1
|
von Schacky C. Assessing omega-3 fatty acids-critically weighing options and relevance. J Clin Lipidol 2025; 19:208-214. [PMID: 40050195 DOI: 10.1016/j.jacl.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 04/20/2025]
|
2
|
Liu X, Gong M, Wu N. Research progress on the relationship between free fatty acid profile and type 2 diabetes complicated by coronary heart disease. Front Endocrinol (Lausanne) 2024; 15:1503704. [PMID: 39713052 PMCID: PMC11658973 DOI: 10.3389/fendo.2024.1503704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/14/2024] [Indexed: 12/24/2024] Open
Abstract
Patients with type 2 diabetes mellitus (T2DM) have a 2 to 3 times higher risk of cardiovascular disease compared to non-diabetic individuals, and cardiovascular disease has consistently been a leading cause of death among diabetic patients. Therefore, preventing cardiovascular disease in diabetic patients remains a significant challenge. In addition to classic indicators such as cholesterol and lipoproteins, previous studies have demonstrated that plasma level of free fatty acid (FFA) is closely related to the occurrence of atherosclerosis, particularly in T2DM patients. In recent years, with further research and advancements in testing technologies, the FFA profile has garnered widespread attention. The FFA profile includes many different types of FFAs, and changes in the plasma FFA profile and concentrations in T2DM patients may lead to the development of insulin resistance, causing damage to vascular endothelial cells and promoting the occurrence and progression of atherosclerosis. Furthermore, some FFAs have shown potential in predicting cardiovascular complications in T2DM and are associated with the severity of these complications. Here, we aim to review the changes in the FFA profile in T2DM and discuss the relationship between the FFA profile and the occurrence of vascular complications in T2DM.
Collapse
Affiliation(s)
- Xiuyan Liu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ming Gong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Na Wu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Sharma S, Dong Q, Haid M, Adam J, Bizzotto R, Fernandez-Tajes JJ, Jones AG, Tura A, Artati A, Prehn C, Kastenmüller G, Koivula RW, Franks PW, Walker M, Forgie IM, Giordano G, Pavo I, Ruetten H, Dermitzakis M, McCarthy MI, Pedersen O, Schwenk JM, Tsirigos KD, De Masi F, Brunak S, Viñuela A, Mari A, McDonald TJ, Kokkola T, Adamski J, Pearson ER, Grallert H. Role of human plasma metabolites in prediabetes and type 2 diabetes from the IMI-DIRECT study. Diabetologia 2024; 67:2804-2818. [PMID: 39349772 PMCID: PMC11604760 DOI: 10.1007/s00125-024-06282-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/29/2024] [Indexed: 11/29/2024]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is a chronic condition that is caused by hyperglycaemia. Our aim was to characterise the metabolomics to find their association with the glycaemic spectrum and find a causal relationship between metabolites and type 2 diabetes. METHODS As part of the Innovative Medicines Initiative - Diabetes Research on Patient Stratification (IMI-DIRECT) consortium, 3000 plasma samples were measured with the Biocrates AbsoluteIDQ p150 Kit and Metabolon analytics. A total of 911 metabolites (132 targeted metabolomics, 779 untargeted metabolomics) passed the quality control. Multivariable linear and logistic regression analysis estimates were calculated from the concentration/peak areas of each metabolite as an explanatory variable and the glycaemic status as a dependent variable. This analysis was adjusted for age, sex, BMI, study centre in the basic model, and additionally for alcohol, smoking, BP, fasting HDL-cholesterol and fasting triacylglycerol in the full model. Statistical significance was Bonferroni corrected throughout. Beyond associations, we investigated the mediation effect and causal effects for which causal mediation test and two-sample Mendelian randomisation (2SMR) methods were used, respectively. RESULTS In the targeted metabolomics, we observed four (15), 34 (99) and 50 (108) metabolites (number of metabolites observed in untargeted metabolomics appear in parentheses) that were significantly different when comparing normal glucose regulation vs impaired glucose regulation/prediabetes, normal glucose regulation vs type 2 diabetes, and impaired glucose regulation vs type 2 diabetes, respectively. Significant metabolites were mainly branched-chain amino acids (BCAAs), with some derivatised BCAAs, lipids, xenobiotics and a few unknowns. Metabolites such as lysophosphatidylcholine a C17:0, sum of hexoses, amino acids from BCAA metabolism (including leucine, isoleucine, valine, N-lactoylvaline, N-lactoylleucine and formiminoglutamate) and lactate, as well as an unknown metabolite (X-24295), were associated with HbA1c progression rate and were significant mediators of type 2 diabetes from baseline to 18 and 48 months of follow-up. 2SMR was used to estimate the causal effect of an exposure on an outcome using summary statistics from UK Biobank genome-wide association studies. We found that type 2 diabetes had a causal effect on the levels of three metabolites (hexose, glutamate and caproate [fatty acid (FA) 6:0]), whereas lipids such as specific phosphatidylcholines (PCs) (namely PC aa C36:2, PC aa C36:5, PC ae C36:3 and PC ae C34:3) as well as the two n-3 fatty acids stearidonate (18:4n3) and docosapentaenoate (22:5n3) potentially had a causal role in the development of type 2 diabetes. CONCLUSIONS/INTERPRETATION Our findings identify known BCAAs and lipids, along with novel N-lactoyl-amino acid metabolites, significantly associated with prediabetes and diabetes, that mediate the effect of diabetes from baseline to follow-up (18 and 48 months). Causal inference using genetic variants shows the role of lipid metabolism and n-3 fatty acids as being causal for metabolite-to-type 2 diabetes whereas the sum of hexoses is causal for type 2 diabetes-to-metabolite. Identified metabolite markers are useful for stratifying individuals based on their risk progression and should enable targeted interventions.
Collapse
Affiliation(s)
- Sapna Sharma
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Qiuling Dong
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Ludwig-Maximilians-University München, Munich, Germany
| | - Mark Haid
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jonathan Adam
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
| | - Roberto Bizzotto
- Institute of Neuroscience, National Research Council, Padova, Italy
| | | | - Angus G Jones
- Department of Clinical and Biomedical Sciences, University of Exeter College of Medicine & Health, Exeter, UK
| | - Andrea Tura
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Anna Artati
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Robert W Koivula
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Paul W Franks
- Department of Clinical Science, Genetic and Molecular Epidemiology, Lund University Diabetes Centre, Malmö, Sweden
| | - Mark Walker
- Translational and Clinical Research Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle upon Tyne, UK
| | - Ian M Forgie
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Giuseppe Giordano
- Department of Clinical Science, Genetic and Molecular Epidemiology, Lund University Diabetes Centre, Malmö, Sweden
| | - Imre Pavo
- Eli Lilly Regional Operations GmbH, Vienna, Austria
| | - Hartmut Ruetten
- Sanofi Partnering, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - Manolis Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Oluf Pedersen
- Center for Clinical Metabolic Research, Herlev and Gentofte University Hospital, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jochen M Schwenk
- Science for Life Laboratory, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | | | - Federico De Masi
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Soren Brunak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ana Viñuela
- Biosciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle upon Tyne, UK
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padova, Italy
| | | | - Tarja Kokkola
- Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jerzy Adamski
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Experimental Genetics, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ewan R Pearson
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), München Neuherberg, Germany.
| |
Collapse
|
4
|
Shakiba E, Pasdar Y, Asoudeh F, Najafi F, Saber A, Shakiba MH, Bagheri A. The relationship of dietary omega-3 fatty acid and omega-6 to omega-3 ratio intake and likelihood of type 2 diabetes in a cross-sectional study. BMC Endocr Disord 2024; 24:259. [PMID: 39614340 DOI: 10.1186/s12902-024-01787-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/14/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND There is a belief that consuming a significant amount of omega-3 and omega-6 fatty acids can positively affect chronic diseases. However, the connection between these fatty acids and type 2 diabetes mellitus (T2DM) risk remains unclear. To explore this further, we conducted a study to investigate the relationship between dietary intake of omega-3 and omega-6 fatty acids (FA), as well as omega-6 to omega-3 ratio, and the odds of T2DM. METHODS Our research involved a cross-sectional analysis of data from the Ravansar Non-Communicable Disease (RaNCD) cohort. We evaluated their dietary habits using a comprehensive 118-item food frequency questionnaire (FFQ). To determine the aforementioned association, we employed logistic regression analysis to calculate odds ratios (OR) and 95% confidence intervals (CIs). RESULTS The prevalence of T2DM among 8744 qualified participants was 751 (8.6%). After considering all the possible factors that could affect the outcome, high dietary omega-3 intake was associated with a 58% lower likelihood of T2DM (OR: 0.42; 95% CI: 0.32, 0.56; P-trend: <0.001). In contrast, participants at the fourth quartile of the dietary omega 6 to omega 3 FA ratio had a higher odd of T2DM (OR: 1.42; 95%CI: 1.11, 1.84; P-trend: 0.01). Nevertheless, there was no significant connection between the highest and lowest quartile of dietary omega-6 intake (OR: 0.91; 95% CI: 0.71, 1.17; P-trend: 0.80). CONCLUSION According to the study, consuming omega-3 fatty acids through diet was linked with lower odds of type 2 diabetes. Conversely, an elevated omega-6 to omega-3 ratio was associated with a greater likelihood of T2DM.
Collapse
Affiliation(s)
- Ebrahim Shakiba
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yahya Pasdar
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farzaneh Asoudeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Najafi
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Saber
- Department of Nutritional Sciences, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Amir Bagheri
- Department of Nutritional Sciences, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
5
|
Qian F, Guo Y, Li C, Liu Y, Luttmann-Gibson H, Gomelskaya N, Demler OV, Cook NR, Lee IM, Buring JE, Larsen J, Boring J, McPhaul MJ, Manson JE, Pradhan AD, Mora S. Biomarkers of glucose-insulin homeostasis and incident type 2 diabetes and cardiovascular disease: results from the Vitamin D and Omega-3 trial. Cardiovasc Diabetol 2024; 23:393. [PMID: 39488682 PMCID: PMC11531120 DOI: 10.1186/s12933-024-02470-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/13/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Dysglycemia and insulin resistance increase type 2 diabetes (T2D) and cardiovascular disease (CVD) risk, yet associations with specific glucose-insulin homeostatic biomarkers have been inconsistent. Vitamin D and marine omega-3 fatty acids (n-3 FA) may improve insulin resistance. We sought to examine the association between baseline levels of insulin, C-peptide, HbA1c, and a novel insulin resistance score (IRS) with incident cardiometabolic diseases, and whether randomized vitamin D or n-3 FA modify these associations. METHODS VITamin D and OmegA-3 TriaL (NCT01169259) was a randomized clinical trial testing vitamin D and n-3 FA for the prevention of CVD and cancer over a median of 5.3 years. Incident cases of T2D and CVD (including cardiovascular death, myocardial infarction, stroke, and coronary revascularization) were matched 1:1 on age, sex, and fasting status to controls. Conditional logistic regressions adjusted for demographic, clinical, and adiposity-related factors were used to assess the adjusted odds ratio (aOR) per-standard deviation (SD) and 95%CI of baseline insulin, C-peptide, HbA1c, and IRS (Insulin×0.0295 + C-peptide×0.00372) with risk of T2D, CVD, and coronary heart disease (CHD). RESULTS We identified 218 T2D case-control pairs and 715 CVD case-control pairs including 423 with incident CHD. Each of the four biomarkers at baseline was separately associated with incident T2D, aOR (95%CI) per SD increment: insulin 1.46 (1.03, 2.06), C-peptide 2.04 (1.35, 3.09), IRS 1.72 (1.28, 2.31) and HbA1c 7.00 (3.76, 13.02), though only HbA1c remained statistically significant with mutual adjustments. For cardiovascular diseases, we only observed significant associations of HbA1c with CVD (1.19 [1.02, 1.39]), and IRS with CHD (1.25 [1.04, 1.50]), which persisted after mutual adjustment. Randomization to vitamin D and/or n-3 FA did not modify the association of these biomarkers with the endpoints. CONCLUSIONS Each of insulin, C-peptide, IRS, and HbA1c were associated with incident T2D with the strongest association noted for HbA1c. While HbA1c was significantly associated with CVD risk, a novel IRS appears to be associated with CHD risk. Neither vitamin D nor n-3 FA modified the associations between these biomarkers and cardiometabolic outcomes.
Collapse
Affiliation(s)
- Frank Qian
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Section of Cardiovascular Medicine, Boston Medical Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Yanjun Guo
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chunying Li
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yanyan Liu
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heike Luttmann-Gibson
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Natalya Gomelskaya
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Olga V Demler
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nancy R Cook
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - I-Min Lee
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Julie E Buring
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Julia Larsen
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA, USA
| | - Jennifer Boring
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA, USA
| | | | - JoAnn E Manson
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Aruna D Pradhan
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Bristol Myers Squibb, Cambridge, MA, USA
| | - Samia Mora
- Division of Preventive Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Divisions of Preventive and Cardiovascular Medicine, Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
6
|
Schuchardt JP, Hahn A, Greupner T, Tintle NL, Westra J, Harris WS. Higher docosahexaenoic acid proportions in blood are inversely associated with the prevalence of prediabetes: Evidence from the UK Biobank. Nutr Res 2024; 131:62-70. [PMID: 39368287 DOI: 10.1016/j.nutres.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
Prediabetes and type 2 diabetes mellitus are growing global health concerns, predisposing individuals to various vascular complications. Lifestyle modifications, including dietary interventions, offer promising avenues for prevention and management. Using a multivariable-adjusted model, we analyzed the cross-sectional associations between plasma proportions (% of total fatty acids) of omega-3 polyunsaturated fatty acids (n3 PUFA, including total n3 PUFA, docosahexaenoic acid [DHA], non-DHA n3 PUFA), and glycated hemoglobin A1c (HbA1c) as well as the prevalence of prediabetes in a sample from the UK Biobank cohort. Our hypothesis was that proportions of n3 PUFA, especially DHA, would by inversely associated with the prediabetes prevalence. The sample (n = 92,762; 54.5% females) had an average age of 56 years and was overweight (mean body mass index = 27). The mean plasma DHA proportion in the sample was 2.03% (standard deviation [SD] = 0.67%), non-DHA n3 PUFA was 2.41% (SD = 1.02%) and total n3 PUFA was 4.43% (SD = 1.56%). Prediabetic individuals were identified by blood HbA1c proportions between 5.7% and 6.4% (39-46 mmol/mol) according to American Diabetes Association criteria. Each of the three n3 PUFA biomarkers was inversely associated with HbA1c proportions. In particular, DHA showed the strongest inverse association, with an OR of 0.62 (95% confidence intervals: 0.58, 0.67; P < .001) when comparing quintiles 5 to 1 in a fully adjusted model. These findings suggest a potential protective role of n3 PUFA, particularly DHA, in mitigating the risk of having prediabetes. Further prospective investigations are needed to clarify whether long-chain n3 PUFA could function as modifiable factors for prediabetes.
