1
|
Wang Y, Wu G, Wang Y, Xiao F, Yin H, Yu L, Shehzad Q, Zhang H, Jin Q, Wang X. Association of erythrocyte fatty acid compositions with the risk of pancreatic cancer: A case-control study. Lipids 2024. [PMID: 39397372 DOI: 10.1002/lipd.12420] [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: 07/01/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024]
Abstract
Pancreatic cancer (PC) is one of the most fatal malignancies, which has attracted scientists to investigate its etiology and pathogenesis. Nevertheless, the association between erythrocyte fatty acids and PC risk remains unclear. This study aimed to evaluate the association between levels of erythrocyte fatty acids and PC risk. The erythrocyte fatty acid compositions of 105 PC patients and 120 controls were determined by gas chromatography. Cases and controls were frequency matched by age and sex. Multivariable conditional logistic regression model and restricted cubic spline were applied to estimate the odds ratio with 95% confidence interval (OR, 95% CI) of erythrocyte fatty acids and PC risk. Our main findings indicated a significant negative association between levels of erythrocyte total monounsaturated fatty acids (MUFA) and n-3 polyunsaturated fatty acids (n-3 PUFA) and the risk of PC (ORT3-T1 = 0.30 [0.14, 0.63] and ORT3-T1 = 0.15 [0.06, 0.33], respectively). In contrast, erythrocyte n-6 polyunsaturated fatty acids, specifically linoleic acid (LA) and arachidonic acid (AA) levels, were positively associated with PC incidence (RT1-T3 = 4.24 [1.97, 9.46] and ORT1-T3 = 4.53 [2.09, 10.20]). Total saturated fatty acid (SFA), especially high levels of palmitic acid (16:0), was positively associated with the risk of PC (ORT3-T1 = 3.25 [1.53, 7.08]). Our findings suggest that levels of different types of fatty acids in erythrocytes may significantly alter PC susceptibility. Protective factors against PC include unsaturated fatty acids such as n-3 PUFA and MUFA.
Collapse
Affiliation(s)
- Yongjin Wang
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Gangcheng Wu
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yandan Wang
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
- Department of Research and Development, Jiahe Foods Industry Co., Ltd, Suzhou, China
| | - Feng Xiao
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Hongming Yin
- Department of Laboratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Le Yu
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
- Department of Research and Development, Jiahe Foods Industry Co., Ltd, Suzhou, China
- Department of Dairy Technology and Equipment Research, National Center of Technology Innovation for Dairy, Hohhot, China
| | - Qayyum Shehzad
- School of Fundamental Sciences, Massey University, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Hui Zhang
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qingzhe Jin
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xingguo Wang
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
2
|
Schmieta HML, Greupner T, Schneider I, Wrobel S, Christa V, Kutzner L, Hahn A, Harris WS, Schebb NH, Schuchardt JP. Plasma levels of EPA and DHA after ingestion of a single dose of EPA and DHA ethyl esters. Lipids 2024. [PMID: 39299684 DOI: 10.1002/lipd.12417] [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: 05/16/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024]
Abstract
Omega-3 polyunsaturated fatty acids (n3 PUFA), specifically eicosapentaenoic acid (EPA, 20:5n3), and docosahexaenoic acid (DHA, 22:6n3), are essential for maintaining health. To better understand their biology, it is important to define their bioavailability. The aim of this cross-over study was to investigate and compare the acute effects on plasma EPA and DHA levels after single doses of EPA oil (99% pure) and DHA (97% pure) ethyl esters. Twelve men aged 20-40 years with a body-mass-index of 20-27 kg/m2 and low fish consumption were recruited. Several measures (e.g., 4-week run-in period, standardized diet, and blood collection protocols) were taken to reduce the inter-individual variability of plasma fatty acids levels. Using a cross-over design, the subjects received 2.2 g of EPA in the first test period and 2.3 g of DHA in the second. The test periods were separated by 2 weeks. Blood samples were taken before dosing and after 2, 4, 6, 8, 12, 24, 48, and 72 h. The mean ± SE maximum concentrations for EPA were higher than for DHA (115 ± 11 μg/mL vs. 86 ± 12 μg/mL; p = 0.05). The mean ± SE incremented area under the plasma concentration curve over 72 h for EPA (2461 ± 279 μg/mL) was 2.4 times higher (p < 0.001) than that for DHA (1021 ± 170 μg/mL). The mean ± SE half-life was for EPA and DHA was 45 ± 8 and 66 ± 12 h. Our results indicate that EPA administration in single doses leads to higher circulating plasma levels of EPA compared to an effect of an equivalent dose of DHA on DHA plasma levels.
Collapse
Affiliation(s)
| | - Theresa Greupner
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany
| | - Inga Schneider
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany
| | - Sonja Wrobel
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Vanessa Christa
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Laura Kutzner
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Andreas Hahn
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany
| | - William S Harris
- The 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
| | - Nils Helge Schebb
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Jan Philipp Schuchardt
- Institute of Food and One Health, Leibniz University Hannover, Hannover, Germany
- The Fatty Acid Research Institute, Sioux Falls, South Dakota, USA
| |
Collapse
|
3
|
Abedimanesh N, Motlagh B, Hejazi J, Eskandari MR, Asghari-Jafarabadi M, Mazloomzadeh S. Biomarker-based validation of a food frequency questionnaire for the assessment of omega-3 fatty acid status in a healthy Iranian population. Sci Rep 2023; 13:14813. [PMID: 37684272 PMCID: PMC10491660 DOI: 10.1038/s41598-023-41623-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
There is no valid instrument to assess n-3 polyunsaturated fatty acids (n-3 PUFAs) intake in Iran. This study aims to develop a food frequency questionnaire (FFQ) that estimates the intake of n-3 PUFA and validate it in a healthy Iranian population based on the n-3 PUFA content of red blood cells (RBCs) and a 3-day food record (FR). A healthy population (n = 221) was recruited between February and July 2021. Participants completed the new FFQ and 3-day FR to evaluate the average intake of n-3 PUFAs. We used gas chromatography to assess the n-3 PUFA content of RBCs. To validate the FFQ based on FR and biomarker as references, the correlation coefficient was calculated. According to the Bland-Altman plots, a good agreement was found between the new FFQ and FR. Moreover, absolute intake values of ALA, EPA, DPA, DHA, and total n-3 PUFAs based on FFQ were positively correlated to their respective RBC membrane levels (coefficients between 0.205 and 0.508, p < 0.005) and FR (coefficients between 0.771 and 0.827, p < 0.001). This new FFQ is a valid instrument that can be applied to estimate the n-3 PUFA status of healthy Iranian adults.
