1
|
Rabehl M, Wei Z, Leineweber CG, Enssle J, Rothe M, Jung A, Schmöcker C, Elbelt U, Weylandt KH, Pietzner A. Effect of FADS1 SNPs rs174546, rs174547 and rs174550 on blood fatty acid profiles and plasma free oxylipins. Front Nutr 2024; 11:1356986. [PMID: 39021601 PMCID: PMC11253720 DOI: 10.3389/fnut.2024.1356986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 06/06/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Previous studies have indicated that activity of fatty acid desaturase 1 (FADS1), is involved in cardiometabolic risk. Recent experimental data have shown that FADS1 knockdown can promote lipid accumulation and lipid droplet formation in liver cells. In this study, we aimed to characterize whether different FADS1 genotypes affect liver fat content, essential fatty acid content and free oxylipin mediators in the blood. Methods We analyzed the impact of FADS1 single-nucleotide polymorphisms (SNPs) rs174546, rs174547, and rs174550 on blood fatty acids and free oxylipins in a cohort of 85 patients from an academic metabolic medicine outpatient center. Patients were grouped based on their genotype into the homozygous major (derived) allele group, the heterozygous allele group, and the homozygous minor (ancestral) allele group. Omega-3 polyunsaturated fatty acids (n-3 PUFA) and omega-6 polyunsaturated fatty acids (n-6 PUFA) in the blood cell and plasma samples were analyzed by gas chromatography. Free Oxylipins in plasma samples were analyzed using HPLC-MS/MS. Liver fat content and fibrosis were evaluated using Fibroscan technology. Results Patients with the homozygous ancestral (minor) FADS1 genotype exhibited significantly lower blood levels of the n-6 PUFA arachidonic acid (AA), but no significant differences in the n-3 PUFAs eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). There were no significant differences in liver fat content or arachidonic acid-derived lipid mediators, such as thromboxane B2 (TXB2), although there was a trend toward lower levels in the homozygous ancestral genotype group. Discussion Our findings suggest that FADS1 genotypes influence the blood levels of n-6 PUFAs, while not significantly affecting the n-3 PUFAs EPA and DHA. The lack of significant differences in liver fat content and arachidonic acid-derived lipid mediators suggests that the genotype-related variations in fatty acid levels may not directly translate to differences in liver fat or inflammatory lipid mediators in this cohort. However, the trend towards lower levels of certain lipid mediators in the homozygous ancestral genotype group warrants further investigation to elucidate the underlying mechanisms of different FADS1 genotypes and potential implications for cardiometabolic risk.
Collapse
Affiliation(s)
- Miriam Rabehl
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | - Zeren Wei
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Can G. Leineweber
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | - Jörg Enssle
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | | | - Adelheid Jung
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
| | - Christoph Schmöcker
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | - Ulf Elbelt
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Karsten H. Weylandt
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | - Anne Pietzner
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| |
Collapse
|
2
|
Loukil I, Mutch DM, Plourde M. Genetic association between FADS and ELOVL polymorphisms and the circulating levels of EPA/DHA in humans: a scoping review. GENES & NUTRITION 2024; 19:11. [PMID: 38844860 PMCID: PMC11157910 DOI: 10.1186/s12263-024-00747-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) are two omega-3 fatty acids that can be synthesized out of their precursor alpha-linolenic acid (ALA). FADS and ELOVL genes encode the desaturase and elongase enzymes required for EPA and DHA synthesis from ALA; however, single nucleotide polymorphisms (SNPs) in FADS and ELOVL genes could modify the levels of EPA and DHA synthesized from ALA although there is no consensus in this area. This review aims to investigate EPA and DHA circulating levels in human blood and their association with FADS or ELOVL. METHODS PubMed, Cochrane, and Scopus databases were used to identify research articles. They were subsequently reviewed by two independent investigators. RESULTS Initially, 353 papers were identified. After removing duplicates and articles not meeting inclusion criteria, 98 full text papers were screened. Finally, this review included 40 studies investigating FADS and/or ELOVL polymorphisms. A total of 47 different SNPs in FADS genes were reported. FADS1 rs174537, rs174547, rs174556 and rs174561 were the most studied SNPs, with minor allele carriers having lower levels of EPA and DHA. SNPs in the FADS genes were in high linkage disequilibrium. SNPs in FADS were correlated with levels of EPA and DHA. No conclusion could be drawn with the ELOVL polymorphisms since the number of studies was too low. CONCLUSION Specific SNPs in FADS gene, such as rs174537, have strong associations with circulating levels of EPA and DHA. Continued investigation regarding the impact of genetic variants related to EPA and DHA synthesis is warranted.
Collapse
Affiliation(s)
- Insaf Loukil
- Research Center on Aging, Health, and Social Sciences Center, Department of Medicine, Sherbrooke University Geriatrics Institute, University of Sherbrooke, Sherbrooke, QC, J1G 1B1, Canada
- Department de Medicine, Faculty of Medicine and health sciences, University of Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada
| | - David M Mutch
- Department of Human Health and Nutritional Sciences, Guelph, ON, N1G 2W1, Canada
| | - Mélanie Plourde
- Research Center on Aging, Health, and Social Sciences Center, Department of Medicine, Sherbrooke University Geriatrics Institute, University of Sherbrooke, Sherbrooke, QC, J1G 1B1, Canada.
- Department de Medicine, Faculty of Medicine and health sciences, University of Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada.
| |
Collapse
|
3
|
Ghooray DT, Xu M, Shi H, McClain CJ, Song M. Hepatocyte-Specific Fads1 Overexpression Attenuates Western Diet-Induced Metabolic Phenotypes in a Rat Model. Int J Mol Sci 2024; 25:4836. [PMID: 38732052 PMCID: PMC11084797 DOI: 10.3390/ijms25094836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/01/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Fatty acid desaturase 1 (FADS1) is a rate-limiting enzyme in long-chain polyunsaturated fatty acid (LCPUFA) synthesis. Reduced activity of FADS1 was observed in metabolic dysfunction-associated steatotic liver disease (MASLD). The aim of this study was to determine whether adeno-associated virus serotype 8 (AAV8) mediated hepatocyte-specific overexpression of Fads1 (AAV8-Fads1) attenuates western diet-induced metabolic phenotypes in a rat model. Male weanling Sprague-Dawley rats were fed with a chow diet, or low-fat high-fructose (LFHFr) or high-fat high-fructose diet (HFHFr) ad libitum for 8 weeks. Metabolic phenotypes were evaluated at the endpoint. AAV8-Fads1 injection restored hepatic FADS1 protein levels in both LFHFr and HFHFr-fed rats. While AAV8-Fads1 injection led to improved glucose tolerance and insulin signaling in LFHFr-fed rats, it significantly reduced plasma triglyceride (by ~50%) and hepatic cholesterol levels (by ~25%) in HFHFr-fed rats. Hepatic lipidomics analysis showed that FADS1 activity was rescued by AAV8-FADS1 in HFHFr-fed rats, as shown by the restored arachidonic acid (AA)/dihomo-γ-linolenic acid (DGLA) ratio, and that was associated with reduced monounsaturated fatty acid (MUFA). Our data suggest that the beneficial role of AAV8-Fads1 is likely mediated by the inhibition of fatty acid re-esterification. FADS1 is a promising therapeutic target for MASLD in a diet-dependent manner.
Collapse
Affiliation(s)
- Dushan T. Ghooray
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (D.T.G.); (M.X.); (C.J.M.)
| | - Manman Xu
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (D.T.G.); (M.X.); (C.J.M.)
| | - Hongxue Shi
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Craig J. McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (D.T.G.); (M.X.); (C.J.M.)
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA
| | - Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (D.T.G.); (M.X.); (C.J.M.)
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Capra ME, Stanyevic B, Giudice A, Monopoli D, Decarolis NM, Esposito S, Biasucci G. Long-Chain Polyunsaturated Fatty Acids Effects on Cardiovascular Risk in Childhood: A Narrative Review. Nutrients 2023; 15:nu15071661. [PMID: 37049503 PMCID: PMC10096679 DOI: 10.3390/nu15071661] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Long-chain polyunsaturated fatty acids (LCPUFAs) are semi-essential fatty acids widely studied in adult subjects for their healthy-heart effects, especially on secondary prevention in patients who already experienced a cardiac event. LCPUFAs consumption is safe, without adverse effects, and they are usually well-tolerated; they can be taken either in foods or as nutritional supplements. LCPUFAs' positive effect on global health has been worldwide recognized also for pediatric patients. In childhood and adolescence, research has mainly focused on LCPUFAs' effects on neurodevelopment, brain and visual functions and on maternal-fetal medicine, yet their cardiovascular effects in childhood are still understudied. Atherosclerosis is a multifactorial process that starts even before birth and progresses throughout life; thus, cardiovascular prevention is advisable and effective from the very first years of life. Nutritional and lifestyle interventions are the main factors that can interfere with atherosclerosis in childhood, and the consumption of specific nutrients, such as LCPUFAs, can enhance positive nutritional effects. The aim of our narrative review is to analyze the effect of LCPUFAs on cardiovascular risk factors and on cardiovascular risk prevention in developmental age, focusing on specific conditions such as weight excess and dyslipidemia.
Collapse
Affiliation(s)
- Maria Elena Capra
- Pediatrics and Neonatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy
- Società Italiana di Nutrizione Pediatrica, 20126 Milan, Italy
| | - Brigida Stanyevic
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Antonella Giudice
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Delia Monopoli
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Nicola Mattia Decarolis
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Giacomo Biasucci
- Pediatrics and Neonatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy
- Società Italiana di Nutrizione Pediatrica, 20126 Milan, Italy
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
5
|
Huang P, Cheng H, Su Y, Huang M, Hsu C, Hwang S, Shin S, Chang W. 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. J Diabetes Investig 2022; 14:297-308. [PMID: 36412559 PMCID: PMC9889619 DOI: 10.1111/jdi.13944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/10/2022] [Accepted: 10/28/2022] [Indexed: 11/23/2022] Open
Abstract
AIMS/INTRODUCTION Fatty acid desaturase (FADS) genetic polymorphisms are strongly correlated with the risk of dyslipidemia and cardiovascular disease. In this study, we examined the impact of FADS1 and FADS2 genetic variants on plasma lipid status, and assessed interactions between FADS genetic polymorphisms and plasma n-3/n-6 fatty acids regarding lipid status within a population of 816 Taiwanese patients with type 2 diabetes. MATERIALS AND METHODS Selected tag single-nucleotide polymorphisms (FADS1 rs174546 [T/C]; FADS2 rs174602 [A/G] and rs2072114 [A/G]) were genotyped (n = 816). RESULTS The distribution of genotypes were compared with reports publicly available in the Genome Aggregation Database for East Asian populations (https://gnomad.broadinstitute.org). In the subgroup of patients not taking lipid-lowering medications (n = 192), we observed that the G allele of FADS2 rs174602 was statistically significantly correlated with lower low-density lipoprotein cholesterol (LDL-C) concentrations (P = 0.001), whereas the G allele of rs2072114 was marginally associated with LDL-C concentrations (P = 0.091). Using a general linear model adjusted for confounding factors, statistically significant interactions (P = 0.016) between single-nucleotide polymorphisms in rs2072114 and a low alpha-linolenic acid (18:3n-3)/linoleic acid (18:2n-6) ratio; the G allele correlated with lower LDL-C levels among individuals with a low alpha-linolenic acid/linoleic acid ratio. Interaction between rs174602 single-nucleotide polymorphisms and low alpha-linolenic acid/linoleic acid values on LDL-C was only marginally significant (P = 0.063). CONCLUSIONS Our results show the role of n-3/n-6 dietary polyunsaturated fatty acids in modifying the effects of genetic susceptibility on lipoprotein concentrations in patients with type 2 diabetes. Our findings highlight the potential of interventions with dietary polyunsaturated fatty acids regarding developing individualized prevention strategies for type 2 diabetes presenting with co-occurring dyslipidemia and cardiovascular diseases.