Collapse
Affiliation(s)
- Jan Philipp Schuchardt
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany; The Fatty Acid Research Institute, Sioux Falls, SD, USA.
| | - Andreas Hahn
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany
| | - Theresa Greupner
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany
| | - Nathan L Tintle
- The Fatty Acid Research Institute, Sioux Falls, SD, USA; Department of Population Health Nursing Science, College of Nursing, University of Illinois - Chicago, Chicago, IL, USA
| | - Jason Westra
- The Fatty Acid Research Institute, Sioux Falls, SD, USA
| | - William S Harris
- The Fatty Acid Research Institute, Sioux Falls, SD, USA; Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| |
Collapse
|
7
|
Harris WS, Westra J, Tintle NL, Sala-Vila A, Wu JH, Marklund M. Plasma n6 polyunsaturated fatty acid levels and risk for total and cause-specific mortality: A prospective observational study from the UK Biobank. Am J Clin Nutr 2024; 120:936-942. [PMID: 39181205 DOI: 10.1016/j.ajcnut.2024.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND The potential role of n-6 PUFAs in major health outcomes remains controversial. OBJECTIVES To examine the relationship between the major plasma n6 PUFA, linoleic acid (LA), as well as the non-LA n6 PUFAs, and total and cause-specific mortality. METHODS This was a prospective, observational, biomarker-based study in the UK Biobank. Individuals with complete information on baseline demographic, covariate and plasma PUFA levels (percent ot total fatty acids) and mortality outcomes were included (n=257,925). Multivariable-adjusted, Cox-proportional hazards models were used to predict risk of death from all-causes, and from cardiovascular disease (CVD), cancer, and other causes as a function of plasma LA and non-LA n6 levels, both continuously and by PUFA quintile (Q). RESULTS Comparing LA Q5 to Q1, the hazard ratio (HR, 95% CI) for total mortality was 0.80 (0.76, 0.84; p<0.001), and this was similar for all three cause-specific death categories. On the other hand, mortality HR for non-LA n6 was 1.12 (1.08,1.17; p<0.001), and this was primarily due to increased risk for non-CVD, noncancer deaths [HR 1.29 (1.19,1.40; p<0.001)]. Exploratory analyses among the eight next most common other causes of death suggested that both the decreased risk associated with higher LA and the increased risk associated with non-LA n6 were confined to deaths from respiratory and digestive diseases. CONCLUSIONS These findings highlight the profound differences in mortality risk related to LA and non-LA n6 PUFA levels and underscore the inappropriateness of treating n-6 PUFAs as a homogenous class with respect to health outcomes. They also support recommendations to maintain (if not increase) current LA intakes.
Collapse
Affiliation(s)
- William S Harris
- Fatty Acid Research Institute, Sioux Falls, SD, United States; Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, United States.
| | - Jason Westra
- Fatty Acid Research Institute, Sioux Falls, SD, United States
| | - Nathan L Tintle
- Fatty Acid Research Institute, Sioux Falls, SD, United States; Department of Population Health Nursing Science, College of Nursing, University of Illinois-Chicago, Chicago, IL, United States
| | - Aleix Sala-Vila
- Fatty Acid Research Institute, Sioux Falls, SD, United States; Hospital del Mar Research Institute, Barcelona, Spain
| | - Jason Hy Wu
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Australia; The School of Population Health, UNSW Sydney, Sydney, Australia
| | - Matti Marklund
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Australia; The School of Population Health, UNSW Sydney, Sydney, Australia; Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden; The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
8
|
Sato A, Oomori Y, Nakano R, Matsuura T. Metabolic Dysfunction-Associated Steatotic Liver Disease in Japan: Prevalence Trends and Clinical Background in the 10 Years before the Coronavirus Disease 2019 Pandemic. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1330. [PMID: 39202611 PMCID: PMC11356294 DOI: 10.3390/medicina60081330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/03/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: The trends in metabolic dysfunction-associated steatotic liver disease (MASLD) and related metabolic dysfunctions in Japan are unknown. Thus, we aimed to clarify these trends before the novel coronavirus disease 2019 pandemic in Japan. Materials and Methods: We included Japanese individuals aged 25-79 years who underwent health examinations at our center. We analyzed anthropometry, lifestyle-related disease, and nutritional intake in relation to MASLD trends from 2010-2019. Results: The prevalence of MASLD increased in all ages and body mass index (BMI) classes, reaching 30.3% in males and 16.1% in females, with MASLD accounting for 75% of steatotic liver cases and more than half of all type 2 diabetes mellitus (T2DM) and high waist circumference (HWC) cases. The increase in the prevalence of MASLD was thought to be largely attributable to an increase in that of the incidence of steatotic liver itself, and there was no increase in the prevalence of other factors, such as overweight, T2DM, hypertension, and dyslipidemia. The prevalence of glucose metabolic disorders (GMDs) and hypertension decreased. National nutritional data showed an increase in energy intake, total fat, saturated fatty acids, monounsaturated fatty acids, and polyunsaturated fatty acids, which correlated with a decrease in GMDs. Salt intake also decreased, which correlated with hypertension. The MASLD group had a higher prevalence of all related metabolic factors than the non-MASLD group, especially HWC, T2DM, and hyperlipidemia. Conclusions: The prevalence of MASLD increased with that of steatotic liver, regardless of age or BMI. A relationship between increased dietary fat, increased steatotic liver, and decreased GMDs was suggested.
Collapse
Affiliation(s)
- Akira Sato
- Department of Health Management, St. Marianna University Yokohama Seibu Hospital, 1197-1 Yasashicho Asahi-ku, Yokohama 241-0811, Kanagawa, Japan
- Medical Department, Sasaki Foundation Shonan Health Examination Center, 10-4 Takaracho, Hiratsuka 254-0034, Kanagawa, Japan
| | - Yumiko Oomori
- Department of Clinical Examination, Sasaki Foundation Shonan Health Examination Center, 10-4 Takaracho, Hiratsuka 254-0034, Kanagawa, Japan
| | - Rika Nakano
- Department of Radiology, Sasaki Foundation Shonan Health Examination Center, 10-4 Takaracho, Hiratsuka 254-0034, Kanagawa, Japan;
| | - Tomokazu Matsuura
- Medical Department, Sasaki Foundation Shonan Health Examination Center, 10-4 Takaracho, Hiratsuka 254-0034, Kanagawa, Japan
| |
Collapse
|
9
|
Wang Y, Wu G, Xiao F, Yin H, Yu L, Chen Y, Shehzad Q, Xu L, Zhang H, Jin Q, Wang X. Fatty acid composition in erythrocytes and coronary artery disease risk: a case-control study in China. Food Funct 2024; 15:7174-7188. [PMID: 38895817 DOI: 10.1039/d4fo00016a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background and aims: There is limited and conflicting evidence about the association of erythrocyte fatty acids with coronary artery disease (CAD), particularly in China where the CAD rates are high. Our study aimed to explore the association between erythrocyte fatty acid composition and CAD risk in Chinese adults. Methods: Erythrocyte fatty acids of 314 CAD patients and 314 matched controls were measured by gas chromatography. Multivariable conditional logistic regression and restricted cubic spline models were used to explore the odds ratio with 95% confidence interval (OR, 95% CI) and potential association between erythrocyte fatty acids and CAD risk. Principal component analysis (PCA) was used to analyze further the potential role of various erythrocyte fatty acid patterns in relation to CAD risk. Results: Significant inverse associations were observed between high levels of erythrocyte total n-3 polyunsaturated fatty acids (n-3 PUFA) [ORT3-T1 = 0.18 (0.12, 0.28)], monounsaturated fatty acids (MUFA) [ORT3-T1 = 0.21 (0.13, 0.32)], and the risk of CAD. Conversely, levels of saturated fatty acids (SFAs) and n-6 polyunsaturated fatty acids (n-6 PUFAs) were positively associated with CAD risk [ORT3-T1 = 3.33 (2.18, 5.13), ORT3-T1 = 1.61 (1.06, 2.43)]. No significant association was observed between CAD risk and total trans fatty acids. Additionally, the PCA identifies four new fatty acid patterns (FAPs). The risk of CAD was significantly positively associated with FAP1 and FAP2, while being negatively correlated with FAP3 and FAP4. Conclusion: The different types of erythrocyte fatty acids may significantly alter susceptibility to CAD. Elevated levels of n-3-PUFAs and MUFAs are considered as protective biomarkers against CAD, while SFAs and n-6 PUFAs may be associated with higher CAD risk in Chinese adults. The risk of CAD was positively associated with FAP1 and FAP2, and negatively associated with FAP3 and FAP4. Combinations of erythrocyte fatty acids may be more important markers of CAD development than individual fatty acids or their subgroups.
Collapse
Affiliation(s)
- Yongjin Wang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Gangcheng Wu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| | - Feng Xiao
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Hongming Yin
- Department of Laboratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Le Yu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
- National Center of Technology Innovation for Dairy, Hohhot 010000, China
| | - Yujia Chen
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Qayyum Shehzad
- School of Fundamental Sciences, Massey University, Palmerston North 4410, New Zealand
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand
| | - Lirong Xu
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, China
| | - Hui Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| | - Qingzhe Jin
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| | - Xingguo Wang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
10
|
Zhang L, Liu J, Cao Y, Liu S, Zhao W, Wang C, Banzhao S, Liu Z, Liu L. Association between circulating levels of unsaturated fatty acids and risk for prediabetes in the NHANES 2003-2004 and 2011-2012. Diabetes Res Clin Pract 2024; 213:111728. [PMID: 38838943 DOI: 10.1016/j.diabres.2024.111728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/11/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
AIMS This study aimed to investigate the association between serum levels of common and uncommon unsaturated fatty acids and prediabetes risk. METHODS Data were collected from the National Health and Nutrition Examination Survey for 2003-2004 and 2011-2012. Weighted proportional and multivariate logistic regression analyses were performed to assess the association of serum PUFAs and MUFAs with prediabetes risk after adjusting for potential confounders. RESULTS A total of 3575 individuals were enrolled in this study. Serum levels of PUFAs EPA (20:5 n3) and GLA (18:3 n6) were associated with increased prediabetes risk (EPA (20:5 n3): OR = 1.878, 95% CI: 1.177-2.996, Ptrend = 0.002; GLA (18:3 n6): 1.702, 95% CI: 1.140-2.541, Ptrend = 0.016). The MUFAs PA (16:1 n7) and EA (20:1 n9) were associated with the risk of prediabetes (OR in quintile5: PA (16:1 n7): 1.780, 95% CI: 1.056-3.001, Ptrend = 0.003; EA (20:1 n9): 0.587, 95% CI: 0.347-0.994, Ptrend = 0.010). Moreover, nonlinear analysis revealed that serum levels of EPA (20:5 n3) and EA (20:1 n-9) were nonlinearly associated with prediabetes risk. CONCLUSION Some serum n-3 PUFAs are positively associated with prediabetes, several serum n-6 PUFAs are inversely associated with prediabetes. Regulating individual serum USFA levels may help prevent prediabetes, thereby providing evidence for clinical and nutritional practices.
Collapse
Affiliation(s)
- Liwen Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Jiayi Liu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Yuxuan Cao
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Shan Liu
- Department of Endocrinology, The Second Hospital of Shijiazhuang, Shijiazhuang, Hebei 050000, China
| | - Weili Zhao
- Hebei Key Laboratory of Basic Medicine for Diabetes, The Second Hospital of Shijiazhuang, Shijiazhuang, Hebei 050000, China
| | - Ci Wang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Shangfang Banzhao
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Zanchao Liu
- Hebei Key Laboratory of Basic Medicine for Diabetes, The Second Hospital of Shijiazhuang, Shijiazhuang, Hebei 050000, China; Shijiazhuang Diabetes Precision Diagnosis and Treatment Technology Innovation Center, Shijiazhuang, Hebei 050000, China.
| | - Lipeng Liu
- Hebei Key Laboratory of Basic Medicine for Diabetes, The Second Hospital of Shijiazhuang, Shijiazhuang, Hebei 050000, China; College of Veterinary, Hebei Agricultural University, Baoding 071000, China.
| |
Collapse
|
11
|
Dicklin MR, Anthony JC, Winters BL, Maki KC. ω-3 Polyunsaturated Fatty Acid Status Testing in Humans: A Narrative Review of Commercially Available Options. J Nutr 2024; 154:1487-1504. [PMID: 38522783 DOI: 10.1016/j.tjnut.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
There is an increasing body of evidence supporting a link between low intakes of ω-3 long-chain polyunsaturated fatty acids (LCPUFA) and numerous diseases and health conditions. However, few people are achieving the levels of fish/seafood or eicosapentaenoic acid and docosahexaenoic acid intake recommended in national and international guidelines. Knowledge of a person's ω-3 LCPUFA status will benefit the interpretation of research results and could be expected to lead to an increased effort to increase intake. Dietary intake survey methods are often used as a surrogate for measuring ω-3 PUFA tissue status and its impact on health and functional outcomes. However, because individuals vary widely in their ability to digest and absorb ω-3 PUFA, analytical testing of biological samples is desirable to accurately evaluate ω-3 PUFA status. Adipose tissue is the reference biospecimen for measuring tissue fatty acids, but less-invasive methods, such as measurements in whole blood or its components (e.g., plasma, serum, red blood cell membranes) or breast milk are often used. Numerous commercial laboratories provide fatty acid testing of blood and breast milk samples by different methods and present their results in a variety of reports such as a full fatty acid profile, ω-3 and ω-6 fatty acid profiles, fatty acid ratios, as well as the Omega-3 Index, the Holman Omega-3 Test, OmegaScore, and OmegaCheck, among others. This narrative review provides information about the different ways to measure ω-3 LCPUFA status (including both dietary assessments and selected commercially available analytical tests of blood and breast milk samples) and discusses evidence linking increased ω-3 LCPUFA intake or status to improved health, focusing on cardiovascular, neurological, pregnancy, and eye health, in support of recommendations to increase ω-3 LCPUFA intake and testing.
Collapse
Affiliation(s)
| | | | | | - Kevin C Maki
- Midwest Biomedical Research, Addison, IL, United States; Indiana University School of Public Health, Bloomington, IN, United States.
| |
Collapse
|
12
|
Napiórkowska-Baran K, Treichel P, Czarnowska M, Drozd M, Koperska K, Węglarz A, Schmidt O, Darwish S, Szymczak B, Bartuzi Z. Immunomodulation through Nutrition Should Be a Key Trend in Type 2 Diabetes Treatment. Int J Mol Sci 2024; 25:3769. [PMID: 38612580 PMCID: PMC11011461 DOI: 10.3390/ijms25073769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/14/2024] Open
Abstract
An organism's ability to function properly depends not solely on its diet but also on the intake of nutrients and non-nutritive bioactive compounds that exert immunomodulatory effects. This principle applies both to healthy individuals and, in particular, to those with concomitant chronic conditions, such as type 2 diabetes. However, the current food industry and the widespread use of highly processed foods often lead to nutritional deficiencies. Numerous studies have confirmed the occurrence of immune system dysfunction in patients with type 2 diabetes. This article elucidates the impact of specific nutrients on the immune system function, which maintains homeostasis of the organism, with a particular emphasis on type 2 diabetes. The role of macronutrients, micronutrients, vitamins, and selected substances, such as omega-3 fatty acids, coenzyme Q10, and alpha-lipoic acid, was taken into consideration, which outlined the minimum range of tests that ought to be performed on patients in order to either directly or indirectly determine the severity of malnutrition in this group of patients.
