IMR Press / RCM / Volume 25 / Issue 4 / DOI: 10.31083/j.rcm2504142
Open Access Review
Relationship between Polyunsaturated Fatty Acid Metabolism and Atherosclerosis
Show Less
1 Department of Vascular Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
2 Department of Vascular Surgery, Central Hospital Affiliated to Shandong First Medical University, 250013 Jinan, Shandong, China
*Correspondence: sdws188@163.com (Sheng Wang)
Rev. Cardiovasc. Med. 2024, 25(4), 142; https://doi.org/10.31083/j.rcm2504142
Submitted: 12 September 2023 | Revised: 4 November 2023 | Accepted: 10 November 2023 | Published: 9 April 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Multiple factors cause atherosclerosis, meaning its pathogenesis is complex, and has not been fully elucidated. Polyunsaturated fatty acids are a member of the fatty acid family, which are critical nutrients for mammalian growth and development. The types of polyunsaturated fatty acids ingested, their serum levels, and fatty acid desaturase can influence the atherosclerotic disease progression. The fatty acid desaturase gene cluster can regulate fatty acid desaturase activity and further affect atherosclerosis. This study reviewed the research progress on the effects of polyunsaturated fatty acids on atherosclerosis regulated by fatty acid desaturase and the relationship between genetic variants of the fatty acid desaturase gene cluster and atherosclerotic cardiovascular disease.

Keywords
atherosclerosis
polyunsaturated fatty acids metabolism
fatty acid desaturase gene cluster
fatty acid desaturase
1. Introduction

Atherosclerosis (AS) is a chronic inflammatory disease. Systemic or localized inflammation plays a central role in the onset and progression of AS, while inflammatory markers have been shown to predict cardiovascular disease (CVD) independently of traditional risk factors [1, 2, 3]. The downstream metabolites of polyunsaturated fatty acids (PUFAs), such as arachidonic acid (AA), prostaglandins (PG), thromboxanes (TXs), leukotriene (LTs), and other inflammatory factors, have an important impact on the development of AS. PUFAs and fatty acid desaturase (FADS), a key enzyme affecting its metabolism, play an equally important role in the pathogenesis of AS [4, 5].

As one of the essential dietary fatty acids in the human body, the content of PUFAs in the body reflects both the dietary intake and the fatty acid desaturase activity [5]. Previous studies have found that the type and amount of PUFAs being consumed are closely related to atherosclerotic cardiovascular disease (ASCVD). Prospective, observational studies support the role of omega-3 PUFAs in the primary prevention of ASCVD [6], although randomized controlled trials (RCTs) have often reached neutral conclusions [7, 8]. The potential impact of the intake of omega-6 PUFAs on ASCVD is also controversial, with previous studies suggesting that higher intakes of omega-6 PUFAs (predominantly linoleic acid) are associated with a lower risk of ASCVD [9, 10]. However, clinical studies have shown that excessive intake of ω-6 PUFAs (predominantly linoleic acid) leads to increased production of proinflammatory factors, which can lead to a higher risk of developing ASVCD [11, 12]. Thus, the roles of the omega-3 and omega-6 PUFAs in AS are complex and remain inconclusive. Studies on lipid metabolism disorders and inflammatory responses due to the regulation of gene expression have shown that there is as yet an undefined association between fatty acid desaturases, they are regulated by the FADS gene cluster, and AS, whereby fatty acid desaturase expression levels, as well as its activity, can differentially affect AS [13, 14, 15]. Based on existing studies, the effect of polyunsaturated fatty acid metabolism on atherosclerosis remains in the exploratory stage. This article aims to illustrate the relationship between polyunsaturated fatty acids, the regulation of fatty acid desaturases by the fatty acid desaturase gene cluster, and atherosclerosis.

2. Classification of Polyunsaturated Fatty Acids

Fatty acids are components of cell membrane phospholipids with specific functions, metabolism, and signaling roles. As a member of the fatty acid family, PUFAs are a crucial nutrient for mammalian growth and development, they are biologically active cellular components of membrane phospholipids, a substrate for signaling molecules, and a direct regulator of gene expression that can directly affect cellular function and the responsiveness of cells and tissues to signals [16, 17]. Moreover, PUFAs can regulate inflammatory processes by modulating signaling pathways [18, 19]. Fatty acids can be classified into short-chain, medium-chain, and long-chain fatty acids according to the number of carbon atoms they contain and into saturated fatty acids, monounsaturated fatty acids, and polyunsaturated fatty acids according to the number of carbon–carbon bonds they possess. PUFAs can be classified into two categories—ω-3 PUFA and ω-6 PUFA—according to the position of the double bond in their chemical structure and the principle of counting the position of the first double bond following the methyl carbon atom. The chemical structure of fatty acids is usually expressed as the number of carbon atoms, double bonds, and the position of the first double bond. For example, eicosapentaenoic acid (EPA) is expressed as 20:5 ω-3, meaning it contains 20 carbon atoms, five double bonds, and belongs to the ω-3 PUFA. In addition, fatty acids also have the Δ-coding system, which is different from the ω-coding system because the double bond position is counted from the carboxyl carbon atom.

3. Endogenous Metabolism of Polyunsaturated Fatty Acids

The human body cannot synthesize the amount of PUFAs needed for the body’s metabolism, meaning the ω-3 PUFA and ω-6 PUFA need to be supplemented through the diet, thus, they are referred to as essential fatty acids [20, 21]. The proportion of PUFAs in the diet is dominated by linoleic acid (LA) and α-linolenic acid (ALA), which are precursors of short-chain PUFAs that can be converted to biologically active long-chain PUFAs by Δ5 and Δ6 desaturase and elongase enzymes. The metabolism and metabolites of PUFAs will be summarized below according to the different types of PUFAs.

3.1 Endogenous Metabolism and Metabolites of Omega-3 Polyunsaturated Fatty Acids

ALA can be gradually converted to stearidonic acid (SDA), EPA, docosapentaenoic acid (DPA), and docosahexaenoic acid (DHA) in vivo by the action of the desaturase system and the elongation enzyme. The downstream metabolites of EPA and DHA are physiologically crucial for the organism. Specifically, EPA and DHA are catalyzed by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 oxidase (CYP450) to produce a series of specialized pro-resolving mediators (SPMs). SPMs include separate families of molecules: resolvins, protectins, and maresins. These act as stimulatory cell agonists, arresting neutrophil infiltration and enhancing macrophage uptake of apoptotic cells [22]. Resolvins can inhibit neutrophil infiltration, inhibit platelet aggregation, and reduce the production of proinflammatory factors [23]. Protectins can promote the expression and activity of antiapoptotic proteins and inhibit the expression and activity of proapoptotic proteins [24]. In addition, EPA is catalyzed by COX to produce thromboxane A3, which inhibits platelet aggregation, prostacyclin I3, which promotes vasodilatation, as well as prostaglandin E3 and leukotriene LT5, which has anti-inflammatory properties. Thus, both EPA and DHA metabolites can exert anti-inflammatory effects (Fig. 1).

Fig. 1.

