serum and colonic fatty acid profiles( Reference Porenta, Ko and Gruber 42 ). In a 6-month intervention (n 108) and genotyping for four FADS SNP, a significant diet by genotype interaction for AA concentrations in the colon was observed; subjects with FADS major alleles following the Mediterranean diet had 18 % lower AA concentrations than subjects on the control diet (healthy eating diet)( Reference Porenta, Ko and Gruber 42 ). There were no significant diets by genotype interactions for other colonic or serum fatty acids. Overall, it is clear that further research is necessary to determine the potential of the diet, particularly dietary fatty acids, to modify the relationship between the FADS genotype and fatty acid status. An investigation of diet composition × FADS genotype × fatty acid status represents a secondary objective of the recently completed NU-AGE intervention.

Table 3. Diet–gene interactions: impact of dietary intakes/interventions on associations between fatty acid desaturase genotype and fatty acid status and cardiovascular health

TC, total cholesterol; LC, long chained; αLNA, alpha-linolenic acid; LA, linoleic acid; DPA, docosapentaenoic acid; hs-CRP, high sensitivity C-reactive protein; HR, hazard ratio; D5D, Δ-5-desaturase; D6D, Δ-6-desaturase; LDL-C, LDL-cholesterol.

NU-AGE: a focus on older adults

The NU-AGE (New dietary strategies addressing the specific needs of the elderly population for healthy ageing in Europe) study investigated the impact of a whole-diet intervention on markers of chronic inflammation in older adults (aged 65–79 years)( Reference Berendsen, Santoro and Pini 43 ). The NU-AGE recommendations for the consumption of oily fish, as well as the provision of an αLNA-rich spread, aimed to increase total n-3 PUFA intakes and the dietary n-6 : n-3 PUFA ratio of study participants. As previously discussed, although a small number of dietary interventions have been shown to modify the relationship between the FADS genotype and PUFA status( Reference Gillingham, Harding and Rideout 40 , Reference Porenta, Ko and Gruber 42 , Reference Cormier, Rudkowska and Lemieux 44 ), none have examined the impact of a 1-year whole-diet (including significant fatty acid manipulation) intervention in older adults, a group who are likely to be in a higher state of chronic inflammation and CVD risk relative to healthy general adult population. Therefore, we aim to examine whether the NU-AGE diet could influence the relationship between the FADS genotype and plasma PUFA status in our study population. Specifically, we wish to establish if the NU-AGE diet can overcome any identified negative impacts of FADS minor alleles on EPA and DHA status, as well as the potential negative effect that the major allele has on AA status. We will also examine the interactive impact of diet and FADS genotype on CVD risk biomarkers, including inflammatory and plasma lipid status and measures of vascular function and arterial stiffness( Reference Martinelli, Girelli and Malerba 18 , Reference Kwak, Paik and Kim 27 , Reference Li, Lin and Ma 28 ).

Conclusion

Current estimates indicate that for most countries, average population intakes of EPA and DHA are 0·2 g/d, and <0·05 g/d in non-fish consumers( Reference Minihane 16 ). In this latter large population subgroup, the efficacy of endogenous synthesis from αLNA determines the tissue EPA and DHA status. A comprehensive understanding of the determinants of the regulation of the desaturation and elongation pathway is lacking. Although common FADS variants have been consistently associated with LC-PUFA status, the exact size of the effect is relatively unquantified and the FADS functional gene variant(s) has not been identified. A recent study by Li et al.( Reference Li, Lin and Ma 28 ) (described in Table 1) reported a difference of 8·3 % in plasma EPA and DHA combined between those homozygous for the major allele and those homozygous for the minor allele of the rs174537 FADS genotype( Reference Li, Wang and Yang 45 ). This is clinically significant as previous research, which showed that EPA and DHA status was associated with sudden cardiac death in US males, reported 9·0 % lower blood EPA and DHA concentrations in the sudden death group compared with controls( Reference Albert, Ma and Rifai 46 ). Modest dietary intakes of EPA and DHA could overcome this genotype effect; supplementation of 300 mg EPA and DHA or 90 g salmon per week has been shown to increase combined plasma EPA plus DHA by about 30 %( Reference Flock, Skulas-Ray and Harris 47 , Reference Raatz, Rosenberger and Johnson 48 ). The mechanistic basis of the relationship between the FADS genotype and LC n-3 PUFA interactions are also poorly understood. The impact of FADS genotype on PUFA status should be carefully considered when using plasma and tissue EPA and DHA concentrations as biomarkers of dietary EPA and DHA exposure in randomised controlled trials and epidemiological studies, with a greater contribution of endogenously synthesised EPA, and to a lesser extent DHA, to the total pool likely in FADS major allele carriers. Furthermore, FADS genotype could contribute to future stratification and targeting of dietary advice with additional EPA and DHA intakes recommended for those carrying the FADS minor allele.