Collapse
Affiliation(s)
- Pei‐Chi Huang
- Department of Public Health and Environmental Medicine, School of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan,Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Hsuan Cheng
- Department of Public Health and Environmental Medicine, School of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Yu‐Ting Su
- Department of Public Health and Environmental Medicine, School of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan,Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Meng‐Chuan Huang
- Department of Public Health and Environmental Medicine, School of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan,Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan,Department of Nutrition and DieteticsKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Chih‐Cheng Hsu
- Institute of Population Health SciencesNational Health Research InstitutesZhunanTaiwan,Department of Health Services AdministrationChina Medical UniversityTaichungTaiwan
| | - Shang‐Jyh Hwang
- Department of Internal MedicineKaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiungTaiwan
| | - Shyi‐Jang Shin
- Department of Internal MedicineKaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiungTaiwan
| | - Wen‐Tsan Chang
- Division of General and Digestive Surgery, Department of SurgeryKaohsiung Medical University HospitalKaohsiungTaiwan,Department of Surgery, School of Medicine, College of MedicineKaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiungTaiwan,Department of Biotechnology, College of Life ScienceKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
6
|
Žák A, Jáchymová M, Burda M, Staňková B, Zeman M, Slabý A, Vecka M, Šeda O. FADS Polymorphisms Affect the Clinical and Biochemical Phenotypes of Metabolic Syndrome. Metabolites 2022; 12:metabo12060568. [PMID: 35736500 PMCID: PMC9228863 DOI: 10.3390/metabo12060568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 12/04/2022] Open
Abstract
Long-chain polyunsaturated fatty acids (LC-PUFAs) play important roles in human health, from controlling inflammation to lipid and glucose homeostasis. In our previous study, which employed a cluster analysis of a plasma fatty acid (FA) pattern, we identified two clusters of metabolic syndrome (MetS) independent of clinical and biochemical parameters within the whole study group (controls together with metabolic syndrome (MetS) patients). FA desaturase (FADS) genes are the key regulators of LC-PUFA metabolism. The aim of this study was to analyze associations between FADS polymorphisms and clusters of MetS. The study group consisted of 188 controls and 166 patients with MetS. The first cluster contained 71 controls (CON1) and 109 MetS patients (MetS1). The second cluster consisted of 117 controls (CON2) and 57 MetS patients (MetS2). In comparison with MetS2, cluster MetS1 displayed a more adverse risk profile. Cluster CON1 had, in comparison with CON2, higher body weight and increased triacylglycerol levels (p < 0.05). We found that the FADS rs174537 (p < 0.001), rs174570 (p < 0.01), and rs174602 (p < 0.05) polymorphisms along with two inferred haplotypes had statistically significant genotype associations with the splitting of MetS into MetS1 and MetS2. Conversely, we observed no significant differences in the distribution of FADS polymorphisms between MetS and CON subjects, or between CON1 and CON2. These associations between FADS polymorphisms and two clusters of MetS (differing in waist circumference, HOMA-IR, lipolysis, and oxidative stress) implicate the important influence of genetic factors on the phenotypic manifestation of MetS.
Collapse
Affiliation(s)
- Aleš Žák
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
| | - Marie Jáchymová
- Institute of Clinical Chemistry and Laboratory Diagnostics, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic;
| | - Michal Burda
- Institute for Research and Applications of Fuzzy Modeling, University of Ostrava, 701 03 Ostrava, Czech Republic;
| | - Barbora Staňková
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
| | - Miroslav Zeman
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
| | - Adolf Slabý
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
| | - Marek Vecka
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
- Institute of Clinical Chemistry and Laboratory Diagnostics, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic;
- Correspondence:
| | - Ondřej Šeda
- Institute of Biology and Medical Genetics, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 00 Prague, Czech Republic;
| |
Collapse
|
7
|
Huang J, Shao Y, Zong X, Zhang H, Zhang X, Zhang Z, Shi H. FADS1 overexpression promotes fatty acid synthesis and triacylglycerol accumulation via inhibiting the AMPK/SREBP1 pathway in goat mammary epithelial cells. Food Funct 2022; 13:5870-5882. [PMID: 35548952 DOI: 10.1039/d2fo00246a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Delta-5 desaturase (D5D), encoded by the fatty acid desaturase 1 (FADS1) gene, is a rate-limiting enzyme in polyunsaturated fatty acid (PUFA) synthesis that influences the PUFA levels in milk fat. However, the function and molecular mechanism of FADS1 in milk fat metabolism remain largely unknown. The FADS1 overexpression increased the triglyceride content, lipid droplet size, and expression of genes related to fatty acid de novo synthesis (SREBP1 and ACC), intracellular fatty acid transporters (FABP3 and FABP4) and triacylglycerol synthesis gene (DGAT2). It also significantly promoted the SREBP1 nuclear translocation by inhibiting the AMPK activation. In addition, FADS1 overexpression inhibited cell proliferation and arrested cell cycle at the G1 phase. These findings reveal a novel FADS1-AMPK-SREBP1 pathway regulating milk fat production in the goat mammary gland.
Collapse
Affiliation(s)
- Jiangtao Huang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Yuexin Shao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Xueyang Zong
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Huawen Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Xian Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Zhifei Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Huaiping Shi
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
8
|
Seral-Cortes M, Larruy-García A, De Miguel-Etayo P, Labayen I, Moreno LA. Mediterranean Diet and Genetic Determinants of Obesity and Metabolic Syndrome in European Children and Adolescents. Genes (Basel) 2022; 13:genes13030420. [PMID: 35327974 PMCID: PMC8954235 DOI: 10.3390/genes13030420] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/25/2022] Open
Abstract
Childhood obesity and metabolic syndrome (MetS) are multifactorial diseases influenced by genetic and environmental factors. The Mediterranean Diet (MD) seems to modulate the genetic predisposition to obesity or MetS in European adults. The FTO gene has also been shown to have an impact on the MD benefits to avoid obesity or MetS. Since these interaction effects have been scarcely analyzed in European youth, the aim was to describe the gene–MD interplay, analyzing the impact of the genetic factors to reduce the obesity and MetS risk through MD adherence, and the MD impact in the obesity and MetS genetic profile. From the limited evidence on gene–MD interaction studies in European youth, a study showed that the influence of high MD adherence on adiposity and MetS was only observed with a limited number of risk alleles; the gene–MD interplay showed sex-specific differences, being higher in females. Most results analyzed in European adults elucidate that, the relationship between MD adherence and both obesity and MetS risk, could be modulated by obesity genetic variants and vice versa. Further research is needed, to better understand the inter-individual differences in the association between MD and body composition, and the integration of omics and personalized nutrition considering MD.
Collapse
Affiliation(s)
- Miguel Seral-Cortes
- Growth, Exercise, NUtrition and Development (GENUD) Research Group, Faculty of Health Sciences, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.S.-C.); (A.L.-G.); (L.A.M.)
| | - Alicia Larruy-García
- Growth, Exercise, NUtrition and Development (GENUD) Research Group, Faculty of Health Sciences, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.S.-C.); (A.L.-G.); (L.A.M.)
| | - Pilar De Miguel-Etayo
- Growth, Exercise, NUtrition and Development (GENUD) Research Group, Faculty of Health Sciences, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.S.-C.); (A.L.-G.); (L.A.M.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence:
| | - Idoia Labayen
- Department of Health Sciences, Public University of Navarra, 31006 Pamplona, Spain;
| | - Luis A. Moreno
- Growth, Exercise, NUtrition and Development (GENUD) Research Group, Faculty of Health Sciences, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.S.-C.); (A.L.-G.); (L.A.M.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
9
|
Keathley J, Garneau V, Marcil V, Mutch DM, Robitaille J, Rudkowska I, Sofian GM, Desroches S, Vohl MC. Nutrigenetics, omega-3 and plasma lipids/lipoproteins/apolipoproteins with evidence evaluation using the GRADE approach: a systematic review. BMJ Open 2022; 12:e054417. [PMID: 35193914 PMCID: PMC8867311 DOI: 10.1136/bmjopen-2021-054417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 01/28/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Despite the uptake of nutrigenetic testing through direct-to-consumer services and healthcare professionals, systematic reviews determining scientific validity are limited in this field. The objective of this review was to: retrieve, synthesise and assess the quality of evidence (confidence) for nutrigenetic approaches related to the effect of genetic variation on plasma lipid, lipoprotein and apolipoprotein responsiveness to omega-3 fatty acid intake. DESIGN A systematic review was conducted using three search engines (Embase, Web of Science and Medline) for articles published up until 1 August 2020. We aimed to systematically search, identify (select) and provide a narrative synthesis of all studies that assessed nutrigenetic associations/interactions for genetic variants (comparators) influencing the plasma lipid, lipoprotein and/or apolipoprotein response (outcomes) to omega-3 fatty acid intake (intervention/exposure) in humans-both paediatric and adult populations (population). We further aimed to assess the overall quality of evidence for specific priority nutrigenetic associations/interactions based on the following inclusion criteria: nutrigenetic associations/interactions reported for the same genetic variants (comparators) influencing the same plasma lipid, lipoprotein and/or apolipoprotein response (outcomes) to omega-3 fatty acid intake (intervention/exposure) in humans-both paediatric and adult populations (population) in at least two independent studies, irrespective of the findings. Risk of bias was assessed in individual studies. Evidence was evaluated using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach with a modification to further consider biological plausibility. RESULTS Out of 1830 articles screened, 65 met the inclusion criteria for the narrative synthesis (n=23 observational, n=42 interventional); of these, 25 met the inclusion criteria for GRADE evidence evaluation. Overall, current evidence is insufficient for gene-diet associations related to omega-3 fatty acid intake on plasma apolipoproteins, total cholesterol, high-density lipoprotein-cholesterol, low-density lipoprotein (LDL)-cholesterol and LDL particle size. However, there is strong (GRADE rating: moderate quality) evidence to suggest that male APOE-E4 carriers (rs429358, rs7412) exhibit significant triglyceride reductions in response to omega-3-rich fish oil with a dose-response effect. Moreover, strong (GRADE rating: high quality) evidence suggests that a 31-SNP nutrigenetic risk score can predict plasma triglyceride responsiveness to omega-3-rich fish oil in adults with overweight/obesity from various ethnicities. CONCLUSIONS Most evidence in this area is weak, but two specific nutrigenetic interactions exhibited strong evidence, with generalisability limited to specific populations. PROSPERO REGISTRATION NUMBER CRD42020185087.
