1
|
Kim J, Cho HJ, Kim Z, Youn HJ, Cho J, Min JW, Kim YS, Lee JE. Intakes of saturated and unsaturated fat and circulating levels of inflammatory markers among breast cancer survivors. Sci Rep 2025; 15:9481. [PMID: 40108240 PMCID: PMC11923270 DOI: 10.1038/s41598-025-92951-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
We investigated whether dietary intakes of saturated fat (SFA), monounsaturated fat (MUFA), or polyunsaturated fat (PUFA) were associated with plasma inflammatory markers among breast cancer survivors in Korea. This cross-sectional study included 419 female breast cancer survivors aged 30 to 78 years. Dietary intake was assessed using 3-day dietary records (DRs) or food frequency questionnaires (FFQs). Plasma levels of adiponectin, high-sensitivity C-reactive protein (hs-CRP), interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α were measured. We calculated an inflammatory composite score by summing the z-scores of each inflammatory marker, with adiponectin assigned a negative sign. Least-squares means (LS-means) and 95% confidence intervals (CIs) of inflammatory markers were estimated according to SFA, MUFA, and PUFA intakes using the generalized linear models. We found that increasing dietary MUFA intake was associated with increasing levels of adiponectin, but decreasing levels of hs-CRP (p for trend = 0.042 and 0.032, respectively). Similarly, higher dietary PUFA intake was associated with higher levels of adiponectin (p for trend = 0.023), but lower levels of hs-CRP and inflammatory composite score (p for trend < 0.001 and 0.036, respectively). However, no significant associations were found between SFA intake and plasma inflammatory markers. In conclusion, our results suggest that a higher intake of MUFA or PUFA is associated with a more favorable inflammatory profile among Korean female breast cancer survivors, which may potentially help in managing chronic inflammation.
Collapse
Affiliation(s)
- Jiwoo Kim
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Hyun Jeong Cho
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Zisun Kim
- Department of Surgery, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Hyun Jo Youn
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Jihyoung Cho
- Department of Surgery, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Jun Won Min
- Department of Surgery, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Yoo Seok Kim
- Department of Surgery, Chosun University College of Medicine, Gwangju, Republic of Korea
| | - Jung Eun Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Albardan L, Platat C, Kalupahana NS. Role of Omega-3 Fatty Acids in Improving Metabolic Dysfunctions in Polycystic Ovary Syndrome. Nutrients 2024; 16:2961. [PMID: 39275277 PMCID: PMC11397015 DOI: 10.3390/nu16172961] [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: 08/12/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/16/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder that impacts women of reproductive age. In addition to reproductive and psychological complications, women with PCOS are also at a higher risk of developing metabolic diseases such as obesity, type 2 diabetes and cardiovascular disease. While weight reduction can help manage these complications in overweight or obese women, many weight loss interventions have been ineffective due to weight stigma and its psychological impact on women with PCOS. Therefore, exploring alternative dietary strategies which do not focus on weight loss per se is of importance. In this regard, omega-3 polyunsaturated fatty acids of marine origin (n-3 PUFAs), which are known for their hypotriglyceridemic, cardioprotective and anti-inflammatory effects, have emerged as a potential therapy for prevention and reversal of metabolic complications in PCOS. Several clinical trials showed that n-3 PUFAs can improve components of metabolic syndrome in women with PCOS. In this review, we first summarize the available clinical evidence for different dietary patterns in improving PCOS complications. Next, we summarize the clinical evidence for n-3 PUFAs for alleviating metabolic complications in PCOS. Finally, we explore the mechanisms by which n-3 PUFAs improve the metabolic disorders in PCOS in depth.
Collapse
Affiliation(s)
| | | | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
3
|
Seike M, Ashida H, Yamashita Y. Dietary flaxseed oil induces production of adiponectin in visceral fat and prevents obesity in mice. Nutr Res 2024; 121:16-27. [PMID: 38039598 DOI: 10.1016/j.nutres.2023.11.004] [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: 08/30/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 12/03/2023]
Abstract
Induction of obesity by dietary fats and oils differs according to the type of fat. Adiponectin is believed to be related to obesity prevention. We hypothesized that flaxseed oil is important for preventing obesity and producing adiponectin. To clarify this hypothesis, we investigated the relationship between obesity and different fat sources in mice fed diets with 6 types of fat and oils. C57BL/6J mice were given a control diet containing 5% corn oil or a high-fat diet containing 20% of either lard, palm oil, rapeseed oil, oleate-rich safflower oil, corn oil, or flaxseed oil for 14 weeks. In another experiment, mice were given a control diet and rosiglitazone (10 mg/kg body weight) by oral gavage for 1 week. At the end of study, plasma adiponectin and expression of fatty acid metabolism-related factors in white and brown adipose tissue and the liver were measured. Dietary flaxseed oil, which is rich in α-linolenic acid, did not induce obesity. Flaxseed oil resulted in increased β-oxidation-related factors in epididymal white adipose tissue, decreased fatty acid synthesis-related factors in the liver, and thermogenesis-related factor in brown adipose tissue following increase of plasma adiponectin. The results suggested that increase in plasma adiponectin after intake of flaxseed oil may be due to altered expression of AdipoQ and peroxisome proliferator-activated receptor γ in epididymal white adipose tissue. Flaxseed oil increased expression of adiponectin in visceral fat and regulated obesity-controlling fatty acid metabolism-related factors in white adipose tissue and liver, and thermogenesis-related factor in brown adipose tissue.
Collapse
Affiliation(s)
- Midori Seike
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan
| | - Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, 657-8501, Japan.
| |
Collapse
|
4
|
Lu LW, Quek SY, Lu SP, Chen JH. Potential Benefits of Omega-3 Polyunsaturated Fatty Acids (N3PUFAs) on Cardiovascular Health Associated with COVID-19: An Update for 2023. Metabolites 2023; 13:630. [PMID: 37233671 PMCID: PMC10222821 DOI: 10.3390/metabo13050630] [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: 04/04/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
The accumulating literature demonstrates that omega-3 polyunsaturated fatty acid (n-3 polyunsaturated fatty acid, N3PUFA) can be incorporated into the phospholipid bilayer of cell membranes in the human body to positively affect the cardiovascular system, including improving epithelial function, decreasing coagulopathy, and attenuating uncontrolled inflammatory responses and oxidative stress. Moreover, it has been proven that the N3PUFAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are precursors of some potent endogenous bioactive lipid mediators that mediate some favorable effects attributed to their parent substances. A dose-response relationship between increased EPA and DHA intake and reduced thrombotic outcomes has been reported. The excellent safety profile of dietary N3PUFAs makes them a prospective adjuvant treatment for people exposed to a higher risk of cardiovascular problems associated with COVID-19. This review presented the potential mechanisms that might contribute to the beneficial effects of N3PUFA and the optimal form and dose applied.
Collapse
Affiliation(s)
- Louise Weiwei Lu
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Siew-Young Quek
- Food Science, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand;
- Riddet Institute, New Zealand Centre of Research Excellence for Food Research, Palmerston North 4474, New Zealand
| | - Shi-Ping Lu
- Pharma New Zealand PNZ Limited, Hamilton 3210, New Zealand;
| | - Jie-Hua Chen
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China;
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
5
|
Zhao J, Sui X, Shi Q, Su D, Lin Z. Effects of antioxidant intervention in patients with polycystic ovarian syndrome: A systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e30006. [PMID: 35960093 PMCID: PMC9371494 DOI: 10.1097/md.0000000000030006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The role of antioxidant intervention in polycystic ovary syndrome (PCOS) patients has been increasingly investigated in recent years. In order to further clarify whether antioxidant therapy is beneficial for PCOS patients and the emphasis of its effects, this study provides a systematic review and meta-analysis of randomized controlled trials examining the effect of antioxidant intervention on PCOS. METHODS Enrolled study designs related to antioxidant interventions and PCOS, published from 1999 to 2020, were searched from EMBASE, PubMed, and Web of Science databases to sort out proven studies on antioxidant interventions and PCOS. Data were reported as weighted mean difference (WMD) or standard mean difference with associated confidence intervals of 95%. The analysis was conducted using Stata version 16.0. RESULTS Twenty-three studies were included in total. Antioxidant intervention had a positive impact on homeostasis model assessment of insulin resistance (WMD = -0.37, P = .011) and Triglycerides (WMD = -25.51, P < .001). And antioxidant intervention did not improve testosterone levels significantly (WMD = -0.20, P = .2611). Subgroup analysis showed that except for the D-chiro-inosito subgroup, no difference in body mass index was observed between the intervention group and the control group. CONCLUSIONS This meta-analysis demonstrates the efficacy of antioxidant intervention in patients with PCOS, demonstrating that antioxidant intervention has a significant effect on insulin resistance and lipid metabolism improvement. However, antioxidant intervention therapy has no discernible impact on testosterone levels or body mass index. Omega-3 may be a more effective antioxidant intervention for PCOS. In addition, this meta-analysis provides important reference opinions and treatment recommendations for PCOS.