Collapse
Affiliation(s)
- Katarzyna Napiórkowska-Baran
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland;
| | - Paweł Treichel
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland; (P.T.); (M.C.); (M.D.); (K.K.); (A.W.); (O.S.); (S.D.); (B.S.)
| | - Marta Czarnowska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland; (P.T.); (M.C.); (M.D.); (K.K.); (A.W.); (O.S.); (S.D.); (B.S.)
| | - Magdalena Drozd
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland; (P.T.); (M.C.); (M.D.); (K.K.); (A.W.); (O.S.); (S.D.); (B.S.)
| | - Kinga Koperska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland; (P.T.); (M.C.); (M.D.); (K.K.); (A.W.); (O.S.); (S.D.); (B.S.)
| | - Agata Węglarz
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland; (P.T.); (M.C.); (M.D.); (K.K.); (A.W.); (O.S.); (S.D.); (B.S.)
| | - Oskar Schmidt
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland; (P.T.); (M.C.); (M.D.); (K.K.); (A.W.); (O.S.); (S.D.); (B.S.)
| | - Samira Darwish
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland; (P.T.); (M.C.); (M.D.); (K.K.); (A.W.); (O.S.); (S.D.); (B.S.)
| | - Bartłomiej Szymczak
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland; (P.T.); (M.C.); (M.D.); (K.K.); (A.W.); (O.S.); (S.D.); (B.S.)
| | - Zbigniew Bartuzi
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland;
| |
Collapse
|
13
|
Li Y, Liu X, Zhuang P, Zhang L, Wu Y, Wu S, Zhang Y, Jiao J. Fish oil supplementation and risk of dementia among diabetic patients: a prospective study of 16,061 older patients. J Nutr Health Aging 2024; 28:100176. [PMID: 38341308 DOI: 10.1016/j.jnha.2024.100176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/12/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND Although n-3 Polyunsaturated fatty acids (PUFAs) may benefit cognitive performance, the association of n-3 PUFA intake with dementia risk under dysglycemia has not been examined. We aimed to evaluate the relationship between fish oil supplement use or fish consumption and dementia risk among older patients with diabetes. METHOD A total of 16,061 diabetic patients aged over 60 years were followed up in the UK Biobank. Fish oil supplements use (yes or no) was collected by the touch screen questionnaire. The diagnosis of dementia was ascertained by the UK Biobank Outcome Adjudication Group. The hazard ratios (HRs) and 95% confidence intervals (95% CIs) were estimated using Cox proportional hazards models. RESULTS A total of 337 cases of dementia were confirmed after a mean duration of 7.7 years (123,486 person-years) of follow-up. Habitual use of fish oil supplements showed a 24% lower dementia risk among older diabetic patients [HRs (95% CIs): 0.76 (0.60-0.98) (P = 0.031)] compared with non-users. Such inverse association was not modified by the APOE ε4 genotype. However, the consumption of both oily fish (≥2 times/week) and non-oily fish (≥2 times/week) had no significant association with dementia risk (p-trend = 0.271 and p-trend = 0.065) compared with non-consumers. CONCLUSION In summary, fish oil supplementation may play a protective role in cognitive function across all APOE genotypes, while non-oily fish and oily fish consumption have no protective association among older diabetic patients.
Collapse
Affiliation(s)
- Yin Li
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Xiaohui Liu
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Pan Zhuang
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Lange Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yuqi Wu
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Shanyun Wu
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yu Zhang
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jingjing Jiao
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW The very-long chain (VLC) omega-3 polyunsaturated fatty acids (PUFAs) eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) promote optimal development, physiological function and healthy ageing and help to manage disease. EPA and DHA are sourced mainly from fish, which is not sustainable. This review explores alternative sustainable sources. RECENT FINDINGS Recent research confirms that higher intake and status of EPA and DHA are associated with health benefits including lower risk of incident type-2 diabetes and cardiovascular disease mortality. Meta-analyses confirm benefits of intravenous EPA and DHA in hospitalized adults. Algal oils and seed oils from some genetically modified (GM) plants are sources of EPA and DHA. An oil from GM camelina showed equivalence with fish oil in human trials. Ahiflower oil, a source of stearidonic acid, had biological effects in experimental studies that might translate into health benefits. An intravenous lipid emulsion based on Ahiflower oil has been tested in experimental research. Pine nut oil (PNO) is a source of pinolenic acid, which is not an omega-3 PUFA but has similar actions. SUMMARY Algal oils, oils from GM seed crops, Ahiflower oil and other sources of stearidonic acid, and nonomega-3 oils including PNO, are plant-sourced sustainable alternatives to fish-sourced VLC omega-3 PUFAs.
Collapse
Affiliation(s)
- Ella J Baker
- School of Human Development and Health, Faculty of Medicine
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| |
Collapse
|
15
|
Retterstøl K, Rosqvist F. Fat and fatty acids - a scoping review for Nordic Nutrition Recommendations 2023. Food Nutr Res 2024; 68:9980. [PMID: 38327998 PMCID: PMC10845901 DOI: 10.29219/fnr.v68.9980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/05/2023] [Accepted: 08/30/2023] [Indexed: 02/09/2024] Open
Abstract
Two de novo NNR2022 systematic reviews (SRs) as well as 21 qualified SRs (qSRs) were available. A literature search yielded an additional ~70 SRs, meta-analyses and biomarker papers. Diets lower in total fat are associated with reductions in body weight and blood pressure compared with diets higher in total fat in adults. Partial replacement of saturated fatty acid (SFA) with n-6 polyunsaturated fatty acid (PUFA) improves blood lipid profile, decreases the risk of cardiovascular disease (CVD), improves glucose-insulin homeostasis and may decrease the risk of total mortality. Long-chain n-3 PUFAs (eicosapentaenoic acid [EPA] and docosahexaenoic acid [DHA]) decrease triglycerides and are associated with lower risk of CVD. Dietary PUFAs, both n-3 and n-6, may be associated with reduced risk of type 2 diabetes (T2D). There is inconclusive evidence to suggest that the type of dietary fat is associated with blood pressure, risk of hypertension or musculoskeletal health. Higher intake of total PUFA is associated with lower mortality from any cancer. Long-chain n-3 PUFA is associated with reduced risk of breast cancer, whereas biomarker levels of n-6 PUFA are associated with lower risk of any cancer. Intake of long-chain n-3 PUFA during pregnancy increases length of gestation and child birth weight and reduces the risk of preterm delivery, but there is inconclusive evidence to suggest that it may influence child neurodevelopment, growth or development of allergic disease. In studies with higher versus lower dietary cholesterol intake levels, total blood cholesterol increased or were unaffected by the dietary cholesterol, resulting in inconclusive results. Trans fatty acid (TFA), regardless of source, impairs blood lipid profile compared to unsaturated fat. In observational studies, TFA is positively associated with CVD and total mortality but whether associations differ by source is inconclusive. Ruminant TFA, as well as biomarker levels of odd-chain fatty acids, might be associated with lower risk of T2D.
Collapse
Affiliation(s)
- Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Fredrik Rosqvist
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
von Schacky C, Kuipers RS, Pijl H, Muskiet FAJ, Grobbee DE. Omega-3 fatty acids in heart disease-why accurately measured levels matter. Neth Heart J 2023; 31:415-423. [PMID: 36795219 PMCID: PMC10602979 DOI: 10.1007/s12471-023-01759-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 02/17/2023] Open
Abstract
Current guidelines barely support marine omega‑3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in cardiology, mainly because results of large trials were equivocal. Most large trials have tested EPA alone or EPA + DHA combined as a drug, thereby disregarding the relevance of their blood levels. These levels are frequently assessed with the Omega‑3 Index (percentage of EPA + DHA in erythrocytes), which is determined using a specific standardised analytical procedure. EPA and DHA are present in every human being at unpredictable levels (even in the absence of intake), and their bioavailability is complex. Both facts need to be incorporated into trial design and should direct clinical use of EPA and DHA. An Omega‑3 Index in the target range of 8-11% is associated with lower total mortality, fewer major adverse cardiac and other cardiovascular events. Moreover, functions of organs such as the brain benefit from an Omega‑3 Index in the target range, while untoward effects, such as bleeding or atrial fibrillation, are minimised. In pertinent intervention trials, several organ functions were improved, with improvements correlating with the Omega‑3 Index. Thus, the Omega‑3 Index is relevant in trial design and clinical medicine, which calls for a widely available standardised analytical procedure and a discussion on possible reimbursement of this test.
Collapse
Affiliation(s)
| | - R S Kuipers
- Heart Centre, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
- Department of Cardiology, Dijklander Hospital, Purmerend/Hoorn, The Netherlands
| | - H Pijl
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, The Netherlands
| | - F A J Muskiet
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - D E Grobbee
- Julius Global Health, Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
17
|
Miao Z, Zeng FF, Tian Y, Xiao C, Yan Y, Jiang Z, Fu Y, Chen YM, Zheng JS. Furan fatty acid metabolite CMPF is associated with lower risk of type 2 diabetes, but not chronic kidney disease: a longitudinal population-based cohort study. Am J Clin Nutr 2023; 118:637-645. [PMID: 37482300 DOI: 10.1016/j.ajcnut.2023.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Furan fatty acid metabolite 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF) is a strong biomarker of fish and n-3 polyunsaturated fatty acid (PUFA) intake. The relationship of CMPF with human health has been controversial, especially for type 2 diabetes and chronic kidney disease. OBJECTIVE We performed a prospective cohort study to examine the association of serum CMPF with incident type 2 diabetes and chronic kidney disease. METHODS In the Guangzhou Nutrition and Health Study, during a median follow-up of 8.8 y, we used a multivariable-adjusted Poisson regression model to investigate the association of baseline serum CMPF with the incidence of type 2 diabetes (1470 participants and 170 incident cases) and chronic kidney disease (1436 participants and 112 incident cases). We also examined the association of serial measures of serum CMPF with glycemic and renal function biomarkers. Mediation analysis was also performed to examine the contribution of CMPF in the association between marine n-3 PUFAs and risk of type 2 diabetes or chronic kidney disease. RESULTS Each standard deviation increase in baseline serum CMPF was associated with an 18% lower risk of type 2 diabetes (relative risk: 0.82, 95% confidence interval [CI]: 0.68, 0.99) but was not associated with chronic kidney disease (relative risk: 0.95; 95% CI: 0.77-1.16). Correlation analyses of CMPF with glycemic and renal function biomarkers showed similar results. Mediation analysis suggested that serum CMPF contributed to the inverse association between erythrocyte marine n-3 PUFAs and incident type 2 diabetes (proportion mediated 37%, P-mediation = 0.022). CONCLUSIONS Our findings suggest that serum CMPF was associated with a lower risk of type 2 diabetes but not chronic kidney disease. This study also suggests that CMPF may be a functional metabolite underlying the protective relationship between marine n-3 PUFA intake and type 2 diabetes.
Collapse
Affiliation(s)
- Zelei Miao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Fang-Fang Zeng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Yunyi Tian
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China; Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Congmei Xiao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China; Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Yan Yan
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zengliang Jiang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China; Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Yuanqing Fu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China; Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Yu-Ming Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Ju-Sheng Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China; Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China.
| |
Collapse
|
18
|
Liu SH, Lin WH, Tzeng HP, Chiang MT. Attenuation of diabetes-mediated muscle atrophy in rats by fish oil enriched omega-3 polyunsaturated fatty acids supplementation. J Food Drug Anal 2023; 31:458-472. [PMID: 39666277 PMCID: PMC10629917 DOI: 10.38212/2224-6614.3468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/12/2023] [Indexed: 12/13/2024] Open
Abstract
Diabetes is associated with an increased risk of muscle wasting/atrophy, which adversely affects quality of life. We hypothesized that long term supplementation of fish oil may have protective effects against sarcopenia or muscle atrophy in streptozotocin (STZ) and high-fat (HF) diet-induced diabetic rat model. Wistar rats at age of 7 weeks were injected with saline or STZ to induce hyperglycemia. After one week, they were fed on a normal control diet or HF diet with/without supplementation of fish oil for 18 weeks. Feeding diabetic rats with a fish oil-enriched diet alleviated body weight loss and the impaired glucose tolerance using OGTT test. Although fish oil did not improve the decreased muscle mass, the muscle atrophy induced by diabetes was attenuated by fish oil in gastrocnemius, soleus, tibialis anterior, and extensor digitorum longus muscles. Fish oil supplementation reversed the decreased expression of phospho (p)-AKT, pmTOR, and p-p70s6k, which are molecules related to protein synthesis. Besides, protein degradation-related signaling pathways were inhibited by fish oil, such as increasing p-FoxO1 and decreasing Atrogin-1 and MURF1 protein expression. Fish oil down-regulated the expression of autophagy-related molecules including ATG5, p62, and LC3B II/I ratio, which may result in less muscle atrophy. Inflammation-related signaling regulators including TNF-α, NF-κB, AGEs, and RAGE were suppressed by fish oil supplementation as well. Moreover, the down-regulated p-AMPKα, SIRT1, and PGC-1 in diabetic rats were counteracted by fish oil, which may improve mitochondrial function and further block FoxO action. These data suggest that long-term fish oil supplementation exerts protective effects against diabetes-induced muscle atrophy, which may in turn ameliorate insulin resistance and impaired glucose tolerance.
Collapse
Affiliation(s)
- Shing-Hwa Liu
- Institute of Toxicology, National Taiwan University, Taipei,
Taiwan
- Department of Pediatrics, College of Medicine and Hospital, National Taiwan University, Taipei,
Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung,
Taiwan
| | - Wei-Hsuan Lin
- Department of Food Science, National Taiwan Ocean University, Keelung,
Taiwan
| | - Huei-Ping Tzeng
- Institute of Toxicology, National Taiwan University, Taipei,
Taiwan
| | - Meng-Tsan Chiang
- Department of Food Science, National Taiwan Ocean University, Keelung,
Taiwan
| |
Collapse
|
19
|
Su J, Luo Y, Hu S, Tang L, Ouyang S. Advances in Research on Type 2 Diabetes Mellitus Targets and Therapeutic Agents. Int J Mol Sci 2023; 24:13381. [PMID: 37686185 PMCID: PMC10487533 DOI: 10.3390/ijms241713381] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Diabetes mellitus is a chronic multifaceted disease with multiple potential complications, the treatment of which can only delay and prolong the terminal stage of the disease, i.e., type 2 diabetes mellitus (T2DM). The World Health Organization predicts that diabetes will be the seventh leading cause of death by 2030. Although many antidiabetic medicines have been successfully developed in recent years, such as GLP-1 receptor agonists and SGLT-2 inhibitors, single-target drugs are gradually failing to meet the therapeutic requirements owing to the individual variability, diversity of pathogenesis, and organismal resistance. Therefore, there remains a need to investigate the pathogenesis of T2DM in more depth, identify multiple therapeutic targets, and provide improved glycemic control solutions. This review presents an overview of the mechanisms of action and the development of the latest therapeutic agents targeting T2DM in recent years. It also discusses emerging target-based therapies and new potential therapeutic targets that have emerged within the last three years. The aim of our review is to provide a theoretical basis for further advancement in targeted therapies for T2DM.
Collapse
Affiliation(s)
- Jingqian Su
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Y.L.); (S.H.); (L.T.)
- Provincial University Key Laboratory of Microbial Pathogenesis and Interventions, Fujian Normal University, Fuzhou 350117, China
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Fujian Normal University, Fuzhou 350117, China
| | - Yingsheng Luo
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Y.L.); (S.H.); (L.T.)
- Provincial University Key Laboratory of Microbial Pathogenesis and Interventions, Fujian Normal University, Fuzhou 350117, China
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Fujian Normal University, Fuzhou 350117, China
| | - Shan Hu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Y.L.); (S.H.); (L.T.)
- Provincial University Key Laboratory of Microbial Pathogenesis and Interventions, Fujian Normal University, Fuzhou 350117, China
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Fujian Normal University, Fuzhou 350117, China
| | - Lu Tang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Y.L.); (S.H.); (L.T.)