Collapse
Affiliation(s)
- Justine Keathley
- Université Laval, Centre Nutrition, Santé et Société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Quebec, Quebec City, Quebec, Canada
- Universite Laval, School of Nutrition, Quebec City, Quebec, Canada
| | - Véronique Garneau
- Université Laval, Centre Nutrition, Santé et Société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Quebec, Quebec City, Quebec, Canada
- Universite Laval, School of Nutrition, Quebec City, Quebec, Canada
| | - Valérie Marcil
- Department of Nutrition, University of Montréal, Montréal, Quebec, Canada
- Research Centre, Sainte-Justine University Health Centre, Montréal, Quebec, Canada
| | - David M Mutch
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Julie Robitaille
- Université Laval, Centre Nutrition, Santé et Société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Quebec, Quebec City, Quebec, Canada
- Universite Laval, School of Nutrition, Quebec City, Quebec, Canada
| | - Iwona Rudkowska
- Department of Kinesiology, Université Laval, Quebec City, Quebec, Canada
- Endocrinology and Nephrology Unit, CHU de Québec - Université Laval Research Center, Quebec City, Quebec, Canada
| | | | - Sophie Desroches
- Université Laval, Centre Nutrition, Santé et Société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Quebec, Quebec City, Quebec, Canada
- Universite Laval, School of Nutrition, Quebec City, Quebec, Canada
| | - Marie-Claude Vohl
- Université Laval, Centre Nutrition, Santé et Société (NUTRISS), Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Quebec, Quebec City, Quebec, Canada
- Universite Laval, School of Nutrition, Quebec City, Quebec, Canada
| |
Collapse
|
10
|
Ahluwalia MK. Nutrigenetics and nutrigenomics-A personalized approach to nutrition. ADVANCES IN GENETICS 2021; 108:277-340. [PMID: 34844714 DOI: 10.1016/bs.adgen.2021.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The prevalence of non-communicable diseases has been on an upward trajectory for some time and this puts an enormous burden on the healthcare expenditure. Lifestyle modifications including dietary interventions hold an immense promise to manage and prevent these diseases. Recent advances in genomic research provide evidence that focussing these efforts on individual variations in abilities to metabolize nutrients (nutrigenetics) and exploring the role of dietary compounds on gene expression (nutrigenomics and nutri-epigenomics) can lead to more meaningful personalized dietary strategies to promote optimal health. This chapter aims to provide examples on these gene-diet interactions at multiple levels to support the need of embedding targeted dietary interventions as a way forward to prevent, avoid and manage diseases.
Collapse
|
11
|
Iglesias-Carres L, Neilson AP. Utilizing preclinical models of genetic diversity to improve translation of phytochemical activities from rodents to humans and inform personalized nutrition. Food Funct 2021; 12:11077-11105. [PMID: 34672309 DOI: 10.1039/d1fo02782d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mouse models are an essential tool in different areas of research, including nutrition and phytochemical research. Traditional inbred mouse models have allowed the discovery of therapeutical targets and mechanisms of action and expanded our knowledge of health and disease. However, these models lack the genetic variability typically found in human populations, which hinders the translatability of the results found in mice to humans. The development of genetically diverse mouse models, such as the collaborative cross (CC) or the diversity outbred (DO) models, has been a useful tool to overcome this obstacle in many fields, such as cancer, immunology and toxicology. However, these tools have not yet been widely adopted in the field of phytochemical research. As demonstrated in other disciplines, use of CC and DO models has the potential to provide invaluable insights for translation of phytochemicals from rodents to humans, which are desperately needed given the challenges and numerous failed clinical trials in this field. These models may prove informative for personalized use of phytochemicals in humans, including: predicting interindividual variability in phytochemical bioavailability and efficacy, identifying genetic loci or genes governing response to phytochemicals, identifying phytochemical mechanisms of action and therapeutic targets, and understanding the impact of genetic variability on individual response to phytochemicals. Such insights would prove invaluable for personalized implementation of phytochemicals in humans. This review will focus on the current work performed with genetically diverse mouse populations, and the research opportunities and advantages that these models can offer to phytochemical research.
Collapse
Affiliation(s)
- Lisard Iglesias-Carres
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA.
| | - Andrew P Neilson
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA.
| |
Collapse
|
12
|
Metelcová T, Vaňková M, Zamrazilová H, Hovhannisyan M, Staňková B, Tvrzická E, Hill M, Hainer V, Včelák J, Kunešová M. FADS1 gene polymorphism(s) and fatty acid composition of serum lipids in adolescents. Lipids 2021; 56:499-508. [PMID: 34189740 DOI: 10.1002/lipd.12317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 11/09/2022]
Abstract
Polyunsaturated fatty acids (PUFA) influence many physiological functions. Associations have been found between single nucleotide polymorphisms (SNP) in the FADS1 (Fatty acid desaturase 1) gene and the relative abundance of PUFA in serum lipids. This study examines the relationship between two SNPs in the FADS1 gene (rs174546, rs174537) and the fatty acid (FA) composition of serum lipids in adolescents (13-18 years). We used DNA samples (670 children; 336 girls and 334 boys) from the Childhood Obesity Prevalence and Treatment (COPAT) project. Genomic DNA was extracted from peripheral blood leukocytes in whole blood samples. For genotype analysis, TaqMan SNP Genotyping assays (Applied Biosystems) were used. Fatty acid composition of serum lipids was assessed using gas chromatography. The T-statistic and regression were used for statistical evaluations. Minor allele T carriers in both SNPs had significant lower level of palmitic acid (16:0, phospholipids) and arachidonic acid (20:4[n-6], phospholipids) in both sexes. In girls, we found a significant positive association between minor allele T carriers and eicosadienoic acid (20:2[n-6], cholesteryl esters) in both SNPs. Being a minor allele T carrier was significantly positively associated with dihomo-γ-linolenic acid (20:3[n-6], phospholipids) in boys in both SNPs. SNPs (including rs174546, rs174537) in the FADS gene cluster should have impacted desaturase activity, which may contribute to different efficiency of PUFA synthesis.
Collapse
Affiliation(s)
- Tereza Metelcová
- Institute of Endocrinology, Prague, The Czech Republic.,1st Medical Faculty, Charles University, Prague, The Czech Republic
| | | | | | | | - Barbora Staňková
- 4th Department of Internal Medicine, 1st Medical Faculty, Charles University, Prague, The Czech Republic
| | - Eva Tvrzická
- 4th Department of Internal Medicine, 1st Medical Faculty, Charles University, Prague, The Czech Republic
| | - Martin Hill
- Institute of Endocrinology, Prague, The Czech Republic
| | | | - Josef Včelák
- Institute of Endocrinology, Prague, The Czech Republic
| | - Marie Kunešová
- Institute of Endocrinology, Prague, The Czech Republic.,4th Department of Internal Medicine, 1st Medical Faculty, Charles University, Prague, The Czech Republic
| |
Collapse
|
13
|
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. Lancet Diabetes Endocrinol 2020; 8:915-930. [PMID: 32949497 DOI: 10.1016/s2213-8587(20)30148-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 12/12/2022]
Abstract
Prospective observational studies support the use of long-chain omega-3 polyunsaturated fatty acids (PUFAs) in the primary prevention of atherosclerotic cardiovascular disease; however, randomised controlled trials, have often reported neutral findings. There is a long history of debate about the potential harmful effects of a high intake of omega-6 PUFAs, although this idea is not supported by prospective observational studies or randomised controlled trials. Health effects of PUFAs might be influenced by Δ-5 and Δ-6 desaturases, the key enzymes in the metabolism of PUFAs. The activity of these enzymes and modulation by variants in encoding genes (FADS1-2-3 gene cluster) are linked to several cardiometabolic traits. This Review will further consider non-genetic determinants of desaturase activity, which have the potential to modify the availability of PUFAs to tissues. Finally, we discuss the consequences of altered desaturase activity in the context of PUFA intake, that is, gene-diet interactions and their clinical and public health implications.
Collapse
Affiliation(s)
- Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany; German Center for Diabetes Research, Neuherberg, Germany.
| | - Anne Marie Minihane
- Department of Nutrition and Preventive Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
| | - Rasha Noureldin M Saleh
- Department of Nutrition and Preventive Medicine, Norwich Medical School, University of East Anglia, Norwich, UK; Clinical Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
14
|
Murphy C, Johnson AP, Koenekoop RK, Seiple W, Overbury O. The Relationship Between Cognitive Status and Known Single Nucleotide Polymorphisms in Age-Related Macular Degeneration. Front Aging Neurosci 2020; 12:586691. [PMID: 33178008 PMCID: PMC7596199 DOI: 10.3389/fnagi.2020.586691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/15/2020] [Indexed: 11/13/2022] Open
Abstract
Recent literature has reported a higher occurrence of cognitive impairment among individuals with Age-related Macular Degeneration (AMD) compared to older adults with normal vision. This pilot study explored potential links between single nucleotide polymorphisms (SNPs) in AMD and cognitive status. Individuals with AMD (N = 21) and controls (N = 18) were genotyped for the SNPs CFHY402H, ARMS2A69S and FADS1 rs174547. Cognitive status was evaluated using the Montreal Cognitive Assessment. The two groups differed significantly on which subscales were most difficult. The control group had difficulty with delayed recall while those with AMD had difficulty on delayed recall in addition to abstraction and orientation. Homozygous carriers of the FADS1 rs174547 SNP had significantly lower scores than heterozygotes or non-carriers on the MoCA. The results suggest that the FADS1 SNP may play a role in visual impairment/cognitive impairment comorbidity as reflected in the poorer cognitive scores among homozygotes with AMD compared to those carrying only one, or no copies of the SNP.