Collapse
Affiliation(s)
- Nasim Abedimanesh
- Social Determinants of Health Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Behrooz Motlagh
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Jalal Hejazi
- Department of Nutrition, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohammad Reza Eskandari
- Department of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohammad Asghari-Jafarabadi
- Cabrini Research, Cabrini Health, Malvern, VIC, 3144, Australia
- School of Public Health and Preventative Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3800, Australia
- Department of Psychiatry, School of Clinical Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3168, Australia
| | | |
Collapse
|
4
|
Chong SY, Wang X, van Bloois L, Huang C, Syeda NS, Zhang S, Ting HJ, Nair V, Lin Y, Lou CKL, Benetti AA, Yu X, Lim NJY, Tan MS, Lim HY, Lim SY, Thiam CH, Looi WD, Zharkova O, Chew NWS, Ng CH, Bonney GK, Muthiah M, Chen X, Pastorin G, Richards AM, Angeli V, Storm G, Wang JW. Injectable liposomal docosahexaenoic acid alleviates atherosclerosis progression and enhances plaque stability. J Control Release 2023; 360:344-364. [PMID: 37406819 DOI: 10.1016/j.jconrel.2023.06.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 06/12/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory vascular disease that is characterized by the accumulation of lipids and immune cells in plaques built up inside artery walls. Docosahexaenoic acid (DHA, 22:6n-3), an omega-3 polyunsaturated fatty acid (PUFA), which exerts anti-inflammatory and antioxidant properties, has long been purported to be of therapeutic benefit to atherosclerosis patients. However, large clinical trials have yielded inconsistent data, likely due to variations in the formulation, dosage, and bioavailability of DHA following oral intake. To fully exploit its potential therapeutic effects, we have developed an injectable liposomal DHA formulation intended for intravenous administration as a plaque-targeted nanomedicine. The liposomal formulation protects DHA against chemical degradation and increases its local concentration within atherosclerotic lesions. Mechanistically, DHA liposomes are readily phagocytosed by activated macrophages, exert potent anti-inflammatory and antioxidant effects, and inhibit foam cell formation. Upon intravenous administration, DHA liposomes accumulate preferentially in atherosclerotic lesional macrophages and promote polarization of macrophages towards an anti-inflammatory M2 phenotype, resulting in attenuation of atherosclerosis progression in both ApoE-/- and Ldlr-/- experimental models. Plaque composition analysis demonstrates that liposomal DHA inhibits macrophage infiltration, reduces lipid deposition, and increases collagen content, thus improving the stability of atherosclerotic plaques against rupture. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) further reveals that DHA liposomes can partly restore the complex lipid profile of the plaques to that of early-stage plaques. In conclusion, DHA liposomes offer a promising approach for applying DHA to stabilize atherosclerotic plaques and attenuate atherosclerosis progression, thereby preventing atherosclerosis-related cardiovascular events.
Collapse
Affiliation(s)
- Suet Yen Chong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Xiaoyuan Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Louis van Bloois
- Department of Pharmaceutics, Faculty of Science, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Chenyuan Huang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Nilofer Sayed Syeda
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Sitong Zhang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Hui Jun Ting
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Vaarsha Nair
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Yuanzhe Lin
- Department of Biomedical Engineering, National University of Singapore, 117583 Singapore, Singapore
| | - Charles Kang Liang Lou
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Ayca Altay Benetti
- Department of Pharmacy, Faculty of Science, National University of Singapore, 117543 Singapore, Singapore
| | - Xiaodong Yu
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Nicole Jia Ying Lim
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Michelle Siying Tan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Sheau Yng Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Chung Hwee Thiam
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Wen Donq Looi
- Bruker Daltonics, Bruker Singapore Pte. Ltd., 138671 Singapore, Singapore
| | - Olga Zharkova
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Nicholas W S Chew
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Department of Cardiology, National University Heart Centre, National University Hospital, 119074 Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Glenn Kunnath Bonney
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, National University Hospital, 119074 Singapore, Singapore
| | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, 119074 Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, 119074 Singapore, Singapore
| | - Xiaoyuan Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 119074 Singapore, Singapore; Departments of Chemical and Biomolecular Engineering, and Biomedical Engineering, Faculty of Engineering, National University of Singapore, 117575 Singapore, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy, Faculty of Science, National University of Singapore, 117543 Singapore, Singapore
| | - A Mark Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Veronique Angeli
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Pharmaceutics, Faculty of Science, Utrecht University, 3584 CG Utrecht, the Netherlands; Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, the Netherlands.
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Physiology, National University of Singapore, 117593 Singapore, Singapore.
| |
Collapse
|
5
|
Xu L, Wang S, Tian A, Liu T, Benjakul S, Xiao G, Ying X, Zhang Y, Ma L. Characteristic Volatile Compounds, Fatty Acids and Minor Bioactive Components in Oils from Green Plum Seed by HS-GC-IMS, GC-MS and HPLC. Food Chem X 2022; 17:100530. [DOI: 10.1016/j.fochx.2022.100530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/10/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
|
6
|
Ding L, Liu Y, Kang M, Wei X, Geng C, Liu W, Han L, Yuan F, Wang P, Wang B, Sun Y. UPLC-QTOF/MS Metabolomics and Biochemical Assays Reveal Changes in Hepatic Nutrition and Energy Metabolism during Sexual Maturation in Female Rainbow Trout ( Oncorhynchus mykiss). BIOLOGY 2022; 11:1679. [PMID: 36421392 PMCID: PMC9687450 DOI: 10.3390/biology11111679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 09/20/2024]
Abstract
Mobilization and repartition of nutrients and energy are prerequisites for the normal sexual maturity of broodstock. However, there are few studies on the mechanisms of hepatic nutrients and energy metabolism during sexual maturation in female rainbow trout (Oncorhynchus mykiss). This study investigated hepatic metabolite changes and explored the potential nutritional regulation mechanism between mature and immature female rainbow trout by combining UPLC-QTOF/MS metabolomics and biochemical assays. It was observed that hepatic biochemical assays differed considerably between the two groups, such as glucose, triglycerides, hexokinase, lipase, and aspartate aminotransferase. Liver metabolomics showed that various differential metabolites involved in amino acid and lipid metabolism markedly increased, suggesting the enhancement of lipid metabolism and amino acid anabolism in the liver provides the necessary material basis for ovarian development. Meanwhile, glycogen catabolism and glycolysis hold the key to maintaining organismal energy homeostasis with normal sexual maturation of female rainbow trout. Overall, the results from this study suggested that the liver undergoes drastic reprogramming of the metabolic profile in response to mobilization and repartition of nutrients and energy during the sexual maturation of female rainbow trout. This study further deepened the understanding of the reproductive biology of rainbow trout, and provided the theoretical basis and practical ramifications for nutritional requirements of breeding high-quality broodstock in the artificial propagation of rainbow trout.
