Skip to main content
Erschienen in: European Journal of Nutrition 7/2022

Open Access 14.06.2022 | Original Contribution

The FADS1 genotypes modify the effect of linoleic acid-enriched diet on adipose tissue inflammation via pro-inflammatory eicosanoid metabolism

verfasst von: Maija Vaittinen, Maria A. Lankinen, Pirjo Käkelä, Jyrki Ågren, Craig E. Wheelock, Markku Laakso, Ursula Schwab, Jussi Pihlajamäki

Erschienen in: European Journal of Nutrition | Ausgabe 7/2022

Abstract

Purpose

Fatty acid desaturase (FADS) variants associate with fatty acid (FA) and adipose tissue (AT) metabolism and inflammation. Thus, the role of FADS1 variants in the regulation of dietary linoleic acid (LA)-induced effects on AT inflammation was investigated.

Methods

Subjects homozygotes for the TT and CC genotypes of the FADS1-rs174550 (TT, n = 25 and CC, n = 28) or -rs174547 (TT, n = 42 and CC, n = 28), were either recruited from the METabolic Syndrome In Men cohort to participate in an intervention with LA-enriched diet (FADSDIET) or from the Kuopio Obesity Surgery (KOBS) study. GC and LC–MS for plasma FA proportions and eicosanoid concentrations and AT gene expression for AT inflammatory score (AT-InSc) was determined.

Results

We observed a diet-genotype interaction between LA-enriched diet and AT-InSc in the FADSDIET. In the KOBS study, interleukin (IL)1 beta mRNA expression in AT was increased in subjects with the TT genotype and highest LA proportion. In the FADSDIET, n-6/LA proportions correlated positively with AT-InSc in those with the TT genotype but not with the CC genotype after LA-enriched diet. Specifically, LA- and AA-derived pro-inflammatory eicosanoids related to CYP450/sEH-pathways correlated positively with AT-InSc in those with the TT genotype, whereas in those with the CC genotype, the negative correlations between pro-inflammatory eicosanoids and AT-InSc related to COX/LOX-pathways.

Conclusions

LA-enriched diet increases inflammatory AT gene expression in subjects with the TT genotype, while CC genotype could play a protective role against LA-induced AT inflammation. Overall, the FADS1 variant could modify the dietary LA-induced effects on AT inflammation through the differential biosynthesis of AA-derived eicosanoids.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s00394-022-02922-y.

Introduction

Dietary fatty acids (FA) have important roles in the regulation of adipose tissue (AT) metabolism, gene expression and inflammation [1, 2]. The dietary intake of the essential omega-6 (n-6) polyunsaturated fatty acid (PUFA) linoleic acid (LA) associates with reduced inflammation [3, 4] whereas arachidonic acid (AA) intake has been shown to associate with increased inflammation [5, 6]. This is suggested to be due to the fact, that AA is a major substrate for pro-inflammatory eicosanoid lipid mediators [7, 8]. Free LA and AA, released from cell membrane by the phospholipase A2 (PLA2), are substrates for the eicosanoid biosynthetic enzymes, including cyclooxygenases (COX), lipoxygenases (LOX), cytochrome P450s (CYP450) and soluble epoxide hydrolase (sEH) [9]. The COX-pathway is associated with the production of pro-inflammatory eicosanoids while eicosanoids produced by some LOXs and CYP450/sEHs are anti-inflammatory [1012] which could explain why the increased intake of LA or AA has been associated both with increased and reduced inflammation [13].
Two key enzymes responsible for the production of n-6 PUFA AA are delta-5 (D5D) and delta-6 desaturases (D6D), encoded by the fatty acid desaturase (FADS) 1/2 genes, respectively [1416]. Recent studies suggest that genetic variations in the FADS genes, that reside in near complete linkage disequilibrium in the same locus, associate with glucose homeostasis, PUFA biosynthesis and the proportions of PUFAs in plasma, serum pro-inflammatory markers, hepatic lipid accumulation and inflammation, dyslipidemias [14, 15, 1719], lipid mediators [18, 20, 21] and also with the risk of type 2 diabetes (T2D) [22].
Previous studies by others and us have shown that the health outcomes of dietary LA intake are regulated by the FADS1 variants [2326]. Dietary LA intake has shown to modify the association between FADS1 variants and clinical phenotypes, including HDL and anthropometric measures [23, 24]. Additionally, we recently demonstrated in a dietary intervention study that the effect of dietary LA intake on serum high-sensitive C-reactive protein (hs-CRP) concentrations depended on the FADS1-rs174550 variant (FADSDIET study) [25]. In another study, we showed that AT inflammation correlates positively with the plasma and AT proportion of AA and estimated D6D enzyme activity [27]. This correlation was modified by the FADS1/2 genetic variation in obese individuals participating in the Kuopio Obesity Surgery (KOBS) study [27]. This suggests that the variants of FADS genes could regulate the effect of dietary n-6 PUFA on AT inflammation. However, the effect of dietary LA intake in the association between FADS1 variant (rs174550) and AT inflammation is poorly understood. In this study, we aimed to clarify the role of FADS1-rs174550 in the interaction of dietary LA intake and eicosanoids with AT inflammation. Thus, we investigated in the FADSDIET study (1) if the dietary LA-enriched diet modifies the expression of inflammatory genes in AT and (2) if the regulation of plasma eicosanoid and the expression of inflammatory genes in AT differs between subjects homozygous (CC vs. TT) for FADS1-rs174550 variant.

Experimental section

Subjects: FADSDIET

The study design of the FADSDIET intervention (NCT02543216) has been previously described [25]. Briefly, 255 healthy normal weight or borderline overweight men homozygous for the FADS1-rs174550 (minor allele frequency 0.2977 [28]) were invited from the METabolic Syndrome In Men (METSIM) study to participate in the FADSDIET intervention study. The subjects using an anticoagulant treatment and having severe chronic diseases were excluded from the study. Altogether, 62 men participated in the intervention and 59 of them completed the intervention. AT biopsies (major TT: n = 25, minor CC: n = 28) and blood samples were taken after overnight fasting at the end of the run-in period (baseline: 0 week) and after LA-enriched dietary intervention (4 week).

Dosage information

During the 4-week run-in period they consumed habitual diet but stopped consumption of oil supplements, e.g. fish oil supplements, or plant stanol or sterol enriched products, which were not allowed during the study. During the 4-week intervention period, participants consumed their habitual diet with supplemental LA 30, 40, or 50 mL (27–45 g) in the form of sunflower oil (62% of LA) daily depending on BMI with the increase of ~ 6% of energy intake.

Subjects: KOBS

Patients accepted to obesity surgery at the Kuopio University Hospital have been recruited to the ongoing Kuopio Obesity Surgery (KOBS) study since 2005 [29]. The present analysis contains cross‐sectional baseline data from a sub-cohort of 70 individuals (males: n = 23 and females: n = 47), having FADS1-rs174547 variant (minor allele frequency 0.2979 [28]) genotyped (only CC: n = 28 and TT: n = 42) genotypes included in the analysis), gene expression data available from AT and plasma FA composition (clinical characteristics presented in Supplementary Table S1). Furthermore, the FADS1-rs174547 is in near complete linkage disequilibrium with the FADS1-rs174550.

Genotyping and gene expression

The study participants were genotyped for the FADS1 variants rs174550 (FADSDIET) and rs174547 (KOBS), that are in complete linkage disequilibrium, using the TaqMan SNP Genotyping Assay according to the protocol provided by the manufacturer (Applied Biosystems, Foster City, CA, USA). For the gene expression, AT samples were immediately frozen in liquid nitrogen. TruSeq Targeted RNA Expression (TREx) platform with the MiSeq system (Illumina, San Diego, CA) was used for measuring gene expression levels in AT, as previously described [27, 30]. Additionally, the interleukin 1 beta (IL1β) gene expression in obese subjects (n = 70) of the KOBS study was measured, as described previously [30]. To investigate the effect of dietary LA intake on AT inflammation and whether the FADS1-rs174550 could modify this interaction, we measured the expression of genes which are increased in the AT of subjects with obesity and have a major role in the development of insulin resistance, and T2D (specific associations with inflammation are shown in Supplementary Table S2 with references).