Collapse
Affiliation(s)
- Junde Zhao
- Shandong University of Traditional Chinese Medicine, JinanShandong, China
| | - Xiaohui Sui
- Shandong University of Traditional Chinese Medicine, JinanShandong, China
| | - Qingyu Shi
- Shandong University of Finance and Economics, JinanShandong, China
| | - Dan Su
- People’s Hospital of Lixia District of Jinan, Jinan, Shandong, China
| | - Zhiheng Lin
- Shandong University of Traditional Chinese Medicine, JinanShandong, China
- *Correspondence: Zhiheng Lin, Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Lixia District, Jinan, Shandong, China (e-mail: )
| |
Collapse
|
6
|
Shi H, Ter Horst R, Nielen S, Bloemendaal M, Jaeger M, Joosten I, Koenen H, Joosten LAB, Schweren LJS, Vasquez AA, Netea MG, Buitelaar J. The gut microbiome as mediator between diet and its impact on immune function. Sci Rep 2022; 12:5149. [PMID: 35338162 PMCID: PMC8956630 DOI: 10.1038/s41598-022-08544-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/18/2022] [Indexed: 12/22/2022] Open
Abstract
Dietary habits may affect inflammatory status in humans. Here we explore this interaction as well as the potential mediating role of the gut microbiome (GM), given that the GM is both involved in processing of dietary components and influences the immune system. A cross-sectional analysis of a sample of 482 healthy participants (207 males and 275 females) was performed. Dietary intake was assessed by a semiquantitative food questionnaire. Adipokines and soluble inflammatory mediators were assayed with multiple immunoassays and ELISA. Microbial DNA was extracted from frozen stool samples of 471 participants. Polychoric correlation analysis was used to establish dietary patterns, and joint multivariate associations between these dietary patterns and immune biomarkers were studied using regression analyses with adjustment for sex, age, BMI, smoking, education levels and physical exercise and other dietary patterns. Non-parametric entropy mediation was applied to investigate whether diet-immune relationships are mediated by abundance of microbial species. In this cohort, we identified three dietary patterns, characterized as “high-meat” (meat and sweetened drink), “prudent diet” (fish, fruit, legumes and vegetables) and “high alcohol” (higher alcohol consumption). Higher adherence to prudent diet was associated with a higher adiponectin level. The high alcohol pattern was associated with high concentrations of circulating concentrations of pro-inflammatory markers (CRP, IL-6, VEGF). Dialister invisus was found to mediate the relationship between a prudent dietary pattern and adiponectin, AAT, CRP, IL-6, and VEGF. In conclusion, a meat-based diet and a diet with high alcohol consumption were associated with high concentrations of biomarkers of chronic low-grade inflammation, and conversely, a prudent diet was associated with anti-inflammatory biomarkers. Diet-inflammation regulation may differ between sexes. Mediation analyses revealed that the association between prudent diet and immune function was partially mediated by the GM. The study adds to our understanding of the associations between diet, the immune system and the GM in a healthy population.
Collapse
Affiliation(s)
- Huiqing Shi
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Radboudumc, Kapittelweg 29, 6525 EN, Nijmegen, Gelderland, The Netherlands.
| | - Rob Ter Horst
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Suzanne Nielen
- Department of Psychiatry, and Department of Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, 6525 EN, Nijmegen, Gelderland, The Netherlands
| | - Mirjam Bloemendaal
- Department of Psychiatry, and Department of Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, 6525 EN, Nijmegen, Gelderland, The Netherlands
| | - Martin Jaeger
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Irma Joosten
- Department of Laboratory Medicine, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Hans Koenen
- Department of Laboratory Medicine, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands
| | - Lizanne J S Schweren
- Interdisciplinary Center Psychopathology and Emotion Regulation, University Medical Center Groningen, University of Groningen, 9700 RB, Hanzeplein 1, Groningen, The Netherlands
| | - Alejandro Arias Vasquez
- Department of Psychiatry, and Department of Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, 6525 EN, Nijmegen, Gelderland, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboudumc, 6500 HB, Nijmegen, Gelderland, The Netherlands.,Department of Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Jan Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Radboudumc, Kapittelweg 29, 6525 EN, Nijmegen, Gelderland, The Netherlands.,Karakter Child and Adolescent Psychiatry University Centre, 6525 GC, Nijmegen, Gelderland, The Netherlands
| |
Collapse
|
7
|
Sepidarkish M, Rezamand G, Qorbani M, Heydari H, Estêvão MD, Omran D, Morvaridzadeh M, Roffey DM, Farsi F, Ebrahimi S, Shokri F, Heshmati J. Effect of omega-3 fatty acids supplementation on adipokines: a systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 2021; 62:7561-7575. [PMID: 33998914 DOI: 10.1080/10408398.2021.1915743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Although a large body of literature reported the beneficial effects of omega-3 fatty acids (omega-3 FAs) consumption on adipokines levels, but recent findings from clinical trials are not univocal. The aim of this systematic review and meta-analysis was to evaluate the effect of omega-3 FAs supplements on adipokines. METHODS We searched Medline, Web of Science, Scopus, Embase, and Cochrane Library from inception to August 2020 without any particular language limitations. Outcomes were summarized as standardized mean difference (SMD) with 95% confidence intervals (CIs) estimated from Hedge's g and random effects modeling. RESULTS Fifty-two trials involving 4,568 participants were included. Omega-3 FAs intake was associated with a significant increase in plasma adiponectin levels (n = 43; 3,434 participants; SMD: 0.21, 95% CI: 0.04, 0.37; p = 0.01; I2= 80.14%). This meta-analysis indicates that supplementing participants with omega-3 fatty acids more than 2000 mg daily and more than 10 weeks resulted in a significant and more favorable improvement in plasma adiponectin levels. However, omega-3 FAs intake had no significant effect on leptin levels (SMD: -0.02, 95% CI: -0.20, 0.17, I2= 54.13%). CONCLUSION The evidence supports a beneficial effect of omega-3 FAs intake on serum adiponectin levels but does not appear to impact on leptin concentrations. Larger well-designed RCTs are still required to evaluate the effect of omega-3 FAs on leptin in specific diseases.
Collapse
Affiliation(s)
- Mahdi Sepidarkish
- Department of Biostatistics and Epidemiology, School of Public Health, Babol University of Medical Sciences, Babol, Iran
| | - Gholamreza Rezamand
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hafez Heydari
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - M Dulce Estêvão
- Universidade do Algarve, Escola Superior de Saúde, Campus de Gambelas, Faro, Portugal
| | - Dalia Omran
- Department of Endemic Medicine and Hepatology, Thabet hospital for Endemic diseases, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mojgan Morvaridzadeh
- Department of Nutritional Science, School of Nutritional Science and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Darren M Roffey
- Vancouver General Hospital, Vancouver Coastal Health, Vancouver, Canada
| | - Farnaz Farsi
- Student Research Committee, Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Ebrahimi
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Fatemeh Shokri
- Department of Health Education and Promotion, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Heshmati
- Department of Nutritional Science, School of Nutritional Science and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
8
|
Qian F, Ardisson Korat AV, Imamura F, Marklund M, Tintle N, Virtanen JK, Zhou X, Bassett JK, Lai H, Hirakawa Y, Chien KL, Wood AC, Lankinen M, Murphy RA, Samieri C, Pertiwi K, de Mello VD, Guan W, Forouhi NG, Wareham N, Hu ICFB, Riserus U, Lind L, Harris WS, Shadyab AH, Robinson JG, Steffen LM, Hodge A, Giles GG, Ninomiya T, Uusitupa M, Tuomilehto J, Lindström J, Laakso M, Siscovick DS, Helmer C, Geleijnse JM, Wu JH, Fretts A, Lemaitre RN, Micha R, Mozaffarian D, Sun Q. n-3 Fatty Acid Biomarkers and Incident Type 2 Diabetes: An Individual Participant-Level Pooling Project of 20 Prospective Cohort Studies. Diabetes Care 2021; 44:1133-1142. [PMID: 33658295 PMCID: PMC8132316 DOI: 10.2337/dc20-2426] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Prospective associations between n-3 fatty acid biomarkers and type 2 diabetes (T2D) risk are not consistent in individual studies. We aimed to summarize the prospective associations of biomarkers of α-linolenic acid (ALA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and docosahexaenoic acid (DHA) with T2D risk through an individual participant-level pooled analysis. RESEARCH DESIGN AND METHODS For our analysis we incorporated data from a global consortium of 20 prospective studies from 14 countries. We included 65,147 participants who had blood measurements of ALA, EPA, DPA, or DHA and were free of diabetes at baseline. De novo harmonized analyses were performed in each cohort following a prespecified protocol, and cohort-specific associations were pooled using inverse variance-weighted meta-analysis. RESULTS A total of 16,693 incident T2D cases were identified during follow-up (median follow-up ranging from 2.5 to 21.2 years). In pooled multivariable analysis, per interquintile range (difference between the 90th and 10th percentiles for each fatty acid), EPA, DPA, DHA, and their sum were associated with lower T2D incidence, with hazard ratios (HRs) and 95% CIs of 0.92 (0.87, 0.96), 0.79 (0.73, 0.85), 0.82 (0.76, 0.89), and 0.81 (0.75, 0.88), respectively (all P < 0.001). ALA was not associated with T2D (HR 0.97 [95% CI 0.92, 1.02]) per interquintile range. Associations were robust across prespecified subgroups as well as in sensitivity analyses. CONCLUSIONS Higher circulating biomarkers of seafood-derived n-3 fatty acids, including EPA, DPA, DHA, and their sum, were associated with lower risk of T2D in a global consortium of prospective studies. The biomarker of plant-derived ALA was not significantly associated with T2D risk.