Collapse
Affiliation(s)
- Lu Ding
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
- Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Yingjie Liu
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
- Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Meng Kang
- Heilongjiang Provincial Fishery Extension Center, Harbin 150080, China
| | - Xiaofeng Wei
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
- Department of Food Science and Engineering, College of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China
| | - Chuanye Geng
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
- Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Wenzhi Liu
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
- Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Lin Han
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
- Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Fangying Yuan
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
- Department of Chemical Engineering and Technology, College of Materials and Chemical Engineering, Harbin University of Science and Technology, Harbin 150080, China
| | - Peng Wang
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
| | - Bingqian Wang
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
| | - Yanchun Sun
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Ministry of Agriculture and Rural Areas, Harbin 150070, China
- Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Department of Food Science and Engineering, College of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China
- Department of Chemical Engineering and Technology, College of Materials and Chemical Engineering, Harbin University of Science and Technology, Harbin 150080, China
| |
Collapse
|
7
|
Udumula MP, Poisson LM, Dutta I, Tiwari N, Kim S, Chinna-Shankar J, Allo G, Sakr S, Hijaz M, Munkarah AR, Giri S, Rattan R. Divergent Metabolic Effects of Metformin Merge to Enhance Eicosapentaenoic Acid Metabolism and Inhibit Ovarian Cancer In Vivo. Cancers (Basel) 2022; 14:cancers14061504. [PMID: 35326656 PMCID: PMC8946838 DOI: 10.3390/cancers14061504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/07/2022] [Accepted: 03/12/2022] [Indexed: 02/01/2023] Open
Abstract
Metformin is being actively repurposed for the treatment of gynecologic malignancies including ovarian cancer. We investigated if metformin induces analogous metabolic changes across ovarian cancer cells. Functional metabolic analysis showed metformin caused an immediate and sustained decrease in oxygen consumption while increasing glycolysis across A2780, C200, and SKOV3ip cell lines. Untargeted metabolomics showed metformin to have differential effects on glycolysis and TCA cycle metabolites, while consistent increased fatty acid oxidation intermediates were observed across the three cell lines. Metabolite set enrichment analysis showed alpha-linolenic/linoleic acid metabolism as being most upregulated. Downstream mediators of the alpha-linolenic/linoleic acid metabolism, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), were abundant in all three cell lines. EPA was more effective in inhibiting SKOV3 and CaOV3 xenografts, which correlated with inhibition of inflammatory markers and indicated a role for EPA-derived specialized pro-resolving mediators such as Resolvin E1. Thus, modulation of the metabolism of omega-3 fatty acids and their anti-inflammatory signaling molecules appears to be one of the common mechanisms of metformin's antitumor activity. The distinct metabolic signature of the tumors may indicate metformin response and aid the preclinical and clinical interpretation of metformin therapy in ovarian and other cancers.
Collapse
Affiliation(s)
- Mary P. Udumula
- Department of Women’s Health Services, Henry Ford Hospital, Henry Ford Cancer Institute, Detroit, MI 48202, USA; (M.P.U.); (N.T.); (J.C.-S.); (M.H.); (A.R.M.)
| | - Laila M. Poisson
- Center for Bioinformatics, Department of Public Health Services, Henry Ford Cancer Institute, Detroit, MI 48202, USA; (L.M.P.); (I.D.)
| | - Indrani Dutta
- Center for Bioinformatics, Department of Public Health Services, Henry Ford Cancer Institute, Detroit, MI 48202, USA; (L.M.P.); (I.D.)
| | - Nivedita Tiwari
- Department of Women’s Health Services, Henry Ford Hospital, Henry Ford Cancer Institute, Detroit, MI 48202, USA; (M.P.U.); (N.T.); (J.C.-S.); (M.H.); (A.R.M.)
| | - Seongho Kim
- Biostatistics and Bioinformatics Core, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA;
| | - Jasdeep Chinna-Shankar
- Department of Women’s Health Services, Henry Ford Hospital, Henry Ford Cancer Institute, Detroit, MI 48202, USA; (M.P.U.); (N.T.); (J.C.-S.); (M.H.); (A.R.M.)
| | - Ghassan Allo
- Department of Pathology, Henry Ford Hospital, Henry Ford Cancer Institute, Detroit, MI 48202, USA;
| | - Sharif Sakr
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA;
| | - Miriana Hijaz
- Department of Women’s Health Services, Henry Ford Hospital, Henry Ford Cancer Institute, Detroit, MI 48202, USA; (M.P.U.); (N.T.); (J.C.-S.); (M.H.); (A.R.M.)
| | - Adnan R. Munkarah
- Department of Women’s Health Services, Henry Ford Hospital, Henry Ford Cancer Institute, Detroit, MI 48202, USA; (M.P.U.); (N.T.); (J.C.-S.); (M.H.); (A.R.M.)
| | - Shailendra Giri
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA;
| | - Ramandeep Rattan
- Department of Women’s Health Services, Henry Ford Hospital, Henry Ford Cancer Institute, Detroit, MI 48202, USA; (M.P.U.); (N.T.); (J.C.-S.); (M.H.); (A.R.M.)