- Provincial University Key Laboratory of Microbial Pathogenesis and Interventions, Fujian Normal University, Fuzhou 350117, China
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Fujian Normal University, Fuzhou 350117, China
| | - Songying Ouyang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Y.L.); (S.H.); (L.T.)
- Provincial University Key Laboratory of Microbial Pathogenesis and Interventions, Fujian Normal University, Fuzhou 350117, China
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Fujian Normal University, Fuzhou 350117, China
- Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| |
Collapse
|
20
|
Ballantyne CM, Jia X. Omega-3 Fatty Acids and Risk for Atrial Fibrillation: Big Fish or Small Fry? J Am Coll Cardiol 2023; 82:350-352. [PMID: 37468190 DOI: 10.1016/j.jacc.2023.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 07/21/2023]
Affiliation(s)
| | - Xiaoming Jia
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
21
|
Qian F, Tintle N, Jensen PN, Lemaitre RN, Imamura F, Feldreich TR, Nomura SO, Guan W, Laguzzi F, Kim E, Virtanen JK, Steur M, Bork CS, Hirakawa Y, O'Donoghue ML, Sala-Vila A, Ardisson Korat AV, Sun Q, Rimm EB, Psaty BM, Heckbert SR, Forouhi NG, Wareham NJ, Marklund M, Risérus U, Lind L, Ärnlöv J, Garg P, Tsai MY, Pankow J, Misialek JR, Gigante B, Leander K, Pester JA, Albert CM, Kavousi M, Ikram A, Voortman T, Schmidt EB, Ninomiya T, Morrow DA, Bayés-Genís A, O'Keefe JH, Ong KL, Wu JHY, Mozaffarian D, Harris WS, Siscovick DS. Omega-3 Fatty Acid Biomarkers and Incident Atrial Fibrillation. J Am Coll Cardiol 2023; 82:336-349. [PMID: 37468189 DOI: 10.1016/j.jacc.2023.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND The relationship between omega-3 fatty acids and atrial fibrillation (AF) remains controversial. OBJECTIVES This study aimed to determine the prospective associations of blood or adipose tissue levels of eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and docosahexaenoic acid (DHA) with incident AF. METHODS We used participant-level data from a global consortium of 17 prospective cohort studies, each with baseline data on blood or adipose tissue omega-3 fatty acid levels and AF outcomes. Each participating study conducted a de novo analyses using a prespecified analytical plan with harmonized definitions for exposures, outcome, covariates, and subgroups. Associations were pooled using inverse-variance weighted meta-analysis. RESULTS Among 54,799 participants from 17 cohorts, 7,720 incident cases of AF were ascertained after a median 13.3 years of follow-up. In multivariable analysis, EPA levels were not associated with incident AF, HR per interquintile range (ie, the difference between the 90th and 10th percentiles) was 1.00 (95% CI: 0.95-1.05). HRs for higher levels of DPA, DHA, and EPA+DHA, were 0.89 (95% CI: 0.83-0.95), 0.90 (95% CI: 0.85-0.96), and 0.93 (95% CI: 0.87-0.99), respectively. CONCLUSIONS In vivo levels of omega-3 fatty acids including EPA, DPA, DHA, and EPA+DHA were not associated with increased risk of incident AF. Our data suggest the safety of habitual dietary intakes of omega-3 fatty acids with respect to AF risk. Coupled with the known benefits of these fatty acids in the prevention of adverse coronary events, our study suggests that current dietary guidelines recommending fish/omega-3 fatty acid consumption can be maintained.
Collapse
Affiliation(s)
- Frank Qian
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathan Tintle
- Department of Mathematics and Statistics, Dordt University, Sioux Center, Iowa, USA; Fatty Acid Research Institute, Sioux Falls, South Dakota, USA
| | - Paul N Jensen
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Fumiaki Imamura
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Tobias Rudholm Feldreich
- School of Health and Social Sciences, Dalarna University, Falun, Sweden; Center for Clinical Research Dalarna, Region Dalarna, Falun, Sweden
| | - Sarah Oppeneer Nomura
- Department of Lab Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Federica Laguzzi
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eunjung Kim
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jyrki K Virtanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Marinka Steur
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Christian S Bork
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Yoichiro Hirakawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Michelle L O'Donoghue
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Aleix Sala-Vila
- Fatty Acid Research Institute, Sioux Falls, South Dakota, USA; Cardiovascular Risk and Nutrition - Hospital Del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Andres V Ardisson Korat
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eric B Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Systems and Population Health, University of Washington, Seattle, Washington, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Nita G Forouhi
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Matti Marklund
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden; Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts, USA; The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Johan Ärnlöv
- Center for Clinical Research Dalarna, Region Dalarna, Falun, Sweden; School of Health and Social Studies, Dalarna University, Falun, Sweden; Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institute, Stockholm, Sweden
| | - Parveen Garg
- Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Michael Y Tsai
- Department of Lab Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - James Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jeffrey R Misialek
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Bruna Gigante
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Karin Leander
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Julie A Pester
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Christine M Albert
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands
| | - Trudy Voortman
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands; Division of Human Nutrition and Health, Wageningen University and Research, Wageningenn, the Netherlands
| | - Erik B Schmidt
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - David A Morrow
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Antoni Bayés-Genís
- Department of Cardiology, Heart Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - James H O'Keefe
- Saint Luke's Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Kwok Leung Ong
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Jason H Y Wu
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts, USA; Division of Cardiology, Tufts Medical Center, Boston, Massachusetts, USA
| | - William S Harris
- Fatty Acid Research Institute, Sioux Falls, South Dakota, USA; Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota, USA
| | | |
Collapse
|
22
|
Hu C, Zhou Y, Wu X, Jia X, Zhu Y, Zheng R, Wang S, Lin L, Qi H, Lin H, Li M, Wang T, Zhao Z, Xu M, Xu Y, Chen Y, Ning G, Borges MC, Wang W, Zheng J, Bi Y, Lu J. Evaluating the distinct pleiotropic effects of omega-3 fatty acids on type 2 diabetes mellitus: a mendelian randomization study. J Transl Med 2023; 21:370. [PMID: 37286992 DOI: 10.1186/s12967-023-04202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/14/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND Observational studies and conventional Mendelian randomization (MR) studies showed inconclusive evidence to support the association between omega-3 fatty acids and type 2 diabetes. We aim to evaluate the causal effect of omega-3 fatty acids on type 2 diabetes mellitus (T2DM), and the distinct intermediate phenotypes linking the two. METHODS Two-sample MR was performed using genetic instruments derived from a recent genome-wide association study (GWAS) of omega-3 fatty acids (N = 114,999) from UK Biobank and outcome data obtained from a large-scale T2DM GWAS (62,892 cases and 596,424 controls) in European ancestry. MR-Clust was applied to determine clustered genetic instruments of omega-3 fatty acids that influences T2DM. Two-step MR analysis was used to identify potential intermediate phenotypes (e.g. glycemic traits) that linking omega-3 fatty acids with T2DM. RESULTS Univariate MR showed heterogenous effect of omega-3 fatty acids on T2DM. At least two pleiotropic effects between omega-3 fatty acids and T2DM were identified using MR-Clust. For cluster 1 with seven instruments, increasing omega-3 fatty acids reduced T2DM risk (OR: 0.52, 95%CI 0.45-0.59), and decreased HOMA-IR (β = - 0.13, SE = 0.05, P = 0.02). On the contrary, MR analysis using 10 instruments in cluster 2 showed that increasing omega-3 fatty acids increased T2DM risk (OR:1.10; 95%CI 1.06-1.15), and decreased HOMA-B (β = - 0.04, SE = 0.01, P = 4.52 × 10-5). Two-step MR indicated that increasing omega-3 fatty acid levels decreased T2DM risk via decreasing HOMA-IR in cluster 1, while increased T2DM risk via decreasing HOMA-B in cluster 2. CONCLUSIONS This study provides evidence to support two distinct pleiotropic effects of omega-3 fatty acids on T2DM risk influenced by different gene clusters, which could be partially explained by distinct effects of omega-3 fatty acids on insulin resistance and beta cell dysfunction. The pleiotropic feature of omega-3 fatty acids variants and its complex relationships with T2DM need to be carefully considered in future genetic and clinical studies.
Collapse
Affiliation(s)
- Chunyan Hu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yulin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojing Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyue Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyan Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maria-Carolina Borges
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Epidemiological and mechanistic studies have reported relationships between blood lipids, mostly measured by traditional method in clinical settings, and gestational diabetes mellitus (GDM). Recent advances of high-throughput lipidomics techniques have made available more comprehensive lipid profiling in biological samples. This review aims to summarize evidence from prospective studies in assessing relations between blood lipids and GDM, and discuss potential underlying mechanisms. RECENT FINDINGS Mass spectrometry and nuclear magnetic resonance spectroscopy-based analytical platforms are extensively used in lipidomics research. Epidemiological studies have identified multiple novel lipidomic biomarkers that are associated with risk of GDM, such as certain types of fatty acids, glycerolipids, glycerophospholipids, sphingolipids, cholesterol, and lipoproteins. However, the findings are inconclusive mainly due to the heterogeneities in study populations, sample sizes, and analytical platforms. Mechanistic evidence indicates that abnormal lipid metabolism may be involved in the pathogenesis of GDM by impairing pancreatic β-cells and inducing insulin resistance through several etiologic pathways, such as inflammation and oxidative stress. SUMMARY Lipidomics is a powerful tool to study pathogenesis and biomarkers for GDM. Lipidomic biomarkers and pathways could help to identify women at high risk for GDM and could be potential targets for early prevention and intervention of GDM.
Collapse
Affiliation(s)
- Yi Wang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiong-Fei Pan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University
- Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu, China
| | - An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| |
Collapse
|
24
|
Bhat S, Sarkar S, Zaffar D, Dandona P, Kalyani RR. Omega-3 Fatty Acids in Cardiovascular Disease and Diabetes: a Review of Recent Evidence. Curr Cardiol Rep 2023; 25:51-65. [PMID: 36729217 DOI: 10.1007/s11886-022-01831-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE OF REVIEW Omega-3 fatty acids (n-3 FA) lower triglycerides, have anti-inflammatory properties, and improve metabolism. Clinical evidence of cardiovascular benefit with omega-3 fatty acids is mixed. We discuss mechanisms providing biological plausibility of benefit of omega-3 fatty acids in cardiovascular risk reduction and review clinical trials investigating the benefits of prescription omega-3 fatty acids in dyslipidemia, atherosclerotic cardiovascular disease (ASCVD), and diabetes. RECENT FINDINGS Although early trials showed no benefit of omega-3 fatty acids in ASCVD, the REDUCE-IT trial noted significant risk reduction in ASCVD events with highly purified EPA (icosapent ethyl) use which has changed the landscape for currently available therapeutic options. However, other large trials like STRENGTH and VITAL, which used different formulations of prescription omega-3 fatty acids, did not note significant cardiovascular risk reduction. Thus the effectiveness of omega-3 fatty acids for cardiovascular disease prevention is an ongoing topic of debate. A relative paucity of studies examining benefits for glycemic outcomes in persons with diabetes exists; however, few studies have suggested lack of benefit to date. Significant residual cardiovascular risk exists for individuals with hypertriglyceridemia. Prescription omega-3 fatty acids are more commonly used for CV risk reduction in these patients. Clinical guideline statements now recommend icosapent ethyl use for selected individuals with hypertriglyceridemia to reduce cardiovascular events given recent evidence from the REDUCE-IT trial. Nonetheless, data from other large scale trials has been mixed, and future research is needed to better understand how different preparations of omega-3 may differ in their cardiovascular and metabolic effects, and the mechanisms for their benefit.
Collapse
Affiliation(s)
- Salman Bhat
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sudipa Sarkar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Duha Zaffar
- Department of Internal Medicine, University of Maryland Midtown Campus, Baltimore, MD, USA
| | - Paresh Dandona
- Division of Endocrinology, Diabetes and Metabolism, University at Buffalo, Buffalo, NY, USA
| | - Rita R Kalyani
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
25
|
Ong KL, Marklund M, Huang L, Rye KA, Hui N, Pan XF, Rebholz CM, Kim H, Steffen LM, van Westing AC, Geleijnse JM, Hoogeveen EK, Chen YY, Chien KL, Fretts AM, Lemaitre RN, Imamura F, Forouhi NG, Wareham NJ, Birukov A, Jäger S, Kuxhaus O, Schulze MB, de Mello VD, Tuomilehto J, Uusitupa M, Lindström J, Tintle N, Harris WS, Yamasaki K, Hirakawa Y, Ninomiya T, Tanaka T, Ferrucci L, Bandinelli S, Virtanen JK, Voutilainen A, Jayasena T, Thalamuthu A, Poljak A, Bustamante S, Sachdev PS, Senn MK, Rich SS, Tsai MY, Wood AC, Laakso M, Lankinen M, Yang X, Sun L, Li H, Lin X, Nowak C, Ärnlöv J, Risérus U, Lind L, Le Goff M, Samieri C, Helmer C, Qian F, Micha R, Tin A, Köttgen A, de Boer IH, Siscovick DS, Mozaffarian D, Wu JH. Association of omega 3 polyunsaturated fatty acids with incident chronic kidney disease: pooled analysis of 19 cohorts. BMJ 2023; 380:e072909. [PMID: 36653033 PMCID: PMC9846698 DOI: 10.1136/bmj-2022-072909] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To assess the prospective associations of circulating levels of omega 3 polyunsaturated fatty acid (n-3 PUFA) biomarkers (including plant derived α linolenic acid and seafood derived eicosapentaenoic acid, docosapentaenoic acid, and docosahexaenoic acid) with incident chronic kidney disease (CKD). DESIGN Pooled analysis. DATA SOURCES A consortium of 19 studies from 12 countries identified up to May 2020. STUDY SELECTION Prospective studies with measured n-3 PUFA biomarker data and incident CKD based on estimated glomerular filtration rate. DATA EXTRACTION AND SYNTHESIS Each participating cohort conducted de novo analysis with prespecified and consistent exposures, outcomes, covariates, and models. The results were pooled across cohorts using inverse variance weighted meta-analysis. MAIN OUTCOME MEASURES Primary outcome of incident CKD was defined as new onset estimated glomerular filtration rate <60 mL/min/1.73 m2. In a sensitivity analysis, incident CKD was defined as new onset estimated glomerular filtration rate <60 mL/min/1.73 m2 and <75% of baseline rate. RESULTS 25 570 participants were included in the primary outcome analysis and 4944 (19.3%) developed incident CKD during follow-up (weighted median 11.3 years). In multivariable adjusted models, higher levels of total seafood n-3 PUFAs were associated with a lower incident CKD risk (relative risk per interquintile range 0.92, 95% confidence interval 0.86 to 0.98; P=0.009, I2=9.9%). In categorical analyses, participants with total seafood n-3 PUFA level in the highest fifth had 13% lower risk of incident CKD compared with those in the lowest fifth (0.87, 0.80 to 0.96; P=0.005, I2=0.0%). Plant derived α linolenic acid levels were not associated with incident CKD (1.00, 0.94 to 1.06; P=0.94, I2=5.8%). Similar results were obtained in the sensitivity analysis. The association appeared consistent across subgroups by age (≥60 v <60 years), estimated glomerular filtration rate (60-89 v ≥90 mL/min/1.73 m2), hypertension, diabetes, and coronary heart disease at baseline. CONCLUSIONS Higher seafood derived n-3 PUFA levels were associated with lower risk of incident CKD, although this association was not found for plant derived n-3 PUFAs. These results support a favourable role for seafood derived n-3 PUFAs in preventing CKD.