Biochemical analyses (lipids, glucose, insulin, FA and lipid mediator profiling)

Commercial kits (Thermo Fisher Scientific) were used for measuring concentrations of serum total, LDL, and HDL cholesterol and total triglycerides. The Konelab 20XTi Clinical Chemistry Analyzer (Thermo Fisher Scientific) and Enzymatic photometric (glucose hexokinase) method (Thermo Fisher Scientific) was used for the measurement of plasma glucose concentration. Chemiluminometric immunoassay method (DiaSorin Liaison Analyzer; DiaSorin GmbH) was used for the analysis of plasma insulin concentration.
FA composition in plasma lipid fractions was analyzed according to previously described method [30]. Briefly, plasma samples were extracted with chloroform–methanol (2:1), and the different lipid fractions, cholesteryl esters (CE), triglycerides (TG), and phospholipids (PL) were separated by solid phase extraction with an aminopropyl column. Samples were transmethylated with 14% boron trifluoride in methanol and were analyzed by 7890A gas chromatograph (Agilent Technologies, Inc., Wilmington, DE, USA) equipped with a 25‐m FFAP column. Cholesteryl nonadecanoate (Nu Chek Prep, Inc., Elysian, MA, USA), trinonadecanoin, and phosphatidylcholine dinonadecanoyl (Larodan Fine Chemicals, Malmö, Sweden) served as internal standards. Enzyme activities in different lipid fractions were estimated as product-to-precursor ratios of individual FAs as follows: D5D = 20:4 n-6/20:3 n-6 and D6D = 18:3 n-6/18:2 n-6. Total n-6 PUFA and total n-3 PUFA composition was calculated as a sum of corresponding individual FA proportion. Plasma PL fraction was chosen due to its rapid response to dietary changes in fat, and the fact that CEs are produced by the transfer of FAs from PLs [31]. Thus, PL fraction was chosen for further analyses. Lipid mediator profiling in plasma was performed by liquid chromatography–mass spectrometry, as previously described [32].

Statistics

Statistical analyses were conducted with the SPSS software (version 25, IBM Corp., Armonk, NY, USA) and GraphPad Prism 5.03 for Windows (GraphPad Software, San Diego, CA, USA). Independent sample’s t test was used to compare differences between the genotypes. Within group differences between the two timepoints were analyzed with paired sample’s t test. Repeated-measures general linear model (GLM) was used for the assessment of diet-genotype interaction. One-way analysis of variance (ANOVA) with Bonferroni’s multiple comparison test was used for testing differences in plasma PL-LA tertiles in different genotype groups of the FADS1-rs174547 in the KOBS study. The mRNA expression data of inflammatory genes in AT were normalized using Z-transformation, Z-score, which is a standardized variable with standard deviation of one and a mean of zero. Next, a combined inflammation Z-score was created by calculating the average from the individual Z-scores of inflammatory genes in AT. Combining inflammatory genes using a Z-score provides a more accurate characterization of inflammatory status in tissue [33]. Furthermore, the mean Z-score of individual serum inflammatory factors has been previously used as a surrogate marker for low-grade inflammation [33, 34]. A Shapiro–Wilk’s test was used to examine the normal distribution of variables and the variables were logarithmically (log) transformed when needed. Correlations between the variables were analyzed either using partial correlations adjusted for age for log-transformed variables or using Spearman’s nonparametric correlation. For the grouped correlation analyses regarding eicosanoids, the Spearman’s correlation coefficients were transformed using Fisher's r to z transformation and standard error of correlation was calculated (\(\sqrt{(1-r)^ \wedge 2}/\sqrt{(n-2)}\)), where r is correlation coefficient and n is the number of samples. The z-transformed results of the Spearman’s correlation analysis between AT-InSc and individual eicosanoid concentration in plasma were pooled and the deviation of the average Spearman’s correlations between AT-InSc and eicosanoid parameters from the null hypothesis (correlation coefficient = 0) was tested using one sample t test. Additionally, the z-transformed correlations were used for analyzing the differences between the two timepoints (baseline vs. follow-up) within the genotype group by using paired samples t test. In the correlation analyses, we report significant results both with and without Bonferroni correction for multiple testing. For the TREx analysis, the expression levels for each gene per sample in the gene panel were normalized based on the total number of aligned reads of the corresponding sample. The results are shown as percentage of total transcript reads. Descriptive statistics are presented as mean ± SD or SEM (indicated in table and figures) and p ≤ 0.05 was considered as statistically significant.

Results

Clinical characteristics

The results of clinical characteristics in the whole study population (TT: n = 26, CC: n = 33) of the FADSDIET intervention have been published previously, and the adherence to the consumption of sunflower oil was excellent [25]. The parameters for participants from which AT biopsies were available (TT: n = 25, CC: n = 28) are shown in Table 1. The only significant difference at baseline was that the participants with the CC genotype were older than those with the TT genotype (CC: 58.8 ± 3.1 years vs TT: 55.1 ± 2.2 years, p < 0.0001). Fasting plasma glucose concentration decreased in participants with the CC genotype (paired-samples t test, p = 0.01; diet-genotype interaction, p = 0.012) but not in participants with the TT genotype during the intervention (Table 1). Additionally, serum total cholesterol concentration decreased in participants with the CC genotype (p = 0.017) but not in those with TT genotype. Serum LDL cholesterol concentration decreased in both genotype groups (p = 0.011, p = 0.010) during the intervention. There were no differences in the clinical variables between the genotypes of FADS1-rs174547 in the KOBS study (Supplementary Table S1).
Table 1
Clinical characteristics of the FADSDIET participants according to the FADS1-rs174550 variant at baseline and after LA-enriched diet
 
0 week
4 weeks
pa
pb
pc
TT (n = 25)
CC (n = 28)
TT (n = 25)
CC (n = 28)
TT
CC
Age, years
55.1 ± 2.2
58.8 ± 3.1
  
7 × 10–5
   
Weight, kg
80.9 ± 10.5
78.7 ± 9.2
80.9 ± 10.5
78.7 ± 9.1
0.416
0.981
0.713
0.828
BMI, kg/m2
25.6 ± 2.4
24.6 ± 2.3
25.7 ± 2.4
24.6 ± 2.3
0.101
0.817
0.735
0.972
Systolic blood pressure, mmHg
128 ± 12
129 ± 13
126 ± 9
127 ± 13
0.749
0.207
0.061
0.986
Diastolic blood pressure, mmHg
82 ± 7
84 ± 8
82 ± 7
82 ± 8
0.486
0.878
0.066
0.188
Waist circumference, cm
94.4 ± 8.5
93.0 ± 6.9
94.3 ± 8.5
92.9 ± 6.8
0.532
0.513
0.235
0.631
Plasma glucosed, mmol/L
5.6 ± 0.4
5.7 ± 0.3
5.6 ± 0.5
5.6 ± 0.3
0.144
0.385
0.007
0.012
Plasma insulind, mU/L
7.4 ± 3.2
7.9 ± 3.5
7.8 ± 3.1
7.9 ± 4.4
0.578
0.500
0.991
0.651
Serum total cholesterold, mmol/L
5.2 ± 0.8
5.4 ± 1.2
5.0 ± 0.6
5.1 ± 1.0
0.506
0.156
0.017
0.451
Serum HDL cholesterold, mmol/L
1.4 ± 0.4
1.4 ± 0.3
1.4 ± 0.4
1.4 ± 0.3
0.580
0.439
0.531
0.878
Serum LDL cholesterold, mmol/L
3.2 ± 0.6
3.3 ± 1.0
3.0 ± 0.5
3.0 ± 0.8
0.663
0.011
0.010
0.741
Serum triglyceridesd, mmol/L
1.2 ± 0.6
1.4 ± 0.8
1.3 ± 0.7
1.5 ± 1.1
0.184
0.298
0.309
0.856
Plasma free fatty acidsd, mmol/L
0.4 ± 0.1
0.5 ± 0.1
0.5 ± 0.2
0.5 ± 0.2
0.196
0.223
0.620
0.213
Values are means ± SDs. Bold values are statistically significant
FADS1 fatty acid desaturase 1, LA linoleic acid
aIndependent samples t test for differences between genotypes at baseline
bPaired-samples t test for the change within the genotype
cDiet-genotype interaction, repeated-measures general linear model
dFasting

FADS1-rs174550 modifies the effect of LA intake on AT inflammation

The results showed a significant diet-genotype interaction (repeated-measures GLM) between dietary LA-enriched diet and AT-InSc (p = 0.019, Fig. 1A) in the FADSDIET. There was a non-significant trend towards a reduced AT-InSc in subjects with the CC genotype (p = 0.067), while the AT-InSc did not change in subjects with the TT genotype (p = 0.130) of the FADS1-rs174550 in response to a LA-enriched diet (Fig. 1A). In line with this, we could additionally demonstrate that IL-1β mRNA expression in subcutaneous AT was statistically different between the groups of LA proportion in plasma PL fraction in subjects with the TT genotype (ANOVA, p = 0.019) (Fig. 1B). After adjustment for Bonferroni’s multiple comparison, obese subjects with the highest LA proportion in plasma PL fraction had increased IL-1β expression in subcutaneous AT when compared to the group with the lowest LA proportion in plasma PL fraction in subjects with the TT genotype (p = 0.017) but not in those with the other genotype (CC) of FADS1-rs174547 at baseline of the KOBS study (Fig. 1B).