- Department of Oncology, Wayne State School of Medicine, Detroit, MI 48201, USA
- Correspondence: ; Tel.: +313-876-7381; Fax: +313-876-3415
| |
Collapse
|
8
|
Maki KC, Bays HE, Ballantyne CM, Underberg JA, Kastelein JJP, Johnson JB, Ferguson JJ. A Head-to-Head Comparison of a Free Fatty Acid Formulation of Omega-3 Pentaenoic Acids Versus Icosapent Ethyl in Adults With Hypertriglyceridemia: The ENHANCE-IT Study. J Am Heart Assoc 2022; 11:e024176. [PMID: 35232215 PMCID: PMC9075326 DOI: 10.1161/jaha.121.024176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/23/2021] [Indexed: 01/08/2023]
Abstract
Background MAT9001 is an omega-3 free fatty acid (FFA) formulation containing mainly eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA). Compared with icosapent ethyl (EPA-ethyl esters [EE]), EPA+DPA-FFA previously showed enhanced triglyceride lowering and higher plasma EPA when both were administered once daily with a very-low fat diet. This trial compared pharmacodynamic responses and plasma omega-3 levels following twice daily dosing, with meals, of EPA+DPA-FFA and EPA-EE in hypertriglyceridemic subjects consuming a Therapeutic Lifestyle Changes diet. Methods and Results This open-label, randomized, 2-way crossover trial, with 28-day treatment periods separated by ≥28-day washout, was conducted at 8 US centers and included 100 subjects with fasting triglycerides 1.70 to 5.64 mmol/L (150-499 mg/dL) (median 2.31 mmol/L [204 mg/dL]; 57% women, average age 60.3 years). The primary end point was least squares geometric mean percent change from baseline plasma triglycerides. In the 94 subjects with analyzable data for both treatment periods, EPA+DPA-FFA and EPA-EE reduced least squares geometric mean triglycerides from baseline: 20.9% and 18.3%, respectively (P=not significant). EPA+DPA-FFA reduced least squares geometric mean high-sensitivity C-reactive protein by 5.8%; EPA-EE increased high-sensitivity C-reactive protein by 8.5% (P=0.034). EPA+DPA-FFA increased least squares geometric mean plasma EPA, DPA, and total omega-3 (EPA+docosahexaenoic acid+DPA) concentrations by 848%, 177%, and 205%, respectively, compared with corresponding changes with EPA-EE of 692%, 140%, and 165% (all P<0.001). EPA+DPA-FFA increased docosahexaenoic acid by 1.7%; EPA-EE decreased docosahexaenoic acid by 3.3% (P=0.011). Lipoprotein cholesterol and apolipoprotein responses did not differ between treatments. Conclusions EPA+DPA-FFA raised plasma EPA, DPA, and total omega-3 significantly more than did EPA-EE. EPA+DPA-FFA also reduced triglycerides and high-sensitivity C-reactive protein without increasing low-density lipoprotein cholesterol. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT04177680.
Collapse
Affiliation(s)
- Kevin C. Maki
- Midwest Biomedical ResearchAddisonIL
- Department of Applied Health ScienceIndiana University School of Public HealthBloomingtonIN
| | - Harold E. Bays
- Louisville Metabolic and Atherosclerosis Research Center, Inc.LouisvilleKY
| | | | - James A. Underberg
- NYU School of Medicine and NYU Center for Prevention of Cardiovascular DiseaseNew YorkNY
| | | | | | | |
Collapse
|
9
|
Ljungblad L, Bergqvist F, Tümmler C, Madawala S, Olsen TK, Andonova T, Jakobsson PJ, Johnsen JI, Pickova J, Strandvik B, Kogner P, Gleissman H, Wickström M. Omega-3 fatty acids decrease CRYAB, production of oncogenic prostaglandin E 2 and suppress tumor growth in medulloblastoma. Life Sci 2022; 295:120394. [PMID: 35157910 DOI: 10.1016/j.lfs.2022.120394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 12/09/2022]
Abstract
AIMS Medulloblastoma (MB) is one of the most common malignant central nervous system tumors of childhood. Despite intensive treatments that often leads to severe neurological sequelae, the risk for resistant relapses remains significant. In this study we have evaluated the effects of the ω3-long chain polyunsaturated fatty acids (ω3-LCPUFA) docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on MB cell lines and in a MB xenograft model. MAIN METHODS Effects of ω3-LCPUFA treatment of MB cells were assessed using the following: WST-1 assay, cell death probes, clonogenic assay, ELISA and western blot. MB cells were implanted into nude mice and the mice were randomized to DHA, or a combination of DHA and EPA treatment, or to control group. Treatment effects in tumor tissues were evaluated with: LC-MS/MS, RNA-sequencing and immunohistochemistry, and tumors, erythrocytes and brain tissues were analyzed with gas chromatography. KEY FINDINGS ω3-LCPUFA decreased prostaglandin E2 (PGE2) secretion from MB cells, and impaired MB cell viability and colony forming ability and increased apoptosis in a dose-dependent manner. DHA reduced tumor growth in vivo, and both PGE2 and prostacyclin were significantly decreased in tumor tissue from treated mice compared to control animals. All ω3-LCPUFA and dihomo-γ-linolenic acid increased in tumors from treated mice. RNA-sequencing revealed 10 downregulated genes in common among ω3-LCPUFA treated tumors. CRYAB was the most significantly altered gene and the downregulation was confirmed by immunohistochemistry. SIGNIFICANCE Our findings suggest that addition of DHA and EPA to the standard MB treatment regimen might be a novel approach to target inflammation in the tumor microenvironment.