Collapse
Affiliation(s)
- Caitlin Murphy
- Low Vision Lab, School of Optometry, University of Montreal, Montreal, QC, Canada
- Concordia Vision Labs, Department of Psychology, Concordia University, Montreal, QC, Canada
- Centre for Interdisciplinary Research in Rehabilitation of Greater Montreal (CRIR)/Centre de Réadaptation Lethbridge-Layton-Mackay du Centre Intégré Universitaire de Santé et de Services Sociaux du Centre-Ouest-de-l’Ile-de-Montréal (CIUSSS) du Centre-Ouest-de-l’Île-de-Montréal, Montreal, QC, Canada
| | - Aaron P. Johnson
- Concordia Vision Labs, Department of Psychology, Concordia University, Montreal, QC, Canada
- Centre for Interdisciplinary Research in Rehabilitation of Greater Montreal (CRIR)/Centre de Réadaptation Lethbridge-Layton-Mackay du Centre Intégré Universitaire de Santé et de Services Sociaux du Centre-Ouest-de-l’Ile-de-Montréal (CIUSSS) du Centre-Ouest-de-l’Île-de-Montréal, Montreal, QC, Canada
| | - Robert K. Koenekoop
- Paediatric Surgery and Human Genetics and Ophthalmology, Faculty of Medicine, McGill University Health Centre, Montreal QC, Canada
| | - William Seiple
- Arlene R. Gordon Research Institute, Lighthouse Guild, New York, NY, United States
- School of Medicine, New York University, New York, NY, United States
| | - Olga Overbury
- Low Vision Lab, School of Optometry, University of Montreal, Montreal, QC, Canada
- Lady Davis Institute of Medical Research, Montreal, QC, Canada
| |
Collapse
|
15
|
Li P, Shan B, Jia K, Hu F, Xiao Y, Zheng J, Gao YT, Wang H, Gao Y. Plasma omega-3 polyunsaturated fatty acids and recurrence of endometrial cancer. BMC Cancer 2020; 20:576. [PMID: 32563240 PMCID: PMC7305622 DOI: 10.1186/s12885-020-07035-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 06/03/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Omega-3 polyunsaturated fatty acids (PUFAs) were proposed to have potential effects against inflammation and cancer. However, results from epidemiology studies remain inconsistent. We aimed to explore the associations of plasma PUFAs with EC recurrence and all-cause mortality. METHOD Women diagnosed with endometrial cancer (EC) between 2008 and 2013 and underwent surgery at Fudan University Shanghai Cancer Center of China were recruited. Survival status was followed up through September 2017. EC recurrence and total cause deaths were identified through medical record and telephone interview. In total, 202 patients with enough plasma samples at time of surgery were included. There were 195 patients who provided baseline plasma and survival information included in the current study. Plasma omega-3 PUFAs were measured by GC-FID. Cox Proportional Hazard model adjusted for potential cofounders was used to estimate HRs and 95% CIs. RESULTS Median follow-up time for patients was 58 months after surgery. A total of 13 recurrences and 11 all-cause deaths, of which, 2 deaths from EC, were identified. Level of plasma EPA was higher in recurrent patients than total patients (0.78% vs 0.51%, P = 0.015). Higher plasma eicosapentaenoic acid (EPA) level trended to have positive association with EC recurrence (P-trend = 0.04), although comparing to the lowest tertile, the highest tertile of EPA level was not significantly associated with increased risk of EC recurrence (HRT3vsT1 = 6.02; 95%CI = 0.7-52.06). The association between total omega-3 PUFA and EC recurrence tended to be stronger among patients with deeper myometrial invasion (OR = 3.41; 95%CI = 1.06-10.95; P-interaction = 0.04). CONCLUSIONS Higher plasma EPA level was significantly associated with EC recurrence. Further studies are warranted to confirm these findings. TRIAL REGISTRATION ChiCTR1900025418; Retrospectively registered (26 August 2019); Chinses Clinical Trial Registry.
Collapse
Affiliation(s)
- Peiqin Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Boer Shan
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Keyu Jia
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Fan Hu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ying Xiao
- Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Jusheng Zheng
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | - Huaying Wang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Ying Gao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China.
| |
Collapse
|
16
|
Czumaj A, Śledziński T. Biological Role of Unsaturated Fatty Acid Desaturases in Health and Disease. Nutrients 2020; 12:E356. [PMID: 32013225 PMCID: PMC7071289 DOI: 10.3390/nu12020356] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/20/2020] [Accepted: 01/28/2020] [Indexed: 12/21/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are considered one of the most important components of cells that influence normal development and function of many organisms, both eukaryotes and prokaryotes. Unsaturated fatty acid desaturases play a crucial role in the synthesis of PUFAs, inserting additional unsaturated bonds into the acyl chain. The level of expression and activity of different types of desaturases determines profiles of PUFAs. It is well recognized that qualitative and quantitative changes in the PUFA profile, resulting from alterations in the expression and activity of fatty acid desaturases, are associated with many pathological conditions. Understanding of underlying mechanisms of fatty acid desaturase activity and their functional modification will facilitate the development of novel therapeutic strategies in diseases associated with qualitative and quantitative disorders of PUFA.
Collapse
Affiliation(s)
- Aleksandra Czumaj
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, Dębinki, 80-211 Gdansk, Poland;
| | | |
Collapse
|
17
|
Coltell O, Sorlí JV, Asensio EM, Barragán R, González JI, Giménez-Alba IM, Zanón-Moreno V, Estruch R, Ramírez-Sabio JB, Pascual EC, Ortega-Azorín C, Ordovas JM, Corella D. Genome-Wide Association Study for Serum Omega-3 and Omega-6 Polyunsaturated Fatty Acids: Exploratory Analysis of the Sex-Specific Effects and Dietary Modulation in Mediterranean Subjects with Metabolic Syndrome. Nutrients 2020; 12:E310. [PMID: 31991592 PMCID: PMC7071282 DOI: 10.3390/nu12020310] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Many early studies presented beneficial effects of polyunsaturated fatty acids (PUFA) on cardiovascular risk factors and disease. However, results from recent meta-analyses indicate that this effect would be very low or nil. One of the factors that may contribute to the inconsistency of the results is that, in most studies, genetic factors have not been taken into consideration. It is known that fatty acid desaturase (FADS) gene cluster in chromosome 11 is a very important determinant of plasma PUFA, and that the prevalence of the single nucleotide polymorphisms (SNPs) varies greatly between populations and may constitute a bias in meta-analyses. Previous genome-wide association studies (GWAS) have been carried out in other populations and none of them have investigated sex and Mediterranean dietary pattern interactions at the genome-wide level. Our aims were to undertake a GWAS to discover the genes most associated with serum PUFA concentrations (omega-3, omega-6, and some fatty acids) in a scarcely studied Mediterranean population with metabolic syndrome, and to explore sex and adherence to Mediterranean diet (MedDiet) interactions at the genome-wide level. Serum PUFA were determined by NMR spectroscopy. We found strong robust associations between various SNPs in the FADS cluster and omega-3 concentrations (top-ranked in the adjusted model: FADS1-rs174547, p = 3.34 × 10-14; FADS1-rs174550, p = 5.35 × 10-14; FADS2-rs1535, p = 5.85 × 10-14; FADS1-rs174546, p = 6.72 × 10-14; FADS2-rs174546, p = 9.75 × 10-14; FADS2- rs174576, p = 1.17 × 10-13; FADS2-rs174577, p = 1.12 × 10-12, among others). We also detected a genome-wide significant association with other genes in chromosome 11: MYRF (myelin regulatory factor)-rs174535, p = 1.49 × 10-12; TMEM258 (transmembrane protein 258)-rs102275, p = 2.43 × 10-12; FEN1 (flap structure-specific endonuclease 1)-rs174538, p = 1.96 × 10-11). Similar genome-wide statistically significant results were found for docosahexaenoic fatty acid (DHA). However, no such associations were detected for omega-6 PUFAs or linoleic acid (LA). For total PUFA, we observed a consistent gene*sex interaction with the DNTTIP2 (deoxynucleotidyl transferase terminal interacting protein 2)-rs3747965 p = 1.36 × 10-8. For adherence to MedDiet, we obtained a relevant interaction with the ME1 (malic enzyme 1) gene (a gene strongly regulated by fat) in determining serum omega-3. The top-ranked SNP for this interaction was ME1-rs3798890 (p = 2.15 × 10-7). In the regional-wide association study, specifically focused on the FADS1/FASD2/FADS3 and ELOVL (fatty acid elongase) 2/ELOVL 5 regions, we detected several statistically significant associations at p < 0.05. In conclusion, our results confirm a robust role of the FADS cluster on serum PUFA in this population, but the associations vary depending on the PUFA. Moreover, the detection of some sex and diet interactions underlines the need for these associations/interactions to be studied in all specific populations so as to better understand the complex metabolism of PUFA.
Collapse
Affiliation(s)
- Oscar Coltell
- Department of Computer Languages and Systems, Universitat Jaume I, 12071 Castellón, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
| | - Jose V. Sorlí
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Eva M. Asensio
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Rocío Barragán
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - José I. González
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Ignacio M. Giménez-Alba
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Vicente Zanón-Moreno
- Area of Health Sciences, Valencian International University, 46002 Valencia, Spain;
- Red Temática de Investigación Cooperativa en Patología Ocular (OFTARED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Ophthalmology Research Unit “Santiago Grisolia”, Dr. Peset University Hospital, 46017 Valencia, Spain
| | - Ramon Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Internal Medicine, Hospital Clinic, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | | | - Eva C. Pascual
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
- Assisted Reproduction Unit of the University Hospital of Valencia, 46010 Valencia, Spain
| | - Carolina Ortega-Azorín
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Jose M. Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111 USA;
- Department of Cardiovascular Epidemiology and Population Genetics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- IMDEA Alimentación, 28049 Madrid, Spain
| | - Dolores Corella
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| |
Collapse
|
18
|
Lauritzen L, Amundsen ID, Damsgaard CT, Lind MV, Schnurr TM, Hansen T, Michaelsen KF, Vogel U. FADS and PPARG2 Single Nucleotide Polymorphisms are Associated with Plasma Lipids in 9-Mo-Old Infants. J Nutr 2019; 149:708-715. [PMID: 31050749 DOI: 10.1093/jn/nxy323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/18/2018] [Accepted: 12/21/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Dietary intake of polyunsaturated fatty acids (PUFAs), e.g., linoleic acid and n-3 (ω-3) long-chain PUFAs, has been shown in adults to affect plasma cholesterol and triglycerides (TGs), respectively. Little is known about the effects of PUFAs on plasma lipids in early life. OBJECTIVE The aim of this study was to explore the associations between plasma concentrations of total, LDL, and HDL cholesterol and TGs in infants and 2 single nucleotide polymorphisms (SNPs) in the fatty acid desaturase genes (FADS) oppositely associated with docosahexaenoic acid (rs1535 and rs174448) and potential effect modification by a functional peroxisome proliferator-activated receptor-γ2 gene variant (PPARG2 Pro12Ala). METHODS In 9-mo-old infants (n = 561) from 3 Danish cohorts, we analyzed associations between plasma lipids, erythrocyte PUFAs, and FADS SNPs, and interactions with PPARG2 Pro12Ala genotype, by multiple linear regression. We also examined potential effect modification by breastfeeding, as 46% of the infants were still being breastfed. RESULTS Minor allele carriage of rs174448 was associated with lower total cholesterol (difference: -0.22 mmol/L; 95% CI: -0.37, -0.06 mmol/L; P = 0.006) and LDL cholesterol (difference: -0.15 mmol/L; 95% CI: -0.29, -0.01 mmol/L; P = 0.035), but no associations were observed with TGs or for rs1535. Minor allele carriage of both FADS SNPs was associated with 1 SD lower HDL cholesterol, but only in currently breastfed infants (rs174448 × breastfeeding, P = 0.080; rs1535 × breastfeeding, P = 0.030) and PPARG2 minor allele carriers (rs174448 × PPARG2, P = 0.001; rs1535 × PPARG2, P = 0.004). Erythrocyte arachidonic acid and eicosapentaenoic acid were inversely associated with LDL cholesterol [estimated effect (β): -0.3 mmol/L; 95% CI: -0.06, -0.00 mmol/L per percentage of fatty acids (FA%); P = 0.035] and TGs (β: -0.23 mmol/L; 95% CI: -0.41, -0.05 mmol/L per FA%; P = 0.015), respectively. CONCLUSIONS The observed associations with FADS variants indicate that PUFAs are involved in plasma lipid regulation in 9-mo-old infants. Observed FADS SNP differences and interactions with breastfeeding and PPARG2 warrant additional studies to explore the effects of individual FADS SNPs on PUFA status and potential genetic modification of dietary PUFA effects.