AT-InSc correlates differently with plasma n-6 and LA proportions and eicosanoid levels between individuals with the different FADS1 genotypes

Correlations between AT-InSc and n-6 and LA proportions in plasma PL fraction

Since there was a difference in age between the FADS1 genotypes at baseline, the correlations between AT-InSc and plasma FA proportions in PL fraction were adjusted for age. The results showed that AT-InSc correlated negatively with total n-6 PUFA proportion in plasma PL fraction at baseline (r = − 0.449, p = 0.028) but positively after LA-enriched diet (r = 0.494, p = 0.014) in subjects with the TT genotype of the FADS1-rs174550 (Supplementary Table S3). Additionally, AT-InSc correlated positively with the proportion of LA in plasma PL fraction after LA-enriched diet (r = 0.544, p = 0.006) in subjects with the TT genotype of the FADS1-rs174550 (Fig. 2, Supplementary Table S3). In contrast, there were no significant correlations in subjects with the CC genotype of the FADS1-rs174550 between AT-InSc and n-6 or LA (Fig. 2, Supplementary Table S3). The correlation results between AT-InSc and other individual FA proportions in plasma PL fraction are shown in Supplementary Table S3.

Correlations between AT-InSc and eicosanoids

We investigated whether correlations between AT-InSc and plasma LA- or AA-derived eicosanoid compositions differed between the subjects with TT and CC genotypes. There was on average a positive correlation (r = 0.153, p = 0.008) between the AT-InSc and all LA-derived eicosanoids at the end of the intervention in subjects with the TT genotype of the FADS1-rs174550 (Fig. 3). There was also a significant difference in the correlation between the AT-InSc and LA-derived lipid mediators between the two timepoints (baseline vs. the end of the intervention) within TT genotype (p = 0.024), but not within CC genotype, of the FADS1-rs174550 (Fig. 3). There was on average a positive correlation (r = 0.081, p = 0.008) between AT-InSc and all AA-derived eicosanoids at the end of the intervention in subjects with the TT genotype of the FADS1-rs174550 (Fig. 4). In contrast, there was a positive correlation at baseline (r = 0.116, p = 0.003, corrected for multiple comparison p = 0.042) and a negative correlation (r = − 0.138, p = 0.011) at the end of the intervention between AT-InSc and all AA-derived eicosanoids in subjects with the CC genotype (Fig. 4). Moreover, there was a significant difference in the correlation between the AT-InSc and AA-derived eicosanoids between the two timepoints (baseline vs. the end of the intervention) within TT genotype (p = 0.078) and within CC genotype (p = 0.010), of the FADS1-rs174550 (Fig. 4).
Next, we explored whether there are differences in the mean correlations between AT-InSc and eicosanoids synthesized via COX, LOX, CYP450/sEH pathways between the individuals with the different FADS1 genotypes. This mean correlation summarizes the overall direction of correlation related to the specific pathway. LA-derived eicosanoids related to the CYP450/sEH-pathway correlated positively (r = 0.257, p = 0.016) with the AT-InSc at the end of the intervention in subjects with the TT, but not with the CC genotype of FADS1-rs174550 (Fig. 3). On the other hand, LA-derived eicosanoids related to the LOX-pathway correlated negatively (r = − 0.197, p = 0.046) with the AT-InSc at the end of the intervention in subjects with the CC, but not with the TT genotype of FADS1-rs174550 (Fig. 3). Furthermore, AA-derived eicosanoids related to the CYP450/sEH-pathway correlated positively (r = 0.168, p = 0.003) with the AT-InSc at the end of the intervention in subjects with the TT, but not with the CC genotype of FADS1-rs174550 (Fig. 4). Additionally, AA-derived eicosanoids related to the LOX-pathway correlated negatively at baseline but positively at the end of intervention (r = − 0.214, p = 0.012; r = 0.081, p = 0.011, respectively) with the AT-InSc in subjects with the TT genotype. Moreover, the correlation between AT-InSc and eicosanoids related to the LOX-pathway differed significantly between the two timepoints (baseline vs. the end of the intervention) within TT genotype (p = 0.031), but not within CC genotype, of the FADS1-rs174550 (Fig. 4). There was also a positive correlation (r = 0.164, p = 0.040) between the AT-InSc and eicosanoids related to the LOX-pathway at baseline in subjects with the CC genotype (Fig. 4). Eicosanoids related to COX-pathway correlated positively (r = 0.099, p = 0.021) with the AT-InSc at the end of intervention in subjects the TT genotype. In contrast, eicosanoids related to the COX-pathway correlated positively at baseline but negatively at the end of intervention (r = 0.284, p = 0.012; r = − 0.377, p = 0.006, respectively) with the AT-InSc in subjects with the CC genotype.