Collapse
Affiliation(s)
- Linda Ljungblad
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| | - Filip Bergqvist
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Conny Tümmler
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Samanthi Madawala
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Thale Kristin Olsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Teodora Andonova
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Jana Pickova
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Birgitta Strandvik
- Department of Biosciences and Nutrition Karolinska Institutet, NEO, Flemingsberg, Stockholm, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Pediatric Oncology, Astrid Lindgrens Childrens Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Helena Gleissman
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Malin Wickström
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Liu H, Wang F, Liu X, Xie Y, Xia H, Wang S, Sun G. Effects of marine-derived and plant-derived omega-3 polyunsaturated fatty acids on erythrocyte fatty acid composition in type 2 diabetic patients. Lipids Health Dis 2022; 21:20. [PMID: 35144649 PMCID: PMC8832668 DOI: 10.1186/s12944-022-01630-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/28/2022] [Indexed: 12/15/2022] Open
Abstract
Background Dietary fatty acids intake affects the composition of erythrocyte fatty acids, which is strongly correlated with glycolipid metabolism disorders. This study aimed at investigating the different effects of marine-derived and plant-derived omega-3 polyunsaturated fatty acid (n-3 PUFA) on the fatty acids of erythrocytes and glycolipid metabolism in patients with type 2 diabetes mellitus (T2DM). Methods The randomized double-blinded trial that was performed on 180 T2DM patients. The participants were randomly assigned to three groups for the six-month intervention. The participants were randomly assigned to three groups for the six-month intervention. The fish oil (FO) group was administered with FO at a dose of 3 g/day containing eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), the perilla oil (PO) group was administered with PO at a dose of 3 g/day containing α-linolenic (ALA), the linseed and fish oil (LFO) group was administered with mixed linseed and fish oil at a dose of 3 g/day containing EPA, DHA and ALA. Demographic information were collected and anthropometric indices, glucose and lipid metabolism indexes, erythrocyte fatty acid composition were measured. Statistical analyses were performed using two-way ANOVA. Results A total of 150 patients finished the trial, with 52 of them in the FO group, 50 in the PO group and 48 in the LFO group. There were significant effects of time × treatment interaction on fast blood glucose (FBG), insulin, HOMA-IR and C-peptide, TC and triglyceride (TG) levels (P < 0.001). Glucose and C-peptide in PO and LFO groups decreased significantly and serum TG in FO group significantly decreased (P < 0.001) after the intervention. Erythrocyte C22: 5 n-6, ALA, DPA, n-6/n-3 PUFA, AA/EPA levels in the PO group were significantly higher than FO and LFO groups, while EPA, total n-3 PUFA and Omega-3 index were significantly higher in the FO and LFO groups compared to PO group. Conclusion Supplementation with perilla oil decreased FBG while fish oil supplementation decreased the TG level. Marine-based and plant-based n-3 PUFAs exhibit different effects on fatty acid compositions of erythrocytes and regulated glycolipid metabolism. Trial registration This trial was recorded under Chinese Clinical Trial Registry Center (NO: ChiCTR-IOR-16008435) on May 28 2016.
Collapse
Affiliation(s)
- Hechun Liu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Feng Wang
- Tianjin Institute of Environmental and Operational Medicine, 1 Da Li Road, Tianjin, 300050, China
| | - Xiaosong Liu
- Guanlin Hospital, 17 Wenwei Road, Yixing, 214251, China
| | - Yulan Xie
- Zhongda Hospital Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Hui Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Shaokang Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, 87 Ding Jia Qiao Road, Nanjing, 210009, China.
| |
Collapse
|
11
|
Guo XF, Wang C, Yang T, Ma WJ, Zhai J, Zhao T, Xu TC, Li J, Liu H, Sinclair AJ, Li D. The effects of fish oil plus vitamin D3 intervention on non-alcoholic fatty liver disease: a randomized controlled trial. Eur J Nutr 2022; 61:1931-1942. [DOI: 10.1007/s00394-021-02772-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/03/2021] [Indexed: 01/10/2023]
|
12
|
Review: Improving the nutritional, sensory and market value of meat products from sheep and cattle. Animal 2021; 15 Suppl 1:100356. [PMID: 34600858 DOI: 10.1016/j.animal.2021.100356] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
This paper focuses on improving the sensory, health attributes and meat yield of beef and lamb meats. Value for meat is defined as the weight of meat × price/kg received with price linked to eating quality. To maximise value across the supply chain, accurate carcass grading systems for eating quality and yield are paramount. Grading data can then be used to target consumers' needs at given price points and then to tailor appropriate production and genetic directions. Both the grading methodologies and key phenotypes are complex and still under intensive research with international collaboration to maximise opportunities. In addition, there is value in promoting the health aspects of red meats served as whole trimmed meats. Typically, the total fat content is relatively low (less than 5%) and for forage systems, they deliver a very significant content of long-chain n-3 fatty acids. Further research is needed to clarify the healthiness or otherwise of ground beef served as burgers given the fat content is typically 20% or more. It is important to continue to improve the feedback to producers regarding the quantity and quality of the products they produce to target new value opportunities in a transparent and quantitative manner.
Collapse
|
13
|
Heileson JL, Anzalone AJ, Carbuhn AF, Askow AT, Stone JD, Turner SM, Hillyer LM, Ma DWL, Luedke JA, Jagim AR, Oliver JM. The effect of omega-3 fatty acids on a biomarker of head trauma in NCAA football athletes: a multi-site, non-randomized study. J Int Soc Sports Nutr 2021; 18:65. [PMID: 34579748 PMCID: PMC8477477 DOI: 10.1186/s12970-021-00461-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/12/2021] [Indexed: 12/18/2022] Open
Abstract
Background American-style football (ASF) athletes are at risk for cardiovascular disease (CVD) and exhibit elevated levels of serum neurofilament light (Nf-L), a biomarker of axonal injury that is associated with repetitive head impact exposure over the course of a season of competition. Supplementation with the w-3 fatty acid (FA) docosahexaenoic acid (DHA) attenuates serum Nf-L elevations and improves aspects of CVD, such as the omega-3 index (O3I). However, the effect of combining the w-3 FA eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA) with DHA on, specifically, serum Nf-L in ASF athletes is unknown. Therefore, this study assessed the effect of supplemental w-3 FA (EPA+DPA+DHA) on serum Nf-L, plasma w-3 FAs, the O3I, and surrogate markers of inflammation over the course of a season. Methods A multi-site, non-randomized design, utilizing two American football teams was employed. One team (n = 3 1) received supplementation with a highly bioavailablew-3 FA formulation (2000mg DHA, 560mg EPA, 320mg DPA, Mindset®, Struct Nutrition, Missoula, MT) during pre-season and throughout the regular season, while the second team served as the control (n = 35) and did not undergo supplementation. Blood was sampled at specific times throughout pre- and regular season coincident w ith changes in intensity, physical contact, and changes in the incidence and severity of head impacts. Group differences were determined via a mixed-model between-within subjects ANOVA. Effect sizes were calculated using Cohen’s dfor all between-group differences. Significance was set a priori at p< .05. Results Compared to the control group, ASF athletes in the treatment group experienced large increases in plasma EPA (p < .001, d = 1.71) and DHA (p < .001, d = 2.10) which contributed to increases in the O3I (p < .001, d = 2.16) and the EPA:AA ratio (p = .001, d = 0.83) and a reduction in the w-6: w-3 ratio (p < .001, d = 1.80). w-3 FA supplementation attenuated elevations in Nf-L (p = .024). The control group experienced a significant increase in Nf-L compared to baseline at several measurement time points (T2, T3, and T4 [p range < .001 – .005, drange = 0.59-0.85]). Conclusions These findings suggest a cardio- and neuroprotective effect of combined EPA+DPA+DHA w-3 FA supplementation in American-style football athletes. Trial registration This trial was registered with the ISRCTN registry (ISRCTN90306741).
