1
|
Lu J, Liu R, Ren H, Wang S, Hu C, Shi Z, Li M, Liu W, Wan Q, Su Q, Li Q, Zheng H, Qu S, Yang F, Ji H, Lin H, Qi H, Wu X, Wu K, Chen Y, Xu Y, Xu M, Wang T, Zheng J, Ning G, Zheng R, Bi Y, Zhong H, Wang W. Impact of omega-3 fatty acids on hypertriglyceridemia, lipidomics, and gut microbiome in patients with type 2 diabetes. MED 2025; 6:100496. [PMID: 39163858 DOI: 10.1016/j.medj.2024.07.024] [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: 12/05/2023] [Revised: 05/14/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND Fish oil (FO), a mixture of omega-3 fatty acids mainly comprising docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), has been recommended for patients with type 2 diabetes (T2D) and hypertriglyceridemia. However, its effects on lipidomic profiles and gut microbiota and the factors influencing triglyceride (TG) reduction remain unclear. METHODS We conducted a 12-week, randomized, double-blind, placebo-controlled trial in 309 Chinese patients with T2D with hypertriglyceridemia (ClinicalTrials.gov: NCT03120299). Participants were randomly assigned (1:1) to receive either 4 g FO or corn oil for 12 weeks. The primary outcome was changes in serum TGs and the lipidomic profile, and the secondary outcome included changes in the gut microbiome and other metabolic variables. FINDINGS The FO group had significantly better TG reduction (mean [95% confidence interval (CI)]: -1.51 [-2.01, -1.01] mmol/L) compared to the corn oil group (-0.66 [-1.15, -0.16] mmol/L, p = 0.02). FO significantly altered the serum lipid profile by reducing low-unsaturated TG species and increasing those containing DHA or EPA. FO had minor effects on gut microbiota, while baseline microbial features predicted the TG response to FO better than phenotypic or lipidomic features, potentially mediated by specific lipid metabolites. A total of 9 lipid metabolites significantly mediated the link between 4 baseline microbial variables and the TG response to FO supplementation. CONCLUSIONS Our findings demonstrate differential impacts of omega-3 fatty acids on lipidomic and microbial profiles in T2D and highlight the importance of baseline gut microbiota characteristics in predicting the TG-lowering efficacy of FO. FUNDING This study was funded by the National Nature Science Foundation.
Collapse
Affiliation(s)
- Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 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
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 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
| | - Huahui Ren
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, 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, Shanghai, 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
| | - Chunyan Hu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 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
| | - Zhun Shi
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, 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, Shanghai, 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
| | - Wei Liu
- Department of Endocrinology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Wan
- Department of Endocrine and Metabolic Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qifu Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Fangming Yang
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, 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, Shanghai, 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, Shanghai, 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, Shanghai, 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
| | - Kui Wu
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, 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, Shanghai, 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, Shanghai, 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, Shanghai, 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, Shanghai, 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, Shanghai, 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, Shanghai, 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, Shanghai, 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
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 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
| | - Huanzi Zhong
- BGI Research, Shenzhen, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, China.
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 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
|
2
|
Mullins VA, Snider JM, Michael B, Porter LR, Brinton RD, Chilton FH. Impact of fish oil supplementation on plasma levels of highly unsaturated fatty acid-containing lipid classes and molecular species in American football athletes. Nutr Metab (Lond) 2024; 21:43. [PMID: 38978004 PMCID: PMC11232345 DOI: 10.1186/s12986-024-00815-x] [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: 04/02/2024] [Accepted: 06/18/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Previous studies have linked sports-related concussions and repeated subconcussive head impacts in contact sport athletes to elevated brain injury biomarkers. Docosahexaenoic acid (DHA), the primary omega-3 (n-3) highly unsaturated fatty acid (HUFA) in the brain, has shown neuroprotective effects in animal models after brain injury, but clinical research has shown mixed results. METHODS We conducted a randomized, double-blind, placebo-controlled study on 29 Division 1 collegiate American football players, exploring the impact of DHA (2.5 g) and eicosapentaenoic acid (EPA) (1.0 g) supplied as ethyl esters, on levels of plasma lipids shown to cross the blood-brain barrier. Dietary intake data was collected using food frequency questionnaires (FFQ). Complex lipids and unesterified fatty acids were isolated from plasma, separated via reversed-phase liquid chromatography and analyzed by targeted lipidomics analysis. RESULTS FFQ results indicated that participants had low dietary n-3 HUFA intake and high omega-6 (n-6):n-3 polyunsaturated fatty acids (PUFA) and HUFA ratios at baseline. After DHA + EPA supplementation, plasma lysophosphatidylcholine (LPC) containing DHA and EPA significantly increased at all timepoints (weeks 17, 21, and 26; p < 0.0001), surpassing placebo at Weeks 17 (p < 0.05) and 21 (p < 0.05). Phosphatidylcholine (PC) molecular species containing DHA or EPA, PC38:6 PC36:6, PC38:7, PC40:6, and PC40:8, increased significantly in the DHA + EPA treatment group at Weeks 17 (and 21. Plasma concentrations of non-esterified DHA and EPA rose post-supplementation in Weeks 17 and 21. CONCLUSIONS This study demonstrates that n-3 HUFA supplementation, in the form of ethyl esters, increased the DHA and EPA containing plasma lipid pools the have the capacity to enrich brain lipids and the potential to mitigate the effects of sports-related concussions and repeated subconcussive head impacts. TRIAL REGISTRATION All deidentified data are available at ClinicalTrials.gov #NCT0479207.