Collapse
Affiliation(s)
- Frank Qian
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Andres V. Ardisson Korat
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Fumiaki Imamura
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - Matti Marklund
- Clinical Nutrition and Metabolism, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Nathan Tintle
- Department of Mathematics and Statistics, Dordt University, Sioux Center, IA
- Fatty Acid Research Institute, Sioux Falls, SD
| | - Jyrki K. Virtanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Xia Zhou
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | | | - Heidi Lai
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
- Imperial College London, London, U.K
| | - Yoichiro Hirakawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Alexis C. Wood
- Children’s Nutrition Research Center, U.S. Department of Agriculture/Agricultural Research Service, Houston, TX
| | - Maria Lankinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Rachel A. Murphy
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Cecilia Samieri
- INSERM, UMR 1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
| | - Kamalita Pertiwi
- Division of Human Nutrition, Wageningen University, Wageningen, the Netherlands
| | - Vanessa D. de Mello
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota, Minneapolis, MN
| | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - InterAct Consortium, Frank B. Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Ulf Riserus
- Clinical Nutrition and Metabolism, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Clinical Nutrition and Metabolism, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - William S. Harris
- Fatty Acid Research Institute, Sioux Falls, SD
- Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD
| | - Aladdin H. Shadyab
- Department of Family Medicine and Public Health, University of California San Diego School of Medicine, La Jolla, CA
| | | | - Lyn M. Steffen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Allison Hodge
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, Australia
| | - Graham G. Giles
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Jaakko Tuomilehto
- Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jaana Lindström
- Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | | | - Catherine Helmer
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Johanna M. Geleijnse
- INSERM, UMR 1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
| | - Jason H.Y. Wu
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Amanda Fretts
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
| | - Rozenn N. Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Renata Micha
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
- Division of Cardiology, Tufts Medical Center, Boston, MA
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Balstad TR, Brunelli C, Pettersen CH, Schønberg SA, Skorpen F, Fallon M, Kaasa S, Bye A, Laird BJA, Stene GB, Solheim TS. Power Comparisons and Clinical Meaning of Outcome Measures in Assessing Treatment Effect in Cancer Cachexia: Secondary Analysis From a Randomized Pilot Multimodal Intervention Trial. Front Nutr 2021; 7:602775. [PMID: 33585533 PMCID: PMC7874039 DOI: 10.3389/fnut.2020.602775] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Background: New clinical trials in cancer cachexia are essential, and outcome measures with high responsiveness to detect meaningful changes are crucial. This secondary analysis from a multimodal intervention trial estimates sensitivity to change and between treatment effect sizes (ESs) of outcome measures associated with body composition, physical function, metabolism, and trial intervention. Methods: The study was a multicenter, open-label, randomized pilot study investigating the feasibility of a 6-week multimodal intervention [exercise, non-steroidal anti-inflammatory drugs, and oral nutritional supplements containing polyunsaturated fatty acids (n−3 PUFAs)] vs. standard cancer care in non-operable non-small-cell lung cancer and advanced pancreatic cancer. Body composition measures from computerized tomography scans and circulating biomarkers were analyzed. Results: Forty-six patients were randomized, and the analysis included 22 and 18 patients in the treatment and control groups, respectively. The between-group ESs were high for body weight (ES = 1.2, p < 0.001), small for body composition and physical function [handgrip strength (HGS)] measures (ES < 0.25), moderate to high for n-3 PUFAs and 25-hydroxyvitamin D (25-OH vitamin D) (ES range 0.64–1.37, p < 0.05 for all), and moderate for serum C-reactive protein (ES = 0.53, p = 0.12). Analysis within the multimodal treatment group showed high sensitivity to change for adiponectin (ES = 0.86, p = 0.001) and n-3 PUFAs (ES > 0.8, p < 0.05 for all) and moderate for 25-OH vitamin D (ES = 0.49, p = 0.03). In the control group, a moderate sensitivity to change for body weight (ES = −0.84, p = 0.002) and muscle mass (ES = −0.67, p = 0.016) and a high sensitivity to change for plasma levels of 25-OH vitamin D (ES = −0.88, p = 0.002) were found. Conclusion: Demonstrating high sensitivity to change and between treatment ES and body composition measures, body weight still stands out as a clinical and relevant outcome measure in cancer cachexia. Body composition and physical function measures clearly are important to address but demand large sample sizes to detect treatment group differences. Trial registration:ClinicalTrials.gov identifier: NCT01419145.
Collapse
Affiliation(s)
- Trude R Balstad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.,Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Cinzia Brunelli
- Palliative Care, Pain Therapy and Rehabilitation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Caroline H Pettersen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Svanhild A Schønberg
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Frank Skorpen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Marie Fallon
- Edinburgh Cancer Research Centre (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Stein Kaasa
- Department of Oncology, European Palliative Care Research Centre (PRC), Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Asta Bye
- Department of Oncology, European Palliative Care Research Centre (PRC), Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Nursing and Health Promotion, Faculty of Health Sciences, OsloMet-Oslo Metropolitan University, Oslo, Norway
| | - Barry J A Laird
- St. Columba's Hospice, Edinburgh, United Kingdom.,Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Guro B Stene
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.,Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tora S Solheim
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.,Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
10
|
Bardova K, Funda J, Pohl R, Cajka T, Hensler M, Kuda O, Janovska P, Adamcova K, Irodenko I, Lenkova L, Zouhar P, Horakova O, Flachs P, Rossmeisl M, Colca J, Kopecky J. Additive Effects of Omega-3 Fatty Acids and Thiazolidinediones in Mice Fed a High-Fat Diet: Triacylglycerol/Fatty Acid Cycling in Adipose Tissue. Nutrients 2020; 12:nu12123737. [PMID: 33291653 PMCID: PMC7761951 DOI: 10.3390/nu12123737] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/20/2022] Open
Abstract
Long-chain n-3 polyunsaturated fatty acids (Omega-3) and anti-diabetic drugs thiazolidinediones (TZDs) exhibit additive effects in counteraction of dietary obesity and associated metabolic dysfunctions in mice. The underlying mechanisms need to be clarified. Here, we aimed to learn whether the futile cycle based on the hydrolysis of triacylglycerol and re-esterification of fatty acids (TAG/FA cycling) in white adipose tissue (WAT) could be involved. We compared Omega-3 (30 mg/g diet) and two different TZDs—pioglitazone (50 mg/g diet) and a second-generation TZD, MSDC-0602K (330 mg/g diet)—regarding their effects in C57BL/6N mice fed an obesogenic high-fat (HF) diet for 8 weeks. The diet was supplemented or not by the tested compound alone or with the two TZDs combined individually with Omega-3. Activity of TAG/FA cycle in WAT was suppressed by the obesogenic HF diet. Additive effects in partial rescue of TAG/FA cycling in WAT were observed with both combined interventions, with a stronger effect of Omega-3 and MSDC-0602K. Our results (i) supported the role of TAG/FA cycling in WAT in the beneficial additive effects of Omega-3 and TZDs on metabolism of diet-induced obese mice, and (ii) showed differential modulation of WAT gene expression and metabolism by the two TZDs, depending also on Omega-3.
Collapse
Affiliation(s)
- Kristina Bardova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Jiri Funda
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Radek Pohl
- NMR Spectroscopy, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemmingovo Namesti 542/2, 160 00 Prague 6, Czech Republic;
| | - Tomas Cajka
- Laboratory of Metabolomics, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic;
- Laboratory of Translational Metabolism, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic
| | - Michal Hensler
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Ondrej Kuda
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic;
| | - Petra Janovska
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Katerina Adamcova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Ilaria Irodenko
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Lucie Lenkova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Petr Zouhar
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Olga Horakova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Pavel Flachs
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Martin Rossmeisl
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
| | - Jerry Colca
- Cirius Therapeutics, Kalamazoo, MI 490 07, USA;
| | - Jan Kopecky
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic; (K.B.); (J.F.); (M.H.); (P.J.); (K.A.); (I.I.); (L.L.); (P.Z.); (O.H.); (P.F.); (M.R.)
- Correspondence: ; Tel.: +420-296442554; Fax: +420-296442599
| |
Collapse
|
11
|
Rausch J, Gillespie S, Orchard T, Tan A, McDaniel JC. Systematic review of marine-derived omega-3 fatty acid supplementation effects on leptin, adiponectin, and the leptin-to-adiponectin ratio. Nutr Res 2020; 85:135-152. [PMID: 33482602 DOI: 10.1016/j.nutres.2020.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 10/22/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
Increasing evidence suggests that adipokines, leptin and adiponectin, produced and secreted by adipocytes, are involved in regulating systemic inflammation and may be important targets for interventions to reduce the chronic systemic inflammation linked to some conditions common in aging (e.g., atherosclerosis). Lower leptin levels and higher adiponectin levels in peripheral circulation have been associated with less systemic inflammation. While some studies have shown that marine-derived omega-3 fatty acids (eicosapentaenoic acid [EPA] and/or docosahexaenoic acid [DHA]) have effects on leptin and adiponectin in the context of inflammation, the extent of their effects remain unclear. The purpose of this systematic review was to summarize findings from randomized, controlled trials that measured effects of EPA+DHA supplementation on circulating levels of leptin and adiponectin to determine the state of the science. PubMed, CINAHL, Web of Science, Scopus, and Cochrane Trials were searched up to June 2018 for studies meeting inclusion criteria. Thirty-one studies included in this review were conducted in 16 countries. Eighteen studies reported lower leptin and/or higher adiponectin levels with EPA+DHA supplementation versus placebo at study end point (9 reported statistically significant differences), but doses, supplementation duration, and population characteristics varied across studies. In 9 studies reporting significantly lower leptin and/or higher adiponectin levels the EPA+DHA dose was 0.52 to 4.2 g/day for 4 to 24 weeks. Additional studies are warranted which assess dose parameters and patient populations similar to studies reporting significant effects of EPA+DHA on leptin or adiponectin in order to evaluate the extent of reproducibility before recommending EPA+DHA as a therapy to target these adipokines.
Collapse
Affiliation(s)
- Jamie Rausch
- Indiana University Fort Wayne, School of Nursing, Fort Wayne, IN, 46805, USA.
| | | | - Tonya Orchard
- Ohio State University, College of Education and Human Ecology, Columbus, OH, 43210, USA
| | - Alai Tan
- Ohio State University, College of Nursing, Columbus, OH, 43210, USA
| | - Jodi C McDaniel
- Ohio State University, College of Nursing, Columbus, OH, 43210, USA
| |
Collapse
|
12
|
Gura K, Premkumar MH, Calkins KL, Puder M. Intravenous Fish Oil Monotherapy as a Source of Calories and Fatty Acids Promotes Age-Appropriate Growth in Pediatric Patients with Intestinal Failure-Associated Liver Disease. J Pediatr 2020; 219:98-105.e4. [PMID: 32059815 DOI: 10.1016/j.jpeds.2019.12.065] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/15/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To compare growth in children with intestinal failure-associated liver disease (IFALD) who received a fish oil intravenous lipid emulsion (FOLE) to those who received a soybean oil intravenous lipid emulsion (SOLE). STUDY DESIGN This multisite, retrospective study pair-matched FOLE (n = 82) to SOLE recipients (n = 41) using baseline serum direct bilirubin levels and postmenstrual age. Study subjects received open-label FOLE (1 g/kg/day) until IFALD resolved or parenteral nutrition was stopped. Historical control subjects received SOLE (up to 3 g/kg/day). Growth measures (changes in body weight, height/length, and head circumference), prealbumin, triglycerides, and glucose were compared between groups over time using the Wilcoxon rank-sum test. RESULTS Although changes in all of the growth measures were similar for both groups (P > .05), FOLE recipients demonstrated an overall improved growth trajectory. After 28 weeks, FOLE recipients had a mean body weight within a z score range of -1 to 1 indicating age-appropriate growth. FOLE recipients consistently had higher prealbumin, lower triglyceride, and more normal glucose concentrations over time compared with SOLE recipients. CONCLUSIONS Children with IFALD who received FOLE had similar growth and fewer metabolic abnormalities compared with those who received SOLE. TRIAL REGISTRATION Clinicaltrials.gov: NCT00910104 and NCT00738101.