Discussion

We observed that the FADS1-rs174550 variant modifies the effect of dietary LA intake on AT inflammation in normal/overweight subjects in our study (p = 0.019 for gene-diet interaction). We found an association between an increased proportion of LA in plasma PL fraction, reflecting dietary LA intake, and AT inflammation in individuals with obesity with the TT, but not with the CC genotype in the KOBS study. Correlation analyses demonstrated that there was a genotype-dependent difference between LA proportions in plasma PL fraction and AT inflammation after LA-enriched diet. Interestingly, the correlations between LA- or AA-derived eicosanoids and AT inflammation differed between the participants homozygous (CC vs. TT) for the FADS1-rs174550 variant, suggesting that the FADS1 genotypes may modify the shuttling of eicosanoid synthesis to the CYP450/sEH, COX and LOX pathways and could partly explain differential responses to LA-enriched diet between the individuals with the FADS1 genotypes.
Dietary LA intake has shown to associate with a reduced risk for cardiovascular diseases [3]. However, there also are evidence that LA could induce pro-inflammatory events [35, 36]. Our main finding was that dietary LA-enriched diet decreased AT inflammation, as expected based on the known anti-inflammatory effects of LA [3, 4, 37], but only in subjects with the CC genotype of FADS1-rs174550 in the FADSDIET study. On the other hand, the AT inflammation was increased in subjects with the TT genotype of FADS1-rs174550/rs174547 either in response to LA-enriched diet (FADSDIET) or in individuals in the highest tertile of LA in plasma PL fraction (KOBS). Furthermore, our correlation analyses between LA proportions and AT-InSc demonstrated a positive correlation after LA-enriched diet only in subjects with the TT genotype of FADS1-rs174550. Obesity is associated with increased AT inflammation which is considered as major factors for the development of insulin resistance, T2D, and cardiovascular diseases [38]. Thus, the increased AT inflammation after dietary LA-enriched diet may predispose the subjects with the TT genotype of FADS1-rs174550 at higher risk for developing dysfunctional AT and insulin resistance. These results-suggest that the effect of dietary LA on AT inflammation depends on the FADS1 genotype.
Our following analyses demonstrated that the differences between the genotypes could also be related to the differential metabolism of LA- and AA-derived eicosanoids in plasma. The results demonstrated a positive correlation between AT-InSc and the average concentration of all LA- or AA-derived eicosanoids after the LA-enriched diet in subjects with the TT genotype of FADS1-rs174550. In contrast, there was a negative correlation between AT-InSc and the average level of all AA-derived eicosanoids after dietary LA intake in subjects with the CC genotype of FADS1-rs174550. It is suggested that LA-derived metabolites can mediate inflammation [39, 40], and that AA-derived eicosanoids promote inflammation [10, 12]. Thus, it is possible that high intake of dietary LA and consequent changes in eicosanoid metabolism could contribute to low-grade inflammation in AT depending on the FADS1 variant.
When investigating further the pathways through which the eicosanoids are synthesized, we could demonstrate that the dietary LA-enriched diet resulted in a positive correlation between AT-InSc and the average of LA- and AA-derived eicosanoids related to the CYP450/sEH-pathway in subjects with the TT genotype, but not the CC genotype of FADS1-rs174550. In contrast, there was a negative correlation between AT-InSc and the average of LA-derived eicosanoids related to the LOX-pathway in subjects with the CC genotype; whereas, AA-derived eicosanoids related to the LOX-pathway correlated positively in subjects with the TT genotype of FADS1-rs174550 after LA-enriched diet. Additionally, AA-derived eicosanoids related to the COX-pathway correlated positively in subjects with the TT genotype of FADS1-rs174550 after LA-enriched diet. Most of the individual lipid mediators included in the analyses have pro-inflammatory effects. For example, the LA-derived lipid mediators of hydroxyoctadecadienoic acids (HODEs, LOX-pathway) are markers of oxidative stress and the regulators of inflammation [41], and epoxy-octadecenoic acids (EpOMEs, CYP450-pathway) and dihydroxy-octadecenoic acids (DiHOMEs, sEH-pathway) can act as protoxins [42], induce oxidative stress and NF-KB and promote inflammation [43, 44]. In addition, many AA-derived eicosanoids, like prostaglandins (COX-pathway), hydroxy-eicosatetraenoic acids (HETEs, LOX pathway) and dihydroxy-eicosatrienoic acids (DHETs, sEH-pathway), are known to induce inflammation [45]. Previous studies have shown that the FADS1 variation contributes to inflammation via differential enzyme activity and eicosanoid biosynthesis. Hester et al. [20] have shown that the FADS1 variation could modify LOX-mediated eicosanoid levels in human blood, indicating that FADS1 genotypes could contribute to inflammation through differential eicosanoid biosynthesis. Additionally, AA-derived pro-inflammatory eicosanoids mediated via LOX- and COX-pathways in intestine have shown to be decreased in a rodent model of FADS1 null mice, reflecting the decreased FADS1 function, like the CC genotype of the FADS1-rs174550 [21]. Furthermore, Gromovsky et al. [18] have shown that the FADS1 knockdown induced a differential expression of COX/LOX genes. On the other hand, the FADS1 overexpression has shown to induce pro-inflammatory prostaglandin E2 (PGE2) level which is mediated by the COX [46]. Taken together, since the FADS1 has shown to regulate the activity of enzymes or their eicosanoids these results suggest that LA-enriched diet induces AT inflammation in those with the TT genotype, the genotype known to be associated with the T2D [47], possibly via the increased levels of enzymes responsible for the biosynthesis of pro-inflammatory eicosanoids. In contrast, the CC genotype of FADS1-rs174550 could have a protective role against dietary LA-induced effects on AT inflammation, and this may be related to the decreased levels of enzymes responsible for the biosynthesis of pro-inflammatory eicosanoids. We fully acknowledge that further studies are needed to clarify the mechanisms underlying these genotype-specific relationships between AT inflammation and LA/AA-related eicosanoid metabolism.
The previous study of our group demonstrated that hs-CRP level in serum was decreased after LA-enriched diet in subjects with the TT genotype, while in subjects with the CC genotype hs-CRP level was increased [25]. The discrepancy in the results between the decreasing effect of LA-enriched diet on hs-CRP in our previous study [25], and the increasing effect on AT inflammation in this study in subjects with the TT genotype could be in part explained by the fact that CRP is mainly synthesized by the liver [48] while in AT, that was the main topic of this study, the regulation may be different. This is in line with our previous observation that AA proportion in plasma/AT correlated positively with AT inflammation but negatively with the liver inflammation in subjects with obesity and the major genotype of FADS2-rs174616 (located in the same FADS cluster as rs174550 and rs174547) in the KOBS study [27]. Furthermore, a reduced FADS1 function, due to the CC genotype of FADS1-rs174550 with a lower desaturase activity, has been shown to associate positively with hepatic inflammation, non-alcoholic fatty liver disease and to induce changes in n-6 and n-3 PUFA-derived eicosanoid levels in a mouse model and in a human study [17, 18]. These results suggest that the FADS1-rs174550 variant could modify the association between PUFA metabolism and inflammation tissue dependently. Unfortunately, it is difficult to obtain serial liver biopsies, unlike AT biopsies, during a dietary intervention.
The strengths of our study are the unique study design with the genotype-based recruitment of individuals [25]. Additionally, the availability of AT biopsies and FA composition in plasma enabled to investigate the genetic influence of FADS1 in the relationship between n-6 PUFA metabolism and inflammation in AT. Another strength of this study is that we were able to combine results from normal/overweight individuals (FADSDIET) with obese individuals of the cross-sectional KOBS study. Due to the fact, that the subjects were chosen based in their specific FADS1 variant and were middle-aged men in the FADSDIET, the results cannot be generalized to women or younger population. Additionally, it is well known that gene expression data does not necessarily reflect protein expression or the activity of proteins at the tissue and therefore, the lack of protein data in AT for the inflammatory genes limits the implications of the findings. Unfortunately, protein analysis was not possible due to the sample size limitations in the FADSDIET study. Although, our previous study successfully demonstrated LA-induced changes in the AA proportion in plasma PL fraction according to the FADS1 variant in the same cohort [25]. It is possible that we may not detect individual dietary LA-induced effects on AT inflammation or eicosanoid levels due to a small sample size. Although the chosen single SNP is unlikely to be causative, the FADS1-rs174550 has a high effect size on plasma and tissue FA concentrations and it has been previously used as a marker for the variation in the FADS1/2 locus [22, 49]. Unfortunately, we were able to determine FA and eicosanoid concentrations only in plasma. Knowledge about the tissue FA levels could give further insights in the interaction between PUFA metabolism and inflammation in AT.
In conclusion, the FADS1-rs174550 variant regulates the interaction between dietary LA intake and AT inflammation. Increased dietary LA intake induced a positive relationship between AT inflammation and eicosanoids mediated through CYP450-pathway in subjects with the TT genotype of FADS1-rs174550. On the contrary, there was a negative relationship between AT inflammation and eicosanoids mediated through the COX-pathway in subjects with the CC genotype of FADS1-rs174550. Our results suggest that CC genotype could play a protective role against LA-induced AT inflammation. Overall, the FADS1 variant could modify the dietary LA-induced effects on AT inflammation through differential biosynthesis of AA-derived eicosanoids.

Acknowledgements

We thank Päivi Turunen, Tuomas Onnukka, Matti Laitinen, and Sirkku Karhunen for their technical assistance.

Declarations

Conflict of interest

The authors declare that they have no competing interest.

Ethics approval

The study protocols were approved by the Ethics Committee of the Northern Savo Hospital District (FADSDIET: 75/2014 and KOBS: 1108/2018) and carried out in accordance with the Helsinki Declaration. Written and oral information was given to the participants and an informed written consent was obtained from all participants.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Unsere Produktempfehlungen