Acknowledgement

NU-AGE (New dietary strategies addressing the specific needs of the elderly population for healthy ageing in Europe) is supported by the EU Seventh Framework Program under grant agreement no. 266486. The authors thank all principal investigators and collaborators.

Financial Support

A. M. M.’s research in the area of EPA, DHA and health is partially funded by a BBSRC Institute Strategic Programme grant (BB/J004545/1).

Conflict of Interest

None.

Authorship

C. M. O. N. drafted the outline of the manuscript, conducted the literature search and drafted the manuscript. A. M. M. was responsible for critically reviewing the manuscript. All authors read and approved the final manuscript before submission.

References

1. 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.CrossRefGoogle ScholarPubMed
2. Mente, A, de Koning, L, Shannon, HS et al. (2009) A systematic review of the evidence supporting a causal link between dietary factors and coronary heart disease. Arch Intern Med 169, 659669.CrossRefGoogle ScholarPubMed
3. Wang, Q, Liang, X, Wang, L et al. (2012) Effect of omega-3 fatty acids supplementation on endothelial function: a meta-analysis of randomized controlled trials. Atherosclerosis 221, 536543.CrossRefGoogle ScholarPubMed
4. Wang, C, Harris, WS, Chung, M et al. (2006) n-3 Fatty acids from fish or fish-oil supplements, but not α-linolenic acid, benefit cardiovascular disease outcomes in primary- and secondary-prevention studies: a systematic review. Am J Clin Nutr 84, 517.CrossRefGoogle Scholar
5. Dacks, PA, Shineman, DW & Fillit, HM (2013) Current evidence for the clinical use of long-chain polyunsaturated n-3 fatty acids to prevent age-related cognitive decline and Alzheimer's disease. J Nutr Health Aging 17, 240251.CrossRefGoogle ScholarPubMed
6. Welch, AA, Shakya-Shrestha, S, Lentjes, MA et al. (2010) Dietary intake and status of n-3 polyunsaturated fatty acids in a population of fish-eating and non-fish-eating meat-eaters, vegetarians, and vegans and the precursor-product ratio of α-linolenic acid to long-chain n-3 polyunsaturated fatty acids: results from the EPIC-Norfolk cohort. Am J Clin Nutr 92, 10401051.CrossRefGoogle Scholar
7. León, H, Shibata, MC, Sivakumaran, S et al. (2008) Effect of fish oil on arrhythmias and mortality: systematic review. BMJ 337, a2931.CrossRefGoogle ScholarPubMed
8. 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.CrossRefGoogle ScholarPubMed
9. 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. JAMA 308, 10241033.CrossRefGoogle ScholarPubMed
10. 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 ScholarPubMed
11. Fetterman, JW Jr & Zdanowicz, MM (2009) Therapeutic potential of n-3 polyunsaturated fatty acids in disease. Am J Health Syst Pharm 66, 11691179.CrossRefGoogle ScholarPubMed
12. Janssen, CI & Kiliaan, AJ (2014) Long-chain polyunsaturated fatty acids (LCPUFA) from genesis to senescence: the influence of LCPUFA on neural development, aging, and neurodegeneration. Prog Lipid Res 53, 117.CrossRefGoogle ScholarPubMed
13. Chen, C, Yu, X & Shao, S (2015) Effects of omega-3 fatty acid supplementation on glucose control and lipid levels in Type 2 Diabetes: a meta-analysis. PLoS ONE 10, e0139565.Google ScholarPubMed