Collapse
Affiliation(s)
- Veronica Anne Mullins
- School of Nutritional Sciences and Wellness, Bioscience Research Laboratory (BSRL), University of Arizona, Room 370, 1230 N Cherry Avenue, Tucson, AZ, 85719, USA
| | - Justin M Snider
- School of Nutritional Sciences and Wellness, Bioscience Research Laboratory (BSRL), University of Arizona, Room 370, 1230 N Cherry Avenue, Tucson, AZ, 85719, USA
- Center for Precision Nutrition and Wellness, University of Arizona, 1230 N Cherry Avenue, Tucson, AZ, 85719, USA
| | - Bryce Michael
- School of Nutritional Sciences and Wellness, Bioscience Research Laboratory (BSRL), University of Arizona, Room 370, 1230 N Cherry Avenue, Tucson, AZ, 85719, USA
| | - Lydia Rose Porter
- School of Nutritional Sciences and Wellness, Bioscience Research Laboratory (BSRL), University of Arizona, Room 370, 1230 N Cherry Avenue, Tucson, AZ, 85719, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, The University of Arizona Health Sciences, University of Arizona, 1230 N. Cherry Avenue, Tucson, AZ, 85719, USA
| | - Floyd H Chilton
- School of Nutritional Sciences and Wellness, Bioscience Research Laboratory (BSRL), University of Arizona, Room 370, 1230 N Cherry Avenue, Tucson, AZ, 85719, USA.
- Center for Precision Nutrition and Wellness, University of Arizona, 1230 N Cherry Avenue, Tucson, AZ, 85719, USA.
| |
Collapse
|
3
|
Ayats-Vidal R, Bosque-García M, Cordobilla B, Asensio-De la Cruz O, García-González M, Loureda-Pérez S, Fernández-López E, Robert-Barriocanal E, Valiente-Planas A, Domingo JC. Impact of 1-Year Supplementation with High-Rich Docosahexaenoic Acid (DHA) on Clinical Variables and Inflammatory Biomarkers in Pediatric Cystic Fibrosis: A Randomized Double-Blind Controlled Trial. Nutrients 2024; 16:970. [PMID: 38613004 PMCID: PMC11013158 DOI: 10.3390/nu16070970] [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: 02/14/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
A randomized, double-blind, and placebo-controlled study was conducted to assess the effect of dietary supplementation with high-rich docosahexaenoic acid (DHA) (Tridocosahexanoin-AOX® 70%) at 50 mg/kg/day in pediatric patients with cystic fibrosis (CF) as compared with placebo. The duration of supplementation was 12 months. A total of 22 patients were included, with 11 in the DHA group and 11 in the placebo group. The mean age was 11.7 years. The outcome variables were pulmonary function, exacerbations, sputum cellularity, inflammatory biomarkers in sputum and peripheral blood, and anthropometric variables. In the DHA group, there was a significant increase in FVC (p = 0.004) and FVE1 expressed in liters (p = 0.044) as compared with placebo, and a lower median number of exacerbations (1 vs. 2). Differences in sputum cellularity (predominantly neutrophilic), neutrophilic elastase, and sputum and serum concentrations of resolvin D1 (RvD1), interleukin (IL)-8 (IL-8), and tumor necrosis factor alpha (TNF-α) between the study groups were not found. Significant increases in weight and height were also observed among DHA-supplemented patients. The administration of the study product was safe and well tolerated. In summary, the use of a highly concentrated DHA supplement for 1 year as compared with placebo improved pulmonary function and reduced exacerbations in pediatric CF.
Collapse
Affiliation(s)
- Roser Ayats-Vidal
- Pediatric Allergies, Immunology and Pneumology Unit, Pediatric Medicine Service, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain; (M.B.-G.); (O.A.-D.l.C.); (M.G.-G.); (S.L.-P.)
| | - Montserrat Bosque-García
- Pediatric Allergies, Immunology and Pneumology Unit, Pediatric Medicine Service, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain; (M.B.-G.); (O.A.-D.l.C.); (M.G.-G.); (S.L.-P.)
| | - Begoña Cordobilla
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, E-08028 Barcelona, Spain;
| | - Oscar Asensio-De la Cruz
- Pediatric Allergies, Immunology and Pneumology Unit, Pediatric Medicine Service, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain; (M.B.-G.); (O.A.-D.l.C.); (M.G.-G.); (S.L.-P.)
| | - Miguel García-González
- Pediatric Allergies, Immunology and Pneumology Unit, Pediatric Medicine Service, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain; (M.B.-G.); (O.A.-D.l.C.); (M.G.-G.); (S.L.-P.)