Neuer Inhalt

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat De Boer AA, Monk JM, Robinson LE (2014) Docosahexaenoic acid decreases pro-inflammatory mediators in an in vitro murine adipocyte macrophage co-culture model. PLoS ONE 9:e85037PubMedPubMedCentralCrossRef De Boer AA, Monk JM, Robinson LE (2014) Docosahexaenoic acid decreases pro-inflammatory mediators in an in vitro murine adipocyte macrophage co-culture model. PLoS ONE 9:e85037PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Karbowski M, Youle RJ (2003) Dynamics of mitochondrial morphology in healthy cells and during apoptosis. Cell Death Differ 10:870–880PubMedCrossRef Karbowski M, Youle RJ (2003) Dynamics of mitochondrial morphology in healthy cells and during apoptosis. Cell Death Differ 10:870–880PubMedCrossRef
3.
Zurück zum Zitat Virtanen JK, Mursu J, Voutilainen S, Tuomainen TP (2018) The associations of serum n-6 polyunsaturated fatty acids with serum C-reactive protein in men: the Kuopio Ischaemic Heart Disease Risk Factor Study. Eur J Clin Nutr 72:342–348PubMedCrossRef Virtanen JK, Mursu J, Voutilainen S, Tuomainen TP (2018) The associations of serum n-6 polyunsaturated fatty acids with serum C-reactive protein in men: the Kuopio Ischaemic Heart Disease Risk Factor Study. Eur J Clin Nutr 72:342–348PubMedCrossRef
5.
Zurück zum Zitat Lee H, Park WJ (2014) Unsaturated fatty acids, desaturases, and human health. J Med Food 17:189–197PubMedCrossRef Lee H, Park WJ (2014) Unsaturated fatty acids, desaturases, and human health. J Med Food 17:189–197PubMedCrossRef
7.
Zurück zum Zitat Vucic V (2013) The role of dietary polyunsaturated fatty acids in inflammation. Ser J Exp Clin Res 14(3):93–99CrossRef Vucic V (2013) The role of dietary polyunsaturated fatty acids in inflammation. Ser J Exp Clin Res 14(3):93–99CrossRef
8.
Zurück zum Zitat Teng KT, Chang CY, Chang LF, Nesaretnam K (2014) Modulation of obesity-induced inflammation by dietary fats: mechanisms and clinical evidence. Nutr J 13:122891CrossRef Teng KT, Chang CY, Chang LF, Nesaretnam K (2014) Modulation of obesity-induced inflammation by dietary fats: mechanisms and clinical evidence. Nutr J 13:122891CrossRef
9.
Zurück zum Zitat Mouchlis VD, Dennis EA (2019) Phospholipase A2 catalysis and lipid mediator lipidomics. Biochim Biophys Acta Mol Cell Biol Lipids 1864:766–771PubMedCrossRef Mouchlis VD, Dennis EA (2019) Phospholipase A2 catalysis and lipid mediator lipidomics. Biochim Biophys Acta Mol Cell Biol Lipids 1864:766–771PubMedCrossRef
11.
Zurück zum Zitat Lundstrom SL, Saluja R, Adner M, Haeggstrom JZ, Nilsson G, Wheelock CE (2013) Lipid mediator metabolic profiling demonstrates differences in eicosanoid patterns in two phenotypically distinct mast cell populations. J Lipid Res 54:116–126PubMedPubMedCentralCrossRef Lundstrom SL, Saluja R, Adner M, Haeggstrom JZ, Nilsson G, Wheelock CE (2013) Lipid mediator metabolic profiling demonstrates differences in eicosanoid patterns in two phenotypically distinct mast cell populations. J Lipid Res 54:116–126PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Innes JK, Calder PC (2018) Omega-6 fatty acids and inflammation. Prostaglandins Leukot Essent Fatty Acids 132:41–48PubMedCrossRef Innes JK, Calder PC (2018) Omega-6 fatty acids and inflammation. Prostaglandins Leukot Essent Fatty Acids 132:41–48PubMedCrossRef
14.
Zurück zum Zitat Ameur A, Enroth S, Johansson A, Zaboli G, Igl W, Johansson AC, Rivas MA, Daly MJ, Schmitz G, Hicks AA, Meitinger T, Feuk L, van Duijn C, Oostra B, Pramstaller PP, Rudan I, Wright AF, Wilson JF, Campbell H, Gyllensten U (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 90:809–820PubMedPubMedCentralCrossRef Ameur A, Enroth S, Johansson A, Zaboli G, Igl W, Johansson AC, Rivas MA, Daly MJ, Schmitz G, Hicks AA, Meitinger T, Feuk L, van Duijn C, Oostra B, Pramstaller PP, Rudan I, Wright AF, Wilson JF, Campbell H, Gyllensten U (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 90:809–820PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Al-Hilal M, Alsaleh A, Maniou Z, Lewis FJ, Hall WL, Sanders TA, O’Dell SD, MARINA study team (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 54:542–551PubMedPubMedCentralCrossRef Al-Hilal M, Alsaleh A, Maniou Z, Lewis FJ, Hall WL, Sanders TA, O’Dell SD, MARINA study team (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 54:542–551PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Kathiresan S, Willer CJ, Peloso GM, Demissie S, Musunuru K, Schadt EE, Kaplan L, Bennett D, Li Y, Tanaka T, Voight BF, Bonnycastle LL, Jackson AU, Crawford G, Surti A, Guiducci C, Burtt NP, Parish S, Clarke R, Zelenika D, Kubalanza KA, Morken MA, Scott LJ, Stringham HM, Galan P, Swift AJ, Kuusisto J, Bergman RN, Sundvall J, Laakso M, Ferrucci L, Scheet P, Sanna S, Uda M, Yang Q, Lunetta KL, Dupuis J, de Bakker PI, O’Donnell CJ, Chambers JC, Kooner JS, Hercberg S, Meneton P, Lakatta EG, Scuteri A, Schlessinger D, Tuomilehto J, Collins FS, Groop L, Altshuler D, Collins R, Lathrop GM, Melander O, Salomaa V, Peltonen L, Orho-Melander M, Ordovas JM, Boehnke M, Abecasis GR, Mohlke KL, Cupples LA (2009) Common variants at 30 loci contribute to polygenic dyslipidemia. Nat Genet 41:56–65PubMedCrossRef Kathiresan S, Willer CJ, Peloso GM, Demissie S, Musunuru K, Schadt EE, Kaplan L, Bennett D, Li Y, Tanaka T, Voight BF, Bonnycastle LL, Jackson AU, Crawford G, Surti A, Guiducci C, Burtt NP, Parish S, Clarke R, Zelenika D, Kubalanza KA, Morken MA, Scott LJ, Stringham HM, Galan P, Swift AJ, Kuusisto J, Bergman RN, Sundvall J, Laakso M, Ferrucci L, Scheet P, Sanna S, Uda M, Yang Q, Lunetta KL, Dupuis J, de Bakker PI, O’Donnell CJ, Chambers JC, Kooner JS, Hercberg S, Meneton P, Lakatta EG, Scuteri A, Schlessinger D, Tuomilehto J, Collins FS, Groop L, Altshuler D, Collins R, Lathrop GM, Melander O, Salomaa V, Peltonen L, Orho-Melander M, Ordovas JM, Boehnke M, Abecasis GR, Mohlke KL, Cupples LA (2009) Common variants at 30 loci contribute to polygenic dyslipidemia. Nat Genet 41:56–65PubMedCrossRef
17.
Zurück zum Zitat Nobili V, Alisi A, Liu Z, Liang T, Crudele A, Raponi M, Lin J, Chalasani NP, Liu W (2018) In a pilot study, reduced fatty acid desaturase 1 function was associated with nonalcoholic fatty liver disease and response to treatment in children. Pediatr Res 84:696–703PubMedPubMedCentralCrossRef Nobili V, Alisi A, Liu Z, Liang T, Crudele A, Raponi M, Lin J, Chalasani NP, Liu W (2018) In a pilot study, reduced fatty acid desaturase 1 function was associated with nonalcoholic fatty liver disease and response to treatment in children. Pediatr Res 84:696–703PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Gromovsky AD, Schugar RC, Brown AL, Helsley RN, Burrows AC, Ferguson D, Zhang R, Sansbury BE, Lee RG, Morton RE, Allende DS, Parks JS, Spite M, Brown JM (2018) Delta-5 fatty acid desaturase FADS1 impacts metabolic disease by balancing proinflammatory and proresolving lipid mediators. Arterioscler Thromb Vasc Biol 38:218–231PubMedCrossRef Gromovsky AD, Schugar RC, Brown AL, Helsley RN, Burrows AC, Ferguson D, Zhang R, Sansbury BE, Lee RG, Morton RE, Allende DS, Parks JS, Spite M, Brown JM (2018) Delta-5 fatty acid desaturase FADS1 impacts metabolic disease by balancing proinflammatory and proresolving lipid mediators. Arterioscler Thromb Vasc Biol 38:218–231PubMedCrossRef
19.