14. Jump, DB, Depner, CM, Tripathy, S et al. (2015) Potential for dietary omega-3 fatty acids to prevent nonalcoholic fatty liver disease and reduce the risk of primary liver cancer. Adv Nutr 6, 694702.CrossRefGoogle ScholarPubMed
15. Scientific Advisory Committee on Nutrition. Committee on Toxicity of Chemicals in Food (2004) Advice on Fish Consumption: Benefits and Risks.Google Scholar
16. Minihane, AM (2013) Fish oil omega-3 fatty acids and cardio-metabolic health, alone or with statins. Eur J Clin Nutr 67, 536540.CrossRefGoogle ScholarPubMed
17. Chilton, FH, Murphy, RC, Wilson, BA et al. (2014) Diet-gene interactions and PUFA metabolism: a potential contributor to health disparities and human diseases. Nutrients 6, 19932022.CrossRefGoogle ScholarPubMed
18. Martinelli, N, Girelli, D, Malerba, G et al. (2008) FADS genotypes and desaturase activity estimated by the ratio of arachidonic acid to linoleic acid are associated with inflammation and coronary artery disease. Eur J Clin Nutr 88, 941949.Google ScholarPubMed
19. Sprecher, H (1981) Biochemistry of essential fatty acids. Prog Lipid Res 20, 1322.CrossRefGoogle ScholarPubMed
20. Cho, HP, Nakamura, M & Clarke, SD (1999) Cloning, expression, and fatty acid regulation of the human delta-5 desaturase. J Biol Chem 274, 3733537339.CrossRefGoogle ScholarPubMed
21. Cho, HP, Nakamura, MT & Clarke, SD (1999) Cloning, expression, and nutritional regulation of the mammalian delta-6 desaturase. J Biol Chem 274, 471477.CrossRefGoogle ScholarPubMed
22. Marquardt, A, Stohr, H, White, K et al. (2000) cDNA cloning, genomic structure, and chromosomal localization of three members of the human fatty acid desaturase family. Genomics 66, 175183.CrossRefGoogle ScholarPubMed
23. Burdge, GC (2006) Metabolism of α-linolenic acid in humans. Prostaglandins Leukot Essent Fatty Acids 75, 161168.CrossRefGoogle ScholarPubMed
24. Schaeffer, L, Gohlke, H, Muller, M et al. (2006) Common genetic variants of the FADS1 FADS2 gene cluster and their reconstructed haplotypes are associated with the fatty acid composition in phospholipids. Hum Mol Genet 15, 17451756.CrossRefGoogle ScholarPubMed
25. Ameur, A, Enroth, S, Johansson, A et al. (2012) Genetic adaptation of fatty-acid metabolism: a human-specific haplotype increasing the biosynthesis of long-chain omega-3 and omega-6 fatty acids. Am J Hum Genet 90, 809820.CrossRefGoogle ScholarPubMed
26. Wang, L, Athinarayanan, S, Jiang, G et al. (2015) Fatty acid desaturase 1 gene polymorphisms control human hepatic lipid composition. Hepatology 61, 119128.CrossRefGoogle ScholarPubMed
27. Kwak, JH, Paik, JK, Kim, OY et al. (2011) FADS gene polymorphisms in Koreans: association with omega6 polyunsaturated fatty acids in serum phospholipids, lipid peroxides, and coronary artery disease. Atherosclerosis 214, 94100.CrossRefGoogle Scholar
28. Li, S-W, Lin, K, Ma, P et al. (2013) FADS gene polymorphisms confer the risk of coronary artery disease in a Chinese Han population through the altered desaturase activities: based on high-resolution melting analysis. PLoS ONE 8, e55869.Google Scholar
29. Aulchenko, YS, Ripatti, S, Lindqvist, I et al. (2009) Loci influencing lipid levels and coronary heart disease risk in 16 European population cohorts. Nat genet 41, 4755.Google ScholarPubMed