Collapse
Affiliation(s)
| | | | - Kara L Calkins
- Mattel Children's Hospital, University of California Los Angeles, Los Angeles, CA
| | | |
Collapse
|
13
|
Delarue J. Are marine n-3 fatty acids protective towards insulin resistance? From cell to human. Proc Nutr Soc 2020; 79:1-11. [PMID: 32138806 DOI: 10.1017/s0029665120000087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Marine n-3 fatty acids improve most of the biochemical alterations associated with insulin resistance (IR). Experimental models of dietary-induced IR in rodents have shown their ability (often at a very high dose) to prevent IR, but with sometimes a tissue specific effect. However, in a high sucrose diet-induced IR rat model, they are unable to reverse IR once installed; in other rodent models (dexamethasone, Zucker rats), they are inefficacious perhaps because of the severity of IR. The very low incidence of type-2 diabetes (T2D) in Inuits in the 1960s, which largely increased over the following decades in parallel to the replacement of their traditional marine food for a western diet strongly suggests a protective effect of marine n-3 towards the risk of T2D; this was confirmed by reversal of its incidence in intervention studies reintroducing their traditional food. In healthy subjects and insulin-resistant non-diabetic patients, most trials and meta-analyses conclude to an insulin-sensitising effect and to a very probable preventive or alleviating effect towards IR. Concerning the risk of T2D, concordant data allow us to conclude the protective effect of marine n-3 in Asians while suspicion exists of an aggravation of risk in Westerners, but with the possibility that it could be explained by a high heterogeneity of studies performed in this population. Some longitudinal cohorts in US/European people showed no association or a decreased risk. Further studies using more homogeneous doses, sources of n-3 and assessment of insulin sensitivity methods are required to better delineate their effects in Westerners.
Collapse
Affiliation(s)
- Jacques Delarue
- Department of Nutritional Sciences & Laboratory of Human Nutrition, University Hospital/Faculty of Medicine/University of Brest, Brittany, France
| |
Collapse
|
14
|
Abstract
The effect of dietary fats on cardiometabolic diseases, including cardiovascular diseases and type 2 diabetes mellitus, has generated tremendous interest. Many earlier investigations focused on total fat and conventional fat classes (such as saturated and unsaturated fats) and their influence on a limited number of risk factors. However, dietary fats comprise heterogeneous molecules with diverse structures, and growing research in the past two decades supports correspondingly complex health effects of individual dietary fats. Moreover, health effects of dietary fats might be modified by additional factors, such as accompanying nutrients and food-processing methods, emphasizing the importance of the food sources. Accordingly, the rapidly increasing scientific findings on dietary fats and cardiometabolic diseases have generated debate among scientists, caused confusion for the general public and present challenges for translation into dietary advice and policies. This Review summarizes the evidence on the effects of different dietary fats and their food sources on cell function and on risk factors and clinical events of cardiometabolic diseases. The aim is not to provide an exhaustive review but rather to focus on the most important evidence from randomized controlled trials and prospective cohort studies and to highlight current areas of controversy and the most relevant future research directions for understanding how to improve the prevention and management of cardiometabolic diseases through optimization of dietary fat intake.
Collapse
|
15
|
Wang T, Sha L, Li Y, Zhu L, Wang Z, Li K, Lu H, Bao T, Guo L, Zhang X, Wang H. Dietary α-Linolenic Acid-Rich Flaxseed Oil Exerts Beneficial Effects on Polycystic Ovary Syndrome Through Sex Steroid Hormones-Microbiota-Inflammation Axis in Rats. Front Endocrinol (Lausanne) 2020; 11:284. [PMID: 32670195 PMCID: PMC7326049 DOI: 10.3389/fendo.2020.00284] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/15/2020] [Indexed: 12/24/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) represents a common endocrine-metabolic disorder disease with chronic low-grade inflammation and alteration of intestinal flora. Serving as functional food, flaxseed oil (FO), which is rich in plant-derived α-linolenic acid of omega-3 polyunsaturated fatty acids, has been proven to benefit for chronic metabolic diseases. However, the exact role of dietary FO on PCOS remains largely unclear. In the present study, 6-week-old female Sprague-Dawley rats were randomly divided into four groups (eight rats/group), including (a) pair-fed (PF) control (CON) group (PF/CON), (b) FO-fed CON group (FO/CON), (c) PF with letrozole-induced PCOS model (MOD) group (PF/MOD), and (d) FO-fed MOD group (FO/MOD). All rats were fed a standard diet. After 3 weeks of modeling and subsequent 8 weeks of treatment, the rats in diverse groups were euthanized and associated indications were investigated. The results showed that dietary FO ameliorated the disorder of estrous cycle and ovarian morphology. In parallel, dietary FO improved the sex steroid hormone disturbance (luteinizing hormone/follicle-stimulating hormone, estrogen, testosterone, and progesterone), body weights, dyslipidemia, and insulin resistance. Moreover, FO treatment improved plasma and ovary inflammatory interleukin (IL)-1β, IL-6, IL-10, and IL-17A, tumor necrosis factor-α, and monocyte chemoattractant protein-1. Additionally, FO intervention significantly modulated the composition of gut microbiota and vaginal microbiota by increasing the abundances of Allobaculum, Lactobacillus, Butyrivibrio, Desulfovibrio, Bifidobacterium, Faecalibacterium, Parabacteroides as well as decreasing the abundances of Actinobacteria, Bacteroides, Proteobacteria, and Streptococcus, the ratio of Firmicutes/Bacteroidetes. A decrease in plasma lipopolysaccharide level and an increase in short-chain fatty acids, including acetic acid, propionic acid, butyric acid and pentanoic acid, were determined after dietary FO supplementation. Correlation analysis revealed close relationships among sex steroid hormones, inflammation, and gut/vaginal microbiota. Collectively, this study demonstrated that dietary FO ameliorated PCOS through the sex steroid hormones-microbiota-inflammation axis in rats, which may contribute to the understanding of pathogenesis and potentially serve as an inexpensive intervention in the control of PCOS.
Collapse
Affiliation(s)
- Ting Wang
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Liping Sha
- Endocrinology Department, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yiwei Li
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Lili Zhu
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Zhen Wang
- Endocrinology Department, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ke Li
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Haixia Lu
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Ting Bao
- Endocrinology Department, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Li Guo
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
- *Correspondence: Xiaoxia Zhang
| | - Hao Wang
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Hao Wang
| |
Collapse
|
16
|
Thomas DD, Stockman MC, Yu L, Meshulam T, McCarthy AC, Ionson A, Burritt N, Deeney J, Cabral H, Corkey B, Istfan N, Apovian CM. Effects of medium chain triglycerides supplementation on insulin sensitivity and beta cell function: A feasibility study. PLoS One 2019; 14:e0226200. [PMID: 31869355 PMCID: PMC6927614 DOI: 10.1371/journal.pone.0226200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/20/2019] [Indexed: 12/20/2022] Open
Abstract
Objective Medium chain triglycerides (MCT) have unique metabolic properties which may improve insulin sensitivity (Si) and beta cell function but data in humans are limited. We conducted a 6-week clinical trial of MCT oil supplementation. Methods 22 subjects without diabetes (8 males, 14 females, mean ± standard error age 39±2.9 years, baseline BMI 27.0±1.4 kg/m2) were counseled to maintain their body weight and physical activity (PA) during the trial. Dietary intake, PA data, body composition, and resting energy expenditure (REE) were obtained through dietary recall, international PA questionnaire, dual x-ray absorptiometry, and indirect calorimetry, respectively. MCT prescriptions were given based on REE and PA to replace part of dietary fat with 30 grams of MCT per 2000 kcal daily. Insulin-modified frequently sampled intravenous glucose tolerance tests were performed before and after MCT to measure changes in Si, acute insulin response (AIR), disposition index (DI), and glucose effectiveness (Sg). Results MCT were well tolerated and weight remained stable (mean change 0.3 kg, p = 0.39). Fasting REE, respiratory quotient, and body composition were stable during the intervention. There were no significant changes in mean fasting glucose, insulin, insulin resistance, fasting total ketones, Si, AIR, DI, Sg, leptin, fructosamine, and proinsulin. The mean change in Si was 0.5 10−4 min-1 per mU/L (95% CI: -1.4, 2.4), corresponding to a 12% increase from baseline, and the range was -4.7 to 12.9 10−4 min-1 per mU/L. Mean total adiponectin decreased significantly from 22925 ng/mL at baseline to 17598 ng/mL at final visit (p = 0.02). The baseline clinical and laboratory parameters were not significantly associated with the change in Si. Discussion There were a wide range of changes in the minimal model parameters of glucose and insulin metabolism in subjects following 6 weeks of MCT as an isocaloric substitution for part of usual dietary fat intake. Since this was a single-arm non-randomized study without a control group, it cannot be certain whether these changes were due to MCT so further randomized controlled trials are warranted.
Collapse
Affiliation(s)
- Dylan D. Thomas
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Mary-Catherine Stockman
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
| | - Liqun Yu
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
| | - Tova Meshulam
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Ashley C. McCarthy
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Annaliese Ionson
- Boston University School of Medicine, Boston, MA, United States of America
| | - Nathan Burritt
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
| | - Jude Deeney
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Howard Cabral
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
| | - Barbara Corkey
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
| | - Nawfal Istfan
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
| | - Caroline M. Apovian
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University, MA, United States of America
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition and Weight Management, Nutrition and Weight Management Center, Boston Medical Center, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
17
|
Fish oil supplementation to a high-fat diet improves both intestinal health and the systemic obese phenotype. J Nutr Biochem 2019; 72:108216. [DOI: 10.1016/j.jnutbio.2019.07.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/28/2019] [Accepted: 07/19/2019] [Indexed: 12/25/2022]
|
18
|
Adiponectin levels among individuals with varied employment status in Japan: a cross-sectional analysis of the J-SHINE study. Sci Rep 2019; 9:10936. [PMID: 31358891 PMCID: PMC6662719 DOI: 10.1038/s41598-019-47448-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 07/16/2019] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study was to examine the association between employment status and adiponectin levels. This cross-sectional study was a part of the Japanese Stratification, Health, Income, and Neighborhood study, a population-based survey in metropolitan Japan. The analysis included data from 848 individuals. A one-way analysis of variance was used to assess differences in log-transformed adiponectin levels among individuals according to their employment status. Multiple linear regression analysis was used to assess these differences after adjusting for other cardiovascular disease risk factors. The main outcome was log-transformed adiponectin. Of the participants, 6.2% of the men and 15.1% of the women were precarious workers. Mean adiponectin values differed significantly by employment status in men, but not in women. In men, multiple regression analysis showed that precarious workers had significantly lower adiponectin levels than permanent workers (β = −0.16, P = 0.02). However, in women, adiponectin levels were significantly lower only in precarious workers with low household incomes (β = −0.35, P = 0.02). Male precarious workers and their female counterparts with low annual household incomes had significantly lower levels of adiponectin. These results might help us to understand mechanisms underlying the relationship between employment status and cardiovascular disease.