Zurück zum Zitat Wang L, Athinarayanan S, Jiang G, Chalasani N, Zhang M, Liu W (2015) Fatty acid desaturase 1 gene polymorphisms control human hepatic lipid composition. Hepatology 61:119–128PubMedCrossRef Wang L, Athinarayanan S, Jiang G, Chalasani N, Zhang M, Liu W (2015) Fatty acid desaturase 1 gene polymorphisms control human hepatic lipid composition. Hepatology 61:119–128PubMedCrossRef
20.
Zurück zum Zitat Hester AG, Murphy RC, Uhlson CJ, Ivester P, Lee TC, Sergeant S, Miller LR, Howard TD, Mathias RA, Chilton FH (2014) Relationship between a common variant in the fatty acid desaturase (FADS) cluster and eicosanoid generation in humans. J Biol Chem 289:22482–22489PubMedPubMedCentralCrossRef Hester AG, Murphy RC, Uhlson CJ, Ivester P, Lee TC, Sergeant S, Miller LR, Howard TD, Mathias RA, Chilton FH (2014) Relationship between a common variant in the fatty acid desaturase (FADS) cluster and eicosanoid generation in humans. J Biol Chem 289:22482–22489PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Monk JM, Turk HF, Fan YY, Callaway E, Weeks B, Yang P, McMurray DN, Chapkin RS (2014) Antagonizing arachidonic acid-derived eicosanoids reduces inflammatory Th17 and Th1 cell-mediated inflammation and colitis severity. Mediators Inflamm 2014:917149PubMedPubMedCentralCrossRef Monk JM, Turk HF, Fan YY, Callaway E, Weeks B, Yang P, McMurray DN, Chapkin RS (2014) Antagonizing arachidonic acid-derived eicosanoids reduces inflammatory Th17 and Th1 cell-mediated inflammation and colitis severity. Mediators Inflamm 2014:917149PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N, Jackson AU, Wheeler E, Glazer NL, Bouatia-Naji N, Gloyn AL, Lindgren CM, Magi R, Morris AP, Randall J, Johnson T, Elliott P, Rybin D, Thorleifsson G, Steinthorsdottir V, Henneman P, Grallert H, Dehghan A, Hottenga JJ, Franklin CS, Navarro P, Song K, Goel A, Perry JR, Egan JM, Lajunen T, Grarup N, Sparso T, Doney A, Voight BF, Stringham HM, Li M, Kanoni S, Shrader P, Cavalcanti-Proenca C, Kumari M, Qi L, Timpson NJ, Gieger C, Zabena C, Rocheleau G, Ingelsson E, An P, O'Connell J, Luan J, Elliott A, McCarroll SA, Payne F, Roccasecca RM, Pattou F, Sethupathy P, Ardlie K, Ariyurek Y, Balkau B, Barter P, Beilby JP, Ben-Shlomo Y, Benediktsson R, Bennett AJ, Bergmann S, Bochud M, Boerwinkle E, Bonnefond A, Bonnycastle LL, Borch-Johnsen K, Bottcher Y, Brunner E, Bumpstead SJ, Charpentier G, Chen YD, Chines P, Clarke R, Coin LJ, Cooper MN, Cornelis M, Crawford G, Crisponi L, Day IN, de Geus EJ, Delplanque J, Dina C, Erdos MR, Fedson AC, Fischer-Rosinsky A, Forouhi NG, Fox CS, Frants R, Franzosi MG, Galan P, Goodarzi MO, Graessler J, Groves CJ, Grundy S, Gwilliam R, Gyllensten U, Hadjadj S, Hallmans G, Hammond N, Han X, Hartikainen AL, Hassanali N, Hayward C, Heath SC, Hercberg S, Herder C, Hicks AA, Hillman DR, Hingorani AD, Hofman A, Hui J, Hung J, Isomaa B, Johnson PR, Jorgensen T, Jula A, Kaakinen M, Kaprio J, Kesaniemi YA, Kivimaki M, Knight B, Koskinen S, Kovacs P, Kyvik KO, Lathrop GM, Lawlor DA, Le Bacquer O, Lecoeur C, Li Y, Lyssenko V, Mahley R, Mangino M, Manning AK, Martinez-Larrad MT, McAteer JB, McCulloch LJ, McPherson R, Meisinger C, Melzer D, Meyre D, Mitchell BD, Morken MA, Mukherjee S, Naitza S, Narisu N, Neville MJ, Oostra BA, Orru M, Pakyz R, Palmer CN, Paolisso G, Pattaro C, Pearson D, Peden JF, Pedersen NL, Perola M, Pfeiffer AF, Pichler I, Polasek O, Posthuma D, Potter SC, Pouta A, Province MA, Psaty BM, Rathmann W, Rayner NW, Rice K, Ripatti S, Rivadeneira F, Roden M, Rolandsson O, Sandbaek A, Sandhu M, Sanna S, Sayer AA, Scheet P, Scott LJ, Seedorf U, Sharp SJ, Shields B, Sigurethsson G, Sijbrands EJ, Silveira A, Simpson L, Singleton A, Smith NL, Sovio U, Swift A, Syddall H, Syvanen AC, Tanaka T, Thorand B, Tichet J, Tonjes A, Tuomi T, Uitterlinden AG, van Dijk KW, van Hoek M, Varma D, Visvikis-Siest S, Vitart V, Vogelzangs N, Waeber G, Wagner PJ, Walley A, Walters GB, Ward KL, Watkins H, Weedon MN, Wild SH, Willemsen G, Witteman JC, Yarnell JW, Zeggini E, Zelenika D, Zethelius B, Zhai G, Zhao JH, Zillikens MC, DIAGRAM Consortium, GIANT Consortium, Global BPgen Consortium, Borecki IB, Loos RJ, Meneton P, Magnusson PK, Nathan DM, Williams GH, Hattersley AT, Silander K, Salomaa V, Smith GD, Bornstein SR, Schwarz P, Spranger J, Karpe F, Shuldiner AR, Cooper C, Dedoussis GV, Serrano-Rios M, Morris AD, Lind L, Palmer LJ, Hu FB, Franks PW, Ebrahim S, Marmot M, Kao WH, Pankow JS, Sampson MJ, Kuusisto J, Laakso M, Hansen T, Pedersen O, Pramstaller PP, Wichmann HE, Illig T, Rudan I, Wright AF, Stumvoll M, Campbell H, Wilson JF, Anders Hamsten on behalf of Procardis Consortium, MAGIC investigators, Bergman RN, Buchanan TA, Collins FS, Mohlke KL, Tuomilehto J, Valle TT, Altshuler D, Rotter JI, Siscovick DS, Penninx BW, Boomsma DI, Deloukas P, Spector TD, Frayling TM, Ferrucci L, Kong A, Thorsteinsdottir U, Stefansson K, van Duijn CM, Aulchenko YS, Cao A, Scuteri A, Schlessinger D, Uda M, Ruokonen A, Jarvelin MR, Waterworth DM, Vollenweider P, Peltonen L, Mooser V, Abecasis GR, Wareham NJ, Sladek R, Froguel P, Watanabe RM, Meigs JB, Groop L, Boehnke M, McCarthy MI, Florez JC, Barroso I (2010) New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet 42:105–116 Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N, Jackson AU, Wheeler E, Glazer NL, Bouatia-Naji N, Gloyn AL, Lindgren CM, Magi R, Morris AP, Randall J, Johnson T, Elliott P, Rybin D, Thorleifsson G, Steinthorsdottir V, Henneman P, Grallert H, Dehghan A, Hottenga JJ, Franklin CS, Navarro P, Song K, Goel A, Perry JR, Egan JM, Lajunen T, Grarup N, Sparso T, Doney A, Voight BF, Stringham HM, Li M, Kanoni S, Shrader P, Cavalcanti-Proenca C, Kumari M, Qi L, Timpson NJ, Gieger C, Zabena C, Rocheleau G, Ingelsson E, An P, O'Connell J, Luan J, Elliott A, McCarroll SA, Payne F, Roccasecca RM, Pattou F, Sethupathy P, Ardlie K, Ariyurek Y, Balkau B, Barter P, Beilby JP, Ben-Shlomo Y, Benediktsson R, Bennett AJ, Bergmann S, Bochud M, Boerwinkle E, Bonnefond A, Bonnycastle LL, Borch-Johnsen K, Bottcher Y, Brunner E, Bumpstead SJ, Charpentier G, Chen YD, Chines P, Clarke R, Coin LJ, Cooper MN, Cornelis M, Crawford G, Crisponi L, Day IN, de Geus EJ, Delplanque J, Dina C, Erdos MR, Fedson AC, Fischer-Rosinsky A, Forouhi NG, Fox CS, Frants R, Franzosi MG, Galan P, Goodarzi MO, Graessler J, Groves CJ, Grundy S, Gwilliam R, Gyllensten U, Hadjadj S, Hallmans G, Hammond N, Han X, Hartikainen AL, Hassanali N, Hayward C, Heath SC, Hercberg S, Herder C, Hicks AA, Hillman DR, Hingorani AD, Hofman A, Hui J, Hung J, Isomaa B, Johnson PR, Jorgensen T, Jula A, Kaakinen M, Kaprio J, Kesaniemi YA, Kivimaki M, Knight B, Koskinen S, Kovacs P, Kyvik KO, Lathrop GM, Lawlor DA, Le Bacquer O, Lecoeur C, Li Y, Lyssenko V, Mahley R, Mangino M, Manning AK, Martinez-Larrad MT, McAteer JB, McCulloch LJ, McPherson R, Meisinger C, Melzer D, Meyre D, Mitchell BD, Morken MA, Mukherjee S, Naitza S, Narisu N, Neville MJ, Oostra BA, Orru M, Pakyz R, Palmer CN, Paolisso G, Pattaro C, Pearson D, Peden JF, Pedersen NL, Perola M, Pfeiffer AF, Pichler I, Polasek O, Posthuma D, Potter SC, Pouta A, Province MA, Psaty BM, Rathmann W, Rayner NW, Rice K, Ripatti S, Rivadeneira F, Roden M, Rolandsson O, Sandbaek A, Sandhu M, Sanna S, Sayer AA, Scheet P, Scott LJ, Seedorf U, Sharp SJ, Shields B, Sigurethsson G, Sijbrands EJ, Silveira A, Simpson L, Singleton A, Smith NL, Sovio U, Swift A, Syddall H, Syvanen AC, Tanaka T, Thorand B, Tichet J, Tonjes A, Tuomi T, Uitterlinden AG, van Dijk KW, van Hoek M, Varma D, Visvikis-Siest S, Vitart V, Vogelzangs N, Waeber G, Wagner PJ, Walley A, Walters GB, Ward KL, Watkins H, Weedon MN, Wild SH, Willemsen G, Witteman JC, Yarnell