30. Standl, M, Lattka, E, Stach, B et al. (2012) FADS1 FADS2 gene cluster, PUFA intake and blood lipids in children: results from the GINIplus and LISAplus studies. PLoS ONE 7, e37780.CrossRefGoogle ScholarPubMed
31. Sabatti, C, Service, SK, Hartikainen, A-L et al. (2009) Genome-wide association analysis of metabolic traits in a birth cohort from a founder population. Nat Genet 41, 3546.CrossRefGoogle Scholar
32. 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
33. Hester, AG, Murphy, RC, Uhlson, CJ et al. (2014) Relationship between a common variant in the fatty acid desaturase (FADS) cluster and eicosanoid generation in humans. J Biol Chem 289, 2248222489.CrossRefGoogle ScholarPubMed
34. Mathias, R, Pani, V & Chilton, F (2014) Genetic variants in the FADS gene: implications for dietary recommendations for fatty acid intake. Curr Nutr Rep 3, 139148.CrossRefGoogle ScholarPubMed
35. Qin, L, Sun, L, Ye, L et al. (2011) A case–control study between the gene polymorphisms of polyunsaturated fatty acids metabolic rate-limiting enzymes and coronary artery disease in a Chinese Han population. Prostaglandins Leukot Essent Fatty Acids 85, 329333.CrossRefGoogle ScholarPubMed
36. Lu, Y, Vaarhorst, A, Merry, AH et al. (2012) Markers of endogenous desaturase activity and risk of coronary heart disease in the CAREMA cohort study. PLoS ONE 7, e41681.Google ScholarPubMed
37. Song, Z, Cao, H, Qin, L et al. (2013) A case–control study between gene polymorphisms of polyunsaturated fatty acid metabolic rate-limiting enzymes and acute coronary syndrome in Chinese Han population. BioMed Res Int 2013, 928178.CrossRefGoogle ScholarPubMed
38. Hellstrand, S, Sonestedt, E, Ericson, U et al. (2012) Intake levels of dietary long-chain PUFAs modify the association between genetic variation in FADS and LDL-C. J Lipid Res 53, 11831189.CrossRefGoogle ScholarPubMed
39. Hellstrand, S, Ericson, U, Gullberg, B et al. (2014) Genetic variation in FADS1 has little effect on the association between dietary PUFA intake and cardiovascular disease. J Nutr 144, 13561363.CrossRefGoogle ScholarPubMed
40. Gillingham, LG, Harding, SV, Rideout, TC et al. (2013) Dietary oils and FADS1-FADS2 genetic variants modulate [13C]α-linolenic acid metabolism and plasma fatty acid composition. Am J Clin Nutr 97, 195207.CrossRefGoogle ScholarPubMed
41. Cormier, H, Rudkowska, I, Paradis, AM et al. (2012) Association between polymorphisms in the fatty acid desaturase gene cluster and the plasma triacylglycerol response to an n-3 PUFA supplementation. Nutrients 4, 10261041.CrossRefGoogle Scholar
42. Porenta, SR, Ko, Y-A, Gruber, SB et al. (2013) Interaction of fatty acid genotype and diet on changes in colonic fatty acids in a Mediterranean diet intervention study. Cancer Prev Res 6, 11.CrossRefGoogle Scholar
43. Berendsen, A, Santoro, A, Pini, E et al. (2013) A parallel randomized trial on the effect of a healthful diet on inflammageing and its consequences in European elderly people: design of the NU-AGE dietary intervention study. Mech Ageing Dev 134, 523530.CrossRefGoogle ScholarPubMed
44. Cormier, H, Rudkowska, I, Lemieux, S et al. (2014) Effects of FADS and ELOVL polymorphisms on indexes of desaturase and elongase activities: results from a pre-post fish oil supplementation. Genes Nutr 9, 437.CrossRefGoogle ScholarPubMed