Classification and metabolism of PUFAs. ALA, α-linolenic acid; SDA, stearidonic acid; ETA, eicosatetraenoic acid; EPA, eicosapentaenoic acid; DPA, docosapentaenoic acid; DHA, docosahexaenoic acid; LA, linoleic acid; GLA, gamma-linoleic acid; DGLA, dihomogamma linoleic acid; AA, arachidonic acid; DTA, docosatetraenoic acid; DPA, docosapentaenoic acid; ELOVL, elongation of very long fatty acids; FADS, fatty acid desaturase; COX, cyclooxygenase; LOX, lipoxygenase; CYP450, cytochrome p450; LT, leukotriene; PGE3, prostaglandin E3; PGI3, prostacyclin I3; TX, thromboxane synthase; PUFAs, polyunsaturated fatty acids; FADS2, fatty acid desaturase 2.

3.2 Endogenous Metabolism and Metabolites of Omega-6 Polyunsaturated Fatty Acids

LA can be converted to gamma-linolenic acid (GLA), double-high gamma-linolenic acid (dihomo-γ-linolenic acid, DGLA), arachidonic acid (AA), and DPA in vivo, under the action of the desaturase system and elongation enzyme. AA has essential biological functions and is catalyzed by COX to produce prostaglandin E2 and thromboxane A2, which promotes the inflammatory response, platelet aggregation, and vasoconstriction. AA can also produce proinflammatory factors from the leukotriene four-family, which, in the presence of LOX, play an essential role in the development and maintenance of the inflammatory response. In addition, AA is also catalyzed by LOX to produce lipoxin A4 (lipoxin) and lipoxin B4, which have a pro-resolving role in the abrogation of inflammatory responses (Fig. 1). However, in cardiovascular diseases, metabolites of AA exert deleterious effects that are proinflammatory, prothrombotic, and proplatelet aggregation to promote the development of atherosclerosis [25].

4. Effect of Polyunsaturated Fatty Acid Intake on Atherosclerotic Cardiovascular Disease

The role of PUFA intake in ASCVD has been controversial [6, 8, 26]. Although it has been demonstrated that ω-3 PUFAs can lower blood triglyceride levels and ω-6 PUFAs can lower blood total cholesterol levels, the results of the effects of ω-3 and ω-6 PUFAs on ASCVD in clinical practice have been inconsistent. Although clinical guidelines point to a positive effect from the use of icosapent ethyl and EPA in preventing ASCVD [27, 28, 29], there is a high degree of clinical heterogeneity in the design of previous studies and the final results. There is no solid evidence for using PUFAs to effectively treat or prevent ASCVD in patients with different backgrounds [30]. Therefore, the effects on ASCVD following the intake of PUFAs will be briefly summarized, as well as the reasons for the different results among the various studies.

4.1 Effect of Omega-3 Polyunsaturated Fatty Acids on Atherosclerotic Cardiovascular Disease

The controversy over the effect of ω-3 PUFAs on ASCVD lies in the fact that different clinical studies have yielded different results. REDUCE-IT, a multicenter RCT that included and followed more than 8000 patients with cardiovascular disease for almost five years, showed that compared to the placebo, patients who ingested EPA (4 g/d) underwent a significant reduction in the risk of cardiovascular death and nonfatal risk of myocardial infarction [31]. However, two similar RCTs (OMEMI (Omega-3 Fatty acids in Elderly with Myocardial Infarction), STRENGTH study (the Long-Term Outcomes Study to Assess Statin Residual Risk with Epanova in High Cardiovascular Risk Patients with Hypertriglyceridemia)) concluded that the intake of omega-3 PUFAs (EPA+DHA) did not reduce the risk of nonfatal myocardial infarction, stroke, or cardiovascular death [32, 33]. A systematic evaluation of the effects of ω-3 PUFAs on cardiovascular health showed that although the increased intake of EPA and DHA lowered plasma triglycerides and increased high-density lipoprotein (HDL) levels, they did not reduce the incidence of coronary artery disease (CAD), stroke, or other ASCVD events, nor the risk of death. In addition, increased intake of ALA reduces the risk of death from CAD, and increased intake of ALA has a preventive effect on ASCVD [34, 35, 36]. An RCT of the CVD population in the United States showed that increased omega-3 PUFAs (EPA+DHA) did not significantly reduce the risk of major cardiovascular events (myocardial infarction, stroke, and cardiovascular death) [8]. Circulating DHA, total omega-3, LA, and total omega-6 concentrations had no protective effect on the risk of cardiovascular disease in a Mendelian randomization study from the UK Biobank [37].

Although the above studies yielded different results, an analysis of these results from the different studies found that the composition of ω-3 PUFAs being consumed by the patients was different in each study, which may be one of the reasons for the controversy over the effects of ω-3 PUFAs on ASCVD. It has been suggested that EPA intake alone may be more effective in reducing the risk of cardiovascular disease than EPA+DHA [38], that serum EPA levels may need to reach a certain threshold to exert a preventive effect on ASCVD, and that a high dose (>1 g/d) significantly reduces the risk of cardiovascular events compared to low-dose EPA levels [39, 40]. In addition, when EPA was combined with DHA, higher DHA levels attenuated the preventive effect of EPA on ASCVD [30]. Therefore, the intake of EPA in ω-3 PUFAs and EPA blood levels in the study population may be important reasons for the observed variability in the effects of ω-3 PUFAs on ASCVD [41]. At present, the effect of omega-3 PUFAs on atherosclerosis remains a focus of clinical research. An ongoing RCT (NCT05365438) from Korea will assess the effects of combination therapy using atorvastatin and omega-3 PUFAs (EPA+DHA) compared with atorvastatin and ezetimibe combination therapy in diabetes mellitus type 2 (T2DM) patients with asymptomatic carotid atherosclerosis. The progression of carotid intima-media thickness and carotid artery plaques will be evaluated by three-dimensional (3D) carotid ultrasound. Another ongoing RCT (NCT05725486) from Croatia will investigate the influence of n-3 PUFAs enriched chicken on vascular and endothelial functions in a population of healthy young subjects and active athletes. Specifically, whether the intake of omega-3 PUFAs affects lipid profiles, oxidative stress, and inflammation. Further elucidating the mechanisms of vascular protection for omega-3 PUFAs may lead to new interventions for atherosclerosis in clinical practice.

4.2 Effect of Omega-6 Polyunsaturated Fatty Acids on Atherosclerotic Cardiovascular Disease

The effect of the intake of ω-6 PUFAs on ASCVD is equally controversial. A meta-analysis involving 44 prospective cohort studies showed that higher LA intake was associated with a reduced risk of death from CAD. It supported the potential long-term benefits of ω-6 PUFAs in reducing the risk of cardiovascular disease [9]. In addition, replacing a saturated fatty acid diet with an ω-6 PUFA (LA) may reduce the risk associated with CAD [42]. However, a systematic evaluation of the effects of ω-6 PUFAs on cardiovascular health suggests that increased intake of ω-6 PUFAs (LA+GLA) may reduce total cholesterol levels in the blood but does not significantly affect the risk of cardiovascular disease or mortality; therefore, the potential benefits in terms of reduced myocardial infarction remain to be demonstrated [43]. Another meta-analysis of randomized controlled trials showed that increasing the intake of omega-6 PUFAs (LA, GLA, DGLA, and AA) did not affect the incidence of myocardial infarction, stroke, CAD, and mortality [44]. An RCT studying the effects following the intake of different types of PUFAs from vegetable oils on cardiovascular disease in a population of hypercholesterolemic adults in China showed that after one year of measuring the intake of oleic acid (saturated fatty acid)-rich peanut oil, LA-rich corn oil, and ALA-rich blended oils, on fasting lipids, glucose, insulin concentration, and high sensitivity C-reactive protein levels of the different populations, the intake of different fatty acids did not affect cardiovascular risk factors [45].