Collapse
Affiliation(s)
- Jeffery L Heileson
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX, USA
| | | | | | - Andrew T Askow
- Nutrition and Exercise Performance Laboratory, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Jason D Stone
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Stephanie M Turner
- Department of Kinesiology, Texas Christian University, Fort Worth, TX, USA
| | - Lyn M Hillyer
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - David W L Ma
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - Joel A Luedke
- Athletics Department, University of Wisconsin - La Crosse, La Crosse, WI, USA
| | - Andrew R Jagim
- Sports Medicine, Mayo Clinic Health Systems, Onalaska, WI, USA
| | - Jonathan M Oliver
- Department of Kinesiology, Texas Christian University, Fort Worth, TX, USA
| |
Collapse
|
14
|
Zhang Z, Xue Z, Yang H, Zhao F, Liu C, Chen J, Lu S, Zou Z, Zhou Y, Zhang X. Differential effects of EPA and DHA on DSS-induced colitis in mice and possible mechanisms involved. Food Funct 2021; 12:1803-1817. [PMID: 33523066 DOI: 10.1039/d0fo02308f] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The anti-inflammatory effect of n-3 PUFAs has been widely documented. Emerging evidence suggests that the main component of n-3 PUFAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), may have differential effects in ulcerative colitis (UC). It was aimed to clarify their differential effects in UC. METHODS Eight-week-old male C57BL/6J mice were randomly divided into 7 groups, namely control, UC model, salicylazosulfapyridine (SASP), low-dose DHA, high-dose DHA, low-dose EPA, and high-dose EPA. DHA, EPA and SASP treatment groups were orally treated accordingly for 9 weeks. During the 5th to 9th week the control group was given distilled water, while other groups were given distilled water with 2% dextran sodium sulfate (DSS) to induce UC. Body weight loss, diarrhea, and stool bleeding were recorded to calculate the disease activity index (DAI). The level of tight junction proteins Claudin-1 and Occludin, and cytokines including TNF-α, IL-6, and IL-1β as well as inflammatory cell markers such as MPO, F4/80, and MCP-1 in the intestinal epithelium were measured using western blotting. Activation of IL-6/STAT3 and NLRP3/IL-1β inflammatory pathways was also assessed. Levels of proliferation-related proteins of the Wnt/β-catenin pathway with c-myc, Cyclin-D1, and PCNA were detected. RESULTS EPA, superior to DHA, significantly attenuated DSS-induced colitis evidenced by reduced DAI scores, cytokine production and inflammatory cell infiltration. Mechanically, EPA triggered a marked up-regulation of Claudin-1 and Occludin with down-regulation of their up-stream Akt and ERK. EPA also inhibited NLRP3/IL-1β and IL-6/STAT3 inflammatory pathways and up-regulated the Wnt/β-catenin pathway. CONCLUSIONS EPA is more suitable to be used for the treatment of UC than DHA.
Collapse
Affiliation(s)
- Zhuangwei Zhang
- Institute of Preventative Medicine and Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, 315211 Zhejiang, China. and Department of Nutrition, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310000 Zhejiang, China
| | - Zhe Xue
- Institute of Preventative Medicine and Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, 315211 Zhejiang, China.
| | - Haitao Yang
- Department of Pathology, Mingzhou Hospital of Zhejiang University, Ningbo, 315040 Zhejiang, China
| | - Feng Zhao
- Institute of Nutrition and Health, Qingdao University, 266071 Qingdao, China
| | - Chundi Liu
- Central South University, Changsha, 410083 Hunan, China
| | - Jiahui Chen
- Institute of Preventative Medicine and Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, 315211 Zhejiang, China.
| | - Songtao Lu
- Institute of Preventative Medicine and Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, 315211 Zhejiang, China.
| | - Zuquan Zou
- Institute of Preventative Medicine and Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, 315211 Zhejiang, China.
| | - Yuping Zhou
- Department of Gastroenterology, Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315040 Zhejiang, China
| | - Xiaohong Zhang
- Institute of Preventative Medicine and Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, School of Medicine, Ningbo University, Ningbo, 315211 Zhejiang, China.
| |
Collapse
|
15
|
Omega-6 DPA and its 12-lipoxygenase-oxidized lipids regulate platelet reactivity in a nongenomic PPARα-dependent manner. Blood Adv 2021; 4:4522-4537. [PMID: 32946570 DOI: 10.1182/bloodadvances.2020002493] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022] Open
Abstract
Arterial thrombosis is the underlying cause for a number of cardiovascular-related events. Although dietary supplementation that includes polyunsaturated fatty acids (PUFAs) has been proposed to elicit cardiovascular protection, a mechanism for antithrombotic protection has not been well established. The current study sought to investigate whether an omega-6 essential fatty acid, docosapentaenoic acid (DPAn-6), and its oxidized lipid metabolites (oxylipins) provide direct cardiovascular protection through inhibition of platelet reactivity. Human and mouse blood and isolated platelets were treated with DPAn-6 and its 12-lipoxygenase (12-LOX)-derived oxylipins, 11-hydroxy-docosapentaenoic acid and 14-hydroxy-docosapentaenoic acid, to assess their ability to inhibit platelet activation. Pharmacological and genetic approaches were used to elucidate a role for DPA and its oxylipins in preventing platelet activation. DPAn-6 was found to be significantly increased in platelets following fatty acid supplementation, and it potently inhibited platelet activation through its 12-LOX-derived oxylipins. The inhibitory effects were selectively reversed through inhibition of the nuclear receptor peroxisome proliferator activator receptor-α (PPARα). PPARα binding was confirmed using a PPARα transcription reporter assay, as well as PPARα-/- mice. These approaches confirmed that selectivity of platelet inhibition was due to effects of DPA oxylipins acting through PPARα. Mice administered DPAn-6 or its oxylipins exhibited reduced thrombus formation following vessel injury, which was prevented in PPARα-/- mice. Hence, the current study demonstrates that DPAn-6 and its oxylipins potently and effectively inhibit platelet activation and thrombosis following a vascular injury. Platelet function is regulated, in part, through an oxylipin-induced PPARα-dependent manner, suggesting that targeting PPARα may represent an alternative strategy to treat thrombotic-related diseases.