JW, Zeggini E, Zelenika D, Zethelius B, Zhai G, Zhao JH, Zillikens MC, DIAGRAM Consortium, GIANT Consortium, Global BPgen Consortium, Borecki IB, Loos RJ, Meneton P, Magnusson PK, Nathan DM, Williams GH, Hattersley AT, Silander K, Salomaa V, Smith GD, Bornstein SR, Schwarz P, Spranger J, Karpe F, Shuldiner AR, Cooper C, Dedoussis GV, Serrano-Rios M, Morris AD, Lind L, Palmer LJ, Hu FB, Franks PW, Ebrahim S, Marmot M, Kao WH, Pankow JS, Sampson MJ, Kuusisto J, Laakso M, Hansen T, Pedersen O, Pramstaller PP, Wichmann HE, Illig T, Rudan I, Wright AF, Stumvoll M, Campbell H, Wilson JF, Anders Hamsten on behalf of Procardis Consortium, MAGIC investigators, Bergman RN, Buchanan TA, Collins FS, Mohlke KL, Tuomilehto J, Valle TT, Altshuler D, Rotter JI, Siscovick DS, Penninx BW, Boomsma DI, Deloukas P, Spector TD, Frayling TM, Ferrucci L, Kong A, Thorsteinsdottir U, Stefansson K, van Duijn CM, Aulchenko YS, Cao A, Scuteri A, Schlessinger D, Uda M, Ruokonen A, Jarvelin MR, Waterworth DM, Vollenweider P, Peltonen L, Mooser V, Abecasis GR, Wareham NJ, Sladek R, Froguel P, Watanabe RM, Meigs JB, Groop L, Boehnke M, McCarthy MI, Florez JC, Barroso I (2010) New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet 42:105–116
24.
Zurück zum Zitat Dumont J, Goumidi L, Grenier-Boley B, Cottel D, Marecaux N, Montaye M, Wagner A, Arveiler D, Simon C, Ferrieres J, Ruidavets JB, Amouyel P, Dallongeville J, Meirhaeghe A (2018) Dietary linoleic acid interacts with FADS1 genetic variability to modulate HDL-cholesterol and obesity-related traits. Clin Nutr 37:1683–1689PubMedCrossRef Dumont J, Goumidi L, Grenier-Boley B, Cottel D, Marecaux N, Montaye M, Wagner A, Arveiler D, Simon C, Ferrieres J, Ruidavets JB, Amouyel P, Dallongeville J, Meirhaeghe A (2018) Dietary linoleic acid interacts with FADS1 genetic variability to modulate HDL-cholesterol and obesity-related traits. Clin Nutr 37:1683–1689PubMedCrossRef
25.
Zurück zum Zitat Lankinen MA, Fauland A, Shimizu BI, Agren J, Wheelock CE, Laakso M, Schwab U, Pihlajamaki J (2019) Inflammatory response to dietary linoleic acid depends on FADS1 genotype. Am J Clin Nutr 109:165–175 PubMedCrossRef Lankinen MA, Fauland A, Shimizu BI, Agren J, Wheelock CE, Laakso M, Schwab U, Pihlajamaki J (2019) Inflammatory response to dietary linoleic acid depends on FADS1 genotype. Am J Clin Nutr 109:165–175 PubMedCrossRef
26.
Zurück zum Zitat Lankinen MA, de Mello VD, Meuronen T, Sallinen T, Agren J, Virtanen KA, Laakso M, Pihlajamaki J, Schwab U (2021) The FADS1 genotype modifies metabolic responses to the linoleic acid and alpha-linolenic acid containing plant oils-genotype based randomized trial FADSDIET2. Mol Nutr Food Res 65:e2001004PubMedCrossRef Lankinen MA, de Mello VD, Meuronen T, Sallinen T, Agren J, Virtanen KA, Laakso M, Pihlajamaki J, Schwab U (2021) The FADS1 genotype modifies metabolic responses to the linoleic acid and alpha-linolenic acid containing plant oils-genotype based randomized trial FADSDIET2. Mol Nutr Food Res 65:e2001004PubMedCrossRef
27.
Zurück zum Zitat Vaittinen M, Mannisto V, Kakela P, Agren J, Tiainen M, Schwab U, Pihlajamaki J (2017) Interorgan cross talk between fatty acid metabolism, tissue inflammation, and FADS2 genotype in humans with obesity. Obesity (Silver Spring) 25:545–552CrossRef Vaittinen M, Mannisto V, Kakela P, Agren J, Tiainen M, Schwab U, Pihlajamaki J (2017) Interorgan cross talk between fatty acid metabolism, tissue inflammation, and FADS2 genotype in humans with obesity. Obesity (Silver Spring) 25:545–552CrossRef
28.
Zurück zum Zitat Buniello A, MacArthur JAL, Cerezo M, Harris LW, Hayhurst J, Malangone C, McMahon A, Morales J, Mountjoy E, Sollis E, Suveges D, Vrousgou O, Whetzel PL, Amode R, Guillen JA, Riat HS, Trevanion SJ, Hall P, Junkins H, Flicek P, Burdett T, Hindorff LA, Cunningham F, Parkinson H (2019) The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res 47:D1005–D1012PubMedCrossRef Buniello A, MacArthur JAL, Cerezo M, Harris LW, Hayhurst J, Malangone C, McMahon A, Morales J, Mountjoy E, Sollis E, Suveges D, Vrousgou O, Whetzel PL, Amode R, Guillen JA, Riat HS, Trevanion SJ, Hall P, Junkins H, Flicek P, Burdett T, Hindorff LA, Cunningham F, Parkinson H (2019) The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res 47:D1005–D1012PubMedCrossRef
29.
Zurück zum Zitat Pihlajamaki J, Kuulasmaa T, Kaminska D, Simonen M, Karja V, Gronlund S, Kakela P, Paakkonen M, Kainulainen S, Punnonen K, Kuusisto J, Gylling H, Laakso M (2012) Serum interleukin 1 receptor antagonist as an independent marker of non-alcoholic steatohepatitis in humans. J Hepatol 56:663–670PubMedCrossRef Pihlajamaki J, Kuulasmaa T, Kaminska D, Simonen M, Karja V, Gronlund S, Kakela P, Paakkonen M, Kainulainen S, Punnonen K, Kuusisto J, Gylling H, Laakso M (2012) Serum interleukin 1 receptor antagonist as an independent marker of non-alcoholic steatohepatitis in humans. J Hepatol 56:663–670PubMedCrossRef
30.
Zurück zum Zitat Vaittinen M, Walle P, Kuosmanen E, Mannisto V, Kakela P, Agren J, Schwab U, Pihlajamaki J (2016) FADS2 genotype regulates delta-6 desaturase activity and inflammation in human adipose tissue. J Lipid Res 57:56–65PubMedPubMedCentralCrossRef Vaittinen M, Walle P, Kuosmanen E, Mannisto V, Kakela P, Agren J, Schwab U, Pihlajamaki J (2016) FADS2 genotype regulates delta-6 desaturase activity and inflammation in human adipose tissue. J Lipid Res 57:56–65PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Furtado J, Beqari J, Campos H (2019) Comparison of the utility of total plasma fatty acids versus those in cholesteryl ester, phospholipid, and triglyceride as biomarkers of fatty acid intake. Nutrients 11(9):2081PubMedCentralCrossRef Furtado J, Beqari J, Campos H (2019) Comparison of the utility of total plasma fatty acids versus those in cholesteryl ester, phospholipid, and triglyceride as biomarkers of fatty acid intake. Nutrients 11(9):2081PubMedCentralCrossRef
32.
Zurück zum Zitat Kolmert J, Fauland A, Fuchs D, Safholm J, Gomez C, Adner M, Dahlen SE, Wheelock CE (2018) Lipid mediator quantification in isolated human and guinea pig airways: an expanded approach for respiratory research. Anal Chem 90:10239–10248PubMedCrossRef Kolmert J, Fauland A, Fuchs D, Safholm J, Gomez C, Adner M, Dahlen SE, Wheelock CE (2018) Lipid mediator quantification in isolated human and guinea pig airways: an expanded approach for respiratory research. Anal Chem 90:10239–10248PubMedCrossRef
33.
Zurück zum Zitat Morrison L, Laukkanen JA, Ronkainen K, Kurl S, Kauhanen J, Toriola AT (2016) Inflammatory biomarker score and cancer: a population-based prospective cohort study. BMC Cancer 16:80-016-2115-6 Morrison L, Laukkanen JA, Ronkainen K, Kurl S, Kauhanen J, Toriola AT (2016) Inflammatory biomarker score and cancer: a population-based prospective cohort study. BMC Cancer 16:80-016-2115-6
34.