| | - Susana Loureda-Pérez
- Pediatric Allergies, Immunology and Pneumology Unit, Pediatric Medicine Service, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain; (M.B.-G.); (O.A.-D.l.C.); (M.G.-G.); (S.L.-P.)
| | - Elena Fernández-López
- Physical Medicine and Rehabilitation Service, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain; (E.F.-L.); (E.R.-B.); (A.V.-P.)
| | - Eva Robert-Barriocanal
- Physical Medicine and Rehabilitation Service, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain; (E.F.-L.); (E.R.-B.); (A.V.-P.)
| | - Andrea Valiente-Planas
- Physical Medicine and Rehabilitation Service, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain; (E.F.-L.); (E.R.-B.); (A.V.-P.)
| | - Joan Carles Domingo
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, E-08028 Barcelona, Spain;
| |
Collapse
|
4
|
González-Meza GM, Elizondo-Luevano JH, Cuellar-Bermudez SP, Sosa-Hernández JE, Iqbal HMN, Melchor-Martínez EM, Parra-Saldívar R. New Perspective for Macroalgae-Based Animal Feeding in the Context of Challenging Sustainable Food Production. PLANTS (BASEL, SWITZERLAND) 2023; 12:3609. [PMID: 37896072 PMCID: PMC10610262 DOI: 10.3390/plants12203609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023]
Abstract
Food production is facing challenging times due to the pandemic, and climate change. With production expected to double by 2050, there is a need for a new paradigm in sustainable animal feed supply. Seaweeds offer a highly valuable opportunity in this regard. Seaweeds are classified into three categories: brown (Phaeophyceae), red (Rhodophyceae), and green (Chlorophyceae). While they have traditionally been used in aquafeed, their demand in the feed market is growing, parallelly increasing according to the food demand. Additionally, seaweeds are being promoted for their nutritional benefits, which contribute to the health, growth, and performance of animals intended for human consumption. Moreover, seaweeds contain biologically active compounds such as polyunsaturated fatty acids, antioxidants (polyphenols), and pigments (chlorophylls and carotenoids), which possess beneficial properties, including antibacterial, antifungal, antiviral, antioxidant, and anti-inflammatory effects and act as prebiotics. This review offers a new perspective on the valorization of macroalgae biomass due to their nutritional profile and bioactive components, which have the potential to play a crucial role in animal growth and making possible new sources of healthy food ingredients.
Collapse
Affiliation(s)
- Georgia M. González-Meza
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico; (G.M.G.-M.); (J.H.E.-L.); (J.E.S.-H.); (H.M.N.I.)
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | - Joel H. Elizondo-Luevano
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico; (G.M.G.-M.); (J.H.E.-L.); (J.E.S.-H.); (H.M.N.I.)
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | - Sara P. Cuellar-Bermudez
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico; (G.M.G.-M.); (J.H.E.-L.); (J.E.S.-H.); (H.M.N.I.)
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | - Juan Eduardo Sosa-Hernández
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico; (G.M.G.-M.); (J.H.E.-L.); (J.E.S.-H.); (H.M.N.I.)
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | - Hafiz M. N. Iqbal
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico; (G.M.G.-M.); (J.H.E.-L.); (J.E.S.-H.); (H.M.N.I.)
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | - Elda M. Melchor-Martínez
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico; (G.M.G.-M.); (J.H.E.-L.); (J.E.S.-H.); (H.M.N.I.)
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | - Roberto Parra-Saldívar
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico; (G.M.G.-M.); (J.H.E.-L.); (J.E.S.-H.); (H.M.N.I.)
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| |
Collapse
|
5
|
Monserrat-Mesquida M, Quetglas-Llabrés MM, Bouzas C, Pastor O, Ugarriza L, Llompart I, Cevallos-Ibarra K, Sureda A, Tur JA. Plasma Fatty Acid Composition, Oxidative and Inflammatory Status, and Adherence to the Mediterranean Diet of Patients with Non-Alcoholic Fatty Liver Disease. Antioxidants (Basel) 2023; 12:1554. [PMID: 37627549 PMCID: PMC10451635 DOI: 10.3390/antiox12081554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex and increasingly prevalent cardiometabolic disorder worldwide. As of today, NAFLD is a pathology without specific pharmacological treatment, with the Mediterranean diet (MedDiet) being the most widely used approach for its management. The objective of this study is to assess the effects of adherence to the Mediterranean diet on fatty acid plasma levels, as well as on the oxidative and inflammatory status of NAFLD patients. A total of 100 adult patients (40-60 years old) diagnosed with NAFLD and from the Balearic Islands, Spain, were classified into three groups according to their adherence to the MedDiet. Consumption was assessed using a validated 143-item semiquantitative Food Frequency Questionnaire. Food items (g/day) were categorised according to their processing using the NOVA system. Anthropometrics, blood pressure, aminotransferases, Dietary Inflammatory Index (DII), inflammatory biomarkers, and fatty acid levels were measured in the plasma of NAFLD patients. High adherence to the MedDiet is associated to a highly plant-based diet, low ultra-processed food (UPF) consumption, low intake of dietary lipids, low intake of animal fats, high intake of monounsaturated fatty acid (MUFA; mainly palmitoleic acid), low intake of saturated fatty acids (SFAs; practically all dietary SFAs), low intake of trans-fatty acids, high intake of omega-3 fatty acids (mainly eicosapentaenoic acid), a higher n-6:n-3 in ratio, low intake of omega-6 fatty acids, and a low level of interleukin-6 (IL-6). High adherence to the MedDiet is related to a better fatty acid profile in the plasma, fewer SFAs and more MUFA and polyunsaturated fatty acids (PUFAs), a plasma biochemical profile, better proinflammatory status, and decreased ultra-processed food consumption of NAFLD patients.