Collapse
|
16
|
Meyer BJ, Sparkes C, Sinclair AJ, Gibson RA, Else PL. Fingertip Whole Blood as an Indicator of Omega-3 Long-Chain Polyunsaturated Fatty Acid Changes during Dose-Response Supplementation in Women: Comparison with Plasma and Erythrocyte Fatty Acids. Nutrients 2021; 13:nu13051419. [PMID: 33922507 PMCID: PMC8147031 DOI: 10.3390/nu13051419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/14/2022] Open
Abstract
The sensitivity of fingertip whole blood to reflect habitual dietary and dose-dependent supplemental omega-3 long-chain polyunsaturated fatty acid (n-3 LCPUFA) intake in premenopausal women was compared to that of venous erythrocytes and plasma fatty acids. Samples were obtained from women in a randomised, double-blind, placebo-controlled trial in which premenopausal women (n = 53) were supplemented with DHA-rich tuna oil capsules and/or placebo (Sunola oil) capsules (6 capsules per day) for 8 weeks to achieve doses of either 0, 0.35, 0.7 or 1.05 g/day n-3 LCPUFA. All blood biomarkers were very similar in their ability to reflect dietary n-3 LCPUFA intake (r = 0.38–0.46 for EPA and DHA intake), and in their dose-dependent increases in n-3 LCPUFA levels after supplementation (R2 = 0.41–0.51 for dose effect on biomarker EPA and DHA levels (mol %)). Fingertip whole blood is an effective alternative to erythrocytes and plasma as a biomarker n-3 LCPUFA intake in premenopausal women.
Collapse
Affiliation(s)
- Barbara J. Meyer
- School of Medicine, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia; (C.S.); (P.L.E.)
- Illawarra Medical Research Institute, Wollongong, NSW 2522, Australia
- Correspondence: ; Tel.: +61-2-4221-3459
| | - Cassandra Sparkes
- School of Medicine, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia; (C.S.); (P.L.E.)
- Illawarra Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Andrew J. Sinclair
- Faculty of Health, Deakin University, Geelong, VIC 3220, Australia;
- Department of Nutrition, Dietetics and Food, Monash University, Notting Hill, VIC 3168, Australia
| | - Robert A. Gibson
- Faculty of Sciences, School of Agriculture, Food and Wine, University of Adelaide, Adelaide, SA 5005, Australia;
| | - Paul L. Else
- School of Medicine, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia; (C.S.); (P.L.E.)
- Illawarra Medical Research Institute, Wollongong, NSW 2522, Australia
| |
Collapse
|
17
|
Ghasemi Fard S, Cameron-Smith D, Sinclair AJ. n - 3 Docosapentaenoic acid: the iceberg n - 3 fatty acid. Curr Opin Clin Nutr Metab Care 2021; 24:134-138. [PMID: 33315722 DOI: 10.1097/mco.0000000000000722] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW Docosapentaenoic acid (DPA) is a minor omega-3 fatty acid (FA) which has been frequently overlooked in lipid research. This review examines the biochemical and physiological outcomes of human trials which have used pure preparations of DPA (n - 3 DPA) and also recent developments in specialized proresolving lipid mediators (SPMs) derived from n - 3 DPA. RECENT FINDINGS There have been only been two human studies and eleven animal studies with pure n - 3 DPA. The doses of n - 3 DPA used in the human trials have been 1-2 g/day. n - 3 DPA abundance is increased in blood lipid fractions within 3-4 days of supplementation. n - 3 DPA has the potential for unique properties, with a greater similarity in biological functioning with docosahexaenoic acid (DHA), than eicosapentaenoic acid (EPA). Despite the typically low levels of n - 3 DPA in most tissue lipids relative to EPA and DHA, unique SPMs, such as resolvins, maresins and protectins of the n - 3 DPA type, are involved in resolution of inflammation and regulating immune function. SUMMARY We suggest that measurement of blood levels of n - 3 DPA gives no indication of its broad biological roles, but that the true functionality of this enigmatic n - 3 polyunsaturated fatty acid (PUFA) remains obscure until more is known about the properties of the unique DPA-derived SPMs.
Collapse
Affiliation(s)
- Samaneh Ghasemi Fard
- School of Life and Environmental Sciences, Deakin University, Melbourne, Victoria, Australia
| | - David Cameron-Smith
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A∗STAR), Brenner Centre for Molecular Medicine, Singapore, Singapore
| | - Andrew J Sinclair
- Department of Nutrition, Dietetics and Food, Monash University
- Faculty of Health, Deakin University, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Lamon-Fava S, So J, Mischoulon D, Ziegler TR, Dunlop BW, Kinkead B, Schettler PJ, Nierenberg AA, Felger JC, Maddipati KR, Fava M, Rapaport MH. Dose- and time-dependent increase in circulating anti-inflammatory and pro-resolving lipid mediators following eicosapentaenoic acid supplementation in patients with major depressive disorder and chronic inflammation. Prostaglandins Leukot Essent Fatty Acids 2021; 164:102219. [PMID: 33316626 PMCID: PMC7855824 DOI: 10.1016/j.plefa.2020.102219] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Eicosapentaenoic acid (EPA) supplementation is an effective treatment option in major depressive disorder (MDD) associated with chronic low-grade inflammation. EPA is the precursor of specialized pro-resolving lipid mediators (SPMs) termed resolvins (Rv), that serve important roles in the resolution of inflammation. The objective of this study was to assess the effects of different doses of EPA on plasma concentrations of EPA metabolites and SPMs in MDD patients. METHODS In a 2-site study, 61 MDD patients with body mass index >25 kg/m2 and serum high-sensitivity C-reactive protein ≥3 μg/mL were enrolled in a 12-week randomized trial comparing EPA 1, 2, and 4 g/d to placebo. Plasma EPA (mol%) and SPMs (pg/mL) were measured in 42 study completers at baseline and at the end of treatment by liquid chromatography/mass spectrometry. RESULTS Plasma EPA and SPM concentrations did not change in the placebo group during 12 weeks of treatment. Plasma EPA and EPA-derived metabolites increased significantly and dose-dependently in all EPA supplementation arms. The increase in 18-hydroxyeicosapentaenoic acid (18-HEPE), the precursor of RvE1-3, was significantly greater with the 4 g/d EPA dose than the other doses from week 4 to 12. RvE1 was undetected in all treatment groups, while RvE2 was detected in half of the subjects both at baseline and after treatment, with dose-dependent increases. RvE3 was detected only after supplementation, dose-dependently. A significant reduction in plasma arachidonic acid (AA), relative to baseline, was observed in all EPA arms. This was in contrast with an increase in AA-derived SPM lipoxin B4 (LXB4) in the 4 g/d arm. CONCLUSIONS Our results show a robust effect of EPA 4 g/d supplementation in increasing plasma EPA and 18-HEPE levels, associated with improved conversion to RvE2-3, and LXB4 levels.