Collapse
|
19
|
Beneficial Effects of Adiponectin on Glucose and Lipid Metabolism and Atherosclerotic Progression: Mechanisms and Perspectives. Int J Mol Sci 2019; 20:ijms20051190. [PMID: 30857216 PMCID: PMC6429491 DOI: 10.3390/ijms20051190] [Citation(s) in RCA: 294] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022] Open
Abstract
Circulating adiponectin concentrations are reduced in obese individuals, and this reduction has been proposed to have a crucial role in the pathogenesis of atherosclerosis and cardiovascular diseases associated with obesity and the metabolic syndrome. We focus on the effects of adiponectin on glucose and lipid metabolism and on the molecular anti-atherosclerotic properties of adiponectin and also discuss the factors that increase the circulating levels of adiponectin. Adiponectin reduces inflammatory cytokines and oxidative stress, which leads to an improvement of insulin resistance. Adiponectin-induced improvement of insulin resistance and adiponectin itself reduce hepatic glucose production and increase the utilization of glucose and fatty acids by skeletal muscles, lowering blood glucose levels. Adiponectin has also β cell protective effects and may prevent the development of diabetes. Adiponectin concentration has been found to be correlated with lipoprotein metabolism; especially, it is associated with the metabolism of high-density lipoprotein (HDL) and triglyceride (TG). Adiponectin appears to increase HDL and decrease TG. Adiponectin increases ATP-binding cassette transporter A1 and lipoprotein lipase (LPL) and decreases hepatic lipase, which may elevate HDL. Increased LPL mass/activity and very low density lipoprotein (VLDL) receptor and reduced apo-CIII may increase VLDL catabolism and result in the reduction of serum TG. Further, adiponectin has various molecular anti-atherosclerotic properties, such as reduction of scavenger receptors in macrophages and increase of cholesterol efflux. These findings suggest that high levels of circulating adiponectin can protect against atherosclerosis. Weight loss, exercise, nutritional factors, anti-diabetic drugs, lipid-lowering drugs, and anti-hypertensive drugs have been associated with an increase of serum adiponectin level.
Collapse
|
20
|
Liu Y, Vu V, Sweeney G. Examining the Potential of Developing and Implementing Use of Adiponectin-Targeted Therapeutics for Metabolic and Cardiovascular Diseases. Front Endocrinol (Lausanne) 2019; 10:842. [PMID: 31920962 PMCID: PMC6918867 DOI: 10.3389/fendo.2019.00842] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiometabolic diseases encompass those affecting the heart and vasculature as well as other metabolic problems, such as insulin resistance, diabetes, and non-alcoholic fatty liver disease. These diseases tend to have common risk factors, one of which is impaired adiponectin action. This may be due to reduced bioavailability of the hormone or resistance to its effects on target tissues. A strong negative correlation between adiponectin levels and cardiometabolic diseases has been well-documented and research shown that adiponectin has cardioprotective, insulin sensitizing and direct beneficial metabolic effects. Thus, therapeutic approaches to enhance adiponectin action are widely considered to be desirable. The complexity of adiponectin structure and function has so far made progress in this area less than ideal. In this article we will review the effects and mechanism of action of adiponectin on cardiometabolic tissues, identify scenarios where enhancing adiponectin action would be of clinical value and finally discuss approaches via which this can be achieved.
Collapse
Affiliation(s)
- Ying Liu
- Metabolic Disease Research Division, iCarbonX Co. Ltd., Shenzhen, China
- *Correspondence: Ying Liu
| | - Vivian Vu
- Department of Biology, York University, Toronto, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
- Gary Sweeney
| |
Collapse
|
21
|
Sedláček P, Plavinová I, Langmajerová J, Dvořáková J, Novák J, Trefil L, Müller L, Buňatová P, Zeman V, Müllerová D. Effect of n-3 fatty acids supplementation during life style modification in women with overweight. Cent Eur J Public Health 2018; 26:265-271. [DOI: 10.21101/cejph.a5259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Indexed: 11/15/2022]
|
22
|
Song J, Hu M, Li C, Yang B, Ding Q, Wang C, Mao L. Dose-dependent effects of fish oil on cardio-metabolic biomarkers in healthy middle-aged and elderly Chinese people: a double-blind randomized controlled trial. Food Funct 2018; 9:3235-3243. [PMID: 29781483 DOI: 10.1039/c7fo01566f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
n-3PUFA consumption has been widely accepted as a nutritional strategy for the secondary prevention of cardiovascular events in patients at high risk of cardiovascular disease (CVD), but little is known about the dose-response relationship between dietary n-3PUFA and serum biomarkers associated with cardiovascular health in the general population. The present study involved a 12-week double-blind, randomized controlled trial to explore the effects of fish oil with different doses (0.31, 0.62 and 1.24 g d-1 of EPA and DHA) on serum fatty acids and cardio-metabolic biomarkers including adiponectin, inflammatory markers, lipid profiles and fasting glucose in healthy middle-aged and elderly Chinese people. 240 volunteers met our inclusion criteria. A total of 39 subjects dropped out and 201 finally completed the intervention. No significant differences in baseline characteristics and daily intakes of dietary nutrients were detected among all groups. After a 12-week intervention, fish oil dose-dependently enhanced serum EPA, DHA, n-3PUFA and adiponectin (except for 0.31 g d-1), but decreased serum n-6/n-3PUFA, TG and fasting glucose. Changes in the above indicators from the baseline to week 12 in fish oil groups significantly differed from those in the control. Meanwhile, all the doses of EPA and DHA led to decreases in serum CRP; only 1.24 g d-1 led to an increase in HDL-C with a concurrent decrease in TC/HDL-C even though these changes were not significantly different among all groups. All the findings suggested that fish oil dose-dependently regulated serum PUFA and cardio-metabolic biomarkers including adiponectin, CRP, lipid profiles and fasting glucose in healthy middle-aged and elderly Chinese people who consumed insufficient dietary n-3PUFA, and the most desirable changes were observed for 1.24 g d-1.
Collapse
Affiliation(s)
- Jia Song
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | | | | | | | | | | | | |
Collapse
|
23
|
Huhmann MB, Yamamoto S, Neutel JM, Cohen SS, Ochoa Gautier JB. Very high-protein and low-carbohydrate enteral nutrition formula and plasma glucose control in adults with type 2 diabetes mellitus: a randomized crossover trial. Nutr Diabetes 2018; 8:45. [PMID: 30158516 PMCID: PMC6115411 DOI: 10.1038/s41387-018-0053-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/23/2018] [Accepted: 06/11/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Standard enteral nutrition (EN) formulas can worsen hyperglycemia in diabetic patients. We hypothesized that altering the proportion of macronutrients in a formula; increasing protein while decreasing carbohydrate concentrations would improve glycemic response. The objective of this study was to demonstrate that an EN formula containing a very high concentration of protein (in the form of whey peptides) and low concentration of carbohydrate provide better control of postprandial blood glucose relative to a very high-protein/higher-carbohydrate formula. SUBJECTS AND METHODS This was a randomized crossover clinical trial of 12 ambulatory adult subjects with type 2 diabetes. The primary outcome was glycemic response following a bolus of isocaloric amounts of two EN formulas; the secondary outcome was insulin response. Subjects were randomized to the experimental or the control formula, on two separate days, 5-7 days apart. RESULTS Mean blood glucose concentrations at 10-180 min post-infusion and mean area under the curve for glucose over 240 min post-infusion were significantly lower with the experimental formula than with the control formula (71.99 ± 595.18 and 452.62 ± 351.38, respectively; p = 0.025). There were no significant differences in the mean insulin concentrations over time, insulinogenic indices, and first-phase insulin measurements. CONCLUSIONS An EN formula containing high-protein and low-carbohydrate loads can significantly improve glucose control in subjects with type 2 diabetes in ambulatory settings as evidenced by observed improved glucose control without significant difference in insulin response.
Collapse
Affiliation(s)
- Maureen B Huhmann
- Clinical Sciences, Nestlé Health Science, 1007 US Highway 202/206, Building JR2, Bridgewater, NJ, 08807, USA.
| | - Shinobu Yamamoto
- Clinical Sciences, Nestlé Health Science, 1007 US Highway 202/206, Building JR2, Bridgewater, NJ, 08807, USA
| | - Joel M Neutel
- Orange County Research Center, 14351 Myford Road, Suite B, Tustin, CA, 92780, USA
| | - Sarah S Cohen
- EpidStat Institute, 2100 Commonwealth Blvd, Suite 203, Ann Arbor, MI, 48105, USA
| | - Juan B Ochoa Gautier
- Clinical Sciences, Nestlé Health Science, 1007 US Highway 202/206, Building JR2, Bridgewater, NJ, 08807, USA.,Department of Critical Care Medicine, Geisinger Medical Center, Danville, PA, 17822, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
24
|
Farimani AR, Hariri M, Azimi-Nezhad M, Borji A, Zarei S, Hooshmand E. The effect of n-3 PUFAs on circulating adiponectin and leptin in patients with type 2 diabetes mellitus: a systematic review and meta-analysis of randomized controlled trials. Acta Diabetol 2018; 55:641-652. [PMID: 29453672 DOI: 10.1007/s00592-018-1110-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/29/2018] [Indexed: 01/01/2023]
Abstract
AIM N-3 PUFAs can potentially influence levels of inflammatory and non-inflammatory adipokines. Given the contradictory effects of n-3 PUFAs on serum levels of adipokines in type 2 diabetes, we conducted a systematic review and meta-analysis study of randomized placebo-controlled clinical trials that examined the effects of n-3 PUFAs on serum levels of leptin and adiponectin in patients with type 2 diabetes. METHODS The electronic databases, without regard to language restrictions including PubMed/Medline, Google Scholar, SCOPUS and ISI Web of Science until August 2017, were used to identify randomized controlled trials that assessed the effect of n-3 PUFAs on serum leptin and adiponectin concentrations in type 2 diabetes. Outcomes were extracted based on the mean ± SD as effect size at baseline and end of the intervention. Between-study heterogeneity was evaluated by the I2 estimates and their 95% CIs. Funnel plot asymmetry was used to investigate the existence of publication bias. Stata software and Review Manager were used for statistical data analysis. RESULTS Data from 10 eligible articles involved 494 subjects with type 2 diabetes mellitus (intervention groups = 254 and control groups = 240), with age between 44 and 70 years, treated with doses of 0.52-7.4 g/day n-3 PUFAs. Adiponectin concentration nonsignificantly increased by a MD = 0.17 µg/mL (95% CI - 0.11, 0.44). Also, leptin concentration nonsignificantly reduced by a MD = - 0.31 ng/mL (95% CI - 0.69, 0.07). CONCLUSION Plant and marine sources of n-3 PUFAs can modify serum leptin and adiponectin levels by increasing adiponectin and decreasing leptin levels in patients with type 2 diabetes. Due to some limitations in this study, further studies are needed to reach a definitive conclusion about the effect of n-3 PUFAs on the levels of leptin and adiponectin in T2DM.