Although some studies supported beneficial outcomes for cardiovascular disease following an increased intake of ω-6 PUFAs, several studies still produced conflicting results. The intake of ω-6 PUFAs may negatively impact ASCVD by causing an increase in downstream metabolites, such as proinflammatory 2-series prostaglandins and 4-series leukotriene. However, it is difficult to show a direct effect of ω-6 PUFAs on ASCVD when the impact of ω-6 PUFA metabolites on ASCVD is studied [42]. Therefore, more rigorous RCTs are needed to elucidate whether ω-6 PUFAs play a preventive or promotional role in ASCVD.

The competitive inhibition of enzymes between ω-3 and ω-6 PUFAs makes balancing the intake ratio between both ω-3 and ω-6 PUFAs complex. In addition, PUFA metabolism is also affected by genetic factors, which lead to alterations in the activity of fatty acid desaturase and the subsequent conversion of its products. Therefore, focusing only on the intake of a particular PUFA without considering the proportion of ω-3/ω-6 PUFAs in the diet and the influence of genetic factors on PUFA metabolism makes it difficult to explain the variability in the results from these studies.

5. Fatty Acid Desaturase and FADS Gene Cluster
5.1 Function and Classification of Fatty Acid Desaturases

The primary function of fatty acid desaturases is to dehydrogenate and introduce a double bond between the carbon atoms of the fatty acyl chain. In humans, membrane-bound fatty acid desaturases are known as “front-end” desaturases, and introduce a nascent double bond between an existing double bond, usually between the carboxyl group and the ninth carbon atom of the terminal methyl group, with front-end desaturation occurring at the Δ4, Δ5, Δ6, and Δ8 positions, while they are responsible for the endogenous biosynthesis of PUFAs. Therefore, fatty acid desaturases are categorized into four different types based on the location where desaturation occurs, namely, Δ4 fatty acid desaturases, Δ5 fatty acid desaturases, Δ6 fatty acid desaturases, and Δ8 fatty acid desaturases [46].

5.2 Structure of the Fatty Acid Desaturase Gene Cluster

The fatty acid desaturase (FADS) gene cluster that encodes fatty acid desaturase is located on human chromosome 11 (11q12-13.1) [47]. Data from the National Center of Biotechnology Information (NCBI) database demonstrates that FADS1, FADS2, and FADS3 are composed of 13 exons and 11 introns, and the total lengths of FADS1, FADS2, and FADS3 are 17.2, 39.1, and 18.7 kb, respectively (Fig. 2). FADS1 encodes Δ5 fatty acid desaturase; FADS2 encodes Δ4, Δ6, and Δ8 fatty acid desaturase; FADS3 encodes Δ9 and Δ13 fatty acid desaturase. Since the gene clusters have the same location and similar structures, it is hypothesized that they have evolved based on gene duplication, meaning they have acquired substrate specificity [46, 48, 49].

Fig. 2.

Function of FADS cluster and location of FADS cluster in chromosome 11. FEN1, flap endonuclease 1; FADS1, fatty acid desaturase 1; FADS2, fatty acid desaturase 2; FADS3, fatty acid desaturase 3; PUFA, polyunsaturated fatty acid; hs-CRP, high-sensitivity C-reactive protein.

5.3 Effect of Variants in the Fatty Acid Desaturase Gene Cluster on the Metabolic Activity of the Organism

Fatty acid desaturase is a critical enzyme in PUFA metabolism, and gene polymorphism in FADS affects the activity and function of fatty acid desaturase [50, 51], which in turn affects metabolic activities in the body, such as lipid concentrations, cardiovascular disease risk, pregnancy, cognitive function, Alzheimer’s disease, overweight, and type 2 diabetes mellitus [20, 52, 53].

The first exploratory study of variants in the FADS gene cluster found that single nucleotide polymorphisms (SNPs) in FADS1 and FADS2affect the composition of serum phospholipid fatty acids in healthy adults, as evidenced by changes in the levels of LA, DGLA, AA, and DPA [54]. Other studies have obtained consistent results. Mid-FADS gene cluster variants in Caucasian populations are associated with higher levels of precursor PUFAs and lower levels of AA and EPA, less inflammation, and lower risks of cardiovascular disease [55, 56]. A recent cohort study on FADS SNPs and fetal growth and development in pregnant women in China’s Han population showed that pregnant women with the FADS1/rs174448G, FADS3/rs174455T, and FADS3/rs174464A allele should be supplemented with DHA-rich ω-3 PUFAs exogenously during pregnancy due to the blockage of the endogenous synthesis of DHA [57].

These findings suggest that FADS gene cluster variants have a direct impact on PUFA metabolism, which in turn affects the risk of inflammation and cardiovascular disease, and that FADS gene cluster variants in pregnant women have a direct effect on PUFA levels in breast milk, which in turn affects infant growth and development. Several studies on the association between variants in the FADS gene cluster and child development have shown that the FADS SNPs are strongly associated with the synthesis of DHA and AA in vivo and can influence intelligence quotient (IQ) as well as cognitive ability in infants and young children, as shown by the fact that genetic restriction of endogenous PUFA synthesis results in poorer cognitive development in infants fed formulas that do not provide DHA and AA. This developmental deficit can be eliminated when infants are fed AA. European legislation mandating the addition of AA and DHA to infant formula may address developmental deficiencies due to insufficient synthesis of endogenous PUFAs in infants or mutations in the FADS gene cluster [20]. A case–control study reported that the FADS1/rs174556 genotype significantly increased the susceptibility to Alzheimer’s disease by regulating the efficiency of AA synthesis in ω-6 PUFAs [58]. However, this study had a small sample size and did not analyze AA derivatives in detail. Another case–control study reported an association between FADS1/rs174556, FADS2/rs174617, and obesity, by demonstrating that plasma levels of omega-6 PUFAs and AA were higher in overweight and obese patients; however, the difference in ω-3 PUFA levels was not significant. The study concluded that mutations in the FADS1/FADS2 locus could cause metabolic disorders and increase the risk of cardiovascular disease [59]. Genome-wide association studies (GWAS) in European and Asian populations have shown that FADS1/rs174546 is associated with reduced Δ5 fatty acid desaturase activity, obesity, and the risk of insulin resistance [60]. A study of the genetic etiology of type 2 diabetes mellitus showed that people with the FADS1/rs174546G allele had higher fasting insulin levels and higher HOMA-IR (an indicator to assess the level of insulin resistance) and that the increase in Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) was more pronounced with elevated plasma DGLA and AA levels [61]. FADS1/rs174547 effects on dyslipidemia have been reported in many races, and a study of the Chinese adult population showed that FADS1/rs174547 was significantly associated with high triglyceride levels in men and negatively associated with low density lipoprotein (LDL) cholesterol levels in women, thereby suggesting that it may be a sex-specific SNP locus [62].

The FADS gene cluster affects a variety of diseases by regulating the metabolism of PUFAs. Genetic variants in the FADS gene cluster are associated with an increased risk of dyslipidemia, obesity, and insulin resistance. These outcomes are strongly associated with atherosclerotic disease and produce a state of hypersecretion of proinflammatory cytokines, which increases the body’s susceptibility to atherosclerotic disease.