Collapse
Affiliation(s)
- Kwok Leung Ong
- Lipid Research Group, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Matti Marklund
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- The Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Liping Huang
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Nicholas Hui
- Lipid Research Group, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Xiong-Fei Pan
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu, Sichuan, China
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lyn M Steffen
- University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Anniek C van Westing
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Johanna M Geleijnse
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Ellen K Hoogeveen
- Department of Nephrology, Jeroen Bosch Hospital, Den Bosch, The Netherlands
- Institute of Epidemiology and Preventive Medicine College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Amanda M Fretts
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | | | - Fumiaki Imamura
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Nita G Forouhi
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Anna Birukov
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Susanne Jäger
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Olga Kuxhaus
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Vanessa Derenji de Mello
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jaakko Tuomilehto
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Saudi Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Matti Uusitupa
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jaana Lindström
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Nathan Tintle
- The Fatty Acid Research Institute, Sioux Falls, SD, USA
- Department of Population Health Nursing Science, College of Nursing, University of Illinois-Chicago, Chicago, IL, USA
| | - William S Harris
- The Fatty Acid Research Institute, Sioux Falls, SD, USA
- Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keisuke Yamasaki
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoichiro Hirakawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, USA
| | | | - Jyrki K Virtanen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ari Voutilainen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tharusha Jayasena
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Anne Poljak
- Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
| | - Sonia Bustamante
- Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | | | - Mackenzie K Senn
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Stephen S Rich
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Alexis C Wood
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Maria Lankinen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Xiaowei Yang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Liang Sun
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Huaixing Li
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xu Lin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Sweden
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Sweden
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mélanie Le Goff
- Bordeaux Population Health Research Centre, INSERM, UMR 1219, University of Bordeaux, Bordeaux, France
| | - Cécilia Samieri
- Bordeaux Population Health Research Centre, INSERM, UMR 1219, University of Bordeaux, Bordeaux, France
| | - Catherine Helmer
- Bordeaux Population Health Research Centre, INSERM, UMR 1219, University of Bordeaux, Bordeaux, France
| | - Frank Qian
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Renata Micha
- Department of Food Science and Nutrition, University of Thessaly, Karditsa, Greece
- The Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Institute of Genetic Epidemiology, Department of Data Driven Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, Freiburg, Germany
| | - Ian H de Boer
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
- Kidney Research Institute, University of Washington, Seattle, WA, USA
- Puget Sound VA Healthcare System, Seattle, WA, USA
| | | | - Dariush Mozaffarian
- The Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Jason Hy Wu
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- School of Population Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
26
|
Salek M, Hosseini Hooshiar S, Salek M, Poorebrahimi M, Jafarnejad S. Omega-3 fatty acids: Current insights into mechanisms of action in systemic lupus erythematosus. Lupus 2023; 32:7-22. [PMID: 36433776 DOI: 10.1177/09612033221140724] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Systemic lupus erythematosus (SLE) is one of the autoimmune diseases characterized by the lack of self-tolerance and the formation of immune complexes and nuclear autoantigens resulting in inflammation in multiple organs. Nowadays, the major aim of SLE therapy is the control of disease activity. However, the biological heterogeneity between patients and the absence of safe and specific targeted treatments complicate the lupus management. Therefore, the potential prophylactic effects of natural therapy considering the potential side effects of classical pharmacology, also the role of diet therapy in decreasing co-morbidities and improving quality of life in SLE patients could be a promising approach to SLE disease. Omega-3 polyunsaturated fatty acids (omega-3 PUFAs) are one of the agents that are considered for their preventive and therapeutic properties in disease activity of SLE and the related complications. The intake of omega-3 PUFAs likely has a direct relationship with improvements in inflammatory, cardiovascular, depressive, and neuromotor symptoms of the patients. The current review summarizes clinical and preclinical studies with comprehensive insights into the mechanisms of action of omega-3 fatty acids (omega-3 FAs) in Systemic Lupus Erythematosus to provide an update on the negative and positive aspects of the intake of omega-3 FAs in SLE patients.
Collapse
Affiliation(s)
- Mina Salek
- Department of Nutrition, School of Public Health, 440827Iran University of Medical Sciences, Tehran, Iran
| | - Saeedeh Hosseini Hooshiar
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, 48462Kashan University of Medical Sciences, Kashan, Iran
| | - Mahsa Salek
- Department of Medicine, 201564Islamic Azad University Najafabad Branch, Najafabad, Iran
| | - Mohsen Poorebrahimi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, 48462Kashan University of Medical Sciences, Kashan, Iran
| | - Sadegh Jafarnejad
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, 48462Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
27
|
Shaikh SR, Virk R, Van Dyke TE. Potential Mechanisms by Which Hydroxyeicosapentaenoic Acids Regulate Glucose Homeostasis in Obesity. Adv Nutr 2022; 13:2316-2328. [PMID: 35709423 PMCID: PMC9776734 DOI: 10.1093/advances/nmac073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/16/2022] [Accepted: 06/13/2022] [Indexed: 01/29/2023] Open
Abstract
Dysregulation of glucose metabolism in response to diet-induced obesity contributes toward numerous complications, such as insulin resistance and hepatic steatosis. Therefore, there is a need to develop effective strategies to improve glucose homeostasis. In this review, we first discuss emerging evidence from epidemiological studies and rodent experiments that increased consumption of EPA (either as oily fish, or dietary/pharmacological supplements) may have a role in preventing impairments in insulin and glucose homeostasis. We then review the current evidence on how EPA-derived metabolites known as hydroxyeicosapentaenoic acids (HEPEs) may be a major mode of action by which EPA exerts its beneficial effects on glucose and lipid metabolism. Notably, cell culture and rodent studies show that HEPEs prevent fat accumulation in metabolic tissues through peroxisome proliferator activated receptor (PPAR)-mediated mechanisms. In addition, activation of the resolvin E1 pathway, either by administration of EPA in the diet or via intraperitoneal administration of resolvin E1, improves hyperglycemia, hyperinsulinemia, and liver steatosis through multiple mechanisms. These mechanisms include shifting immune cell phenotypes toward resolution of inflammation and preventing dysbiosis of the gut microbiome. Finally, we present the next steps for this line of research that will drive future precision randomized clinical trials with EPA and its downstream metabolites. These include dissecting the variables that drive heterogeneity in the response to EPA, such as the baseline microbiome profile and fatty acid status, circadian rhythm, genetic variation, sex, and age. In addition, there is a critical need to further investigate mechanisms of action for HEPEs and to establish the concentration of HEPEs in differing tissues, particularly in response to consumption of oily fish and EPA-enriched supplements.
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School
of Medicine, The University of North Carolina at Chapel Hill, Chapel
Hill, NC, USA
| | - Rafia Virk
- Department of Nutrition, Gillings School of Global Public Health and School
of Medicine, The University of North Carolina at Chapel Hill, Chapel
Hill, NC, USA
| | - Thomas E Van Dyke
- Center for Clinical and Translational Research, The Forsyth
Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection, and Immunity, Harvard School of
Dental Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Rosqvist F, Fridén M, Vessby J, Rorsman F, Lind L, Risérus U. Circulating fatty acids from high-throughput metabolomics platforms as potential biomarkers of dietary fatty acids. Clin Nutr 2022; 41:2637-2643. [PMID: 36308982 DOI: 10.1016/j.clnu.2022.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/27/2022] [Accepted: 10/08/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Some fatty acids, i.e. n-3 and n-6 polyunsaturated fatty acids (PUFA), from metabolomics platforms based on nuclear magnetic resonance imaging (NMR) or liquid chromatography mass-spectrometry (LC-MS) are suggested to reflect dietary exposure. NMR and LC-MS are both relatively fast and cheap, however few studies have investigated their validity. Linoleic acid (LA) and docosahexaenoic acid (DHA), measured using gas chromatography (GC), are established biomarkers of dietary n-6 and n-3 PUFA intake, respectively. OBJECTIVE To examine if circulating fatty acids derived from two commonly applied metabolomics platforms (using NMR and LC-MS) provide similar information compared to GC in two pooled population-based cohorts, one patient cohort, and in a randomized controlled trial (RCT). METHODS Spearman rank correlations were conducted between LA and DHA in cholesteryl esters (CE) from GC and whole serum/plasma LA and DHA from the metabolomics platforms in a pooled population-based cohort of men and women (n ˜ 1100) (primary analysis). Secondary correlation analyses included fatty acid classes such as n-3 PUFA, n-6 PUFA, saturated fatty acids (SFA), monounsaturated fatty acids (MUFA) and total PUFA. Additionally, correlations were investigated for LA, DHA and the five fatty acid classes in phospholipids (PL), triacylglycerols (TAG) and non-esterified fatty acids (NEFA) in a RCT of n = 60 as well as in a population with biopsy-verified non-alcoholic fatty liver disease (NAFLD) (n = 59). Misclassification was examined using cross-tabulation and visualized using alluvial plots. RESULTS Moderate to strong correlations (r = 0.51-0.81) were observed for LA and DHA in multiple lipid fractions in all cohorts using the NMR platform. For the pooled cohort, LA (r = 0.67, P < 0.0001) and DHA (r = 0.68, P < 0.0001) assessed in CE were strongly correlated with LA and DHA derived using NMR. Nearly half (49%) were correctly classified into their respective quartiles. Using LC-MS, only DHA (r = 0.44, P < 0.0001) demonstrated moderate correlations with DHA from GC. CONCLUSIONS Unless fatty acid data from GC analysis is available or feasible, NMR-based technology might be a better option than a LC-MS-based platform, at least for certain PUFA. This should be taken into account in future studies aiming to use circulating fatty acids as dietary biomarkers for the investigation of diet-disease relationships.
Collapse
Affiliation(s)
- Fredrik Rosqvist
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Michael Fridén
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Johan Vessby
- Department of Medical Sciences, Gastroenterology and Hepatology, Uppsala University, Uppsala, Sweden
| | - Fredrik Rorsman
- Department of Medical Sciences, Gastroenterology and Hepatology, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
29
|
Association between Omega-3 Index and Hyperglycemia Depending on Body Mass Index among Adults in the United States. Nutrients 2022; 14:nu14204407. [PMID: 36297090 PMCID: PMC9611386 DOI: 10.3390/nu14204407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/11/2022] [Accepted: 10/18/2022] [Indexed: 12/30/2022] Open
Abstract
There is inconsistency regarding the association between long-chain n-3 polyunsaturated fatty acids such as eicosapentaenoic acid (EPA; 20:5n3) and docosahexaenoic acid (DHA; 22:6n3) and the risk of type 2 diabetes. The present study aimed to investigate the association between the Omega-3 Index (erythrocyte EPA + DHA) and glycemic status as a function of body mass index (BMI). Cross-sectional data from routine clinical laboratory testing with a total of 100,572 people aged over 18 years and BMI ≥ 18.5 kg/m2 were included. Of the patients, 10% were hyperglycemic (fasting plasma glucose levels ≥ 126 mg/dL) and 24.7% were of normal weight, 35.0% were overweight, and 40.3% were obese. Odds ratios (ORs) of being hyperglycemic were inversely associated with the Omega-3 Index, but weakened as BMI increased. Thus, ORs (95% CI) comparing quintile 5 with quintile 1 were 0.54 (0.44-0.66) in the normal weight group, 0.70 (0.61-0.79) in the overweight group, and 0.74 (0.67-0.81) in the obese group. Similar patterns were seen for EPA and DHA separately. The present study suggested that a low Omega-3 Index is associated with a greater risk of disordered glucose metabolism and this is independent of BMI.
Collapse
|
30
|
Abstract
Information on the Omega-3 Index (O3I) in the United Kingdom (UK) are scarce. The UK-Biobank (UKBB) contains data on total plasma omega-3 polyunsaturated fatty acids (n3-PUFA%) and DHA% measured by NMR. The aim of our study was to create an equation to estimate the O3I (eO3I) from these data. We first performed an interlaboratory experiment with 250 random blood samples in which the O3I was measured in erythrocytes by gas chromatography, and total n3% and DHA% were measured in plasma by NMR. The best predictor of eO3I included both DHA% and a derived metric, the total n3%-DHA%. Together these explained 65% of the variability (r=0.832, p<0.0001). We then estimated the O3I in 117,108 UKBB subjects and correlated it with demographic and lifestyle variables in multivariable adjusted models. The mean (SD) eO3I was 5.58% (2.35%) this UKBB cohort. Several predictors were significantly correlated with eO3I (all p<0.0001). In general order of impact and with directionality (- = inverse, + = direct): oily-fish consumption (+), fish oil supplement use (+), female sex (+), older age (+), alcohol use (+), smoking (-), higher waist circumference and BMI (-), lower socioeconomic status and less education (-). Only 20.5% of eO3I variability could be explained by predictors investigated, and oily-fish consumption accounted for 7.0% of that. With the availability of the eO3I in the UKBB cohort we will be in a position to link risk for a variety of diseases with this commonly-used and well-documented marker of n3-PUFA biostatus.
Collapse
|
31
|
Sala-Vila A, Fleming J, Kris-Etherton P, Ros E. Impact of α-Linolenic Acid, the Vegetable ω-3 Fatty Acid, on Cardiovascular Disease and Cognition. Adv Nutr 2022; 13:1584-1602. [PMID: 35170723 PMCID: PMC9526859 DOI: 10.1093/advances/nmac016] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/31/2021] [Accepted: 02/11/2022] [Indexed: 01/28/2023] Open
Abstract
Given the evidence of the health benefits of plant-based diets and long-chain n-3 (ω-3) fatty acids, there is keen interest in better understanding the role of α-linolenic acid (ALA), a plant-derived n-3 fatty acid, on cardiometabolic diseases and cognition. There is increasing evidence for ALA largely based on its major food sources (i.e., walnuts and flaxseed); however, this lags behind our understanding of long-chain n-3 fatty acids. Meta-analyses of observational studies have shown that increasing dietary ALA is associated with a 10% lower risk of total cardiovascular disease and a 20% reduced risk of fatal coronary heart disease. Three randomized controlled trials (RCTs) [AlphaOmega trial, Prevención con Dieta Mediterránea (PREDIMED) trial, and Lyon Diet Heart Study] all showed benefits of diets high in ALA on cardiovascular-related outcomes, but the AlphaOmega trial, designed to specifically evaluate ALA effects, only showed a trend for benefit. RCTs have shown that dietary ALA reduced total cholesterol, LDL cholesterol, triglycerides, and blood pressure, and epidemiologic studies and some trials also have shown an anti-inflammatory effect of ALA, which collectively account for, in part, the cardiovascular benefits of ALA. A meta-analysis reported a trend toward diabetes risk reduction with both dietary and biomarker ALA. For metabolic syndrome and obesity, the evidence for ALA benefits is inconclusive. The role of ALA in cognition is in the early stages but shows promising evidence of counteracting cognitive impairment. Much has been learned about the health benefits of ALA and with additional research we will be better positioned to make strong evidence-based dietary recommendations for the reduction of many chronic diseases.