Collapse
Affiliation(s)
- Azam Rezaei Farimani
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Mitra Hariri
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Mohsen Azimi-Nezhad
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Abasalt Borji
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Sadegh Zarei
- Department of Clinical Biochemistry, Shahid Sadoughi University of Medical Sciences, Yazd, Zip code: 9314634814, Iran
| | - Elham Hooshmand
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
25
|
Becic T, Studenik C. Effects of Omega-3 Supplementation on Adipocytokines in Prediabetes and Type 2 Diabetes Mellitus: Systematic Review and Meta-Analysis of Randomized Controlled Trials. Diabetes Metab J 2018; 42:101-116. [PMID: 29676540 PMCID: PMC5911513 DOI: 10.4093/dmj.2018.42.2.101] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/28/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The objective of this systematic review and meta-analysis was to determine the effects of omega-3 supplementation on adipocytokine levels in adult prediabetic and diabetic individuals. METHODS We searched PubMed, Medline, EMBASE, Scopus, Web of Science, Google Scholar, Cochrane Trial Register, World Health Organization Clinical Trial Registry Platform, and Clinicaltrial.gov Registry from inception to August 1, 2017 for randomized controlled trials. Pooled effects of interventions were assessed as mean difference using random effects model. We conducted a sensitivity, publication bias and subgroup analysis. RESULTS Fourteen studies individuals (n=685) were included in the meta-analysis. Omega-3 supplementation increased levels of adiponectin (0.48 μg/mL; 95% confidence interval [CI], 0.27 to 0.68; P<0.00001, n=10 trials), but effects disappeared after sensitivity analysis. Tumor necrosis factor α (TNF-α) levels were reduced (-1.71; 95% CI, -3.38 to -0.14; P=0.03, n=8 trials). Treatment duration shorter than 12 weeks was associated with greater reduction than longer treatment duration. Levels of other adipocytokines were not significantly affected. Publication bias could generally not be excluded. CONCLUSION Eicosapentaenoic acid and docosahexaenoic acid supplementation may increase adiponectin and reduce TNF-α levels in this population group. However, due to overall study heterogeneity and potential publication bias, a cautious interpretation is needed.
Collapse
Affiliation(s)
- Tarik Becic
- Department of Pharmacology and Toxicology, Faculty of Life Sciences, University of Vienna, Vienna, Austria.
| | - Christian Studenik
- Department of Pharmacology and Toxicology, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
26
|
Gawrieh S, Chalasani N. Emerging Treatments for Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Clin Liver Dis 2018; 22:189-199. [PMID: 29128056 DOI: 10.1016/j.cld.2017.08.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This review discusses completed phase II randomized clinical trials with high-quality published results for compounds that demonstrate effects on nonalcoholic steatohepatitis histology (obeticholic acid, elafibranor, and liraglutide). The authors also review the available preliminary data on cenicriviroc and selonsertib, with or without simtuzumab's phase II studies. Finally, the authors briefly discuss compounds that have been tested but did not achieve the primary end point of histologic improvement and appeared in high-quality published articles (cysteamine bitartrate and long-chain polyunsaturated fatty acids).
Collapse
Affiliation(s)
- Samer Gawrieh
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, 702 Rotary Circle, Suite 225, Indianapolis, IN 46202, USA.
| | - Naga Chalasani
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, 702 Rotary Circle, Suite 225, Indianapolis, IN 46202, USA.
| |
Collapse
|
27
|
Kuda O, Rossmeisl M, Kopecky J. Omega-3 fatty acids and adipose tissue biology. Mol Aspects Med 2018; 64:147-160. [PMID: 29329795 DOI: 10.1016/j.mam.2018.01.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 12/16/2022]
Abstract
This review provides evidence for the importance of white and brown adipose tissue (i.e. WAT and BAT) function for the maintenance of healthy metabolic phenotype and its preservation in response to omega-3 polyunsaturated fatty acids (omega-3 PUFA), namely in the context of diseased states linked to aberrant accumulation of body fat, systemic low-grade inflammation, dyslipidemia and insulin resistance. More specifically, the review deals with (i) the concept of immunometabolism, i.e. how adipose-resident immune cells and adipocytes affect each other and define the immune-metabolic interface; and (ii) the characteristic features of "healthy adipocytes" in WAT, which are relatively small fat cells endowed with a high capacity for mitochondrial oxidative phosphorylation, triacylglycerol/fatty acid (TAG/FA) cycling and de novo lipogenesis (DNL). The intrinsic metabolic features of WAT and their flexible regulations, reflecting the presence of "healthy adipocytes", provide beneficial local and systemic effects, including (i) protection against in situ endoplasmic reticulum stress and related inflammatory response during activation of adipocyte lipolysis; (ii) prevention of ectopic fat accumulation and dyslipidemia caused by increased hepatic VLDL synthesis, as well as prevention of lipotoxic damage of insulin signaling in extra-adipose tissues; and also (iii) increased synthesis of anti-inflammatory and insulin-sensitizing lipid mediators with pro-resolving properties, including the branched fatty acid esters of hydroxy fatty acids (FAHFAs), also depending on the activity of DNL in WAT. The "healthy adipocytes" phenotype can be induced in WAT of obese mice in response to various stimuli including dietary omega-3 PUFA, especially when combined with moderate calorie restriction, and possibly also with other life style (e.g. physical activity) or pharmacological (e.g. thiazolidinediones) interventions. While omega-3 PUFA could exert beneficial systemic effects by improving immunometabolism of WAT without a concomitant induction of BAT, it is currently not clear whether the metabolic effects of the combined intervention using omega-3 PUFA and calorie restriction or thiazolidinediones depend also on the activation of BAT function and/or the induction of brite/beige adipocytes in WAT. It remains to be established why omega-3 PUFA intervention in type 2 diabetic subjects does not improve insulin sensitivity and glucose homeostasis despite inducing various anti-inflammatory mediators in WAT, including the recently discovered docosahexaenoyl esters of hydroxy linoleic acid, the lipokines from the FAHFA family, as well as several endocannabinoid-related anti-inflammatory lipids. To answer the question whether and to which extent omega-3 PUFA supplementation could promote the formation of "healthy adipocytes" in WAT of human subjects, namely in the obese insulin-resistant patients, represents a challenging task that is of great importance for the treatment of some serious non-communicable diseases.
Collapse
Affiliation(s)
- Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic
| | - Martin Rossmeisl
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic
| | - Jan Kopecky
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic.
| |
Collapse
|
28
|
Bahreini M, Ramezani AH, Shishehbor F, Mansoori A. The Effect of Omega-3 on Circulating Adiponectin in Adults With Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Can J Diabetes 2018; 42:553-559. [PMID: 29567080 DOI: 10.1016/j.jcjd.2017.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/23/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023]
Abstract
Whether consumption of omega-3 affects circulating adiponectin has not been established. The objective of this study was to evaluate the effect of omega-3 (food or supplement) on circulating adiponectin in patients with type 2 diabetes through a systematic review of meta-analyses of randomized controlled trials. PubMed, Scopus and Web of Science were searched for relevant studies through May 2016. Two researchers screened and abstracted the literature independently. Pooled estimates were obtained using the random-effects models. Overall, omega-3 increased adiponectin by 0.57 µg/mL (95% confidence interval [CI] 0.15 to 1.31; p=0.01, I-square=74.2% p for heterogeneity <0.001). The source of observed heterogeneity was explored by subgroup analyses. In subgroup analyses, adiponectin levels increased only in those who had consumed omega-3 for more than 8 weeks. This systematic review and meta-analysis of randomized, placebo-controlled clinical trials suggests that omega-3 in patients with type 2 diabetes increases circulating adiponectin. These findings support the potentially beneficial effects of dietary omega-3 in patients with type 2 diabetes on pathways related to adiponectin metabolism.