5.4 Effect of Fatty Acid Desaturase Gene Cluster Variation on Atherosclerotic Cardiovascular Disease

Previous studies on the association between differences in dietary PUFAs and the metabolism of PUFAs in humans and ASCVD suggest that both ω-3 and ω-6 PUFAs and their downstream metabolites can affect ASCVD. However, there are relatively few studies on the effect of the FADS gene cluster on ASCVD.

Genetic studies have shown that variants in FADS1, encoding the Δ5 fatty acid desaturase, and FADS2, encoding the Δ6 fatty acid desaturase, are most directly genetically linked to plasma PUFA levels and that the FADS gene cluster is the most important locus for influencing PUFA metabolism [56, 63, 64, 65]. Therefore, the FADS SNP can alter the accumulation of PUFAs and directly affect ASCVD. Genetic studies have provided a theoretical foundation for exploring the impact of the FADS gene cluster on the pathogenesis of ASCVD. Several studies have found a strong association between FADS gene cluster variants and ASCVD (Table 1, Ref. [10, 51, 66, 67, 68, 69, 70, 71, 72, 73]). For example, a Mendelian randomization study of European and American populations showed that plasma ALA and LA levels were higher in the FADS1/rs174547 sub-allele population and that this population was less likely to have CAD, stroke, and aortic stenosis, thereby suggesting that the FADS1 SNP drives the association between plasma levels of PUFAs and ASCVD [66]. An RCT on the effect of the FADS2 SNP on left ventricular remodeling after acute myocardial infarction (AMI) demonstrated that six months of high doses of ω-3 PUFAs after an AMI resulted in a significant attenuation of adverse left ventricular remodeling, noninfective myocardial fibrosis, and amelioration of the FADS2 rs1535GG-imposed hyperinflammatory response [51]. A case–control study in the Han Chinese population in northern China showed a strong association between the FADS3 SNP and CAD. The recessive G allele of FADS3 rs1000778 was associated with a higher risk of CAD, whereas the minor AA allele was associated with a lower risk of CAD; however, the plasma cholesterol and triglyceride levels remained similar between the two genotypes [67]. Aortic stenosis contributes to cardiovascular mortality and morbidity, and recent studies found that FADS1 SNP is associated with the risk of aortic stenosis. A GWAS of 44703 participants in the Genetic Epidemiology Research on Adult Health and Aging (GERA) cohort, one of the largest collections of aortic stenosis in the world, demonstrated that the FADS1/FADS2 locus variants (rs174547) are associated with aortic stenosis, while higher levels of AA and a higher ratio of AA/LA were associated with increased odds of calcification of the aortic valve leaflets [68]. A study of the relationship between localized PUFA in aortic valves and FADS genotypes by expression quantitative loci (eQTL) found that the minor C allele of rs174547, which corresponds to the protective genotype for aortic stenosis, was associated with higher FADS2 mRNA levels in calcified valve tissues, whereas FADS1 mRNA and other transcripts in proximity of the SNP were unaltered. In contrast, the Δ5 desaturase activity and the Δ6 desaturase activity were decreased. The authors concluded that the association between the FADS1 genotype and lower risk for aortic stenosis may implicate DHA and DHA-derived specialized pro-resolving mediators that contribute to a protective effect [74]. In addition, the diet–gene interaction for ASCVD is crucial. A recent cross-sectional population-based cohort study demonstrated that differential associations between the FADS1 locus variant and carotid–femoral pulse wave velocity (for assessing atherosclerosis) were observed depending on the intake of omega-3 PUFAs, with a high intake of omega-3 PUFAs attenuating the FADS1 locus variant-dependent associations. This suggests that the high intake of omega-3 PUFAs (EPA/DHA) may compensate for an unfavorable FADS1 locus genotype [75].

Table 1.Studies on the association between FADS gene cluster variation and atherosclerotic cardiovascular disease.
First author (year) Study type Intakes/evaluation indicators SNP Outcomes Results
Baylin (2007) [69] Case–control ALA levels in plasma and adipose tissue FADS2 promoter deletion MI No significant effect
Kwak (2011) [70] Case–control Plasma PUFAs and total cholesterol levels FADS1 rs174537 CAD FADS1 rs174537 T allele decreased plasma total cholesterol, AA/LA ratio, and decreased the risk of CAD
Li (2013) [71] Case–control Δ6 fatty acid desaturase activity (AA/LA) FADS1 rs174537 CAD The FADS1 rs174537T allele population has lower Δ6 fatty acid desaturase activity and reduced CAD risk; however, the G allele has increased Δ6 fatty acid desaturase activity and increased CAD risk
Hellstrand (2014) [72] Cohort LA, ALA intakes FADS1 rs174546 ASCVD Negative association of dietary ALA: LA ratio or ALA intake with ASCVD in sub-allele T carriers
Liu (2015) [73] Case–control EPA, DHA intakes FADS1 rs174547 CAD Lower dietary intake of EPA or DHA individuals associated with a higher risk of CAD
Wu (2017) [67] Case–control Blood lipid level FADS3 rs1000778 CAD The secondary allele AA was associated with a lower risk of CAD, whereas the recessive allele G was associated with a higher risk of CAD
Marklund (2019) [10] Meta Plasma LA, AA levels FADS1 rs174547 ASCVD LA was negatively associated with ASCVD in carriers of the common allele in purebloods and not in carriers of the minor allele
Yuan (2019) [66] Mendelian randomization Plasma fatty acid levels FADS1 rs174547 CAD, stroke, AS The FADS1 rs174547 sub-allele is negatively associated with ASCVD
Kwong (2019) [51] RCT ω-3 PUFAs intakes FADS2 rs1535 Left ventricular remodeling after AMI A high omega-3 PUFAs diet ameliorates the heightened inflammatory response associated with FADS2 rs1535GG, significantly attenuating adverse left ventricular remodeling and non-infarcted myocardial fibrosis
Chen (2020) [68] GWAS Plasma LA, AA levels FADS1/FADS2 rs174547 Aortic valve stenosis and calcification FADS1/FADS2 locus variants are associated with aortic stenosis and calcification, and AA level is strongly associated with aortic stenosis

RCT, randomized controlled trial; GWAS, genome-wide association study; MI, myocardial infarction; CAD, coronary artery disease; ASCVD, atherosclerotic cardiovascular disease; AMI, acute myocardial infarction; AS, aortic valve stenosis; FADS, fatty acid desaturase; ALA, α-linolenic acid; PUFAs, polyunsaturated fatty acids; AA, arachidonic acid; LA, linoleic acid; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid; SNP, single nucleotide polymorphism.

The effect of FADS gene cluster variants on ASCVD is mainly due to regulating the metabolism of PUFAs in the body, by altering fatty acid desaturase activity. In addition, exogenous supplementation of PUFAs can reverse the adverse effects of certain FADS gene cluster variants, suggesting that certain FADS gene-deficient disorders can be treated by increasing the intake of PUFAs that can prevent or alter the course of cardiovascular diseases.