Collapse
Affiliation(s)
- Margalida Monserrat-Mesquida
- Research Group on Community Nutrition & Oxidative Stress, University of the Balearic Islands-IUNICS, E-07122 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Health Research Institute of Balearic Islands (IdISBa), E-07120 Palma de Mallorca, Spain
| | - Maria Magdalena Quetglas-Llabrés
- Research Group on Community Nutrition & Oxidative Stress, University of the Balearic Islands-IUNICS, E-07122 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Health Research Institute of Balearic Islands (IdISBa), E-07120 Palma de Mallorca, Spain
| | - Cristina Bouzas
- Research Group on Community Nutrition & Oxidative Stress, University of the Balearic Islands-IUNICS, E-07122 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Health Research Institute of Balearic Islands (IdISBa), E-07120 Palma de Mallorca, Spain
| | - Oscar Pastor
- Service of Clinical Biochemistry, Hospital Universitario Ramon y Cajal-IRYCIS, E-28023 Madrid, Spain (K.C.-I.)
| | - Lucía Ugarriza
- Research Group on Community Nutrition & Oxidative Stress, University of the Balearic Islands-IUNICS, E-07122 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Health Research Institute of Balearic Islands (IdISBa), E-07120 Palma de Mallorca, Spain
- C.S. Camp Redó, IBSalut, E-07010 Palma de Mallorca, Spain
| | - Isabel Llompart
- Research Group on Community Nutrition & Oxidative Stress, University of the Balearic Islands-IUNICS, E-07122 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Health Research Institute of Balearic Islands (IdISBa), E-07120 Palma de Mallorca, Spain
- Clinical Analysis Service, University Hospital Son Espases, E-07198 Palma de Mallorca, Spain
| | - Karla Cevallos-Ibarra
- Service of Clinical Biochemistry, Hospital Universitario Ramon y Cajal-IRYCIS, E-28023 Madrid, Spain (K.C.-I.)
| | - Antoni Sureda
- Research Group on Community Nutrition & Oxidative Stress, University of the Balearic Islands-IUNICS, E-07122 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Health Research Institute of Balearic Islands (IdISBa), E-07120 Palma de Mallorca, Spain
| | - Josep A. Tur
- Research Group on Community Nutrition & Oxidative Stress, University of the Balearic Islands-IUNICS, E-07122 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Health Research Institute of Balearic Islands (IdISBa), E-07120 Palma de Mallorca, Spain
| |
Collapse
|
6
|
Ayats-Vidal R, Bosque-García M, Cordobilla B, Asensio-De la Cruz O, García-González M, Castro-Marrero J, López-Rico I, Domingo JC. Changes of Erythrocyte Fatty Acids after Supplementation with Highly Concentrated Docosahexaenoic Acid (DHA) in Pediatric Cystic Fibrosis: A Randomized Double-Blind Controlled Trial. J Clin Med 2023; 12:jcm12113704. [PMID: 37297899 DOI: 10.3390/jcm12113704] [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: 05/03/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
We characterized the fatty acid profiles in the erythrocyte membrane of pediatric patients with cystic fibrosis (CF) receiving highly concentrated docosahexaenoic acid (DHA) supplementation (Tridocosahexanoin-AOX® 70%) at 50 mg/kg/day (n = 11) or matching placebo (n = 11) for 12 months. The mean age was 11.7 years. The DHA group showed a statistically significant improvement in n-3 polyunsaturated fatty acids (PUFAs), which was observed as early as 6 months and further increased at 12 months. Among the n-3 PUFAs, there was a significant increase in DHA and eicosapentaenoic acid (EPA). Additionally, a statistically significant decrease in n-6 PUFAs was found, primarily due to a decrease in arachidonic acid (AA) levels and elongase 5 activity. However, we did not observe any changes in linoleic acid levels. The long-term administration of DHA over one year was safe and well tolerated. In summary, the administration of a high-rich DHA supplement at a dose of 50 mg/kg/day for one year can correct erythrocyte AA/DHA imbalance and reduce fatty acid inflammatory markers. However, it is important to note that essential fatty acid alterations cannot be fully normalized with this treatment. These data provide timely information of essential fatty acid profile for future comparative research.