Collapse
Affiliation(s)
- Lotte Lauritzen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Ingvild D Amundsen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Camilla T Damsgaard
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Mads V Lind
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Theresia M Schnurr
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Kim F Michaelsen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
| |
Collapse
|
19
|
Taylor K, Davey Smith G, Relton CL, Gaunt TR, Richardson TG. Prioritizing putative influential genes in cardiovascular disease susceptibility by applying tissue-specific Mendelian randomization. Genome Med 2019; 11:6. [PMID: 30704512 PMCID: PMC6354354 DOI: 10.1186/s13073-019-0613-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/08/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The extent to which changes in gene expression can influence cardiovascular disease risk across different tissue types has not yet been systematically explored. We have developed an analysis pipeline that integrates tissue-specific gene expression, Mendelian randomization and multiple-trait colocalization to develop functional mechanistic insight into the causal pathway from a genetic variant to a complex trait. METHODS We undertook an expression quantitative trait loci-wide association study to uncover genetic variants associated with both nearby gene expression and cardiovascular traits. Fine-mapping was performed to prioritize possible causal variants for detected associations. Two-sample Mendelian randomization (MR) was then applied using findings from genome-wide association studies (GWAS) to investigate whether changes in gene expression within certain tissue types may influence cardiovascular trait variation. We subsequently used Bayesian multiple-trait colocalization to further interrogate the findings and also gain insight into whether DNA methylation, as well as gene expression, may play a role in disease susceptibility. Finally, we applied our analysis pipeline genome-wide using summary statistics from large-scale GWAS. RESULTS Eight genetic loci were associated with changes in gene expression and measures of cardiovascular function. Our MR analysis provided evidence of tissue-specific effects at multiple loci, of which the effects at the ADCY3 and FADS1 loci for body mass index and cholesterol, respectively, were particularly insightful. Multiple-trait colocalization uncovered evidence which suggested that changes in DNA methylation at the promoter region upstream of FADS1/TMEM258 may also affect cardiovascular trait variation along with gene expression. Furthermore, colocalization analyses uncovered evidence of tissue specificity between gene expression in liver tissue and cholesterol levels. Applying our pipeline genome-wide using summary statistics from GWAS uncovered 233 association signals at loci which represent promising candidates for further evaluation. CONCLUSIONS Disease susceptibility can be influenced by differential changes in tissue-specific gene expression and DNA methylation. The approach undertaken in our study can be used to elucidate mechanisms in disease, as well as helping prioritize putative causal genes at associated loci where multiple nearby genes may be co-regulated. Future studies which continue to uncover quantitative trait loci for molecular traits across various tissue and cell types will further improve our capability to understand and prevent disease.
Collapse
Affiliation(s)
- Kurt Taylor
- MRC Integrative Epidemiology Unit, Bristol Medical School (Population Health Sciences), University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Bristol Medical School (Population Health Sciences), University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- National Institute for Health Research Biomedical Research Centre, Bristol, UK
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, Bristol Medical School (Population Health Sciences), University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- National Institute for Health Research Biomedical Research Centre, Bristol, UK
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, Bristol Medical School (Population Health Sciences), University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- National Institute for Health Research Biomedical Research Centre, Bristol, UK
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit, Bristol Medical School (Population Health Sciences), University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
| |
Collapse
|
20
|
Lankinen M, Uusitupa M, Schwab U. Genes and Dietary Fatty Acids in Regulation of Fatty Acid Composition of Plasma and Erythrocyte Membranes. Nutrients 2018; 10:nu10111785. [PMID: 30453550 PMCID: PMC6265745 DOI: 10.3390/nu10111785] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/01/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
The fatty acid compositions of plasma lipids and cell membranes of certain tissues are modified by dietary fatty acid composition. Furthermore, many other factors (age, sex, ethnicity, health status, genes, and gene × diet interactions) affect the fatty acid composition of cell membranes or plasma lipid compartments. Therefore, it is of great importance to understand the complexity of mechanisms that may modify fatty acid compositions of plasma or tissues. We carried out an extensive literature survey of gene × diet interaction in the regulation of fatty acid compositions. Most of the related studies have been observational studies, but there are also a few intervention trials that tend to confirm that true interactions exist. Most of the studies deal with the desaturase enzyme cluster (FADS1, FADS2) in chromosome 11 and elongase enzymes. We expect that new genetic variants are being found that are linked with the genetic regulation of plasma or tissue fatty acid composition. This information is of great help to understanding the contribution of dietary fatty acids and their endogenic metabolism to the development of some chronic diseases.
Collapse
Affiliation(s)
- Maria Lankinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Matti Uusitupa
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Ursula Schwab
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland.
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70210 Kuopio, Finland.
| |
Collapse
|
21
|
Hannon BA, Khan NA, Teran-Garcia M. Nutrigenetic Contributions to Dyslipidemia: A Focus on Physiologically Relevant Pathways of Lipid and Lipoprotein Metabolism. Nutrients 2018; 10:E1404. [PMID: 30279335 PMCID: PMC6213032 DOI: 10.3390/nu10101404] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/19/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) remains the number one cause of death worldwide, and dyslipidemia is a major predictor of CVD mortality. Elevated lipid concentrations are the result of multiple genetic and environmental factors. Over 150 genetic loci have been associated with blood lipid levels. However, not all variants are present in pathways relevant to the pathophysiology of dyslipidemia. The study of these physiologically relevant variants can provide mechanistic understanding of dyslipidemia and identify potential novel therapeutic targets. Additionally, dietary fatty acids have been evidenced to exert both positive and negative effects on lipid profiles. The metabolism of both dietary and endogenously synthesized lipids can be affected by individual genetic variation to produce elevated lipid concentrations. This review will explore the genetic, dietary, and nutrigenetic contributions to dyslipidemia.
Collapse
Affiliation(s)
- Bridget A Hannon
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
| | - Naiman A Khan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
| | - Margarita Teran-Garcia
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL 61801, USA.
- Department of Human Development and Family Studies, Cooperative Extension, University of Illinois at Urbana-Champaign, Carle Illinois College of Medicine, Urbana-Champaign, IL 61801, USA.
| |
Collapse
|
22
|
Vazquez-Vidal I, Voruganti VS, Hannon BA, Andrade FCD, Aradillas-García C, Nakamura MT, Terán-García M. Serum Lipid Concentrations and FADS Genetic Variants in Young Mexican College Students: The UP-AMIGOS Cohort Study. Lifestyle Genom 2018; 11:40-48. [PMID: 29847832 DOI: 10.1159/000488085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/01/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Recent genome-wide association studies in the Mexican population have identified several genetic loci associated with blood lipid levels in adults. However, studies focusing on the fatty acid desaturase (FADS) gene cluster have been understudied in this population, even though it seems associated with lipid profiles in other ethnicities. The aim of this study was to test associations between single nucleotide polymorphisms (SNPs) in the FADS cluster (rs174546, rs1535, rs174548, rs174550, rs174450, and rs174618) and serum lipid profiles in young Mexicans. METHODS Anthropometrics, serum lipid profiles, and FADS SNPs were measured in 998 subjects in the UP-AMIGOS cohort study. Genotype-phenotype (total cholesterol [TC], triglyceride [TG], high-density lipoprotein cholesterol [HDL-C], low-density lipoprotein cholesterol [LDL-C], and very-low-density lipoprotein [VLDL]) associations were assessed using PLINK adjusted for sex, age, and body mass index (BMI). RESULTS Among 6 FADS SNPs, we found that carriers of the C-allele of the FADS1-rs174546 showed a significant association with lower TG concentrations (β = -12.6 mg/dL, p = 0.009) and lower VLDL concentrations (β = -2.52 mg/dL, p = 0.005). We found that rs174546, rs1535, and rs174550 were in high linkage disequilibrium (r2 > 0.80). There were no significant associations between rs174550, rs174548, and rs174618 and lipid profiles. CONCLUSION A genetic variant in the FADS1 (rs174546) gene is a major contributor of plasma TG and VLDL concentrations in healthy young Mexicans.
Collapse
Affiliation(s)
- Itzel Vazquez-Vidal
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Nutrition and UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA
| | - V Saroja Voruganti
- Department of Nutrition and UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA
| | - Bridget A Hannon
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | | | - Celia Aradillas-García
- Coordination for the Innovation and Application of Science and Technology, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | - Manabu T Nakamura
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Margarita Terán-García
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Human Development and Family Studies, Cooperative Extension, Family Resiliency Center, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
23
|
Iglesia I, Huybrechts I, González-Gross M, Mouratidou T, Santabárbara J, Chajès V, González-Gil EM, Park JY, Bel-Serrat S, Cuenca-García M, Castillo M, Kersting M, Widhalm K, De Henauw S, Sjöström M, Gottrand F, Molnár D, Manios Y, Kafatos A, Ferrari M, Stehle P, Marcos A, Sánchez-Muniz FJ, Moreno LA. Folate and vitamin B12 concentrations are associated with plasma DHA and EPA fatty acids in European adolescents: the Healthy Lifestyle in Europe by Nutrition in Adolescence (HELENA) study. Br J Nutr 2017; 117:124-133. [PMID: 28098048 DOI: 10.1017/s0007114516004414] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This study aimed to examine the association between vitamin B6, folate and vitamin B12 biomarkers and plasma fatty acids in European adolescents. A subsample from the Healthy Lifestyle in Europe by Nutrition in Adolescence study with valid data on B-vitamins and fatty acid blood parameters, and all the other covariates used in the analyses such as BMI, Diet Quality Index, education of the mother and physical activity assessed by a questionnaire, was selected resulting in 674 cases (43 % males). B-vitamin biomarkers were measured by chromatography and immunoassay and fatty acids by enzymatic analyses. Linear mixed models elucidated the association between B-vitamins and fatty acid blood parameters (changes in fatty acid profiles according to change in 10 units of vitamin B biomarkers). DHA, EPA) and n-3 fatty acids showed positive associations with B-vitamin biomarkers, mainly with those corresponding to folate and vitamin B12. Contrarily, negative associations were found with n-6:n-3 ratio, trans-fatty acids and oleic:stearic ratio. With total homocysteine (tHcy), all the associations found with these parameters were opposite (for instance, an increase of 10 nmol/l in red blood cell folate or holotranscobalamin in females produces an increase of 15·85 µmol/l of EPA (P value <0·01), whereas an increase of 10 nmol/l of tHcy in males produces a decrease of 2·06 µmol/l of DHA (P value <0·05). Positive associations between B-vitamins and specific fatty acids might suggest underlying mechanisms between B-vitamins and CVD and it is worth the attention of public health policies.