5.5 Exploratory Studies of the Effects of FADS1 on Atherosclerosis

Various clinical data indicate that the FADS gene cluster has an essential effect on PUFAs metabolism, ASCVD, and glucose metabolism. Since FADS1 encodes Δ5 fatty acid desaturase and its metabolites, EPA and AA play essential roles in the inflammatory response and atherosclerosis. In a study investigating the effect of FADS1 on atherosclerosis and its mechanism of action, Powell et al. [14] showed that under high-fat dietary conditions, FADS1 knockout mice had weight loss, improved blood glucose, and reduced atherosclerotic plaques compared with wild-type mice. This study hypothesized that the low expression of FADS1 is associated with a reduced inflammatory response in the arterial wall, which is mainly manifested as a reduced AA/LA ratio in plasma and adipose tissue, and plays a positive role in preventing AS. Shuichi et al. [76] found that oral administration of a Δ5 fatty acid desaturase inhibitor to apolipoprotein E (ApoE) KO mice on a high-fat dietary background resulted in a reduction in atherosclerotic plaques, a decrease in the levels of AA and DHA, and an increase in the levels of DGLA. This finding is in general agreement with Powell’s conclusions. Gromovsky et al. [15] used antisense oligonucleotides (ASOs) to specifically knockdown FADS1 in the liver, adipose, and reticuloendothelial systems of low density lipoprotein receptor (LDLR) KO mice. However, they produced a different result, whereby the specific knockdown of the FADS1 in LDLR KO mice aggravated atherosclerosis and the hepatic inflammatory response, which differed from the previous two studies. The authors highlight several possibilities for their differing conclusions. The study by Powell et al. [14] did not validate the hepatic levels of FADS1 expression. Second, the Powell et al. [14] mouse model produced sub-alleles; therefore, the functionality of FADS1 was not completely lost. In addition, the different genetic backgrounds of the two mice may be another reason for the inconsistent results. The study also noted that a diet of ω-3 PUFAs (ALA+SDA) reduced the area of the atherosclerotic plaque in the aortic root of the mice compared with a saturated fatty acid diet.

Studies have shown that mice of different genetic backgrounds present different outcomes after FADS1 knockdown and that the percentage of PUFAs in the diet affects the survival and progression of atherosclerosis in mice. The downstream metabolites and signal transduction pathways regulated by FADS1 have yet to be thoroughly studied. Whether FADS1 can directly affect AS by affecting the production of inflammatory factors, such as PGs, LTs, and TXs is worthy of further exploration.

6. Fatty Acid Metabolism in Macrophages and Atherosclerosis

Monocytes/macrophages play a crucial role in the development and progression of ASCVD and the deterioration of advanced lesions. Intimal infiltration and modification of plasma-derived lipoproteins and their uptake, mainly by macrophages, with the ensuing formation of lipid-filled foam cells, can initiate atherosclerotic lesion formation and alter the efferocytotic removal of apoptotic cells and foam cells, which contributes to the progression of atherosclerotic lesions [77]. One of the properties of macrophages is the ability to dynamically regulate PUFAs metabolism during the acute phase of the inflammatory stimulus-response and inflammatory regression. This dynamic regulation of PUFAs metabolism may contribute to macrophage plasticity, in particular by controlling the balance between pro- and anti-inflammatory mediators [78]. Therefore, PUFAs metabolism in macrophages is closely related to atherosclerosis.

Liver X receptors (LXRs) are nuclear receptors that can participate in regulating cholesterol homeostasis and fatty acid metabolism and are essential in controlling inflammation and innate immune responses. A recent study investigating the treatment of human primary monocyte–macrophages with LXR agonists found that PUFAs synthesis was affected by the significant induction of Δ5 and Δ6 desaturases (FADS1 and FADS2, respectively) after LXR agonist treatment alongside the induction of acyl-CoA synthase long-chain family member 3 (ACSL3) and the fatty acid elongase 5 (ELOVL5). In addition, LXR agonist treatment of ApoE-/- mice led to significant changes in the PUFAs profile in atherosclerotic arteries with increases in both the AA/LA and the DHA/EPA ratios, while also being associated with the decreased expression of proinflammatory genes, such as Cox2 and Il1β. Therefore, local production of PUFAs and derived lipid mediators in macrophages triggered by LXR within the atheromatous plaque could affect inflammation and the development of atherosclerosis [79]. Another study demonstrated that PUFAs can alter the micro RNA (miRNA) profiles of macrophages. When macrophages are enriched with either DHA or AA, they alter the expression of many miRNAs closely associated with inflammation, thus, suggesting that PUFAs are regulators of macrophage phenotypes and the inflammatory response [80]. Macrophages can sense internal and environmental changes and subsequently adapt their phenotype. This sequence is commonly named macrophage activation (classical or alternative) or polarization, and the most common are M1 and M2 polarization. In vitro stimulation of macrophages with interferon-gamma (IFNg) and lipopolysaccharide (LPS) leads to M1 polarization, while interleukin-4 (IL-4) or IL-13 treatment induces alternative activation of M2 polarization. M1 macrophages are characterized by a proinflammatory phenotype with intense bactericidal activities, while M2 macrophages are involved in the resolution of inflammation and in tissue remodeling and repair. The current view is that an unbalanced interplay between M1 and M2 macrophages could contribute to atherogenesis [81, 82]. Fatty acid synthesis in macrophages is activated during M1 polarization, and excess fatty acid and triglyceride synthesis promote foam cell formation with proatherogenic effects [83]. A study demonstrated that peroxisome proliferator-activated receptor gamma (PPARg), a nuclear receptor activated by fatty acid derivatives that control fatty acid oxidation, was required for M2 polarization [84]. A recent study found that FADS1 knockdown in macrophages was associated with a tendency toward M1 and away from M2 polarization. Specifically, FADS1 knockdown resulted in augmented LPS-driven proinflammatory gene expression yet was associated with diminished IL-4-driven alternative activation gene signatures. Therefore, FADS1 reciprocally regulates M1 and M2 polarization programs in the macrophage [15]. The metabolism of fatty acids in macrophages involves M1 and M2 polarization processes, while FADS1 affects the metabolism of PUFAs in macrophages, affecting the dynamic balance between proinflammatory and pro-resolving mediators. Future studies should focus on how macrophages regulate their metabolism of PUFAs, and more studies are needed to assess the impact of FADS1 on atherosclerosis development under various metabolic conditions.

7. Conclusions

Atherosclerosis results from a multifactorial combination of factors. The development and progression of atherosclerosis are associated with PUFAs, fatty acid desaturase, and FADS gene cluster variation (Fig. 3). Clinical trials to establish the necessary ω-3 to ω-6 ratio for optimum CVD health should consider ethnic background, genetic predisposition, biochemical markers, and dietary habits. The effects of dietary intake of ω-3 PUFAs and ω-6 PUFAs on atherosclerotic cardiovascular disease are controversial and need to be validated in more rigorous randomized controlled trials. Moreover, it is imperative to understand the differences between the impact of the formulation and the distinct effects of the ω-3/ω-6 PUFAs on lipid oxidation, inflammation, membrane structure/organization, cholesterol domain formation, and endothelial cell function. FADS gene cluster variants have an essential impact on atherosclerotic cardiovascular diseases and may be potential therapeutic targets to prevent and treat these diseases. FADS genotypes may be helpful for future stratification and targeting of dietary recommendations in individuals carrying the FADS minor allele. PUFAs and their metabolites are regulated by various factors, including FADS gene clusters and dietary background; thus, future in-depth studies of the association between PUFA and atherosclerosis will enrich the knowledge of the pathogenesis of atherosclerosis and provide new therapies for its treatment.

Fig. 3.