Collapse
Affiliation(s)
- Roser Ayats-Vidal
- Pediatric Allergies, Immunology and Pneumology Unit, Pediatric Medicine Service, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain
| | - Montserrat Bosque-García
- Pediatric Allergies, Immunology and Pneumology Unit, Pediatric Medicine Service, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain
| | - Begoña Cordobilla
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, E-08028 Barcelona, Spain
| | - Oscar Asensio-De la Cruz
- Pediatric Allergies, Immunology and Pneumology Unit, Pediatric Medicine Service, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain
| | - Miguel García-González
- Pediatric Allergies, Immunology and Pneumology Unit, Pediatric Medicine Service, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, Parc Taulí 1, E-08208 Sabadell, Spain
| | - Jesús Castro-Marrero
- ME/CFS Research Unit, Division of Rheumatology, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, E-08035 Barcelona, Spain
| | - Irene López-Rico
- Pharmacy Department, Institut d'Investigació I Innovació Parc Taulí (I3PT-CERCA), Parc Taulí Hospital Universitari, Universitat Autònoma de Barcelona, E-08208 Sabadell, Spain
| | - Joan Carles Domingo
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, E-08028 Barcelona, Spain
| |
Collapse
|
7
|
Wang H, Zhao W, Ding B, Zhang Y, Huang X, Liu X, Zuo R, Chang Y, Ding J. Comparative lipidomics profiling of the sea urchin, Strongylocentrotus intermedius. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 40:100900. [PMID: 34418782 DOI: 10.1016/j.cbd.2021.100900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 07/23/2021] [Accepted: 08/09/2021] [Indexed: 01/11/2023]
Abstract
Strongylocentrotus intermedius is an edible sea urchin and well-known for its nutritional value, such as a high content of polyunsaturated fatty acids (PUFAs). We carried out an untargeted lipidomics via high-resolution ultra-high-performance liquid chromatography - mass spectrometry (UPLC-MS) to highlight the features of the lipids profile of sea urchin gonad, which allowed for a more detailed interpretation of the accumulation of PUFAs with different abundances among sea urchins. For the first time, lipidomics profiling of lipid abundances in S. intermedius was demonstrated. We detected 11 PUFAs in sea urchin gonads, which represented >54.13% of the total fatty acid content. A total of 1552 lipid molecular species belonging to 36 lipid classes were identified. Lipidomics profiles data were analyzed using orthogonal partial least squares discriminant analysis (OPLS-DA) model and distinguished the PUFA abundances in both sexes of sea urchins. The significant differences in lipid molecules were highlighted and the major lipid classes identified were phosphatidylcholine (PC [19 species]) among females and triglycerides (TG [11 species]) among males. PC (42: 11) may be used as a potential marker for distinguishing high levels of PUFAs in sea urchin individuals, which as the result of the high level of PC (42:11). These data enrich the lipid profile library of aquatic products and provide a more reliable and refined biomarkers for the further research on fatty acid synthesis and metabolism in aquatic animals.
Collapse
Affiliation(s)
- Heng Wang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Wenfei Zhao
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Beichen Ding
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Yang Zhang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Xiaofang Huang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Xiaoyu Liu
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Rantao Zuo
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Yaqing Chang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Jun Ding
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
8
|
López de Las Hazas MC, Gil-Zamorano J, Cofán M, Mantilla-Escalante DC, Garcia-Ruiz A, Del Pozo-Acebo L, Pastor O, Yañez-Mo M, Mazzeo C, Serra-Mir M, Doménech M, Valls-Pedret C, Rajaram S, Sabaté J, Ros E, Sala-Vila A, Dávalos A. One-year dietary supplementation with walnuts modifies exosomal miRNA in elderly subjects. Eur J Nutr 2021; 60:1999-2011. [PMID: 32979076 DOI: 10.1007/s00394-020-02390-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE Epidemiological studies and clinical trials support the association of nut consumption with a lower risk of prevalent non-communicable diseases, particularly cardiovascular disease. However, the molecular mechanisms underlying nut benefits remain to be fully described. MicroRNAs (miRNAs) are post-transcriptional regulators of gene expression and play a pivotal role in health and disease. Exosomes are extracellular vesicles released from cells and mediate intercellular communication. Whether nut consumption modulates circulating miRNAs (c-miRNAs) transported in exosomes is poorly described. METHODS Cognitively healthy elderly subjects were randomized to either control (n = 110, abstaining from walnuts) or daily supplementation with walnuts (15% of their total energy, ≈30-60 g/day, n = 101) for 1-year. C-miRNAs were screened in exosomes isolated from 10 samples, before and after supplementation, and identified c-miRNA candidates were validated in the whole cohort. In addition, nanoparticle tracking analysis and lipidomics were assessed in pooled exosomes from the whole cohort. RESULTS Exosomal hsa-miR-32-5p and hsa-miR-29b-3p were consistently induced by walnut consumption. No major changes in exosomal lipids, nanoparticle concentration or size were found. CONCLUSION Our results provide novel evidence that certain c-miRNAs transported in exosomes are modulated by walnut consumption. The extent to which this finding contributes to the benefits of walnuts deserves further research.