Collapse
Affiliation(s)
- I Iglesia
- 1Growth Exercise, Nutrition and Development (GENUD) Research Group,Universidad de Zaragoza, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria de Aragón (IIS), 50009 Zaragoza,Spain
| | - I Huybrechts
- 3Department of Public Health, Ghent University,University Hospital, De Pintelaan 185, entrance 42 (building K3),4th floor, B-9000 Ghent,Belgium
| | - M González-Gross
- 5ImFINE Research Group,Department of Health and Human Performance,Universidad Politécnica de Madrid,C/ Martín Fierro, 7, 28040 Madrid,Spain
| | - T Mouratidou
- 1Growth Exercise, Nutrition and Development (GENUD) Research Group,Universidad de Zaragoza, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria de Aragón (IIS), 50009 Zaragoza,Spain
| | - J Santabárbara
- 7Department of Preventive Medicine and Public Health,Universidad de Zaragoza,50009 Zaragoza,Spain
| | - V Chajès
- 4International Agency for Research on Cancer (IARC),150 Cours Albert Thomas,69372 Lyon Cedex 08,France
| | - E M González-Gil
- 1Growth Exercise, Nutrition and Development (GENUD) Research Group,Universidad de Zaragoza, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria de Aragón (IIS), 50009 Zaragoza,Spain
| | - J Y Park
- 4International Agency for Research on Cancer (IARC),150 Cours Albert Thomas,69372 Lyon Cedex 08,France
| | - S Bel-Serrat
- 1Growth Exercise, Nutrition and Development (GENUD) Research Group,Universidad de Zaragoza, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria de Aragón (IIS), 50009 Zaragoza,Spain
| | - M Cuenca-García
- 8Department of Physiology,School of Medicine,University of Granada,Avenida de Madrid 11,18012 Granada,Spain
| | - M Castillo
- 8Department of Physiology,School of Medicine,University of Granada,Avenida de Madrid 11,18012 Granada,Spain
| | - M Kersting
- 10Research Institute of Child Nutrition Dortmund,Pediatric University Clinic,Ruhr-University Bochum,Heinstück 11, D-44225 Dortmund,Germany
| | - K Widhalm
- 11Department of Pediatrics,Division of Clinical Nutrition and Prevention,Medical University of Vienna,1090 Vienna,Austria
| | - S De Henauw
- 3Department of Public Health, Ghent University,University Hospital, De Pintelaan 185, entrance 42 (building K3),4th floor, B-9000 Ghent,Belgium
| | - M Sjöström
- 12Department of Public Health Sciences,Division of Social Medicine,Karolinska Institutet,Norrbacka,level 3,17176 Stockholm,Sweden
| | - F Gottrand
- 15Inserm U995, Faculté de Médecine,Université Lille 2,F-59045 Lille Cedex,France
| | - D Molnár
- 16Department of Paediatrics,University of Pécs,Szigeti str 12, H-7624 Pécs,Hungary
| | - Y Manios
- 17Department of Nutrition and Dietetics,Harokopio University,E. Venizelou 70, 17671 Kallithea, reece, Kallithea-Athens,Greece
| | - A Kafatos
- 18School of Medicine,University of Crete,GR-71033 Crete,Greece
| | - M Ferrari
- 19CREA - Council for Agricultural Research and Economics - Research Center for Food and Nutrition,Via Ardeatina 546 - 00178 Roma,Italy
| | - P Stehle
- 20Department of Nutrition and Food Science,University of Bonn,D-53115 Bonn,Germany
| | - A Marcos
- 21Immunonutrition Research Group,Department of Metabolism and Nutrition,Institute of Food Science,Technology and Nutrition (ICTAN),Spanish National Research Council (CSIC),E-28040 Madrid,Spain
| | - F J Sánchez-Muniz
- 22Departamento de Nutrición, Facultad de Farmacia,Universidad Complutense de Madrid,28040 Madrid,Spain
| | - L A Moreno
- 1Growth Exercise, Nutrition and Development (GENUD) Research Group,Universidad de Zaragoza, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria de Aragón (IIS), 50009 Zaragoza,Spain
| |
Collapse
|
24
|
Rousseaux J, Duhamel A, Dumont J, Dallongeville J, Molnar D, Widhalm K, Manios Y, Sjöström M, Kafatos A, Breidenassel C, Gonzales-Gross M, Cuenca-Garcia M, Censi L, Ascensión M, De Henauw S, Moreno LA, Meirhaeghe A, Gottrand F. The n-3 long-chain PUFAs modulate the impact of the GCKR Pro446Leu polymorphism on triglycerides in adolescents. J Lipid Res 2015; 56:1774-80. [PMID: 26136510 DOI: 10.1194/jlr.m057570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Indexed: 01/19/2023] Open
Abstract
Dietary n-3 long-chain PUFAs (LC-PUFAs) are associated with improvement in the parameters of the metabolic syndrome (MetS). Glucokinase regulatory protein (GCKR) is a key protein regulating intracellular glucose disposal. Our aim was to investigate: i) the relationship between the GCKR rs1260326 (Pro446Leu) polymorphism and parameters of the MetS; and ii) a potential influence of n-3 and n-6 LC-PUFA levels on this relationship in the HELENA study (1,155 European adolescents). Linear regression analyses were performed to study the association between rs1260326 and the outcomes of interest. Interactions between rs1260326 and LC-PUFA levels on outcomes were explored. The T allele of rs1260326 was associated with higher serum TG concentrations compared with the C allele. In contrast to n-6 LC-PUFA levels, a significant interaction (P = 0.01) between rs1260326 and total n-3 LC-PUFA levels on serum TG concentrations was observed. After stratification on the n-3 LC-PUFA median values, the association between rs1260326 and TG concentration was significant only in the group with high n-3 LC-PUFA levels. In conclusion, this is the first evidence that n-3 LC-PUFAs may modulate the impact of the GCKR rs1260326 polymorphism on TG concentrations in adolescents. Several molecular mechanisms, in link with glucose uptake, could explain these findings.
Collapse
Affiliation(s)
- Julien Rousseaux
- Inserm U995, LIRIC, CHU Lille, University Lille, Lille, France Unité de Biostatistiques, CERIM, EA2694, CHU Lille, University Lille, Lille, France
| | - Alain Duhamel
- Unité de Biostatistiques, CERIM, EA2694, CHU Lille, University Lille, Lille, France
| | - Julie Dumont
- Inserm UMR1167, Institut Pasteur de Lille, University Lille, Lille, France
| | - Jean Dallongeville
- Inserm UMR1167, Institut Pasteur de Lille, University Lille, Lille, France
| | - Denes Molnar
- Department of Pediatrics, University of Pécs, Pécs, Hungary
| | - Kurt Widhalm
- Academic Institute for Clinical Nutrition, Vienna, Austria and Private Medical University Salzburg, Salzburg, Austria
| | - Yannis Manios
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Michael Sjöström
- Unit for Preventive Nutrition, Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Anthony Kafatos
- Preventive Medicine and Nutrition Unit, School of Medicine, University of Crete, Heraklion, Greece
| | - Christina Breidenassel
- Institut für Ernährungs-und Lebensmittelwissenschaften, Humanernährung, Rheinische Friedrich-Wilhelms, Universität Bonn, Bonn, Germany
| | - Marcela Gonzales-Gross
- Facultad de la Actividad Física y del Deporte-INEF, Universidad Politécnica de Madrid, Madrid, Spain
| | | | - Laura Censi
- National Research Institute on Food and Nutrition, Rome, Italy
| | - Marcos Ascensión
- Immunonutrition Research Group, Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain
| | - Stefaan De Henauw
- Department of Public Health, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Luis A Moreno
- GENUD (Growth, Exercise, Nutrition and Development) Research Group, Escuela Universitaria de Ciencias de la Salud, Universidad de Zaragoza, Zaragoza, Spain
| | - Aline Meirhaeghe
- Inserm UMR1167, Institut Pasteur de Lille, University Lille, Lille, France
| | | |
Collapse
|
25
|
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. [PMID: 25849351 PMCID: PMC4388373 DOI: 10.1371/journal.pone.0121255] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/29/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Previous studies suggested that dietary fatty acids could affect blood lipids by interacting with genetic variations in fatty acid desaturase 1 (FADS1). However, little is known about their direct effects on coronary artery disease (CAD). The aim of this study was to evaluate whether dietary n-3 long-chain polyunsaturated fatty acids (LCPUFAs)-eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) could modulate the effect of FADS1 rs174547 polymorphism on CAD. METHODS FADS1 single-nucleotide polymorphisms rs174547 genotypes were measured in 440 CAD patients and 838 healthy controls. Dietary EPA and DHA intakes were assessed with a validated quantitative frequency food questionnaire. The association between FADS1 rs174547 and CAD was estimated using logistic regression under both dominant and additive genetic models. The interactions between rs174547 polymorphism and LCPUFAs were analyzed by using multiple logistic regression and the "genotype × n-3 LCPUFAs" interaction term was included into the model. RESULTS We found that the minor T allele of FADS1 rs174547 increased CAD risk (OR = 1.36, 95%CIs 1.03-1.80), and observed significant interaction between rs174547 and dietary EPA intakes on CAD (P-interaction = 0.028). The T-allele was only associated with higher CAD risk among individuals with lower dietary EPA intakes, but not in those with higher EPA intakes. Similarly, significant interaction was also observed between rs174547 and dietary DHA intakes on CAD (P-interaction = 0.020). CONCLUSIONS Dietary n-3 LCPUFA intakes could modulate the association between FADS1 rs174547 polymorphism and CAD. High dietary n-3 LCPUFA intakes could negate the unfavorable effect of genetic variation in FADS1 on CAD in middle-aged and elderly Chinese population.
Collapse
Affiliation(s)
- Fengqiong Liu
- Department of Epidemiology and health statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhongxia Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaofei Lv
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jing Ma
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
- * E-mail:
| |
Collapse
|
26
|
Bonafini S, Antoniazzi F, Maffeis C, Minuz P, Fava C. Beneficial effects of ω-3 PUFA in children on cardiovascular risk factors during childhood and adolescence. Prostaglandins Other Lipid Mediat 2015; 120:72-9. [PMID: 25834924 DOI: 10.1016/j.prostaglandins.2015.03.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/20/2015] [Indexed: 12/19/2022]
Abstract
Omega-3 polyunsatured fatty acids (ω-3 PUFA) are essential nutrients mainly derived from fish and seafood but present also in vegetables such as nuts and seed-oils. Some epidemiological and clinical studies indicate a protection of ω-3 FA against cardiovascular disease and a favourable effect on cardiovascular risk factors control in adults. The evidences of their effects in children and adolescents are scanty but a possible beneficial role, especially for insulin sensitivity and blood pressure control, has been proposed. In this review we want to focus especially on the evidences, which could justify the assumption of ω-3 in children and adolescents, and to underline the aspects which need further investigation. Mechanisms through which ω-3 FA act are manifolds and still a matter of investigation: beside their interaction with ion channel and their influence on plasma membrane fluidity, probably the main effect is acting as competitor for cytochrome P-450 (CYP) with respect to ω-6 FA. Thus, they can modulate the biosynthesis of eicosanoids and other lipid mediators, which likely exert a protective action. Another suggestive hypothesis is that their beneficial effect is not dependent only on the intake of ω-3 FA, but also on the complex interaction between different nutrients including ω-3 and other FAs with polymorphisms in genes involved in ω-3 FA modulation. This complex interaction has seldom been explored in children and adolescents. Further studies are needed to investigate all these points in order to find a better collocation of ω-3 FA on the available armamentarium for preventive, possibly individualized, medicine.