The progression of atherosclerosis is associated with PUFAs intake, fatty acid desaturase, and FADS gene cluster variation. Intake of polyunsaturated fatty acids has an important impact on atherosclerosis, and the effect of FADS gene cluster variants on ASCVD, mainly by regulating the metabolism of PUFAs in the body but more precisely by altering fatty acid desaturase activity. LXRs and FADS1 expression regulate the metabolism of PUFAs in macrophages, while the metabolites of PUFAs in macrophages are also involved in the formation and progression of atherosclerosis. PUFAs, polyunsaturated fatty acids; FADS, fatty acid desaturase; ASCVD, atherosclerotic cardiovascular disease; LXRs, Liver X receptors; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid; AA, arachidonic acid.

Author Contributions

QLL drafted the manuscript and participated in its design and so on. ZXL, DW and SW conceived of the study, and participated in its coordination and helped to draft the manuscript and so on. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We would like to express our gratitude to all those who helped us during the writing of this manuscript. Thanks to all the peer reviewers for their opinions and suggestions.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Henein MY, Vancheri S, Longo G, Vancheri F. The Role of Inflammation in Cardiovascular Disease. International Journal of Molecular Sciences. 2022; 23: 12906.
[2]
Ministrini S, Carbone F, Montecucco F. Updating concepts on atherosclerotic inflammation: From pathophysiology to treatment. European Journal of Clinical Investigation. 2021; 51: e13467.
[3]
Ridker PM, Koenig W, Kastelein JJ, Mach F, Lüscher TF. Has the time finally come to measure hsCRP universally in primary and secondary cardiovascular prevention? European Heart Journal. 2018; 39: 4109–4111.
[4]
Tosi F, Sartori F, Guarini P, Olivieri O, Martinelli N. Delta-5 and delta-6 desaturases: crucial enzymes in polyunsaturated fatty acid-related pathways with pleiotropic influences in health and disease. Advances in Experimental Medicine and Biology. 2014; 824: 61–81.
[5]
Schulze MB, Minihane AM, Saleh RNM, Risérus U. Intake and metabolism of omega-3 and omega-6 polyunsaturated fatty acids: nutritional implications for cardiometabolic diseases. The Lancet. Diabetes & Endocrinology. 2020; 8: 915–930.
[6]
Harris WS, Zotor FB. n-3 Fatty acids and risk for fatal coronary disease. The Proceedings of the Nutrition Society. 2019; 78: 526–531.
[7]
ASCEND Study Collaborative Group, Bowman L, Mafham M, Wallendszus K, Stevens W, Buck G, et al. Effects of n-3 Fatty Acid Supplements in Diabetes Mellitus. The New England Journal of Medicine. 2018; 379: 1540–1550.
[8]
Manson JE, Cook NR, Lee IM, Christen W, Bassuk SS, Mora S, et al. Marine n-3 Fatty Acids and Prevention of Cardiovascular Disease and Cancer. The New England Journal of Medicine. 2019; 380: 23–32.
[9]
Li J, Guasch-Ferré M, Li Y, Hu FB. Dietary intake and biomarkers of linoleic acid and mortality: systematic review and meta-analysis of prospective cohort studies. The American Journal of Clinical Nutrition. 2020; 112: 150–167.
[10]
Marklund M, Wu JHY, Imamura F, Del Gobbo LC, Fretts A, de Goede J, et al. Biomarkers of Dietary Omega-6 Fatty Acids and Incident Cardiovascular Disease and Mortality. Circulation. 2019; 139: 2422–2436.
[11]
DiNicolantonio JJ, O’Keefe JH. Importance of maintaining a low omega-6/omega-3 ratio for reducing inflammation. Open Heart. 2018; 5: e000946.
[12]
DiNicolantonio JJ, O’Keefe JH. Omega-6 vegetable oils as a driver of coronary heart disease: the oxidized linoleic acid hypothesis. Open Heart. 2018; 5: e000898.
[13]
Panda C, Varadharaj S, Voruganti VS. PUFA, genotypes and risk for cardiovascular disease. Prostaglandins, Leukotrienes, and Essential Fatty Acids. 2022; 176: 102377.
[14]
Powell DR, Gay JP, Smith M, Wilganowski N, Harris A, Holland A, et al. Fatty acid desaturase 1 knockout mice are lean with improved glycemic control and decreased development of atheromatous plaque. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy. 2016; 9: 185–199.
[15]
Gromovsky AD, Schugar RC, Brown AL, Helsley RN, Burrows AC, Ferguson D, et al. Δ-5 Fatty Acid Desaturase FADS1 Impacts Metabolic Disease by Balancing Proinflammatory and Proresolving Lipid Mediators. Arteriosclerosis, Thrombosis, and Vascular Biology. 2018; 38: 218–231.
[16]
Calder PC. Functional Roles of Fatty Acids and Their Effects on Human Health. JPEN. Journal of Parenteral and Enteral Nutrition. 2015; 39: 18S–32S.
[17]
Jasani B, Simmer K, Patole SK, Rao SC. Long chain polyunsaturated fatty acid supplementation in infants born at term. The Cochrane Database of Systematic Reviews. 2017; 3: CD000376.
[18]
Oppedisano F, Macrì R, Gliozzi M, Musolino V, Carresi C, Maiuolo J, et al. The Anti-Inflammatory and Antioxidant Properties of n-3 PUFAs: Their Role in Cardiovascular Protection. Biomedicines. 2020; 8: 306.
[19]
Calder PC. n-3 PUFA and inflammation: from membrane to nucleus and from bench to bedside. The Proceedings of the Nutrition Society. 2020; 1–13.
[20]
Koletzko B, Reischl E, Tanjung C, Gonzalez-Casanova I, Ramakrishnan U, Meldrum S, et al. FADS1 and FADS2 Polymorphisms Modulate Fatty Acid Metabolism and Dietary Impact on Health. Annual Review of Nutrition. 2019; 39: 21–44.
[21]
Bokov DO, Makarenko MA, Paleeva MA, Rusinova MB, Bessonov VV. Squid fat as a promising source of polyunsaturated fatty acids. Voprosy Pitaniia. 2022; 91: 116–123.
[22]
Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature. 2014; 510: 92–101.
[23]
Capó X, Martorell M, Busquets-Cortés C, Tejada S, Tur JA, Pons A, et al. Resolvins as proresolving inflammatory mediators in cardiovascular disease. European Journal of Medicinal Chemistry. 2018; 153: 123–130.
[24]
Asatryan A, Bazan NG. Molecular mechanisms of signaling via the docosanoid neuroprotectin D1 for cellular homeostasis and neuroprotection. The Journal of Biological Chemistry. 2017; 292: 12390–12397.
[25]
Yamaguchi A, Botta E, Holinstat M. Eicosanoids in inflammation in the blood and the vessel. Frontiers in Pharmacology. 2022; 13: 997403.
[26]
Hu Y, Hu FB, Manson JE. Marine Omega-3 Supplementation and Cardiovascular Disease: An Updated Meta-Analysis of 13 Randomized Controlled Trials Involving 127 477 Participants. Journal of the American Heart Association. 2019; 8: e013543.
[27]
Kleindorfer DO, Towfighi A, Chaturvedi S, Cockroft KM, Gutierrez J, Lombardi-Hill D, et al. 2021 Guideline for the Prevention of Stroke in Patients With Stroke and Transient Ischemic Attack: A Guideline From the American Heart Association/American Stroke Association [published correction appears in Stroke. 2021; 52: e483–e484]. Stroke. 2021; 52: e364–e467.