Collapse
Affiliation(s)
- María-Carmen López de Las Hazas
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Judit Gil-Zamorano
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Montserrat Cofán
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Diana C Mantilla-Escalante
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Almudena Garcia-Ruiz
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Lorena Del Pozo-Acebo
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain
| | - Oscar Pastor
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
- Servicio de Bioquímica Clínica (UCA-CCM), Hospital Ramón y Cajal-IRYCIS, 28034, Madrid, Spain
| | - María Yañez-Mo
- Department of Molecular Biology, UAM, 28049, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049, Madrid, Spain
| | - Carla Mazzeo
- Department of Molecular Biology, UAM, 28049, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049, Madrid, Spain
| | - Mercè Serra-Mir
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
| | - Monica Doménech
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
| | - Cinta Valls-Pedret
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
| | - Sujatha Rajaram
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Joan Sabaté
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Emilio Ros
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d´Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Aleix Sala-Vila
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, 08003, Spain.
- Hospital del Mar Medical Research Institute, IMIM, Dr. Aiguader 88, 08003, Barcelona, Spain.
| | - Alberto Dávalos
- Laboratory of Epigenetics of Lipid Metabolism, Instituto Madrileño de Estudios Avanzados (IMDEA)-Alimentación, IMDEA Food Institute, CEI UAM+CSIC, Ctra. De Cantoblanco 8, 28049, Madrid, Spain.
| |
Collapse
|
9
|
The Antipsychotic Risperidone Alters Dihydroceramide and Ceramide Composition and Plasma Membrane Function in Leukocytes In Vitro and In Vivo. Int J Mol Sci 2021; 22:ijms22083919. [PMID: 33920193 PMCID: PMC8069118 DOI: 10.3390/ijms22083919] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 01/22/2023] Open
Abstract
Atypical or second-generation antipsychotics are used in the treatment of psychosis and behavioral problems in older persons with dementia. However, these pharmaceutical drugs are associated with an increased risk of stroke in such patients. In this study, we evaluated the effects of risperidone treatment on phospholipid and sphingolipid composition and lipid raft function in peripheral blood mononuclear cells (PBMCs) of older patients (mean age >88 years). The results showed that the levels of dihydroceramides, very-long-chain ceramides, and lysophosphatidylcholines decreased in PBMCs of the risperidone-treated group compared with untreated controls. These findings were confirmed by in vitro assays using human THP-1 monocytes. The reduction in the levels of very-long-chain ceramides and dihydroceramides could be due to the decrease in the expression of fatty acid elongase 3, as observed in THP-1 monocytes. Moreover, risperidone disrupted lipid raft domains in the plasma membrane of PBMCs. These results indicated that risperidone alters phospholipid and sphingolipid composition and lipid raft domains in PBMCs of older patients, potentially affecting multiple signaling pathways associated with these membrane domains.
Collapse
|
10
|
Picklo M, Vallée Marcotte B, Bukowski M, de Toro-Martín J, Rust BM, Guénard F, Vohl MC. Identification of Phenotypic Lipidomic Signatures in Response to Long Chain n-3 Polyunsaturated Fatty Acid Supplementation in Humans. J Am Heart Assoc 2021; 10:e018126. [PMID: 33461307 PMCID: PMC7955441 DOI: 10.1161/jaha.120.018126] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Supplementation with long chain n‐3 polyunsaturated fatty acids is used to reduce total circulating triacylglycerol (TAG) concentrations. However, in about 30% of people, supplementation with long chain n‐3 polyunsaturated fatty acids does not result in decreased plasma TAG. Lipidomic analysis may provide insight into this inter‐individual variability. Methods Lipidomic analyses using targeted, mass spectrometry were performed on plasma samples obtained from a clinical study in which participants were supplemented with 3 g/day of long chain n‐3 in the form of fish oil capsules over a 6‐week period. TAG species and cholesteryl esters (CE) were quantified for 130 participants pre‐ and post‐supplementation. Participants were segregated into 3 potential responder phenotypes: (1) positive responder (Rpos; TAG decrease), (2) non‐responder (Rnon; lacking TAG change), and (3) negative responder (Rneg; TAG increase) representing 67%, 18%, and 15% of the study participants, respectively. Separation of the 3 phenotypes was attributed to differential responses in TAG with 50 to 54 carbons with 1 to 4 desaturations. Elevated TAG with higher carbon number and desaturation were common to all phenotypes following supplementation. Using the TAG responder phenotype for grouping, decreases in total CE and specific CE occurred in the Rpos phenotype versus the Rneg phenotype with intermediate responses in the Rnon phenotype. CE 20:5, containing eicosapentaenoic acid (20:5n‐3), was elevated in all phenotypes. A classifier combining lipidomic and genomic features was built to discriminate triacylglycerol response phenotypes and reached a high predictive performance with a balanced accuracy of 75%. Conclusions These data identify lipidomic signatures, TAG and CE, associated with long chain n‐3 response p henotypes and identify a novel phenotype based upon CE changes. Registration URL: https://www.ClinicalTrials.gov; Unique Identifier: NCT01343342.