45. Li, S-W, Wang, J, Yang, Y et al. (2016) Polymorphisms in FADS1 and FADS2 alter plasma fatty acids and desaturase levels in type 2 diabetic patients with coronary artery disease. J Transl Med 14, 19.CrossRefGoogle ScholarPubMed
46. Albert, CM, Ma, J, Rifai, N et al. (2002) Prospective study of C-reactive protein, homocysteine, and plasma lipid levels as predictors of sudden cardiac death. Circulation 105, 25952599.CrossRefGoogle ScholarPubMed
47. Flock, MR, Skulas-Ray, AC, Harris, WS et al. (2013) Determinants of erythrocyte omega-3 fatty acid content in response to fish oil supplementation: a dose–response randomized controlled trial. J Am Heart Assoc 2, e000513.CrossRefGoogle ScholarPubMed
48. Raatz, SK, Rosenberger, TA, Johnson, LK et al. (2013) Dose-dependent consumption of farmed Atlantic salmon (Salmo salar) increases plasma phospholipid n-3 fatty acids differentially. J Acad Nutr Diet 113, 282287.CrossRefGoogle ScholarPubMed
49. Baylin, A, Ruiz-Narvaez, E, Kraft, P et al. (2007) α-Linolenic acid, Δ6-desaturase gene polymorphism, and the risk of nonfatal myocardial infarction. Am J Clin Nutr 85, 554560.CrossRefGoogle ScholarPubMed
50. Malerba, G, Schaeffer, L, Xumerle, L et al. (2008) SNPs of the FADS gene cluster are associated with polyunsaturated fatty acids in a cohort of patients with cardiovascular disease. Lipids 43, 289299.CrossRefGoogle Scholar
51. Rzehak, P, Heinrich, J, Klopp, N et al. (2009) Evidence for an association between genetic variants of the fatty acid desaturase 1 fatty acid desaturase 2 (FADS1 FADS2) gene cluster and the fatty acid composition of erythrocyte membranes. Br J Nutr 101, 2026.CrossRefGoogle ScholarPubMed
52. Mathias, RA, Vergara, C, Gao, L et al. (2010) FADS genetic variants and omega-6 polyunsaturated fatty acid metabolism in a homogeneous island population. J Lipid Res 51, 27662774.CrossRefGoogle Scholar
53. Zietemann, V, Kröger, J, Enzenbach, C et al. (2010) Genetic variation of the FADS1 FADS2 gene cluster and n-6 PUFA composition in erythrocyte membranes in the European Prospective Investigation into Cancer and Nutrition-Potsdam study. Br J Nutr 104, 17481759.CrossRefGoogle ScholarPubMed
54. Merino, DM, Johnston, H, Clarke, S et al. (2011) Polymorphisms in FADS1 and FADS2 alter desaturase activity in young Caucasian and Asian adults. Mol Genet Metab 103, 171178.CrossRefGoogle ScholarPubMed
55. Freemantle, E, Lalovic, A, Mechawar, N et al. (2012) Age and haplotype variations within FADS1 interact and associate with alterations in fatty acid composition in human male cortical brain tissue. PLoS ONE 7, e42696.CrossRefGoogle ScholarPubMed
56. Hong, SH, Kwak, JH, Paik, JK et al. (2013) Association of polymorphisms in FADS gene with age-related changes in serum phospholipid polyunsaturated fatty acids and oxidative stress markers in middle-aged nonobese men. Clin Interv Aging 8, 585596.Google ScholarPubMed
57. Roke, K, Ralston, JC, Abdelmagid, S et al. (2013) Variation in the FADS1/2 gene cluster alters plasma n-6 PUFA and is weakly associated with hsCRP levels in healthy young adults. Prostaglandins Leukot Essent Fatty Acids 89, 257263.CrossRefGoogle ScholarPubMed
58. Horiguchi, S, Nakayama, K, Iwamoto, S et al. (2016) Associations between a fatty acid desaturase gene polymorphism and blood arachidonic acid compositions in Japanese elderly. Prostaglandins Leukot Essent Fatty Acids 105, 914.CrossRefGoogle ScholarPubMed