[28]
Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, et al. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines [published correction appears in Journal of the American College of Cardiology. 2023; 81: 1551]. Journal of the American College of Cardiology. 2022; 79: e263–e421.
[29]
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk [published correction appears in European Heart Journal. 2020; 41: 4255]. European Heart Journal. 2020; 41: 111–188.
[30]
Li JJ, Dou KF, Zhou ZG, Zhao D, Ye P, Zhao JJ, et al. Role of omega-3 fatty acids in the prevention and treatment of cardiovascular Diseases: A consensus statement from the Experts’ Committee Of National Society Of Cardiometabolic Medicine. Frontiers in Pharmacology. 2022; 13: 1069992.
[31]
Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, et al. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. The New England Journal of Medicine. 2019; 380: 11–22.
[32]
Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, et al. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA. 2020; 324: 2268–2280.
[33]
Kalstad AA, Myhre PL, Laake K, Tveit SH, Schmidt EB, Smith P, et al. Effects of n-3 Fatty Acid Supplements in Elderly Patients After Myocardial Infarction: A Randomized, Controlled Trial. Circulation. 2021; 143: 528–539.
[34]
Abdelhamid AS, Brown TJ, Brainard JS, Biswas P, Thorpe GC, Moore HJ, et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. The Cochrane Database of Systematic Reviews. 2018; 11: CD003177.
[35]
Skulas-Ray AC, Wilson PWF, Harris WS, Brinton EA, Kris-Etherton PM, Richter CK, et al. Omega-3 Fatty Acids for the Management of Hypertriglyceridemia: A Science Advisory From the American Heart Association. Circulation. 2019; 140: e673–e691.
[36]
Lázaro I, Rueda F, Cediel G, Ortega E, García-García C, Sala-Vila A, et al. Circulating Omega-3 Fatty Acids and Incident Adverse Events in Patients With Acute Myocardial Infarction. Journal of the American College of Cardiology. 2020; 76: 2089–2097.
[37]
Borges MC, Haycock PC, Zheng J, Hemani G, Holmes MV, Davey Smith G, et al. Role of circulating polyunsaturated fatty acids on cardiovascular diseases risk: analysis using Mendelian randomization and fatty acid genetic association data from over 114,000 UK Biobank participants. BMC Medicine. 2022; 20: 210.
[38]
Khan SU, Lone AN, Khan MS, Virani SS, Blumenthal RS, Nasir K, et al. Effect of omega-3 fatty acids on cardiovascular outcomes: A systematic review and meta-analysis. eClinicalMedicine. 2021; 38: 100997.
[39]
Jaca A, Durão S, Harbron J. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. South African Medical Journal. 2020; 110: 1158–1159.
[40]
Lombardi M, Chiabrando JG, Vescovo GM, Bressi E, Del Buono MG, Carbone S, et al. Impact of Different Doses of Omega-3 Fatty Acids on Cardiovascular Outcomes: a Pairwise and Network Meta-analysis. Current Atherosclerosis Reports. 2020; 22: 45.
[41]
Sherratt SCR, Libby P, Budoff MJ, Bhatt DL, Mason RP. Role of Omega-3 Fatty Acids in Cardiovascular Disease: the Debate Continues. Current Atherosclerosis Reports. 2023; 25: 1–17.
[42]
Sanders TAB. Omega-6 Fatty Acids and Cardiovascular Disease. Circulation. 2019; 139: 2437–2439.
[43]
Hooper L, Al-Khudairy L, Abdelhamid AS, Rees K, Brainard JS, Brown TJ, et al. Omega-6 fats for the primary and secondary prevention of cardiovascular disease. The Cochrane Database of Systematic Reviews. 2018; 7: CD011094.
[44]
Mazidi M, Shekoohi N, Katsiki N, Banach M. Omega-6 fatty acids and the risk of cardiovascular disease: insights from a systematic review and meta-analysis of randomized controlled trials and a Mendelian randomization study. Archives of Medical Science. 2021; 18: 466–479.
[45]
Chen CG, Wang P, Zhang ZQ, Ye YB, Zhuo SY, Zhou Q, et al. Effects of plant oils with different fatty acid composition on cardiovascular risk factors in moderately hypercholesteremic Chinese adults: a randomized, double-blinded, parallel-designed trial. Food & Function. 2020; 11: 7164–7174.
[46]
Zhang JY, Kothapalli KSD, Brenna JT. Desaturase and elongase-limiting endogenous long-chain polyunsaturated fatty acid biosynthesis. Current Opinion in Clinical Nutrition and Metabolic Care. 2016; 19: 103–110.
[47]
Marquardt A, Stöhr H, White K, Weber BH. cDNA cloning, genomic structure, and chromosomal localization of three members of the human fatty acid desaturase family. Genomics. 2000; 66: 175–183.
[48]
Nakamura MT, Nara TY. Structure, function, and dietary regulation of delta6, delta5, and delta9 desaturases. Annual Review of Nutrition. 2004; 24: 345–376.
[49]
Nagao K, Murakami A, Umeda M. Structure and Function of Δ9-Fatty Acid Desaturase. Chemical & Pharmaceutical Bulletin. 2019; 67: 327–332.
[50]
Tsurutani Y, Inoue K, Sugisawa C, Saito J, Omura M, Nishikawa T. Increased Serum Dihomo-γ-linolenic Acid Levels Are Associated with Obesity, Body Fat Accumulation, and Insulin Resistance in Japanese Patients with Type 2 Diabetes. Internal Medicine. 2018; 57: 2929–2935.
[51]
Kwong RY, Heydari B, Ge Y, Abdullah S, Fujikura K, Kaneko K, et al. Genetic profiling of fatty acid desaturase polymorphisms identifies patients who may benefit from high-dose omega-3 fatty acids in cardiac remodeling after acute myocardial infarction-Post-hoc analysis from the OMEGA-REMODEL randomized controlled trial. PLoS ONE. 2019; 14: e0222061.
[52]
Chen J, Ruan X, Sun Y, Li X, Yuan S, Larsson SC. Plasma phospholipid arachidonic acid in relation to non-alcoholic fatty liver disease: Mendelian randomization study. Nutrition. 2023; 106: 111910.
[53]
Huang PC, Cheng H, Su YT, Huang MC, Hsu CC, Hwang SJ, et al. Interaction among dietary n-3 and n-6 polyunsaturated fatty acid intake, fatty acid desaturase 2 genetic variants, and low-density lipoprotein cholesterol levels in type 2 diabetes patients. Journal of Diabetes Investigation. 2023; 14: 297–308.
[54]
Schaeffer L, Gohlke H, Müller M, Heid IM, Palmer LJ, Kompauer I, et al. Common genetic variants of the FADS1 FADS2 gene cluster and their reconstructed haplotypes are associated with the fatty acid composition in phospholipids. Human Molecular Genetics. 2006; 15: 1745–1756.
[55]
Lemaitre RN, Tanaka T, Tang W, Manichaikul A, Foy M, Kabagambe EK, et al. Genetic loci associated with plasma phospholipid n-3 fatty acids: a meta-analysis of genome-wide association studies from the CHARGE Consortium. PLoS Genetics. 2011; 7: e1002193.
[56]