Collapse
Affiliation(s)
- Aleix Sala-Vila
- Fatty Acid Research Institute, Sioux Falls, SD, USA
- Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Jennifer Fleming
- Department of Nutritional Sciences, College of Health and Human Development, Pennsylvania State University, University Park, PA, USA
| | - Penny Kris-Etherton
- Department of Nutritional Sciences, College of Health and Human Development, Pennsylvania State University, University Park, PA, USA
| | - Emilio Ros
- Lipid Clinic, Endocrinology and Nutrition Service, Hospital Clínic, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
32
|
Bragg F, Kartsonaki C, Guo Y, Holmes M, Du H, Yu C, Pei P, Yang L, Jin D, Chen Y, Schmidt D, Avery D, Lv J, Chen J, Clarke R, Hill MR, Li L, Millwood IY, Chen Z. The role of NMR-based circulating metabolic biomarkers in development and risk prediction of new onset type 2 diabetes. Sci Rep 2022; 12:15071. [PMID: 36064959 PMCID: PMC9445062 DOI: 10.1038/s41598-022-19159-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/25/2022] [Indexed: 11/08/2022] Open
Abstract
Associations of circulating metabolic biomarkers with type 2 diabetes (T2D) and their added value for risk prediction are uncertain among Chinese adults. A case-cohort study included 882 T2D cases diagnosed during 8-years' follow-up and a subcohort of 789 participants. NMR-metabolomic profiling quantified 225 plasma biomarkers in stored samples taken at recruitment into the study. Cox regression yielded adjusted hazard ratios (HRs) for T2D associated with individual biomarkers, with a set of biomarkers incorporated into an established T2D risk prediction model to assess improvement in discriminatory ability. Mean baseline BMI (SD) was higher in T2D cases than in the subcohort (25.7 [3.6] vs. 23.9 [3.6] kg/m2). Overall, 163 biomarkers were significantly and independently associated with T2D at false discovery rate (FDR) controlled p < 0.05, and 138 at FDR-controlled p < 0.01. Branched chain amino acids (BCAA), apolipoprotein B/apolipoprotein A1, triglycerides in VLDL and medium and small HDL particles, and VLDL particle size were strongly positively associated with T2D (HRs 1.74-2.36 per 1 SD, p < 0.001). HDL particle size, cholesterol concentration in larger HDL particles and docosahexaenoic acid levels were strongly inversely associated with T2D (HRs 0.43-0.48, p < 0.001). With additional adjustment for plasma glucose, most associations (n = 147 and n = 129 at p < 0.05 and p < 0.01, respectively) remained significant. HRs appeared more extreme among more centrally adipose participants for apolipoprotein B/apolipoprotein A1, BCAA, HDL particle size and docosahexaenoic acid (p for heterogeneity ≤ 0.05). Addition of 31 selected biomarkers to an established T2D risk prediction model modestly, but significantly, improved risk discrimination (c-statistic 0.86 to 0.91, p < 0.001). In relatively lean Chinese adults, diverse metabolic biomarkers are associated with future risk of T2D and can help improve established risk prediction models.
Collapse
Affiliation(s)
- Fiona Bragg
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yu Guo
- Fuwai Hospital Chinese Academy of Medical Sciences, National Center for Cardiovascular Diseases, Beijing, China
| | - Michael Holmes
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Huaidong Du
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Pei Pei
- Chinese Academy of Medical Sciences, Beijing, 102308, China
| | - Ling Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Donghui Jin
- Hunan Centre for Disease Control and Prevention, Furong Mid Road, Changsha, Hunan, China
| | - Yiping Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dan Schmidt
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
| | - Daniel Avery
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Robert Clarke
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
| | - Michael R Hill
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Iona Y Millwood
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, BDI Building, Old Road Campus, Oxford, OX3 7LF, UK.
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
33
|
Uusitupa M. Sources of animal proteins and type 2 diabetes risk - decreasing the consumption of red meat is well grounded. Diabetes Res Clin Pract 2022; 191:110072. [PMID: 36067914 DOI: 10.1016/j.diabres.2022.110072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 08/31/2022] [Indexed: 11/22/2022]
Affiliation(s)
- Matti Uusitupa
- Professor Emeritus, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Finland
| |
Collapse
|
34
|
Eichelmann F, Sellem L, Wittenbecher C, Jäger S, Kuxhaus O, Prada M, Cuadrat R, Jackson KG, Lovegrove JA, Schulze MB. Deep Lipidomics in Human Plasma: Cardiometabolic Disease Risk and Effect of Dietary Fat Modulation. Circulation 2022; 146:21-35. [PMID: 35422138 PMCID: PMC9241667 DOI: 10.1161/circulationaha.121.056805] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND In blood and tissues, dietary and endogenously generated fatty acids (FAs) occur in free form or as part of complex lipid molecules that collectively represent the lipidome of the respective tissue. We assessed associations of plasma lipids derived from high-resolution lipidomics with incident cardiometabolic diseases and subsequently tested if the identified risk-associated lipids were sensitive to dietary fat modification. METHODS The EPIC Potsdam cohort study (European Prospective Investigation into Cancer and Nutrition) comprises 27 548 participants recruited within an age range of 35 to 65 years from the general population around Potsdam, Germany. We generated 2 disease-specific case cohorts on the basis of a fixed random subsample (n=1262) and all respective cohort-wide identified incident primary cardiovascular disease (composite of fatal and nonfatal myocardial infarction and stroke; n=551) and type 2 diabetes (n=775) cases. We estimated the associations of baseline plasma concentrations of 282 class-specific FA abundances (calculated from 940 distinct molecular species across 15 lipid classes) with the outcomes in multivariable-adjusted Cox models. We tested the effect of an isoenergetic dietary fat modification on risk-associated lipids in the DIVAS randomized controlled trial (Dietary Intervention and Vascular Function; n=113). Participants consumed either a diet rich in saturated FAs (control), monounsaturated FAs, or a mixture of monounsaturated and n-6 polyunsaturated FAs for 16 weeks. RESULTS Sixty-nine lipids associated (false discovery rate<0.05) with at least 1 outcome (both, 8; only cardiovascular disease, 49; only type 2 diabetes, 12). In brief, several monoacylglycerols and FA16:0 and FA18:0 in diacylglycerols were associated with both outcomes; cholesteryl esters, free fatty acids, and sphingolipids were largely cardiovascular disease specific; and several (glycero)phospholipids were type 2 diabetes specific. In addition, 19 risk-associated lipids were affected (false discovery rate<0.05) by the diets rich in unsaturated dietary FAs compared with the saturated fat diet (17 in a direction consistent with a potential beneficial effect on long-term cardiometabolic risk). For example, the monounsaturated FA-rich diet decreased diacylglycerol(FA16:0) by 0.4 (95% CI, 0.5-0.3) SD units and increased triacylglycerol(FA22:1) by 0.5 (95% CI, 0.4-0.7) SD units. CONCLUSIONS We identified several lipids associated with cardiometabolic disease risk. A subset was beneficially altered by a dietary fat intervention that supports the substitution of dietary saturated FAs with unsaturated FAs as a potential tool for primary disease prevention.
Collapse
Affiliation(s)
- Fabian Eichelmann
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (F.E., C.W., S.J., O.K., M.P., R.C., M.B.S.).,German Center for Diabetes Research (DZD), Neuherberg (F.E., S.J., O.K., M.P., R.C., M.B.S.)
| | - Laury Sellem
- Hugh Sinclair Unit of Human Nutrition, and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Science, University of Reading, United Kingdom (L.S., K.G.J., J.A.L.)
| | - Clemens Wittenbecher
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (F.E., C.W., S.J., O.K., M.P., R.C., M.B.S.).,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA (C.W.)
| | - Susanne Jäger
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (F.E., C.W., S.J., O.K., M.P., R.C., M.B.S.).,German Center for Diabetes Research (DZD), Neuherberg (F.E., S.J., O.K., M.P., R.C., M.B.S.)
| | - Olga Kuxhaus
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (F.E., C.W., S.J., O.K., M.P., R.C., M.B.S.).,German Center for Diabetes Research (DZD), Neuherberg (F.E., S.J., O.K., M.P., R.C., M.B.S.)
| | - Marcela Prada
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (F.E., C.W., S.J., O.K., M.P., R.C., M.B.S.).,German Center for Diabetes Research (DZD), Neuherberg (F.E., S.J., O.K., M.P., R.C., M.B.S.)
| | - Rafael Cuadrat
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (F.E., C.W., S.J., O.K., M.P., R.C., M.B.S.).,German Center for Diabetes Research (DZD), Neuherberg (F.E., S.J., O.K., M.P., R.C., M.B.S.)
| | - Kim G. Jackson
- Hugh Sinclair Unit of Human Nutrition, and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Science, University of Reading, United Kingdom (L.S., K.G.J., J.A.L.)
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Science, University of Reading, United Kingdom (L.S., K.G.J., J.A.L.)
| | - Matthias B. Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (F.E., C.W., S.J., O.K., M.P., R.C., M.B.S.).,German Center for Diabetes Research (DZD), Neuherberg (F.E., S.J., O.K., M.P., R.C., M.B.S.).,Institute of Nutritional Science, University of Potsdam, Germany (M.B.S.)
| |
Collapse
|
35
|
Martínez R, Mesas C, Guzmán A, Galisteo M, López-Jurado M, Prados J, Melguizo C, Bermúdez F, Porres JM. Bioavailability and biotransformation of linolenic acid from basil seed oil as a novel source of omega-3 fatty acids tested on a rat experimental model. Food Funct 2022; 13:7614-7628. [PMID: 35731538 DOI: 10.1039/d2fo00672c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Basil is an aromatic herb with a high concentration of bioactive compounds. The oil extracted from its seeds is a good source of α-linolenic acid (ALA) and also provides substantial amounts of linoleic acid (LA). This study aimed to test the bioavailability of the oil derived from basil seeds and its effects on different physiological parameters using 7-15% dietary inclusion levels. Furthermore, the assimilation of LA and ALA and their transformation in long-chain polyunsaturated fatty acids (LC-PUFAs) have been studied. Digestive utilization of total fat from basil seed oil (BSO) was high and similar to that of olive oil used as a control. Consumption of BSO resulted in increased LA and ALA levels of the plasma, liver, and erythrocyte membrane. In addition, the transformation of LA to arachidonic acid (ARA) was decreased by the high dietary intake of ALA which redirected the pathway of the Δ-6 desaturase enzyme towards the transformation of ALA into eicosapentaenoic acid (EPA). No alterations of hematological and plasma biochemical parameters were found for the 7 and 10% dietary inclusion levels of BSO, whereas a decrease in the platelet count and an increase in total- and HDL-cholesterol as well as plasma alkaline phosphatase (ALP) were found for a 15% BSO dose. In conclusion, BSO is a good source of ALA to be transformed into EPA and decrease the precursor of the pro-inflammatory molecule ARA. This effect on the levels of EPA in different tissues offers potential for its use as a dietary supplement, novel functional food, or a constituent of nutraceutical formulations to treat different pathologies.
Collapse
Affiliation(s)
- Rosario Martínez
- Cellbitec S.L., N.I.F. B04847216, Scientific Headquarters of the Almería Technology Park, Universidad de Almería, 04128 La Cañada, Almería, Spain. .,Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Biomedical Research Center (CIBM), Universidad de Granada, 18100 Granada, Spain.
| | - Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), University of Granada, 18100 Granada, Spain. .,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Ana Guzmán
- Cellbitec S.L., N.I.F. B04847216, Scientific Headquarters of the Almería Technology Park, Universidad de Almería, 04128 La Cañada, Almería, Spain. .,Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), University of Granada, 18100 Granada, Spain.
| | - Milagros Galisteo
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071 Granada, Spain.
| | - María López-Jurado
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Biomedical Research Center (CIBM), Universidad de Granada, 18100 Granada, Spain.
| | - José Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), University of Granada, 18100 Granada, Spain. .,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain.,Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Center (CIBM), University of Granada, 18100 Granada, Spain. .,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain.,Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain
| | - Francisco Bermúdez
- Cellbitec S.L., N.I.F. B04847216, Scientific Headquarters of the Almería Technology Park, Universidad de Almería, 04128 La Cañada, Almería, Spain.
| | - Jesus M Porres
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Biomedical Research Center (CIBM), Universidad de Granada, 18100 Granada, Spain.
| |
Collapse
|
36
|
Bhatnagar D, Bhatnagar P. Nutrition and its impact on cardiovascular disease. Curr Opin Lipidol 2022; 33:211-212. [PMID: 35695618 DOI: 10.1097/mol.0000000000000816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Deepak Bhatnagar
- Division of Diabetes, Endocrinology and Gastroenterology, University of Manchester, Manchester
| | - Prachi Bhatnagar
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
37
|
Bragg F, Trichia E, Aguilar-Ramirez D, Bešević J, Lewington S, Emberson J. Predictive value of circulating NMR metabolic biomarkers for type 2 diabetes risk in the UK Biobank study. BMC Med 2022; 20:159. [PMID: 35501852 PMCID: PMC9063288 DOI: 10.1186/s12916-022-02354-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/28/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Effective targeted prevention of type 2 diabetes (T2D) depends on accurate prediction of disease risk. We assessed the role of metabolomic profiling in improving T2D risk prediction beyond conventional risk factors. METHODS Nuclear magnetic resonance (NMR) metabolomic profiling was undertaken on baseline plasma samples in 65,684 UK Biobank participants without diabetes and not taking lipid-lowering medication. Among a subset of 50,519 participants with data available on all relevant co-variates (sociodemographic characteristics, parental history of diabetes, lifestyle-including dietary-factors, anthropometric measures and fasting time), Cox regression yielded adjusted hazard ratios for the associations of 143 individual metabolic biomarkers (including lipids, lipoproteins, fatty acids, amino acids, ketone bodies and other low molecular weight metabolic biomarkers) and 11 metabolic biomarker principal components (PCs) (accounting for 90% of the total variance in individual biomarkers) with incident T2D. These 11 PCs were added to established models for T2D risk prediction among the full study population, and measures of risk discrimination (c-statistic) and reclassification (continuous net reclassification improvement [NRI], integrated discrimination index [IDI]) were assessed. RESULTS During median 11.9 (IQR 11.1-12.6) years' follow-up, after accounting for multiple testing, 90 metabolic biomarkers showed independent associations with T2D risk among 50,519 participants (1211 incident T2D cases) and 76 showed associations after additional adjustment for HbA1c (false discovery rate controlled p < 0.01). Overall, 8 metabolic biomarker PCs were independently associated with T2D. Among the full study population of 65,684 participants, of whom 1719 developed T2D, addition of PCs to an established risk prediction model, including age, sex, parental history of diabetes, body mass index and HbA1c, improved T2D risk prediction as assessed by the c-statistic (increased from 0.802 [95% CI 0.791-0.812] to 0.830 [0.822-0.841]), continuous NRI (0.44 [0.38-0.49]) and relative (15.0% [10.5-20.4%]) and absolute (1.5 [1.0-1.9]) IDI. More modest improvements were observed when metabolic biomarker PCs were added to a more comprehensive established T2D risk prediction model additionally including waist circumference, blood pressure and plasma lipid concentrations (c-statistic, 0.829 [0.819-0.838] to 0.837 [0.831-0.848]; continuous NRI, 0.22 [0.17-0.28]; relative IDI, 6.3% [4.1-9.8%]; absolute IDI, 0.7 [0.4-1.1]). CONCLUSIONS When added to conventional risk factors, circulating NMR-based metabolic biomarkers modestly enhanced T2D risk prediction.