59. Vaittinen, M, Walle, P, Kuosmanen, E et al. (2016) FADS2 genotype regulates delta-6 desaturase activity and inflammation in human adipose tissue. J Lipid Res 57, 5665.CrossRefGoogle ScholarPubMed
60. Schuchardt, JP, Köbe, T, Witte, V et al. (2016) Genetic variants of the FADS gene cluster are associated with erythrocyte membrane LC PUFA levels in patients with mild cognitive impairment. J Nutr Health Aging 20, 611620.CrossRefGoogle ScholarPubMed
61. Gieger, C, Geistlinger, L, Altmaier, E et al. (2008) genetics meets metabolomics: a genome-wide association study of metabolite profiles in human serum. PLoS Genet 4, e1000282.CrossRefGoogle ScholarPubMed
62. Tanaka, T, Shen, J, Abecasis, GR et al. (2009) Genome-wide association study of plasma polyunsaturated fatty acids in the InCHIANTI Study. PLoS Genet 5, e1000338.CrossRefGoogle ScholarPubMed
63. Kathiresan, S, Willer, CJ, Peloso, GM et al. (2009) Common variants at 30 loci contribute to polygenic dyslipidemia. Nat Genet 41, 5665.CrossRefGoogle ScholarPubMed
64. Guan, W, Steffen, BT, Lemaitre, RN et al. (2014) Genome-wide association study of plasma N6 polyunsaturated fatty acids within the cohorts for heart and aging research in genomic epidemiology consortium. Circ Cardiovasc Genet 7, 321331.CrossRefGoogle ScholarPubMed
65. Mozaffarian, D, Kabagambe, EK, Johnson, CO et al. (2015) Genetic loci associated with circulating phospholipid trans fatty acids: a meta-analysis of genome-wide association studies from the CHARGE Consortium. Am J Clin Nutr 101, 398406.CrossRefGoogle ScholarPubMed
66. Lu, Y, Feskens, EJ, Dolle, ME et al. (2010) Dietary n-3 and n-6 polyunsaturated fatty acid intake interacts with FADS1 genetic variation to affect total and HDL-cholesterol concentrations in the Doetinchem Cohort Study. Am J Clin Nutr 92, 258265.Google ScholarPubMed
67. Cormier, H, Rudkowska, I, Paradis, A-M et al. (2012) Association between polymorphisms in the fatty acid desaturase gene cluster and the plasma triacylglycerol response to an n-3 PUFA supplementation. Nutrients 4, 10261041.CrossRefGoogle Scholar
68. Al-Hilal, M, Alsaleh, A, Maniou, Z et al. (2013) Genetic variation at the FADS1-FADS2 gene locus influences delta-5 desaturase activity and LC-PUFA proportions after fish oil supplement. J Lipid Res 54, 542551.CrossRefGoogle ScholarPubMed
69. Roke, K & Mutch, DM (2014) The role of FADS1/2 polymorphisms on cardiometabolic markers and fatty acid profiles in young adults consuming fish oil supplements. Nutrients 6, 22902304.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Synthesis of long-chain PUFA from linoleic acid (LA) and alpha-linolenic acid (αLNA). Both LA (n-6) and αLNA (n-3) are elongated, desaturated and β-oxidised using the same enzyme system. AA, arachidonic acid.

Figure 1

Table 1. Candidate gene studies: associations between fatty acid desaturase SNP and fatty acid status and cardiovascular health

Figure 2

Table 2. Genome wide association studies: associations between fatty acid desaturase SNP and fatty acid status and cardiovascular health

Figure 3

Table 3. Diet–gene interactions: impact of dietary intakes/interventions on associations between fatty acid desaturase genotype and fatty acid status and cardiovascular health