O’Neill CM, Minihane AM. The impact of fatty acid desaturase genotype on fatty acid status and cardiovascular health in adults. The Proceedings of the Nutrition Society. 2017; 76: 64–75.
[57]
Li P, Chen Y, Song J, Yan L, Tang T, Wang R, et al. Maternal DHA-rich n-3 PUFAs supplementation interacts with FADS genotypes to influence the profiles of PUFAs in the colostrum among Chinese Han population: a birth cohort study. Nutrition & Metabolism. 2022; 19: 48.
[58]
Hammouda S, Ghzaiel I, Khamlaoui W, Hammami S, Mhenni SY, Samet S, et al. Genetic variants in FADS1 and ELOVL2 increase level of arachidonic acid and the risk of Alzheimer’s disease in the Tunisian population. Prostaglandins, Leukotrienes, and Essential Fatty Acids. 2020; 160: 102159.
[59]
Khamlaoui W, Mehri S, Hammami S, Hammouda S, Chraeif I, Elosua R, et al. Association Between Genetic Variants in FADS1-FADS2 and ELOVL2 and Obesity, Lipid Traits, and Fatty Acids in Tunisian Population. Clinical and Applied Thrombosis/Hemostasis. 2020; 26: 1076029620915286.
[60]
Albracht-Schulte K, Kalupahana NS, Ramalingam L, Wang S, Rahman SM, Robert-McComb J, et al. Omega-3 fatty acids in obesity and metabolic syndrome: a mechanistic update. The Journal of Nutritional Biochemistry. 2018; 58: 1–16.
[61]
Himanshu D, Ali W, Wamique M. Type 2 diabetes mellitus: pathogenesis and genetic diagnosis. Journal of Diabetes and Metabolic Disorders. 2020; 19: 1959–1966.
[62]
Guo H, Zhang L, Zhu C, Yang F, Wang S, Zhu S, et al. A Single Nucleotide Polymorphism in the FADS1 Gene is Associated with Plasma Fatty Acid and Lipid Profiles and Might Explain Gender Difference in Body Fat Distribution. Lipids in Health and Disease. 2017; 16: 67.
[63]
Juan J, Huang H, Jiang X, Ardisson Korat AV, Song M, Sun Q, et al. Joint effects of fatty acid desaturase 1 polymorphisms and dietary polyunsaturated fatty acid intake on circulating fatty acid proportions. The American Journal of Clinical Nutrition. 2018; 107: 826–833.
[64]
de Toro-Martín J, Guénard F, Rudkowska I, Lemieux S, Couture P, Vohl MC. A common variant in ARHGEF10 alters delta-6 desaturase activity and influence susceptibility to hypertriglyceridemia. Journal of Clinical Lipidology. 2018; 12: 311–320.e3.
[65]
Marklund M, Morris AP, Mahajan A, Ingelsson E, Lindgren CM, Lind L, et al. Genome-Wide Association Studies of Estimated Fatty Acid Desaturase Activity in Serum and Adipose Tissue in Elderly Individuals: Associations with Insulin Sensitivity. Nutrients. 2018; 10: 1791.
[66]
Yuan S, Bäck M, Bruzelius M, Mason AM, Burgess S, Larsson S. Plasma Phospholipid Fatty Acids, FADS1 and Risk of 15 Cardiovascular Diseases: A Mendelian Randomisation Study. Nutrients. 2019; 11: 3001.
[67]
Wu Y, Zeng L, Chen X, Xu Y, Ye L, Qin L, et al. Association of the FADS gene cluster with coronary artery disease and plasma lipid concentrations in the northern Chinese Han population. Prostaglandins, Leukotrienes, and Essential Fatty Acids. 2017; 117: 11–16.
[68]
Chen HY, Cairns BJ, Small AM, Burr HA, Ambikkumar A, Martinsson A, et al. Association of FADS1/2 Locus Variants and Polyunsaturated Fatty Acids With Aortic Stenosis. JAMA Cardiology. 2020; 5: 694–702.
[69]
Baylin A, Ruiz-Narvaez E, Kraft P, Campos H. alpha-Linolenic acid, Delta6-desaturase gene polymorphism, and the risk of nonfatal myocardial infarction. The American Journal of Clinical Nutrition. 2007; 85: 554–560.
[70]
Kwak JH, Paik JK, Kim OY, Jang Y, Lee SH, Ordovas JM, et al. FADS gene polymorphisms in Koreans: association with ω6 polyunsaturated fatty acids in serum phospholipids, lipid peroxides, and coronary artery disease. Atherosclerosis. 2011; 214: 94–100.
[71]
Li SW, Lin K, Ma P, Zhang ZL, Zhou YD, Lu SY, et al. 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. 2013; 8: e55869.
[72]
Hellstrand S, Ericson U, Gullberg B, Hedblad B, Orho-Melander M, Sonestedt E. Genetic variation in FADS1 has little effect on the association between dietary PUFA intake and cardiovascular disease. The Journal of Nutrition. 2014; 144: 1356–1363.
[73]
Liu F, Li Z, Lv X, Ma J. Dietary n-3 polyunsaturated fatty acid intakes modify the effect of genetic variation in fatty acid desaturase 1 on coronary artery disease. PLoS ONE. 2015; 10: e0121255.
[74]
Plunde O, Larsson SC, Artiach G, Thanassoulis G, Carracedo M, Franco-Cereceda A, et al. FADS1 (Fatty Acid Desaturase 1) Genotype Associates With Aortic Valve FADS mRNA Expression, Fatty Acid Content and Calcification. Circulation. Genomic and Precision Medicine. 2020; 13: e002710.
[75]
Bäck M, Xhaard C, Rouget R, Thuillier Q, Plunde O, Larsson SC, et al. Fatty acid desaturase genetic variations and dietary omega-3 fatty acid intake associate with arterial stiffness. European Heart Journal Open. 2022; 2: oeac016.
[76]
Takagahara S, Shinohara H, Itokawa S, Satomi Y, Ando A, Yamamoto T, et al. A Novel Orally Available Delta-5 Desaturase Inhibitor Prevents Atherosclerotic Lesions Accompanied by Changes in Fatty Acid Composition and Eicosanoid Production in ApoE Knockout Mice. The Journal of Pharmacology and Experimental Therapeutics. 2019; 371: 290–298.
[77]
Bäck M, Yurdagul A, Jr, Tabas I, Öörni K, Kovanen PT. Inflammation and its resolution in atherosclerosis: mediators and therapeutic opportunities. Nature Reviews. Cardiology. 2019; 16: 389–406.
[78]
Ménégaut L, Jalil A, Thomas C, Masson D. Macrophage fatty acid metabolism and atherosclerosis: The rise of PUFAs. Atherosclerosis. 2019; 291: 52–61.
[79]
Varin A, Thomas C, Ishibashi M, Ménégaut L, Gautier T, Trousson A, et al. Liver X receptor activation promotes polyunsaturated fatty acid synthesis in macrophages: relevance in the context of atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2015; 35: 1357–1365.
[80]
Roessler C, Kuhlmann K, Hellwing C, Leimert A, Schumann J. Impact of Polyunsaturated Fatty Acids on miRNA Profiles of Monocytes/Macrophages and Endothelial Cells-A Pilot Study. International Journal of Molecular Sciences. 2017; 18: 284.
[81]
Tabas I, Bornfeldt KE. Macrophage Phenotype and Function in Different Stages of Atherosclerosis. Circulation Research. 2016; 118: 653–667.
[82]
Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, et al. Macrophage plasticity, polarization, and function in health and disease. Journal of Cellular Physiology. 2018; 233: 6425–6440.
[83]
Ménégaut L, Thomas C, Lagrost L, Masson D. Fatty acid metabolism in macrophages: a target in cardio-metabolic diseases. Current Opinion in Lipidology. 2017; 28: 19–26.
[84]
Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, et al. Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007; 447: 1116–1120.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top