Collapse
Affiliation(s)
- Mehdi Bahreini
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir-Hossein Ramezani
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farideh Shishehbor
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Nutrition, School of Para-Medical Sciences, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
| | - Anahita Mansoori
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Nutrition, School of Para-Medical Sciences, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran.
| |
Collapse
|
29
|
Saarinen HJ, Husgafvel S, Pohjantähti-Maaroos H, Wallenius M, Palomäki A. Improved insulin sensitivity and lower postprandial triglyceride concentrations after cold-pressed turnip rapeseed oil compared to cream in patients with metabolic syndrome. Diabetol Metab Syndr 2018; 10:38. [PMID: 29755591 PMCID: PMC5935965 DOI: 10.1186/s13098-018-0340-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 04/23/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The aim of this study was to compare acute effects of turnip rapeseed oil rich with mono- and polyunsaturated fatty acids and cream on postprandial triglyceride levels and post-glucose load measures of insulin sensitivity in population of men with metabolic syndrome. METHODS This open-label balanced crossover study included 37 men with metabolic syndrome. They underwent an oral glucose-fat tolerance test where they ingested 75 g of glucose with either 240 mL of cream or 84 mL of turnip rapeseed oil depending on the study arm. Hourly postprandial blood samples were drawn up to 5 h after this oral glucose-fat tolerance test to determine the changes in triglyceride concentrations and to measure insulin sensitivity. Changes in insulin sensitivity were calculated with different insulin sensitivity indices (OGIS, Stumvoll, Gutt and McAuley scores) derived from measured insulin and glucose concentrations. The oral glucose-fat tolerance test was preceded by a period during which the participants consumed a daily portion of either 35 mL of turnip rapeseed oil or 37.5 g of butter depending on the study arm in addition to their habitual diets. Both dietary periods lasted from 6 to 8 weeks. After an 8-week wash-out period the subjects crossed over to the other study arm and underwent the same process with the other fat adjunct. RESULTS The area under the curve for hourly triglyceride concentrations was 16% smaller after turnip rapeseed oil than after cream (13.86 [interquartile range 8.54] vs. 16.41 [9.09] mmol/l, p < 0.001). The insulin sensitivity markers of OGIS (324 [38.97] vs. 377 [68.38] p < 0.001), Stumvoll score (0.079 [0.029] vs. 0.085 [0.029], p = 0.038) and Gutt score (67.0 ± 2.78 vs. 78.8 ± 4.97 p = 0.001) were higher after turnip rapeseed oil period than after butter period. There was a non-significant change in the McAuley score. CONCLUSION Dietary turnip rapeseed oil improved postprandially measured insulin sensitivity and triglyceride concentrations compared to cream and butter. This provides a possible efficient dietary mean to treat cardiovascular risk factors.Trial registration ClinicalTrials.gov NCT01119690 (05-06-2010).
Collapse
Affiliation(s)
| | - Sari Husgafvel
- Central Hospital of Kanta-Häme, Ahvenistontie 20, 13530 Hämeenlinna, Finland
- Linnan Klinikka, Raatihuoneenkatu 10, 13100 Hämeenlinna, Finland
- Central Hospital of Päijät-Häme, Keskussairaalankatu 7, 15850 Lahti, Finland
| | - Hanna Pohjantähti-Maaroos
- Central Hospital of Kanta-Häme, Ahvenistontie 20, 13530 Hämeenlinna, Finland
- Linnan Klinikka, Raatihuoneenkatu 10, 13100 Hämeenlinna, Finland
- Heart Center, Kuopio University Hospital, P.O. Box 1777, 70211 Kuopio, Finland
| | - Marja Wallenius
- Linnan Klinikka, Raatihuoneenkatu 10, 13100 Hämeenlinna, Finland
| | - Ari Palomäki
- Central Hospital of Kanta-Häme, Ahvenistontie 20, 13530 Hämeenlinna, Finland
- Linnan Klinikka, Raatihuoneenkatu 10, 13100 Hämeenlinna, Finland
- Faculty of Medicine and Life Sciences, University of Tampere, 33014 Tampere, Finland
| |
Collapse
|
30
|
Different Dietary Proportions of Fish Oil Regulate Inflammatory Factors but Do Not Change Intestinal Tight Junction ZO-1 Expression in Ethanol-Fed Rats. Mediators Inflamm 2017; 2017:5801768. [PMID: 29386752 PMCID: PMC5745723 DOI: 10.1155/2017/5801768] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/30/2017] [Accepted: 10/15/2017] [Indexed: 12/18/2022] Open
Abstract
Sixty male Wistar rats were fed a control or an ethanol-containing diet in groups C or E. The fat compositions were adjusted with 25% or 57% fish oil substituted for olive oil in groups CF25, CF57, EF25, and EF57. Hepatic thiobarbituric acid-reactive substance (TBARS) levels, cytochrome P450 2E1 protein expression, and tumor necrosis factor- (TNF-) α, interleukin- (IL-) 1β, IL-6, and IL-10 levels, as well as intracellular adhesion molecule (ICAM)-1 levels were significantly elevated, whereas plasma adiponectin level was significantly reduced in group E (p < 0.05). Hepatic histopathological scores of fatty change and inflammation, in group E were significantly higher than those of group C (p < 0.05). Hepatic TBARS, plasma ICAM-1, and hepatic TNF-α, IL-1β, and IL-10 levels were significantly lower, and plasma adiponectin levels were significantly higher in groups EF25 and EF57 than those in group E (p < 0.05). The immunoreactive area of the intestinal tight junction protein, ZO-1, showed no change between groups C and E. Only group CF57 displayed a significantly higher ZO-1 immunoreactive area compared to group C (p = 0.0415). 25% or 57% fish oil substituted for dietary olive oil could prevent ethanol-induced liver damage in rats, but the mechanism might not be related to intestinal tight junction ZO-1 expression.
Collapse
|
31
|
Integrated Immunomodulatory Mechanisms through which Long-Chain n-3 Polyunsaturated Fatty Acids Attenuate Obese Adipose Tissue Dysfunction. Nutrients 2017; 9:nu9121289. [PMID: 29186929 PMCID: PMC5748740 DOI: 10.3390/nu9121289] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is a global health concern with rising prevalence that increases the risk of developing other chronic diseases. A causal link connecting overnutrition, the development of obesity and obesity-associated co-morbidities is visceral adipose tissue (AT) dysfunction, characterized by changes in the cellularity of various immune cell populations, altered production of inflammatory adipokines that sustain a chronic state of low-grade inflammation and, ultimately, dysregulated AT metabolic function. Therefore, dietary intervention strategies aimed to halt the progression of obese AT dysfunction through any of the aforementioned processes represent an important active area of research. In this connection, fish oil-derived dietary long-chain n-3 polyunsaturated fatty acids (PUFA) in the form of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been demonstrated to attenuate obese AT dysfunction through multiple mechanisms, ultimately affecting AT immune cellularity and function, adipokine production, and metabolic signaling pathways, all of which will be discussed herein.
Collapse
|
32
|
Bischoff SC, Boirie Y, Cederholm T, Chourdakis M, Cuerda C, Delzenne NM, Deutz NE, Fouque D, Genton L, Gil C, Koletzko B, Leon-Sanz M, Shamir R, Singer J, Singer P, Stroebele-Benschop N, Thorell A, Weimann A, Barazzoni R. Towards a multidisciplinary approach to understand and manage obesity and related diseases. Clin Nutr 2017; 36:917-938. [DOI: 10.1016/j.clnu.2016.11.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022]
|
33
|
Jacobo-Cejudo MG, Valdés-Ramos R, Guadarrama-López AL, Pardo-Morales RV, Martínez-Carrillo BE, Harbige LS. Effect of n-3 Polyunsaturated Fatty Acid Supplementation on Metabolic and Inflammatory Biomarkers in Type 2 Diabetes Mellitus Patients. Nutrients 2017; 9:nu9060573. [PMID: 28587203 PMCID: PMC5490552 DOI: 10.3390/nu9060573] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/28/2017] [Accepted: 06/01/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is accompanied by chronic low-grade inflammation, with an imbalance in the secretion of adipokines and, worsening insulin resistance. Supplementation with n-3 PUFA in T2DM decreases inflammatory markers, the purpose of the study was to investigate the effect of n-3 PUFA supplementation on adipokines, metabolic control, and lipid profile in T2DM Mexican adults. METHODS In a randomized, single-blind, placebo-controlled pilot study, 54 patients with T2DM received 520 mg of DHA + EPA-enriched fish-oil (FOG) or a placebo (PG) daily. Baseline and 24-week anthropometric and biochemical measurements included glucose, insulin, glycosylated hemoglobin (Hb1Ac), leptin, adiponectin, resistin, and lipid profile; n-3 PUFA intake was calculated in g/day. RESULTS Waist circumference and blood glucose showed significant reductions in the FOG group (p = 0.001 and p = 0.011, respectively). Hb1Ac (p = 0.009 and p = 0.004), leptin (p < 0.000 and p < 0.000), and leptin/adiponectin ratio (p < 0.000 and p < 0.000) decreased significantly in both groups after 24 weeks (FOG and PG respectively). Serum resistin (FOG p < 0.000 and PG p = 0.001), insulin (FOG p < 0.000 and PG p < 0.000), and HOMA-IR (FOG p = 0.000 and PG p < 0.000) increased significantly in both groups. FOG had an overall improvement in the lipid profile with a significant decrease in triacylgycerols (p = 0.002) and atherogenic index (p = 0.031); in contrast, the PG group had increased total cholesterol (p < 0.000), non-HDL cholesterol (p < 0.000), and atherogenic index (p = 0.017). CONCLUSIONS We found a beneficial effect of n-3 PUFA supplementation on waist circumference, glucose, Hb1Ac, leptin, leptin/adiponectin ratio, and lipid profile, without significant changes in adiponectin, and increases in resistin, insulin, and HOMA-IR in both groups.
Collapse
Affiliation(s)
- M Gorety Jacobo-Cejudo
- Faculty of Medicine, Universidad Autónoma del Estado de México, Paseo Tollocan esq. Jesús Carranza, Col. Moderna de la Cruz, Toluca 50180, Mexico.
| | - Roxana Valdés-Ramos
- Faculty of Medicine, Universidad Autónoma del Estado de México, Paseo Tollocan esq. Jesús Carranza, Col. Moderna de la Cruz, Toluca 50180, Mexico.
| | - Ana L Guadarrama-López
- Faculty of Medicine, Universidad Autónoma del Estado de México, Paseo Tollocan esq. Jesús Carranza, Col. Moderna de la Cruz, Toluca 50180, Mexico.
| | - Rosa-Virgen Pardo-Morales
- Instlituto Materno-Infantil del Estado de México, Paseo Colón s/n, Col. Villa Hogar, Toluca 50170, Mexico.
| | - Beatriz E Martínez-Carrillo
- Faculty of Medicine, Universidad Autónoma del Estado de México, Paseo Tollocan esq. Jesús Carranza, Col. Moderna de la Cruz, Toluca 50180, Mexico.
| | - Laurence S Harbige
- Faculty of Life Sciences and Computing, London Metropolitan University, 166-220 Holloway Road, London N7 8DB, UK.