Zurück zum Zitat Wesselink E, Balvers M, Bours MJL, de Wilt JHW, Witkamp RF, van Baar H, Geijsen AJMR, van Halteren H, Keulen ETP, Kok DE, Kouwenhoven EA, van den Ouweland J, van Zutphen M, Weijenberg MP, Kampman E, van Duijnhoven FJB (2020) The association between circulating levels of vitamin D and inflammatory markers in the first 2 years after colorectal cancer diagnosis. Therap Adv Gastroenterol 13:1756284820923922PubMedPubMedCentralCrossRef Wesselink E, Balvers M, Bours MJL, de Wilt JHW, Witkamp RF, van Baar H, Geijsen AJMR, van Halteren H, Keulen ETP, Kok DE, Kouwenhoven EA, van den Ouweland J, van Zutphen M, Weijenberg MP, Kampman E, van Duijnhoven FJB (2020) The association between circulating levels of vitamin D and inflammatory markers in the first 2 years after colorectal cancer diagnosis. Therap Adv Gastroenterol 13:1756284820923922PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Beam J, Botta A, Ye J, Soliman H, Matier BJ, Forrest M, MacLeod KM, Ghosh S (2015) Excess linoleic acid increases collagen I/III ratio and “stiffens” the heart muscle following high fat diets. J Biol Chem 290:23371–23384PubMedPubMedCentralCrossRef Beam J, Botta A, Ye J, Soliman H, Matier BJ, Forrest M, MacLeod KM, Ghosh S (2015) Excess linoleic acid increases collagen I/III ratio and “stiffens” the heart muscle following high fat diets. J Biol Chem 290:23371–23384PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Marchix J, Choque B, Kouba M, Fautrel A, Catheline D, Legrand P (2015) Excessive dietary linoleic acid induces proinflammatory markers in rats. J Nutr Biochem 26:1434–1441PubMedCrossRef Marchix J, Choque B, Kouba M, Fautrel A, Catheline D, Legrand P (2015) Excessive dietary linoleic acid induces proinflammatory markers in rats. J Nutr Biochem 26:1434–1441PubMedCrossRef
37.
Zurück zum Zitat Harris WS, Mozaffarian D, Rimm E, Kris-Etherton P, Rudel LL, Appel LJ, Engler MM, Engler MB, Sacks F (2009) Omega-6 fatty acids and risk for cardiovascular disease: a science advisory from the American Heart Association Nutrition Subcommittee of the Council on Nutrition, Physical Activity, and Metabolism; Council on Cardiovascular Nursing; and Council on Epidemiology and Prevention. Circulation 119:902–907PubMedCrossRef Harris WS, Mozaffarian D, Rimm E, Kris-Etherton P, Rudel LL, Appel LJ, Engler MM, Engler MB, Sacks F (2009) Omega-6 fatty acids and risk for cardiovascular disease: a science advisory from the American Heart Association Nutrition Subcommittee of the Council on Nutrition, Physical Activity, and Metabolism; Council on Cardiovascular Nursing; and Council on Epidemiology and Prevention. Circulation 119:902–907PubMedCrossRef
38.
Zurück zum Zitat Chait A, den Hartigh LJ (2020) Adipose tissue distribution, inflammation and its metabolic consequences, including diabetes and cardiovascular disease. Front Cardiovasc Med 7:22PubMedPubMedCentralCrossRef Chait A, den Hartigh LJ (2020) Adipose tissue distribution, inflammation and its metabolic consequences, including diabetes and cardiovascular disease. Front Cardiovasc Med 7:22PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat DiNicolantonio JJ, O'Keefe JH (2018) Omega-6 vegetable oils as a driver of coronary heart disease: the oxidized linoleic acid hypothesis. Open Heart 5:e000898-2018-000898 (eCollection 2018) DiNicolantonio JJ, O'Keefe JH (2018) Omega-6 vegetable oils as a driver of coronary heart disease: the oxidized linoleic acid hypothesis. Open Heart 5:e000898-2018-000898 (eCollection 2018)
40.
Zurück zum Zitat Schuster S, Johnson CD, Hennebelle M, Holtmann T, Taha AY, Kirpich IA, Eguchi A, Ramsden CE, Papouchado BG, McClain CJ, Feldstein AE (2018) Oxidized linoleic acid metabolites induce liver mitochondrial dysfunction, apoptosis, and NLRP3 activation in mice. J Lipid Res 59:1597–1609PubMedPubMedCentralCrossRef Schuster S, Johnson CD, Hennebelle M, Holtmann T, Taha AY, Kirpich IA, Eguchi A, Ramsden CE, Papouchado BG, McClain CJ, Feldstein AE (2018) Oxidized linoleic acid metabolites induce liver mitochondrial dysfunction, apoptosis, and NLRP3 activation in mice. J Lipid Res 59:1597–1609PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Vangaveti V, Baune BT, Kennedy RL (2010) Hydroxyoctadecadienoic acids: novel regulators of macrophage differentiation and atherogenesis. Ther Adv Endocrinol Metab 1:51–60PubMedPubMedCentralCrossRef Vangaveti V, Baune BT, Kennedy RL (2010) Hydroxyoctadecadienoic acids: novel regulators of macrophage differentiation and atherogenesis. Ther Adv Endocrinol Metab 1:51–60PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Hildreth K, Kodani SD, Hammock BD, Zhao L (2020) Cytochrome P450-derived linoleic acid metabolites EpOMEs and DiHOMEs: a review of recent studies. J Nutr Biochem 86:108484PubMedPubMedCentralCrossRef Hildreth K, Kodani SD, Hammock BD, Zhao L (2020) Cytochrome P450-derived linoleic acid metabolites EpOMEs and DiHOMEs: a review of recent studies. J Nutr Biochem 86:108484PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Levan SR, Stamnes KA, Lin DL, Panzer AR, Fukui E, McCauley K, Fujimura KE, McKean M, Ownby DR, Zoratti EM, Boushey HA, Cabana MD, Johnson CC, Lynch SV (2019) Elevated faecal 12,13-diHOME concentration in neonates at high risk for asthma is produced by gut bacteria and impedes immune tolerance. Nat Microbiol 4:1851–1861PubMedPubMedCentralCrossRef Levan SR, Stamnes KA, Lin DL, Panzer AR, Fukui E, McCauley K, Fujimura KE, McKean M, Ownby DR, Zoratti EM, Boushey HA, Cabana MD, Johnson CC, Lynch SV (2019) Elevated faecal 12,13-diHOME concentration in neonates at high risk for asthma is produced by gut bacteria and impedes immune tolerance. Nat Microbiol 4:1851–1861PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Zhao R, Tian L, Zhao B, Sun Y, Cao J, Chen K, Li F, Li M, Shang D, Liu M (2020) FADS1 promotes the progression of laryngeal squamous cell carcinoma through activating AKT/mTOR signaling. Cell Death Dis 11:272-020-2457-5 Zhao R, Tian L, Zhao B, Sun Y, Cao J, Chen K, Li F, Li M, Shang D, Liu M (2020) FADS1 promotes the progression of laryngeal squamous cell carcinoma through activating AKT/mTOR signaling. Cell Death Dis 11:272-020-2457-5
47.
Zurück zum Zitat Stancakova A, Paananen J, Soininen P, Kangas AJ, Bonnycastle LL, Morken MA, Collins FS, Jackson AU, Boehnke ML, Kuusisto J, Ala-Korpela M, Laakso M (2011) Effects of 34 risk loci for type 2 diabetes or hyperglycemia on lipoprotein subclasses and their composition in 6,580 nondiabetic Finnish men. Diabetes 60:1608–1616PubMedPubMedCentralCrossRef Stancakova A, Paananen J, Soininen P, Kangas AJ, Bonnycastle LL, Morken MA, Collins FS, Jackson AU, Boehnke ML, Kuusisto J, Ala-Korpela M, Laakso M (2011) Effects of 34 risk loci for type 2 diabetes or hyperglycemia on lipoprotein subclasses and their composition in 6,580 nondiabetic Finnish men. Diabetes 60:1608–1616PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Takkunen MJ, de Mello VD, Schwab US, Kuusisto J, Vaittinen M, Agren JJ, Laakso M, Pihlajamaki J, Uusitupa MI (2016) Gene-diet interaction of a common FADS1 variant with marine polyunsaturated fatty acids for fatty acid composition in plasma and erythrocytes among men. Mol Nutr Food Res 60:381–389PubMedCrossRef Takkunen MJ, de Mello VD, Schwab US, Kuusisto J, Vaittinen M, Agren JJ, Laakso M, Pihlajamaki J, Uusitupa MI (2016) Gene-diet interaction of a common FADS1 variant with marine polyunsaturated fatty acids for fatty acid composition in plasma and erythrocytes among men. Mol Nutr Food Res 60:381–389PubMedCrossRef
Metadaten
Titel
The FADS1 genotypes modify the effect of linoleic acid-enriched diet on adipose tissue inflammation via pro-inflammatory eicosanoid metabolism
verfasst von
Maija Vaittinen
Maria A. Lankinen
Pirjo Käkelä
Jyrki Ågren
Craig E. Wheelock
Markku Laakso
Ursula Schwab
Jussi Pihlajamäki
Publikationsdatum
14.06.2022
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Nutrition / Ausgabe 7/2022
Print ISSN: 1436-6207
Elektronische ISSN: 1436-6215
DOI
https://doi.org/10.1007/s00394-022-02922-y

Weitere Artikel der Ausgabe 7/2022

European Journal of Nutrition 7/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.