Collapse
Affiliation(s)
- Stefania Lamon-Fava
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA.
| | - Jisun So
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - David Mischoulon
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | | | - Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA
| | - Becky Kinkead
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA
| | - Pamela J Schettler
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA
| | | | - Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA
| | - Krishna Rao Maddipati
- Department of Pathology, Lipidomics Core Facility, Wayne State University, Detroit, MI
| | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Mark Hyman Rapaport
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA
| |
Collapse
|
19
|
Hsu MC, Huang YS, Ouyang WC. Beneficial effects of omega-3 fatty acid supplementation in schizophrenia: possible mechanisms. Lipids Health Dis 2020; 19:159. [PMID: 32620164 PMCID: PMC7333328 DOI: 10.1186/s12944-020-01337-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/24/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Schizophrenia is a serious long-term psychotic disorder marked by positive and negative symptoms, severe behavioral problems and cognitive function deficits. The cause of this disorder is not completely clear, but is suggested to be multifactorial, involving both inherited and environmental factors. Since human brain regulates all behaviour, studies have focused on identifying changes in neurobiology and biochemistry of brain in schizophrenia. Brain is the most lipid rich organ (approximately 50% of brain dry weight). Total brain lipids is constituted of more than 60% of phospholipids, in which docosahexaenoic acid (DHA, 22:6n-3) is the most abundant (more than 40%) polyunsaturated fatty acid (PUFA) in brain membrane phospholipids. Results from numerous studies have shown significant decreases of PUFAs, in particular, DHA in peripheral blood (plasma and erythrocyte membranes) as well as brain of schizophrenia patients at different developmental phases of the disorder. PUFA deficiency has been associated to psychotic symptoms and cognitive deficits in schizophrenia. These findings have led to a number of clinical trials examining whether dietary omega-3 fatty acid supplementation could improve the course of illness in patients with schizophrenia. Results are inconsistent. Some report beneficial whereas others show not effective. The discrepancy can be attributed to the heterogeneity of patient population. METHODS In this review, results from recent experimental and clinical studies, which focus on illustrating the role of PUFAs in the development of schizophrenia were examined. The rationale why omega-3 supplementation was beneficial on symptoms (presented by subscales of the positive and negative symptom scale (PANSS), and cognitive functions in certain patients but not others was reviewed. The potential mechanisms underlying the beneficial effects were discussed. RESULTS Omega-3 fatty acid supplementation reduced the conversion rate to psychosis and improved both positive and negative symptoms and global functions in adolescents at ultra-high risk for psychosis. Omega-3 fatty acid supplementation could also improve negative symptoms and global functions in the first-episode patients with schizophrenia, but improve mainly total or general PANSS subscales in chronic patients. Patients with low PUFA (particularly DHA) baseline in blood were more responsive to the omega-3 fatty acid intervention. CONCLUSION Omega-3 supplementation is more effective in reducing psychotic symptom severity in young adults or adolescents in the prodromal phase of schizophrenia who have low omega-3 baseline. Omega-3 supplementation was more effective in patients with low PUFA baseline. It suggests that patients with predefined lipid levels might benefit from lipid treatments, but more controlled clinical trials are warranted.
Collapse
Affiliation(s)
- Mei-Chi Hsu
- Department of Nursing, I-Shou University, No.8, Yida Road, Jiaosu Village Yanchao District, Kaohsiung, 82445 Taiwan
| | - Yung-Sheng Huang
- College of Medicine, I-Shou University, No.8, Yida Road, Jiaosu Village Yanchao District, Kaohsiung, 82445 Taiwan
| | - Wen-Chen Ouyang
- Department of Geriatric Psychiatry, Jianan Psychiatric Center, Ministry of Health and Welfare, No.539, Yuzhong Rd., Rende Dist., Tainan City, 71742 Taiwan
- Department of Nursing, Shu-Zen Junior College of Medicine and Management, No.452, Huanqiu Rd. Luzhu Dist, Kaohsiung, 82144 Taiwan
- Department of Psychiatry, College of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist., Kaohsiung, 80708 Taiwan
| |
Collapse
|
20
|
High Variability in Erythrocyte, Plasma and Whole Blood EPA and DHA Levels in Response to Supplementation. Nutrients 2020; 12:nu12041017. [PMID: 32276315 PMCID: PMC7231102 DOI: 10.3390/nu12041017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/15/2022] Open
Abstract
(1) Aim: the aim of this secondary analysis was to report the variability in response to n-3 long chain polyunsaturated fatty acids (LCPUFA) supplementation in erythrocytes, plasma and whole blood of a previously published dose response study. (2) Methods: a randomized, double-blind, placebo-controlled trial of parallel design was conducted, whereby pre-menopausal women were randomly assigned to consume 0, 0.35, 0.7 or 1 g/day of supplemental eicosapentaenoic acid (EPA) plus docosahexaenoic acid (DHA). Fasted blood samples were taken at baseline and after eight weeks intervention. Erythrocyte, plasma and whole blood fatty acids were extracted using the method of Lepage and Roy and analysed using gas chromatography. (3) Results: There were significant increases in EPA plus DHA levels in the 0.7 g and 1 g dose groups, with the highest increase with the 1 g dose notably: in erythrocytes (from 5.69% to 7.59%), plasma (from 2.94% to 5.48%) and in whole blood (from 3.81% to 6.03%). There was high variability in response to the supplement in erythrocytes, plasma and whole blood across the different doses. (4) Conclusion: there is high individual variability in n-3 LCPUFA levels in response to n-3 LCPUFA supplementation, which should be taken into account in clinical trials using n-3 LCPUFA supplements.
Collapse
|