Collapse
Affiliation(s)
- Sara Bonafini
- Department of Medicine, University of Verona, Section of Internal Medicine C, Italy.
| | - Franco Antoniazzi
- University of Verona, Department of Life and Reproduction Science, Italy
| | - Claudio Maffeis
- University of Verona, Department of Life and Reproduction Science, Italy
| | - Pietro Minuz
- Department of Medicine, University of Verona, Section of Internal Medicine C, Italy
| | - Cristiano Fava
- Department of Medicine, University of Verona, Section of Internal Medicine C, Italy
| |
Collapse
|
27
|
Dietary arachidonic acid in perinatal nutrition: a commentary. Pediatr Res 2015; 77:263-9. [PMID: 25314584 DOI: 10.1038/pr.2014.166] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 10/08/2014] [Indexed: 01/31/2023]
Abstract
Arachidonic acid (AA) is supplied together with docosahexaenoic acid (DHA) in infant formulas, but we have limited knowledge about the effects of supplementation with either of these long-chain polyunsaturated fatty acids (LCPUFA) on growth and developmental outcomes. AA is present in similar levels in breast milk throughout the world, whereas the level of DHA is highly diet dependent. Autopsy studies show similar diet-dependent variation in brain DHA, whereas AA is little affected by intake. Early intake of DHA has been shown to affect visual development, but the effect of LCPUFA on neurodevelopment remains to be established. Few studies have found any functional difference between infants supplemented with DHA alone compared to DHA+AA, but some studies show neurodevelopmental advantages in breast-fed infants of mothers supplemented with n-3 LCPUFA alone. It also remains to be established whether the AA/DHA balance could affect allergic and inflammatory outcomes later in life. Disentangling effects of genetic variability and dietary intake on AA and DHA-status and on functional outcomes may be an important step in the process of determining whether AA-intake is of any physiological or clinical importance. However, based on the current evidence we hypothesize that dietary AA plays a minor role on growth and development relative to the impact of dietary DHA.
Collapse
|
28
|
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. J Nutr 2014; 144:1356-63. [PMID: 25008580 PMCID: PMC4130826 DOI: 10.3945/jn.114.192708] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The unclear link between intake of polyunsaturated fatty acids (PUFAs) and risk of cardiovascular disease (CVD) could depend on genetic differences between individuals. Minor alleles of single-nucleotide polymorphisms (SNPs) in the ∆5 fatty acid desaturase (FADS) 1 gene were associated with lower blood concentrations of long-chain ω-3 (n-3) and ω-6 (n-6) PUFAs, indicating an associated loss of function effect. We examined whether the SNP rs174546 in FADS1 modifies the association between PUFA intakes and CVD risk. We included 24,032 participants (62% women, aged 44-74 y) from the Malmö Diet and Cancer cohort without prevalent CVD and diabetes. During a mean follow-up of 14 y, 2648 CVD cases were identified. Diet was assessed by a modified diet history method. A borderline interaction was observed between the α-linolenic acid (ALA) (18:3n-3)-to-linoleic acid (LA) (18:2n-6) intake ratio and FADS1 genotype on CVD incidence (P = 0.06). The ALA-to-LA intake ratio was inversely associated with CVD risk only among participants homozygous for the minor T-allele (HR for quintile 5 vs. quintile 1 = 0.72; 95% CI: 0.50, 1.04; P-trend = 0.049). When excluding participants reporting unstable food habits in the past (35%), the interaction between the ALA-to-LA intake ratio and FADS1 genotype on CVD incidence was strengthened and statistically significant (P = 0.04). Additionally, we observed a significant interaction between ALA and FADS1 genotype on ischemic stroke incidence (P = 0.03). ALA was inversely associated with ischemic stroke only among TT genotype carriers (HR for quintile 5 vs. quintile 1 = 0.50; 95% CI: 0.27, 0.94; P-trend = 0.02). In this large cohort, we found some weak, but not convincing, evidence of effect modification by genetic variation in FADS1 on the associations between PUFA intakes and CVD risk. For the 11% of the population homozygous for the minor T-allele of rs174546 that associates with lower ∆5 FADS activity, high ALA intake and ALA-to-LA intake ratio may be preferable in the prevention of CVD and ischemic stroke.
Collapse
Affiliation(s)
| | - Ulrika Ericson
- Diabetes and Cardiovascular Disease–Genetic Epidemiology
| | | | - Bo Hedblad
- Cardiovascular Epidemiology, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | | | | |
Collapse
|
29
|
Sarkadi Nagy E, Martos É. Present and future of genotype-based personalized nutrition. Orv Hetil 2014; 155:771-7. [DOI: 10.1556/oh.2014.29896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
After the completion of the Human Genome Project, the era of providing personalized dietary advice based on an individual’s genetic profile seemed near. Since then more than a decade has passed and the pace of development has been slower than expected. Genotyping single nucleotide polymorphisms which may determine susceptibility to multifactorial diseases is cheaper and more accessible than it was ten years ago. However, few of them are supported by such solid scientific evidence which would justify their use for personalized dietary advice. The future of genotype-based personalized nutrition depends on whether a sufficient amount of gene–diet-disease interactions are identified and scientifically confirmed. Orv. Hetil., 2014, 155(20), 771–777.
Collapse
Affiliation(s)
- Eszter Sarkadi Nagy
- Országos Élelmezés- és Táplálkozástudományi Intézet Budapest Albert Flórián út 3/A 1097
| | - Éva Martos
- Országos Élelmezés- és Táplálkozástudományi Intézet Budapest Albert Flórián út 3/A 1097
| |
Collapse
|
30
|
Norris JM, Kroehl M, Fingerlin TE, Frederiksen BN, Seifert J, Wong R, Clare-Salzler M, Rewers M. Erythrocyte membrane docosapentaenoic acid levels are associated with islet autoimmunity: the Diabetes Autoimmunity Study in the Young. Diabetologia 2014; 57:295-304. [PMID: 24240437 PMCID: PMC3947295 DOI: 10.1007/s00125-013-3106-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/22/2013] [Indexed: 12/12/2022]
Abstract
AIMS/HYPOTHESES We previously reported that lower n-3 fatty acid intake and levels in erythrocyte membranes were associated with increased risk of islet autoimmunity (IA) but not progression to type 1 diabetes in children at increased risk for diabetes. We hypothesise that specific n-3 fatty acids and genetic markers contribute synergistically to this increased risk of IA in the Diabetes Autoimmunity Study in the Young (DAISY). METHODS DAISY is following 2,547 children at increased risk for type 1 diabetes for the development of IA, defined as being positive for glutamic acid decarboxylase (GAD)65, IA-2 or insulin autoantibodies on two consecutive visits. Using a case-cohort design, erythrocyte membrane fatty acids and dietary intake were measured prospectively in 58 IA-positive children and 299 IA-negative children. RESULTS Lower membrane levels of the n-3 fatty acid, docosapentaenoic acid (DPA), were predictive of IA (HR 0.23; 95% CI 0.09, 0.55), while α-linolenic acid (ALA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) were not, adjusting for HLA and diabetes family history. We examined whether the effect of dietary intake of the n-3 fatty acid ALA on IA risk was modified by fatty acid elongation and desaturation genes. Adjusting for HLA, diabetes family history, ethnicity, energy intake and questionnaire type, ALA intake was significantly more protective for IA in the presence of an increasing number of minor alleles at FADS1 rs174556 (pinteraction = 0.017), at FADS2 rs174570 (pinteraction = 0.016) and at FADS2 rs174583 (pinteraction = 0.045). CONCLUSIONS/INTERPRETATION The putative protective effect of n-3 fatty acids on IA may result from a complex interaction between intake and genetically controlled fatty acid desaturation.
Collapse
Affiliation(s)
- Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, 13001 E. 17th Place, Campus Box B119, Aurora, CO, 80045, USA,
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kokaze A, Ishikawa M, Matsunaga N, Karita K, Yoshida M, Ohtsu T, Ochiai H, Shirasawa T, Nanri H, Hoshino H, Takashima Y. Difference in effects of cigarette smoking or alcohol consumption on serum non-high-density lipoprotein cholesterol levels is related to mitochondrial DNA 5178 C/A polymorphism in middle-aged Japanese men: a cross-sectional study. J Physiol Anthropol 2014; 33:1. [PMID: 24383671 PMCID: PMC3905964 DOI: 10.1186/1880-6805-33-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 12/12/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Mitochondrial DNA 5178 cytosine/adenosine (Mt5178 C/A) polymorphism is associated with longevity in the Japanese. The purpose of this study is to investigate whether Mt5178 C/A polymorphism modifies the effects of habitual smoking or habitual drinking on serum non-high-density lipoprotein (non-HDL) cholesterol levels in middle-aged Japanese men. METHODS A total of 394 male subjects (age 53.9 ± 7.9 years; mean ± SD) were selected from among individuals visiting the hospital for regular medical check-ups. After Mt5178 C/A genotyping, a cross-sectional study assessing the joint effects of Mt5178 C/A polymorphism and cigarette smoking or alcohol drinking on serum non-HDL cholesterol levels was conducted. High levels of serum non-HDL cholesterol were defined as serum non-HDL cholesterol levels ≥160 mg/dl or ≥190 mg/dl. RESULTS For men with Mt5178A, cigarette smoking may increase serum non-HDL cholesterol levels (P for trend < 0.001), as well as the risk of high levels of non-HDL cholesterol (serum non-HDL cholesterol levels ≥160 mg/dl, P for trend < 0.001; serum non-HDL cholesterol levels ≥190 mg/dl, P for trend = 0.004). On the other hand, for men with Mt5178C, after adjusting for age and body mass index, alcohol consumption may decrease serum non-HDL cholesterol levels (P for trend = 0.043) and the risk of high levels of non-HDL cholesterol (serum non-HDL cholesterol level ≥160 mg/dl, P for trend = 0.005). CONCLUSIONS These gene-environment interactions on serum non-HDL cholesterol levels may contribute to the establishment of individualized prevention of the risk of high levels of serum non-HDL cholesterol.
Collapse
Affiliation(s)
- Akatsuki Kokaze
- Department of Public Health, Showa University School of Medicine, 1-5-8 Hatanodai, 142-8555 Shinagawa-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Moltó-Puigmartí C, Jansen E, Heinrich J, Standl M, Mensink RP, Plat J, Penders J, Mommers M, Koppelman GH, Postma DS, Thijs C. Genetic variation in FADS genes and plasma cholesterol levels in 2-year-old infants: KOALA Birth Cohort Study. PLoS One 2013; 8:e61671. [PMID: 23667444 PMCID: PMC3648514 DOI: 10.1371/journal.pone.0061671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 03/12/2013] [Indexed: 12/13/2022] Open
Abstract
Objective Single nucleotide polymorphisms (SNPs) in genes involved in fatty acid metabolism (FADS1 FADS2 gene cluster) are associated with plasma lipid levels. We aimed to investigate whether these associations are already present early in life and compare the relative contribution of FADS SNPs vs traditional (non-genetic) factors as determinants of plasma lipid levels. Methods Information on infants’ plasma total cholesterol levels, genotypes of five FADS SNPs (rs174545, rs174546, rs174556, rs174561, and rs3834458), anthropometric data, maternal characteristics, and breastfeeding history was available for 521 2-year-old children from the KOALA Birth Cohort Study. For 295 of these 521 children, plasma HDLc and non-HDLc levels were also known. Multivariable linear regression analysis was used to study the associations of genetic and non-genetic determinants with cholesterol levels. Results All FADS SNPs were significantly associated with total cholesterol levels. Heterozygous and homozygous for the minor allele children had about 4% and 8% lower total cholesterol levels than major allele homozygotes. In addition, homozygous for the minor allele children had about 7% lower HDLc levels. This difference reached significance for the SNPs rs174546 and rs3834458. The associations went in the same direction for non-HDLc, but statistical significance was not reached. The percentage of total variance of total cholesterol levels explained by FADS SNPs was relatively low (lower than 3%) but of the same order as that explained by gender and the non-genetic determinants together. Conclusions FADS SNPs are associated with plasma total cholesterol and HDLc levels in preschool children. This brings a new piece of evidence to explain how blood lipid levels may track from childhood to adulthood. Moreover, the finding that these SNPs explain a similar amount of variance in total cholesterol levels as the non-genetic determinants studied reveals the potential importance of investigating the effects of genetic variations in early life.