Collapse
Affiliation(s)
- Fiona Bragg
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK. .,Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK.
| | - Eirini Trichia
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Diego Aguilar-Ramirez
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Jelena Bešević
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Sarah Lewington
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK.,Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK.,UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Jonathan Emberson
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK.,Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| |
Collapse
|
38
|
Lai HT, Imamura F, Korat AVA, Murphy RA, Tintle N, Bassett JK, Chen J, Kröger J, Chien KL, Senn M, Wood AC, Forouhi NG, Schulze MB, Harris WS, Vasan RS, Hu F, Giles GG, Hodge A, Djousse L, Brouwer IA, Qian F, Sun Q, Wu JH, Marklund M, Lemaitre RN, Siscovick DS, Fretts AM, Shadyab AH, Manson JE, Howard BV, Robinson JG, Wallace RB, Wareham NJ, Chen YDI, Rotter JI, Tsai MY, Micha R, Mozaffarian D. Trans Fatty Acid Biomarkers and Incident Type 2 Diabetes: Pooled Analysis of 12 Prospective Cohort Studies in the Fatty Acids and Outcomes Research Consortium (FORCE). Diabetes Care 2022; 45:854-863. [PMID: 35142845 PMCID: PMC9114723 DOI: 10.2337/dc21-1756] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/10/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Trans fatty acids (TFAs) have harmful biologic effects that could increase the risk of type 2 diabetes (T2D), but evidence remains uncertain. We aimed to investigate the prospective associations of TFA biomarkers and T2D by conducting an individual participant-level pooled analysis. RESEARCH DESIGN AND METHODS We included data from an international consortium of 12 prospective cohorts and nested case-control studies from six nations. TFA biomarkers were measured in blood collected between 1990 and 2008 from 25,126 participants aged ≥18 years without prevalent diabetes. Each cohort conducted de novo harmonized analyses using a prespecified protocol, and findings were pooled using inverse-variance weighted meta-analysis. Heterogeneity was explored by prespecified between-study and within-study characteristics. RESULTS During a mean follow-up of 13.5 years, 2,843 cases of incident T2D were identified. In multivariable-adjusted pooled analyses, no significant associations with T2D were identified for trans/trans-18:2, relative risk (RR) 1.09 (95% CI 0.94-1.25); cis/trans-18:2, 0.89 (0.73-1.07); and trans/cis-18:2, 0.87 (0.73-1.03). Trans-16:1n-9, total trans-18:1, and total trans-18:2 were inversely associated with T2D (RR 0.81 [95% CI 0.67-0.99], 0.86 [0.75-0.99], and 0.84 [0.74-0.96], respectively). Findings were not significantly different according to prespecified sources of potential heterogeneity (each P ≥ 0.1). CONCLUSIONS Circulating individual trans-18:2 TFA biomarkers were not associated with risk of T2D, while trans-16:1n-9, total trans-18:1, and total trans-18:2 were inversely associated. Findings may reflect the influence of mixed TFA sources (industrial vs. natural ruminant), a general decline in TFA exposure due to policy changes during this period, or the relatively limited range of TFA levels.
Collapse
Affiliation(s)
- Heidi T.M. Lai
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
- Department of Primary Care and Public Health, Imperial College London, London, U.K
| | - Fumiaki Imamura
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | - Andres V. Ardisson Korat
- Department of Nutrition and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Rachel A. Murphy
- School of Population & Public Health, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nathan Tintle
- Department of Mathematics and Statistics, Dordt University, Sioux Center, IA
- Fatty Acid Research Institute, Sioux Falls, SD
| | - Julie K. Bassett
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - Jiaying Chen
- Division of Aging, Brigham and Women's Hospital, Boston, MA
| | - Janine Kröger
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei City, Republic of China
| | - Mackenzie Senn
- U.S. Department of Agriculture/Agriculture Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Alexis C. Wood
- U.S. Department of Agriculture/Agriculture Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | - Matthias B. Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - William S. Harris
- Fatty Acid Research Institute, Sioux Falls, SD
- Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD
| | - Ramachandran S. Vasan
- Boston University School of Medicine, Boston, MA
- The Framingham Heart Study, Framingham, MA
| | - Frank Hu
- Department of Nutrition and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Graham G. Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Parkville, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Allison Hodge
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - Luc Djousse
- Divisions of Aging, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Ingeborg A. Brouwer
- Department of Health Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Frank Qian
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Qi Sun
- Department of Nutrition and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Jason H.Y. Wu
- The George Institute for Global Health, the Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Matti Marklund
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
- The George Institute for Global Health, the Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Rozenn N. Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | | | - Amanda M. Fretts
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
| | - Aladdin H. Shadyab
- Family Medicine and Public Health, School of Medicine, University of California, San Diego, La Jolla, CA
| | - JoAnn E. Manson
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Barbara V. Howard
- Georgetown University Medical Center, Georgetown University, Hyattsville, MD
| | | | | | - Nick J. Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Renata Micha
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | | |
Collapse
|
39
|
Jackson KH, Van Guilder GP, Tintle N, Tate B, McFadden J, Perry CA. Plasma fatty acid responses to a calorie-restricted, DASH-style diet with lean beef. Prostaglandins Leukot Essent Fatty Acids 2022; 179:102413. [PMID: 35395436 DOI: 10.1016/j.plefa.2022.102413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Plasma fatty acid (FA) levels are used as biomarkers of health outcomes and nutritional intake. METHODS This was an exploratory analysis of the plasma FA profile from a parallel-designed, controlled-feeding study in older, obese adults (females, n = 17; males, n = 11) consuming a DASH-based diet with two levels of lean beef (3oz and 6oz per day). Plasma FA levels (as percent composition) were measured by gas chromatography from five timepoints over the 12-week intervention. The primary plasma FA change patterns modeled were sustained (initial change to 'new normal') or homeostatic (initial change, then return toward original baseline). RESULTS The study diet was low in fat (< 60 g/d), especially polyunsaturated FAs (PUFAs; < 5 g/d), compared to the average American diet of obese individuals as described by a nationally representative sample. Participants lost ∼6% of body mass and lowered plasma fasting triglyceride levels by ∼9% over the course of the study. With strong to very strong strength of evidence, the individual FAs displaying a sustained response were C16:1n7t, C18:1n9, C20:1n9, and C18:2n6, and homeostatic response, C18:0, 24:0, C24:1n9, C18:3n6, C20:4n6, and C22:6n3 (Ps < 0.0021, Bonferroni-adjusted). The data suggested that systematic changes in both the PUFA and de novo lipogenesis pathways occurred. CONCLUSIONS Diet can affect plasma FA changes both due to nutritional composition and by affecting metabolic processes.
Collapse
Affiliation(s)
- Kristina Harris Jackson
- OmegaQuant Analytics, LLC. Sioux Falls, SD 57106 USA; Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105 USA
| | - Gary P Van Guilder
- Exercise & Sport Science, Western Colorado University, Gunnison, CO 81230 USA
| | - Nathan Tintle
- Fatty Acid Research Institute (FARI), Sioux Falls, SD 57106 USA
| | - Brianna Tate
- Department of Animal Science, Cornell University, Ithaca, NY 14853 USA
| | - Joseph McFadden
- Department of Animal Science, Cornell University, Ithaca, NY 14853 USA
| | - Cydne A Perry
- Department of Applied Health Science, Indiana University School of Public Health, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
40
|
Chiva-Blanch G, Giró O, Cofán M, Calle-Pascual AL, Delgado E, Gomis R, Jiménez A, Franch-Nadal J, Rojo Martínez G, Ortega E. Low Percentage of Vegetable Fat in Red Blood Cells Is Associated with Worse Glucose Metabolism and Incidence of Type 2 Diabetes. Nutrients 2022; 14:nu14071368. [PMID: 35405981 PMCID: PMC9002701 DOI: 10.3390/nu14071368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022] Open
Abstract
The identification of nutritional patterns associated with the development of type 2 diabetes (T2D) might help lead the way to a more efficient and personalized nutritional intervention. Our study is aimed at evaluating the association between fatty acids (FA) in red blood cell (RBC) membranes, as a quantitative biomarker of regular dietary fat intake, and incident type 2 diabetes in a Spanish population. We included 1032 adult Spaniards (57% women, age 49 ± 15 years, 18% prediabetes), without diabetes at study entry, from the Di@bet.es cohort. Incident diabetes was diagnosed at the end of the study follow-up. The FA percentage in RBC was determined at baseline by gas chromatography. Participants were followed on average 7.5 ± 0.6 years. Lower percentages of linoleic acid (LA), α-linolenic (ALA), and eicosapentaenoic acid (EPA), and higher percentages of docosahexaenoic acid (DHA) in RBC membranes were associated, independently of classical risk factors, with worse glucose metabolism at the end of the study follow-up. In addition, higher percentages of ALA and EPA, and moderate percentages of DHA, were associated with lower risk of diabetes. No significant associations were found for LA and diabetes risk. Dietary patterns rich in vegetables are independently associated with lower risk of both deterioration of glucose regulation and incident diabetes, and should be reinforced for the prevention of diabetes.
Collapse
Affiliation(s)
- Gemma Chiva-Blanch
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute-IDIBAPS, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; (G.C.-B.); (O.G.); (M.C.); (R.G.); (A.J.)
- Spanish Biomedical Research Network in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Oriol Giró
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute-IDIBAPS, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; (G.C.-B.); (O.G.); (M.C.); (R.G.); (A.J.)
- Spanish Biomedical Research Network in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Montserrat Cofán
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute-IDIBAPS, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; (G.C.-B.); (O.G.); (M.C.); (R.G.); (A.J.)
- Spanish Biomedical Research Network in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alfonso L. Calle-Pascual
- Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.L.C.-P.); (J.F.-N.); (G.R.M.)
- Department of Endocrinology and Nutrition, San Carlos University Hospital of Madrid, 28040 Madrid, Spain
| | - Elías Delgado
- Spanish Biomedical Research Network in Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Department of Endocrinology and Nutrition, Central University Hospital of Asturias, University of Oviedo, Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Ramon Gomis
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute-IDIBAPS, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; (G.C.-B.); (O.G.); (M.C.); (R.G.); (A.J.)
- Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.L.C.-P.); (J.F.-N.); (G.R.M.)
| | - Amanda Jiménez
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute-IDIBAPS, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; (G.C.-B.); (O.G.); (M.C.); (R.G.); (A.J.)
- Spanish Biomedical Research Network in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Josep Franch-Nadal
- Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.L.C.-P.); (J.F.-N.); (G.R.M.)
- EAP Raval Sud, Catalan Institute of Health, GEDAPS Network, Primary Care, Research Support Unit (IDIAP-Jordi Gol Foundation), 08001 Barcelona, Spain
| | - Gemma Rojo Martínez
- Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.L.C.-P.); (J.F.-N.); (G.R.M.)
- Biomedical Research Institute of Malaga (IBIMA), Endocrinology and Nutrition Department, Regional University Hospital of Malaga, 29010 Malaga, Spain
| | - Emilio Ortega
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute-IDIBAPS, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; (G.C.-B.); (O.G.); (M.C.); (R.G.); (A.J.)
- Spanish Biomedical Research Network in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
41
|
Zhuang P, Liu X, Li Y, Li H, Zhang L, Wan X, Wu Y, Zhang Y, Jiao J. Circulating Fatty Acids and Genetic Predisposition to Type 2 Diabetes: Gene-Nutrient Interaction Analysis. Diabetes Care 2022; 45:564-575. [PMID: 35089324 DOI: 10.2337/dc21-2048] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To assess the relationship of circulating fatty acids (FA) with risk of type 2 diabetes (T2D) and potential interactions with genetic risk. RESEARCH DESIGN AND METHODS A total of 95,854 participants with complete data on plasma FA from the UK Biobank were enrolled between 2006 and 2010 and were followed up to the end of 2020. Plasma concentrations of saturated fatty acids (SFA), monounsaturated fatty acids (MUFA), and polyunsaturated fatty acids (PUFA) were analyzed by a high-throughput nuclear magnetic resonance-based biomarker profiling platform. The genetic risk scores (GRS) were calculated on the basis of 424 variants associated with T2D. Pathway-specific GRS were calculated based on robust clusters of T2D loci. RESULTS There were 3,052 instances of T2D documented after an average follow-up of 11.6 years. Plasma concentrations of SFA and MUFA were positively associated with T2D risk, while plasma PUFA were inversely associated. After adjustment for major risk factors, hazard ratios (95% CI) of T2D for 1-SD increment were 1.03 (1.02-1.04) for SFA, 1.03 (1.02-1.05) for MUFA, 0.62 (0.56-0.68) for PUFA, 0.67 (0.61-0.73) for n-6 PUFA, 0.90 (0.85-0.95) for n-3 PUFA, and 1.01 (0.98-1.04) for n-6-to-n-3 ratio. Plasma MUFA had significant interactions with the overall GRS and GRS for proinsulin and liver/lipid clusters on T2D risk. The protective associations of n-3 PUFA with T2D risk were weaker among individuals with higher obesity GRS (P interaction = 0.040) and liver/lipid GRS (P interaction = 0.012). Additionally, increased plasma n-3 PUFA concentration was associated with more reductions in T2D risk among participants carrying more docosapentaenoic acid-associated alleles (P interaction = 0.007). CONCLUSIONS Plasma concentrations of SFA and MUFA were associated with a higher T2D risk, whereas plasma PUFA and n-6 and n-3 PUFA were related to a lower risk. Circulating MUFA and n-3 PUFA had significant interactions with genetic predisposition to T2D and FA-associated variants.
Collapse
Affiliation(s)
- Pan Zhuang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaohui Liu
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yin Li
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haoyu Li
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lange Zhang
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xuzhi Wan
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuqi Wu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yu Zhang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Fuli Institute of Food Science, Zhejiang University, Hangzhou, Zhejiang, China.,Ningbo Research Institute, Zhejiang University, Ningbo, Zhejiang, China
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW In contrast to other saturated fatty acids, very long-chain saturated fatty acids (VLSFAs) have received limited attention The purpose of this review is to summarize the associations of VLSFAs, including arachidic acid, behenic acid, and lignoceric acid, with cardiovascular disease outcomes and type 2 diabetes; to discuss the findings implications; and to call for future studies of the VLSFAs. RECENT FINDINGS Increased levels of circulating VLSFAs have been found associated with lower risks of incident heart failure, atrial fibrillation, coronary heart disease, mortality, sudden cardiac arrest, type 2 diabetes, and with better aging. The VLSFA associations are paralleled by associations of plasma ceramide and sphingomyelin species carrying a VLSFA with lower risks of heart failure, atrial fibrillation, and mortality, suggesting VLSFAs affect the biological activity of ceramides and sphingomyelins thereby impacting health. For diabetes, there is no such parallel and the associations of VLSFAs with diabetes may be confounded or mediated by triglyceride and circulating palmitic acid, possible biomarkers of de novo lipogenesis. SUMMARY In many ways, the epidemiology has preceded our knowledge of VLSFAs biology. We hope this review will spur interest from the research community in further studying these potentially beneficial fatty acids.
Collapse
Affiliation(s)
- Rozenn N. Lemaitre
- University of Washington, Department of Medicine, Cardiovascular Health Research Unit, Seattle, Washington
| | | |
Collapse
|
43
|
Bragg F, Kartsonaki C, Guo Y, Holmes M, Du H, Yu C, Pei P, Yang L, Jin D, Chen Y, Schmidt D, Avery D, Lv J, Chen J, Clarke R, Hill M, Li L, Millwood I, Chen Z. Circulating Metabolites and the Development of Type 2 Diabetes in Chinese Adults. Diabetes Care 2022; 45:477-480. [PMID: 34848488 PMCID: PMC7612375 DOI: 10.2337/dc21-1415] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/09/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To assess prospective associations of circulating metabolites with the risk of type 2 diabetes (T2D) among Chinese adults. RESEARCH DESIGN AND METHODS A case-cohort study within the 8-year prospective China Kadoorie Biobank comprised 882 participants with incident T2D and 789 subcohort participants. Nuclear magnetic resonance metabolomic profiling quantified 225 metabolites in stored baseline plasma samples. Cox regression related individual metabolites with T2D risk, adjusting for potential confounders and fasting time. RESULTS After correction for multiple testing, 163 metabolites were significantly associated with the risk of T2D (P < 0.05). There were strong positive associations of VLDL particle size, the ratio of apolipoprotein B to apolipoprotein A-1, branched-chain amino acids, glucose, and triglycerides with T2D, and inverse associations of HDL-cholesterol, HDL particle size, and relative n-3 and saturated fatty acid concentrations. CONCLUSIONS In Chinese adults, metabolites across diverse pathways were independently associated with T2D risk, providing valuable etiological insights and potential to improve T2D risk prediction.