Collapse
Affiliation(s)
- Matthew Picklo
- USDA-ARS Grand Forks Human Nutrition Research Center Grand Forks ND
| | - Bastien Vallée Marcotte
- Centre Nutrition Santé et Société (NUTRISS) Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Université Laval Québec City QC Canada
| | - Michael Bukowski
- USDA-ARS Grand Forks Human Nutrition Research Center Grand Forks ND
| | - Juan de Toro-Martín
- Centre Nutrition Santé et Société (NUTRISS) Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Université Laval Québec City QC Canada
| | - Bret M Rust
- USDA-ARS Grand Forks Human Nutrition Research Center Grand Forks ND
| | - Frédéric Guénard
- Centre Nutrition Santé et Société (NUTRISS) Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Université Laval Québec City QC Canada
| | - Marie-Claude Vohl
- Centre Nutrition Santé et Société (NUTRISS) Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Université Laval Québec City QC Canada
| |
Collapse
|
11
|
Turck D, Castenmiller J, De Henauw S, Hirsch‐Ernst KI, Kearney J, Maciuk A, Mangelsdorf I, McArdle HJ, Naska A, Pelaez C, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Cubadda F, Frenzel T, Heinonen M, Marchelli R, Neuhäuser‐Berthold M, Poulsen M, Maradona MP, Schlatter JR, van Loveren H, Turla E, Knutsen HK. Safety of oil from Schizochytrium limacinum (strain FCC-3204) for use in food supplements as a novel food pursuant to Regulation (EU) 2015/2283. EFSA J 2021; 19:e06345. [PMID: 33505527 PMCID: PMC7814402 DOI: 10.2903/j.efsa.2021.6345] [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] [Indexed: 11/11/2022] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver an opinion on the safety of Schizochytrium sp. oil as a novel food (NF) pursuant to Regulation (EU) 2015/2283. Schizochytrium sp. is a single-cell microalga. The strain FCC-3204, used by the applicant (Fermentalg), belongs to the species Schizochytrium limacinum. The NF, an oil rich in docosahexaenoic acid (DHA), is obtained from microalgae after enzymatic lysis. The applicant proposed to increase the use level of the NF as a food supplement, from 250 mg DHA/day (currently authorised for the general population, excluding pregnant and lactating women) to 3 g DHA/day for adults, excluding pregnant and lactating women. S. limacinum was attributed the qualified presumption of safety (QPS) status with the qualification 'for production purposes only'. Data provided by the applicant demonstrated the absence of viable cells in the NF. No toxicological studies were performed with the NF. However, based on the available toxicological data on oils derived from Schizochytrium sp., the QPS status of the source of the NF, the production process, the composition of the NF and the absence of viable cells in the NF, the Panel considers there are no concerns with regard to toxicity of the NF. The Panel considers that the data provided by the applicant are not sufficient to conclude on the safety of the NF at the proposed uses (3 g DHA/day as a food supplement) in adults. However, in 2012, the Panel concluded that supplemental intakes of DHA alone up to about 1 g/day do not raise safety concerns for the general population. The Panel concludes that the NF is safe for the use in food supplements at the maximum intake level of 1 g DHA/day for the target population (adults, excluding pregnant and lactating women).
Collapse
|
12
|
Amaral MD. How to determine the mechanism of action of CFTR modulator compounds: A gateway to theranostics. Eur J Med Chem 2020; 210:112989. [PMID: 33190956 DOI: 10.1016/j.ejmech.2020.112989] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022]
Abstract
The greatest challenge of 21st century biology is to fully understand mechanisms of disease to drive new approaches and medical innovation. Parallel to this is the huge biomedical endeavour of treating people through personalized medicine. Until now all CFTR modulator drugs that have entered clinical trials have been genotype-dependent. An emerging alternative is personalized/precision medicine in CF, i.e., to determine whether rare CFTR mutations respond to existing (or novel) CFTR modulator drugs by pre-assessing them directly on patient's tissues ex vivo, an approach also now termed theranostics. To administer the right drug to the right person it is essential to understand how drugs work, i.e., to know their mechanism of action (MoA), so as to predict their applicability, not just in certain mutations but also possibly in other diseases that share the same defect/defective pathway. Moreover, an understanding the MoA of a drug before it is tested in clinical trials is the logical path to drug discovery and can increase its chance for success and hence also approval. In conclusion, the most powerful approach to determine the MoA of a compound is to understand the underlying biology. Novel large datasets of intervenients in most biological processes, namely those emerging from the post-genomic era tools, are available and should be used to help in this task.