Collapse
Affiliation(s)
- Carolina Moltó-Puigmartí
- Department of Epidemiology, CAPHRI School for Public Health and Primary Care, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Raffler J, Römisch-Margl W, Petersen AK, Pagel P, Blöchl F, Hengstenberg C, Illig T, Meisinger C, Stark K, Wichmann HE, Adamski J, Gieger C, Kastenmüller G, Suhre K. Identification and MS-assisted interpretation of genetically influenced NMR signals in human plasma. Genome Med 2013; 5:13. [PMID: 23414815 PMCID: PMC3706909 DOI: 10.1186/gm417] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 12/21/2012] [Accepted: 02/15/2013] [Indexed: 12/18/2022] Open
Abstract
Nuclear magnetic resonance spectroscopy (NMR) provides robust readouts of many metabolic parameters in one experiment. However, identification of clinically relevant markers in (1)H NMR spectra is a major challenge. Association of NMR-derived quantities with genetic variants can uncover biologically relevant metabolic traits. Using NMR data of plasma samples from 1,757 individuals from the KORA study together with 655,658 genetic variants, we show that ratios between NMR intensities at two chemical shift positions can provide informative and robust biomarkers. We report seven loci of genetic association with NMR-derived traits (APOA1, CETP, CPS1, GCKR, FADS1, LIPC, PYROXD2) and characterize these traits biochemically using mass spectrometry. These ratios may now be used in clinical studies.
Collapse
Affiliation(s)
- Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany ; Faculty of Biology, Ludwig-Maximilians-Universität, Großhaderner Straße 2, 82152 Planegg-Martinsried, Germany
| | - Werner Römisch-Margl
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Ann-Kristin Petersen
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Philipp Pagel
- numares GmbH, Josef-Engert-Str. 9, 93053 Regensburg, Germany
| | - Florian Blöchl
- numares GmbH, Josef-Engert-Str. 9, 93053 Regensburg, Germany
| | - Christian Hengstenberg
- Klinik und Poliklinik für Innere Medizin II, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Germany
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany ; Hannover Unified Biobank, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Klaus Stark
- Klinik und Poliklinik für Innere Medizin II, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Germany ; Department of Epidemiology and Preventive Medicine, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Germany
| | - H-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Karsten Suhre
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany ; Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. BOX 24144, Doha, Qatar
| |
Collapse
|
34
|
Zhu J, Sun Q, Zong G, Si Y, Liu C, Qi Q, Ye X, Sun L, Sheng H, Li H, Lin X. Interaction between a common variant in FADS1 and erythrocyte polyunsaturated fatty acids on lipid profile in Chinese Hans. J Lipid Res 2013; 54:1477-83. [PMID: 23396965 DOI: 10.1194/jlr.p027516] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Little is known about the associations of FADS1 genetic variants with circulating levels of PUFA and lipids in Asian populations who have a different dietary pattern and dyslipidemia prevalence compared with Western populations. In a population-based sample of 3,210 unrelated Han Chinese living in Beijing and Shanghai, we examined a FADS1 genetic variant, rs174550, in relation to blood PUFA and lipid levels. C-allele of rs174550 was significantly associated with levels of erythrocyte PUFAs in upstream and downstream pathways of delta-5 desaturase (D5D) (P ≤ 0.003). Moreover, rs174550 C-allele was associated with a lower HDL cholesterol level (P = 0.02) in total population and a higher triglyceride level (P = 0.0002) in Beijing residents. Interestingly, erythrocyte levels of 18:2n-6 and 18:3n-3 modified the effect of rs174550 on HDL cholesterol level: stronger associations between rs174550 C-allele and lower HDL cholesterol levels were exhibited when erythrocyte 18:2n-6 or 18:3n-3 level was low (P for interaction = 0.02 and 0.03, respectively). These data suggested that FADS1 genetic variant was associated with circulating PUFA and lipid levels and that its effect on HDL cholesterol might depend on PUFA status in the Han Chinese population.
Collapse
Affiliation(s)
- Jingwen Zhu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate University of Chinese Academy of Sciences, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The rapid technological developments following the Human Genome Project have made possible the availability of personalized genomes. As the focus now shifts from characterizing genomes to making personalized disease associations, in combination with the availability of other omics technologies, the next big push will be not only to obtain a personalized genome, but to quantitatively follow other omics. This will include transcriptomes, proteomes, metabolomes, antibodyomes, and new emerging technologies, enabling the profiling of thousands of molecular components in individuals. Furthermore, omics profiling performed longitudinally can probe the temporal patterns associated with both molecular changes and associated physiological health and disease states. Such data necessitates the development of computational methodology to not only handle and descriptively assess such data, but also construct quantitative biological models. Here we describe the availability of personal genomes and developing omics technologies that can be brought together for personalized implementations and how these novel integrated approaches may effectively provide a precise personalized medicine that focuses on not only characterization and treatment but ultimately the prevention of disease.
Collapse
|
36
|
Adamski J, Suhre K. Metabolomics platforms for genome wide association studies--linking the genome to the metabolome. Curr Opin Biotechnol 2012; 24:39-47. [PMID: 23102864 DOI: 10.1016/j.copbio.2012.10.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/21/2012] [Accepted: 10/03/2012] [Indexed: 01/13/2023]
Abstract
Genome-wide association studies (GWAS) reveal links between genetic variance and predisposition to disease. With the advent of modern 'omics-technologies', GWAS can now identify the genetic factors that influence intermediate traits on pathways to disease, such as blood concentrations of carbohydrates, lipids, amino acids, and secondary metabolites, hormones and signal molecules. At the example of recent GWAS with metabolic traits (mGWAS) we review the high-throughput screening approaches that are available to further advance the field.
Collapse
Affiliation(s)
- Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
| | | |
Collapse
|
37
|
Abstract
Many complex disorders are linked to metabolic phenotypes. Revealing genetic influences on metabolic phenotypes is key to a systems-wide understanding of their interactions with environmental and lifestyle factors in their aetiology, and we can now explore the genetics of large panels of metabolic traits by coupling genome-wide association studies and metabolomics. These genome-wide association studies are beginning to unravel the genetic contribution to human metabolic individuality and to demonstrate its relevance for biomedical and pharmaceutical research. Adopting the most appropriate study designs and analytical tools is paramount to further refining the genotype-phenotype map and eventually identifying the part played by genetic influences on metabolic phenotypes. We discuss such design considerations and applications in this Review.
Collapse
|
38
|
Cormier H, Rudkowska I, Paradis AM, Thifault E, Garneau V, Lemieux S, Couture P, Vohl MC. Association between polymorphisms in the fatty acid desaturase gene cluster and the plasma triacylglycerol response to an n-3 PUFA supplementation. Nutrients 2012; 4:1026-41. [PMID: 23016130 PMCID: PMC3448085 DOI: 10.3390/nu4081026] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 07/14/2012] [Accepted: 08/03/2012] [Indexed: 12/04/2022] Open
Abstract
Eicosapentaenoic and docosahexaenoic acids have been reported to have a variety of beneficial effects on cardiovascular disease risk factors. However, a large inter-individual variability in the plasma lipid response to an omega-3 (n-3) polyunsaturated fatty acid (PUFA) supplementation is observed in different studies. Genetic variations may influence plasma lipid responsiveness. The aim of the present study was to examine the effects of a supplementation with n-3 PUFA on the plasma lipid profile in relation to the presence of single-nucleotide polymorphisms (SNPs) in the fatty acid desaturase (FADS) gene cluster. A total of 208 subjects from Quebec City area were supplemented with 3 g/day of n-3 PUFA, during six weeks. In a statistical model including the effect of the genotype, the supplementation and the genotype by supplementation interaction, SNP rs174546 was significantly associated (p = 0.02) with plasma triglyceride (TG) levels, pre- and post-supplementation. The n-3 supplementation had an independent effect on plasma TG levels and no significant genotype by supplementation interaction effects were observed. In summary, our data support the notion that the FADS gene cluster is a major determinant of plasma TG levels. SNP rs174546 may be an important SNP associated with plasma TG levels and FADS1 gene expression independently of a nutritional intervention with n-3 PUFA.
Collapse
Affiliation(s)
- Hubert Cormier
- Institute of Nutraceuticals and Functional Foods (INAF), Laval University, Quebec G1V 0A6, Canada; (I.R.); (A.-M.P.); (E.T.); (V.G.); (S.L.); (P.C.)
- Authors to whom correspondence should be addressed; (H.C.); (M.-C.V.); Tel.: +1-418-656-2131 (ext. 4676); Fax: +1-418-656-5877
| | - Iwona Rudkowska
- Institute of Nutraceuticals and Functional Foods (INAF), Laval University, Quebec G1V 0A6, Canada; (I.R.); (A.-M.P.); (E.T.); (V.G.); (S.L.); (P.C.)
- Endocrinology and Genomics, Laval University Medical Center, Quebec G1V 0A6, Canada
| | - Ann-Marie Paradis
- Institute of Nutraceuticals and Functional Foods (INAF), Laval University, Quebec G1V 0A6, Canada; (I.R.); (A.-M.P.); (E.T.); (V.G.); (S.L.); (P.C.)
| | - Elisabeth Thifault
- Institute of Nutraceuticals and Functional Foods (INAF), Laval University, Quebec G1V 0A6, Canada; (I.R.); (A.-M.P.); (E.T.); (V.G.); (S.L.); (P.C.)
| | - Véronique Garneau
- Institute of Nutraceuticals and Functional Foods (INAF), Laval University, Quebec G1V 0A6, Canada; (I.R.); (A.-M.P.); (E.T.); (V.G.); (S.L.); (P.C.)
| | - Simone Lemieux
- Institute of Nutraceuticals and Functional Foods (INAF), Laval University, Quebec G1V 0A6, Canada; (I.R.); (A.-M.P.); (E.T.); (V.G.); (S.L.); (P.C.)
| | - Patrick Couture
- Institute of Nutraceuticals and Functional Foods (INAF), Laval University, Quebec G1V 0A6, Canada; (I.R.); (A.-M.P.); (E.T.); (V.G.); (S.L.); (P.C.)
| | - Marie-Claude Vohl
- Institute of Nutraceuticals and Functional Foods (INAF), Laval University, Quebec G1V 0A6, Canada; (I.R.); (A.-M.P.); (E.T.); (V.G.); (S.L.); (P.C.)
- Endocrinology and Genomics, Laval University Medical Center, Quebec G1V 0A6, Canada
- Authors to whom correspondence should be addressed; (H.C.); (M.-C.V.); Tel.: +1-418-656-2131 (ext. 4676); Fax: +1-418-656-5877
| |
Collapse
|
39
|
Walker CG, Jebb SA. Gene–Diet Interactions on Lipid Levels: Current Knowledge in the Era of Genome-Wide Association Studies. Curr Nutr Rep 2012. [DOI: 10.1007/s13668-012-0017-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|