Collapse
Affiliation(s)
- Fiona Bragg
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K.,Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K.,Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Yu Guo
- Fuwai Hospital Chinese Academy of Medical Sciences, National Center for Cardiovascular Diseases, Beijing, China
| | - Michael Holmes
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K.,Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Huaidong Du
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K.,Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Canqing Yu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China.,Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Pei Pei
- Chinese Academy of Medical Sciences, Beijing, China
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K.,Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Donghui Jin
- Hunan Centre for Disease Control and Prevention, Changsha, Hunan, China
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K.,Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Dan Schmidt
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Daniel Avery
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Jun Lv
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China.,Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Robert Clarke
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K.,Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Michael Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Liming Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China.,Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Iona Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K.,Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K.,Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| |
Collapse
|
44
|
Metabolite Signature of Physical Activity and the Risk of Type 2 Diabetes in 7271 Men. Metabolites 2022; 12:metabo12010069. [PMID: 35050191 PMCID: PMC8779070 DOI: 10.3390/metabo12010069] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 12/24/2022] Open
Abstract
Large population-based studies investigating the association of physical activity (PA) with the metabolite signature contribute significantly to the understanding of the effects of PA on metabolic pathways associated with the risk of type 2 diabetes. Our study included 8749 Finnish men without diabetes at baseline recruited from the Metabolic Syndrome in Men (METSIM) cohort. We used a questionnaire to measure leisure-time PA. Metabolites were measured in 7271 men as a part of Metabolon’s untargeted Discovery HD4 platform using ultrahigh-performance liquid chromatography–tandem mass spectrometry. We found 198 metabolites significantly associated with PA. Several of these metabolites were novel including especially steroids, amino acids, imidazoles, carboxylic acids, and hydroxy acids. Increased PA was significantly associated with high levels of choline plasmalogens, lysophosphatidylcholines, polyunsaturated fatty acids, carotenoids, long chain acylcarnitines, imidazoles, bilirubins, aryl sulfates, hydroxy acids, indolepropionate, and indolelactate. Several of these metabolites have been previously associated with a decreased risk of type 2 diabetes and with a healthy diet. Our population-based study shows that the metabolite signature of increased PA includes multiple metabolic pathways and is associated with better adherence to a healthy lifestyle.
Collapse
|
45
|
Takahashi H, Sano H, Hayashi Y, Nishimura R. Eicosapentaenoic acid/arachidonic acid ratio in Japanese junior high school students. Pediatr Int 2022; 64:e14898. [PMID: 34170595 DOI: 10.1111/ped.14898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 06/10/2021] [Accepted: 06/23/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND This epidemiological study investigated the eicosapentaenoic acid/arachidonic acid ratio (EAR) and docosahexaenoic acid/ arachidonic acid ratio (DAR) in ninth-graders in Japan. METHODS A total of 175 ninth-grade children from Tsunan Town (115 boys and 60 girls) were enrolled in the study between 2015 and 2017. All participants were assessed during annual health checkups to determine their EAR and DAR. All data were compared between boys and girls using the Mann-Whitney test. Spearman's rank correlation coefficient was used to describe the strengths of correlations between the EAR, DAR, and relevant parameters. RESULTS The median (minimum, maximum) EAR and DAR of all participants were 0.13 (0.05, 0.46) and 0.47 (0.25, 0.84), respectively. There was no significant difference in the EAR between boys and girls at 0.12 (0.05, 0.46) and 0.13 (0.06, 0.27) (P = 0.375), respectively. The DAR was not significantly different (P = 0.125). There was a significant positive correlation between EAR and triglyceride, body mass index, and homeostasis model assessment but a significantly negative correlation with HbA1c (r = -0.209, P = 0.006). In contrast, there was no significant correlation between DAR and any parameters. CONCLUSIONS This is the first epidemiological study investigating the EAR using a population-based cohort of Japanese adolescents. The EAR in boys/girls in Japan is 0.12 (0.05, 0.46)/0.13 (0.06, 0.27). The study results suggest that children with a healthy diet may generally have a lower average blood glucose level.
Collapse
Affiliation(s)
- Hiroshi Takahashi
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hironari Sano
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Tsunan Town Hospital, Niigata, Japan
| | | | - Rimei Nishimura
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
46
|
Beneficial effects of eicosapentaenoic acid on the metabolic profile of obese female mice entails upregulation of HEPEs and increased abundance of enteric Akkermansia muciniphila. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159059. [PMID: 34619367 PMCID: PMC8627244 DOI: 10.1016/j.bbalip.2021.159059] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/30/2021] [Accepted: 09/24/2021] [Indexed: 01/03/2023]
Abstract
Eicosapentaenoic acid (EPA) ethyl esters are of interest given their clinical approval for lowering circulating triglycerides and cardiometabolic disease risk. EPA ethyl esters prevent metabolic complications driven by a high fat diet in male mice; however, their impact on female mice is less studied. Herein, we first investigated how EPA influences the metabolic profile of female C57BL/6J mice consuming a high fat diet. EPA lowered murine fat mass accumulation, potentially through increased biosynthesis of 8-hydroxyeicosapentaenoic acid (HEPE), as revealed by mass spectrometry and cell culture studies. EPA also reversed the effects of a high fat diet on circulating levels of insulin, glucose, and select inflammatory/metabolic markers. Next, we studied if the improved metabolic profile of obese mice consuming EPA was associated with a reduction in the abundance of key gut Gram-negative bacteria that contribute toward impaired glucose metabolism. Using fecal 16S-ribosomal RNA gene sequencing, we found EPA restructured the gut microbiota in a time-dependent manner but did not lower the levels of key Gram-negative bacteria. Interestingly, EPA robustly increased the abundance of the Gram-negative Akkermansia muciniphila, which controls glucose homeostasis. Finally, predictive functional profiling of microbial communities revealed EPA-mediated reversal of high fat diet-associated changes in a wide range of genes related to pathways such as Th-17 cell differentiation and PI3K-Akt signaling. Collectively, these results show that EPA ethyl esters prevent some of the deleterious effects of a high fat diet in female mice, which may be mediated mechanistically through 8-HEPE and the upregulation of intestinal Akkermansia muciniphila.
Collapse
|
47
|
de Mello VD, Selander T, Lindström J, Tuomilehto J, Uusitupa M, Kaarniranta K. Serum Levels of Plasmalogens and Fatty Acid Metabolites Associate with Retinal Microangiopathy in Participants from the Finnish Diabetes Prevention Study. Nutrients 2021; 13:nu13124452. [PMID: 34960007 PMCID: PMC8703764 DOI: 10.3390/nu13124452] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy (DR) is the most common microvascular complication of diabetes, and retinal microaneurysms (MA) are one of the first detected abnormalities associated with DR. We recently showed elevated serum triglyceride levels to be associated with the development of MA in the Finnish Diabetes Prevention Study (DPS). The purpose of this metabolomics study was to assess whether serum fatty acid (FA) composition, plasmalogens, and low-grade inflammation may enhance or decrease the risk of MA. Originally, the DPS included 522 individuals (mean 55 years old, range 40-64 years) with impaired glucose tolerance who were randomized into an intervention (n = 265) or control group (n = 257). The intervention lasted for a median of four years (active period), after which annual follow-up visits were conducted. At least five years after stopping the intervention phase of DPS, participants classified as MA negative (n = 115) or MA positive (n = 51) were included in the current study. All these participants were free of diabetes at baseline (WHO 1985) and had high-sensitive C-reactive protein (hs-CRP), serum FA composition, and selected lipid metabolites measured during the active study period. Among the markers associated with MA, the serum plasmalogen dm16:0 (p = 0.006), the saturated odd-chain FA 15.0 (pentadecanoic acid; p = 0.015), and omega-3 very long-chain FAs (p < 0.05) were associated with a decreased occurrence of MA. These associations were independent of study group and other risk factors. The association of high serum triglycerides with the MA occurrence was attenuated when these MA-associated serum lipid markers were considered. Our findings suggest that, in addition to n-3 FAs, odd-chain FA 15:0 and plasmalogen dm16:0 may contribute to a lower risk of MA in individuals with impaired glucose tolerance. These putative novel lipid biomarkers have an association with MA independently of triglyceride levels.
Collapse
Affiliation(s)
- Vanessa Derenji de Mello
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, FI-70211 Kuopio, Finland;
- Correspondence:
| | - Tuomas Selander
- Science Service Center, Kuopio University Hospital, FI-70029 Kuopio, Finland;
| | - Jaana Lindström
- Public Health Prevention Unit, Finnish Institute for Health and Welfare, FI-00271 Helsinki, Finland; (J.L.); (J.T.)
| | - Jaakko Tuomilehto
- Public Health Prevention Unit, Finnish Institute for Health and Welfare, FI-00271 Helsinki, Finland; (J.L.); (J.T.)
- Department of Public Health, University of Helsinki, FI-00014 Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Matti Uusitupa
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, FI-70211 Kuopio, Finland;
| | - Kai Kaarniranta
- Department of Ophthalmology, Kuopio University Hospital, FI-70029 Kuopio, Finland;
- Institute of Clinical Medicine, Faculty of Health Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
48
|
Ma MY, Li KL, Zheng H, Dou YL, Han LY, Wang L. Omega-3 index and type 2 diabetes: Systematic review and meta-analysis. Prostaglandins Leukot Essent Fatty Acids 2021; 174:102361. [PMID: 34740031 DOI: 10.1016/j.plefa.2021.102361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/10/2021] [Accepted: 10/27/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND OBJECTIVES The relationship between omega-3 index and type 2 diabetes (T2D) is not well established. It is unclear if the change of omega-3 index will affect T2D. Aiming of the present systematic review was to elucidate the correlation between omega-3 index and T2D. METHODS AND STUDY DESIGN A comprehensive search on PubMed, EMBASE and Web of Science (from 1948 to May 2021) was conducted. The overall effect size (standard mean difference) was combined using a random-effect model. RESULTS Eight eligible case-control studies were identified, and there were 1,357 patients with T2D and 1,616 non-diabetic controls. The result showed that the omega-3 index was significantly lower in diabetic cases than that in controls (SMD= -1.31; 95% confidence interval (CI): -1.40, -1.22), but with significant heterogeneity (I2 = 99.0%). In subgroup analysis based on race, a negative correlation was found in Asians (SMD = -1.71; 95% CI: -1.82, -1.60), and heterogeneity was substantially decreased (I2=0). CONCLUSIONS omega-3 index is negatively correlated with T2D, which indicated that increased dietary intake of omega-3 fatty acids might have beneficial on T2D prevention.
Collapse
Affiliation(s)
- Mu-Yuan Ma
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ke-Lei Li
- Institute of Nutrition & Health, Qingdao University, Qingdao, China
| | - Hui Zheng
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Ya-Li Dou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Li-Yao Han
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ling Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
49
|
Meuronen T, Lankinen MA, Kärkkäinen O, Laakso M, Pihlajamäki J, Hanhineva K, Schwab U. FADS1 rs174550 genotype and high linoleic acid diet modify plasma PUFA phospholipids in a dietary intervention study. Eur J Nutr 2021; 61:1109-1120. [PMID: 34718859 PMCID: PMC8854246 DOI: 10.1007/s00394-021-02722-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/18/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Fatty acid desaturase 1 (FADS1) gene encodes for delta-5 desaturase enzyme which is needed in conversion of linoleic acid (LA) to arachidonic acid (AA). Recent studies have shown that response to dietary PUFAs differs between the genotypes in circulating fatty acids. However, interactions between the FADS1 genotype and dietary LA on overall metabolism have not been studied. OBJECTIVES We aimed to examine the interactions of FADS1 rs174550 genotypes (TT and CC) and high-LA diet to identify plasma metabolites that respond differentially to dietary LA according to the FADS1 genotype. METHODS A total of 59 men (TT n = 26, CC n = 33) consumed a sunflower oil supplemented diet for 4 weeks. Daily dose of 30, 40, or 50 ml was calculated based on body mass index. It resulted in 17-28 g of LA on top of the usual daily intake. Fasting plasma samples at the beginning and at the end of the intervention were analyzed with LC-MS/MS non-targeted metabolomics method. RESULTS At the baseline, the carriers of FADS1 rs174550-TT genotype had higher abundance of long-chain PUFA phospholipids compared to the FADS1 rs174550-CC one. In response to the high-LA diet, LA phospholipids and long-chain acylcarnitines increased and lysophospholipids decreased in fasting plasma similarly in both genotypes. LysoPE (20:4), LysoPC (20:4), and PC (16:0_20:4) decreased and cortisol increased in the carriers of rs174550-CC genotype; however, these genotype-diet interactions were not significant after correction for multiple testing. CONCLUSION Our findings show that both FADS1 rs174550 genotype and high-LA diet modify plasma phospholipid composition. TRIAL REGISTRATION The study was registered to ClinicalTrials: NCT02543216, September 7, 2015 (retrospectively registered).
Collapse
Affiliation(s)
- Topi Meuronen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, PO box 1627, 70211, Kuopio, Finland.
| | - Maria A Lankinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, PO box 1627, 70211, Kuopio, Finland
| | - Olli Kärkkäinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, PO box 1627, 70211, Kuopio, Finland.,Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Kati Hanhineva
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, PO box 1627, 70211, Kuopio, Finland.,Department of Life Technologies, Food Chemistry and Food Development Unit, University of Turku, Turku, Finland.,Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Ursula Schwab
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, PO box 1627, 70211, Kuopio, Finland.,Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW It is accepted that several chronic diseases are associated with inflammation. Dietary habits and the consumption of certain nutrients have been shown to influence inflammation, reflected by inflammatory cytokines. In this narrative review, we discuss currently developed tools to assess the inflammatory potential of diets and compare them with established tools. RECENT FINDINGS Four new indices were recently developed. The Inflammatory Score of the Diet is a modified version of the established Dietary Inflammatory Index. The novel Empirical Dietary Inflammatory Index works without previous dietary intake assessment and the Anti-Inflammatory Diet Index was specifically developed in a northern European population. The Dietary and Lifestyle Inflammation Scores addresses additional confounders. The informative value of dietary indices relies on the accuracy and completeness of dietary intake assessment. SUMMARY Dietary inflammatory indices are important tools to assess, compare and validate the inflammatory potential of diets across populations without the need for biomarker assessments. They allow to investigate associations between an (anti)-inflammatory diet with disease risk and course. Although the Dietary Inflammatory Index remains the most used index worldwide, currently developed indices allow more flexibility, have a different focus or simplify assessment. Additional foods, that were recently shown to modulate inflammation, but are not (fully) considered yet, may deserve more attention in the future.
Collapse
Affiliation(s)
- Lina Samira Bahr
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, ECRC Experimental and Clinical Research Center
| | - Kristina Franz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Research Group on Geriatrics
| | - Anja Mähler
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, ECRC Experimental and Clinical Research Center
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|