Collapse
Affiliation(s)
- Margarida D Amaral
- BioISI - Biosystems & Integrative Sciences Institute, Lisboa, Faculty of Sciences, University of Lisboa, Portugal.
| |
Collapse
|
13
|
Abstract
BACKGROUND Studies suggest that a diet rich in omega-3 essential fatty acids may have beneficial anti-inflammatory effects for chronic conditions such as cystic fibrosis. This is an updated version of a previously published review. OBJECTIVES To determine whether there is evidence that omega-3 polyunsaturated fatty acid supplementation reduces morbidity and mortality and to identify any adverse events associated with supplementation. SEARCH METHODS We searched the Cochrane Cystic Fibrosis and Genetic Disorders Group's Trials Register comprising references identified from comprehensive electronic database searches and handsearches of relevant journals and abstract books of conference proceedings. Date of last search: 01 April 2020. We also searched online study registries and contacted authors. Date of last search: 12 February 2020. SELECTION CRITERIA Randomised controlled trials in people with cystic fibrosis comparing omega-3 fatty acid supplements with placebo. DATA COLLECTION AND ANALYSIS Two authors independently selected studies for inclusion, extracted data and assessed the risk of bias of the studies. The quality of the evidence was assessed using GRADE. MAIN RESULTS The searches identified 23 studies; five studies with 106 participants (children and adults) were included; duration of studies and interventions differed. Two studies compared omega-3 fatty acids to olive oil for six weeks; one study compared omega-3 fatty acids and omega-6 fatty acids to control capsules (customised fatty acid blends) for three months; one study compared a liquid dietary supplement containing omega-3 fatty acids to one without for six months; and one study compared omega-3 fatty acids to a placebo for 12 months. Three studies had a low risk of bias for randomisation, but the risk was unclear in the remaining two studies; all studies had an unclear risk of bias for allocation concealment. Three of the studies adequately blinded participants; the risk of bias for selective reporting was high in one study and unclear for four studies. Two studies reported the number of respiratory exacerbations. At three months, one study (43 participants) reported no change in antibiotic usage. At 12 months the second study (15 participants) reported a reduction in the number of pulmonary exacerbations and cumulative antibiotic days in the supplement group compared to the previous year (no data for the control group); very low-quality evidence means we are unsure whether supplementation has any effect on this outcome. With regards to adverse events, one six-week study (12 participants) reported no difference in diarrhoea between omega-3 or placebo capsules; the very low-quality evidence means we are unsure if supplementation has any effect on this outcome. Additionally, one study reported an increase in steatorrhoea requiring participants to increase their daily dose of pancreatic enzymes, but three studies had already increased pancreatic enzyme dose at study begin so as to reduce the incidence of steatorrhoea. One study (43 participants) reported stomach pains at three months (treatment or control group not specified). One six-week study (19 participants) reported three asthma exacerbations leading to exclusion of participants since corticosteroid treatment could affect affect essential fatty acid metabolism. Four studies reported lung function. One six-week study (19 participants) reported an increase in forced expiratory volume in one second (FEV1) (L) and forced vital capacity (FVC) (L), but the very low-quality evidence means we are unsure if supplementation has any effect on lung function. The remaining studies did not report any difference in lung function at three months (unit of measurement not specified) or at six months and one year (FEV1 % predicted and FVC % predicted). No deaths were reported in any of the five studies. Four studies reported clinical variables. One study reported an increase in Schwachman score and weight alongside a reduction in sputum volume with supplementation compared to placebo at three months (data not analysable). However, three studies reported no differences in either weight at six weeks, in body mass index (BMI) standard deviation (SD) score at six months (very low-quality evidence) or BMI Z score at 12 months. Three studies reported biochemical markers of fatty acid status. One study showed an increase from baseline in both EPA and DHA content of serum phospholipids in the omega-3 group compared to placebo at three months and also a significant decrease in n-6/n-3 ratio in the supplement group compared to placebo; since the quality of the evidence is very low we are not certain that these changes are due to supplementation. One six-month cross-over study showed a higher EPA content of the neutrophil membrane in the supplement group compared to the placebo group, but, no difference in DHA membrane concentration. Furthermore, the leukotriene B4 to leukotriene B5 ratio was lower at six months in the omega-3 group compared to placebo. A one-year study reported a greater increase in the essential fatty acid profile and a decrease in AA levels in the treatment arm compared to placebo. AUTHORS' CONCLUSIONS This review found that regular omega-3 supplements may provide some limited benefits for people with cystic fibrosis with relatively few adverse effects: however, the quality of the evidence across all outcomes was very low. The current evidence is insufficient to draw firm conclusions or recommend routine use of these supplements in people with cystic fibrosis. A large, long-term, multicentre, randomised controlled study is needed to determine any significant therapeutic effect and to assess the influence of disease severity, dosage and duration of treatment. Future researchers should note the need for additional pancreatic enzymes when providing omega-3 supplementation or olive oil placebo capsules. More research is required to determine the exact dose of pancreatic enzyme required.
Collapse
Affiliation(s)
- Helen Watson
- Cambridge University Hospitals NHS Foundation TrustAddenbrookes HospitalCambridge Biomedical Campus, Hills RoadCambridgeUKCB2 0QQ
| | - Caroline Stackhouse
- Royal Papworth Hospital NHS Foundation TrustDietetics and Speech and Language TherapyPapworth RoadCambridge Biomedical CampusCambridgeUKCB2 0AY
| | | |
Collapse
|