| |
Collapse
|
34
|
Poreba M, Mostowik M, Siniarski A, Golebiowska-Wiatrak R, Malinowski KP, Haberka M, Konduracka E, Nessler J, Undas A, Gajos G. Treatment with high-dose n-3 PUFAs has no effect on platelet function, coagulation, metabolic status or inflammation in patients with atherosclerosis and type 2 diabetes. Cardiovasc Diabetol 2017; 16:50. [PMID: 28410617 PMCID: PMC5391604 DOI: 10.1186/s12933-017-0523-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/19/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Despite numerous studies on cardioprotective effects of omega-3 polyunsaturated fatty acids (n-3 PUFAs), there is limited evidence for n-3 PUFA-mediated effects, especially at its higher dose, on cardiovascular risk in patients with type 2 diabetes (DM2) and established atherosclerosis. PURPOSE To investigate the effect of daily treatment with a higher dose (2 g) of n-3 PUFAs on platelet function, coagulation parameters, fibrin clot properties, markers of systemic inflammation and metabolic status, in patients with atherosclerotic vascular disease and DM2 who receive optimal medical therapy. METHODS We conducted a prospective, double-blind, placebo-controlled, randomized, double-center study, in which thrombin generation (plasma thrombogenic potential from automated thrombogram), fibrin clot properties (plasma fibrin clot permeability; lysis time), platelet aggregation (light transmission aggregometry with adenosine diphosphate and arachidonic acid used as agonists), HbA1c, insulin level, lipid profiles, leptin and adiponectin levels, as well as markers of systemic inflammation (i.e., hsCRP, IL-6, TNF-α, ICAM-1, VCAM-1, and myeloperoxidase) were determined at baseline and at 3 months after treatment with 2 g/day of n-3 PUFAs (n = 36) or placebo (n = 38). Moreover, we assessed serum fatty acids of the phospholipid fraction by gas chromatography both at baseline and at the end of the study. RESULTS Majority of patients were treated with optimal medical therapy and achieved recommended treatment targets. Despite higher serum levels of eicosapentaenoic acid (EPA) (by 204%; p < 0.001) and docosahexaenoic acid (DHA) (by 62%; p < 0.0001) in n-3 PUFA group at the end of treatment no changes in platelet aggregation, thrombin generation, fibrin clot properties or markers of systemic inflammation were observed. No intergroup differences in the insulin, HbA1c and lipid levels were found at the end of the study. There was no change in adiponectin and leptin in interventional group, however leptin increased in control group (p = 0.01), therefore after study period leptin levels were lower in the interventional group (p = 0.01). Additionally, resolvin D1 did not differ between interventional and control group. CONCLUSIONS In conclusion, our study demonstrated that in patients with long-standing, well-controlled DM2 and atherosclerotic disease the treatment with a high dose of n-3 PUFAs (namely, 1 g/day of EPA and 1 g/day of DHA for 3 months) does not improve coagulation, metabolic, and inflammatory status when measured with the specified tests. The study was registered in ClinicalTrials.gov; identifier: NCT02178501. Registration date: April 12, 2014.
Collapse
Affiliation(s)
| | - Magdalena Mostowik
- John Paul II Hospital, Pradnicka 80, 31-202, Kraków, Poland.,Department of Medical Education, Jagiellonian University Medical College, Kraków, Poland
| | | | | | - Krzysztof Piotr Malinowski
- Institute of Public Health, Faculty of Health Science, Jagiellonian University Medical College, Kraków, Poland
| | - Maciej Haberka
- Department of Cardiology, School of Health Science, Medical University of Silesia, Katowice, Poland
| | - Ewa Konduracka
- John Paul II Hospital, Pradnicka 80, 31-202, Kraków, Poland.,Department of Coronary Disease and Heart Failure, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, Pradnicka 80, 31-202, Kraków, Poland
| | - Jadwiga Nessler
- John Paul II Hospital, Pradnicka 80, 31-202, Kraków, Poland.,Department of Coronary Disease and Heart Failure, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, Pradnicka 80, 31-202, Kraków, Poland
| | - Anetta Undas
- John Paul II Hospital, Pradnicka 80, 31-202, Kraków, Poland.,Institute of Cardiology, Jagiellonian University Medical College, Pradnicka 80, 31-202, Kraków, Poland
| | - Grzegorz Gajos
- John Paul II Hospital, Pradnicka 80, 31-202, Kraków, Poland. .,Department of Coronary Disease and Heart Failure, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, Pradnicka 80, 31-202, Kraków, Poland.
| |
Collapse
|
35
|
Ebrahimi FA, Samimi M, Foroozanfard F, Jamilian M, Akbari H, Rahmani E, Ahmadi S, Taghizadeh M, Memarzadeh MR, Asemi Z. The Effects of Omega-3 Fatty Acids and Vitamin E Co-Supplementation on Indices of Insulin Resistance and Hormonal Parameters in Patients with Polycystic Ovary Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Exp Clin Endocrinol Diabetes 2017; 125:353-359. [PMID: 28407657 DOI: 10.1055/s-0042-117773] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This study was conducted to determine the effects of omega-3 fatty acids and vitamin E co-supplementation on indices of insulin resistance and hormonal parameters in women with polycystic ovary syndrome (PCOS). This randomized double-blind, placebo-controlled trial was done on 68 women diagnosed with PCOS according to the Rotterdam criteria aged 18-40 years old. Participants were randomly assigned into 2 groups to receive either 1 000 mg omega-3 fatty acids from flaxseed oil containing 400 mg α-Linolenic acid plus 400 IU vitamin E supplements (n=34) or placebo (n=34) for 12 weeks. Hormonal parameters were quantified at the beginning of the study and after 12-week intervention. After 12 weeks of intervention, compared to the placebo, omega-3 fatty acids and vitamin E co-supplementation resulted in a significant decrease in insulin (-1.0±3.5 vs. +2.7±6.6 µIU/mL, P=0.004), homeostasis model of assessment-estimated insulin resistance (-0.2±0.8 vs. +0.6±1.5, P=0.005), homeostasis model of assessment-estimated B cell function (-4.3±14.3 vs. +10.5±24.5, P=0.004) and a significant increase in quantitative insulin sensitivity check index (+0.006±0.02 vs. -0.01±0.04, P=0.008). Supplementation with omega-3 fatty acids plus vitamin E led to significant reductions in serum total testosterone (-0.5±0.7 vs. -0.1±0.5 ng/mL, P=0.008) and free testosterone (-1.2±2.1 vs. -0.2±1.7, P=0.04) compared to the placebo group. We did not observe any significant effect of omega-3 fatty acids and vitamin E co-supplementation on fasting plasma glucose and other hormonal profiles. Omega-3 fatty acids and vitamin E co-supplementation for 12 weeks in PCOS women significantly improved indices of insulin resistance, total and free testosterone.
Collapse
Affiliation(s)
- Faraneh Afshar Ebrahimi
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Mansooreh Samimi
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Fatemeh Foroozanfard
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Mehri Jamilian
- Endocrinology and Metabolism Research Center, Department of Gynecology and Obstetrics, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Hossein Akbari
- Department of Biostatistics, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Elham Rahmani
- Department of Gynecology and Obstetrics, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Shahnaz Ahmadi
- Department of Gynecology and Obstetrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|
36
|
Colussi G, Catena C, Novello M, Bertin N, Sechi LA. Impact of omega-3 polyunsaturated fatty acids on vascular function and blood pressure: Relevance for cardiovascular outcomes. Nutr Metab Cardiovasc Dis 2017; 27:191-200. [PMID: 27692558 DOI: 10.1016/j.numecd.2016.07.011] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/25/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022]
Abstract
AIMS To overview the effects of omega-3 polyunsaturated fatty acids (PUFA) on blood vessels and blood pressure (BP) and their relevance for cardiovascular prevention. DATA SYNTHESIS The importance of omega-3 PUFA for the cardiovascular system has come under the spotlight during the last decades. These fatty acids are present in variable amounts in cell membranes of mammal species, and their content affects a variety of cellular functions. Evidence obtained in animal and human studies suggests that omega-3 PUFA affect many steps of the atherosclerotic process. In blood vessels, omega-3 PUFA improve endothelial function; promote vasodilatation through relaxation of smooth muscle cells; exert antioxidant, anti-inflammatory, and antithrombotic actions; delay development of plaques and increase their stability; and decrease wall stiffening. Omega-3 PUFA might affect BP, and studies conducted with ambulatory monitoring suggest that supplementation with these fatty acids decreases the average 24-h BP levels. This effect on BP is related to the pretreatment membrane content of omega-3 PUFA, and this might explain some inconsistencies among intervention trials. Meta-analyses indicate that omega-3 PUFA have a mild but significant BP lowering effect. While encouraging results were initially obtained with the use of omega-3 PUFA supplements in secondary prevention trials, meta-analyses have not confirmed the ability of these fatty acids to decrease the risk of coronary heart and cerebrovascular disease. CONCLUSIONS Omega-3 PUFA are associated with significant improvement in vascular function and lowering of BP. However, the evidence currently supporting the role of these fatty acids in cardiovascular prevention is weak and needs further investigation.
Collapse
Affiliation(s)
- G Colussi
- Internal Medicine, Department of Experimental and Medical Sciences, University of Udine, Udine, Italy
| | - C Catena
- Internal Medicine, Department of Experimental and Medical Sciences, University of Udine, Udine, Italy
| | - M Novello
- Internal Medicine, Department of Experimental and Medical Sciences, University of Udine, Udine, Italy
| | - N Bertin
- Internal Medicine, Department of Experimental and Medical Sciences, University of Udine, Udine, Italy
| | - L A Sechi
- Internal Medicine, Department of Experimental and Medical Sciences, University of Udine, Udine, Italy.
| |
Collapse
|
37
|
Barbosa MMDAL, Damasceno NRT. The benefits of ω-3 supplementation depend on adiponectin basal level and adiponectin increase after the supplementation: A randomized clinical trial. Nutrition 2017; 34:7-13. [DOI: 10.1016/j.nut.2016.08.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/11/2016] [Accepted: 08/23/2016] [Indexed: 01/28/2023]
|
38
|
Kuda O, Brezinova M, Rombaldova M, Slavikova B, Posta M, Beier P, Janovska P, Veleba J, Kopecky J, Kudova E, Pelikanova T, Kopecky J. Docosahexaenoic Acid-Derived Fatty Acid Esters of Hydroxy Fatty Acids (FAHFAs) With Anti-inflammatory Properties. Diabetes 2016; 65:2580-90. [PMID: 27313314 DOI: 10.2337/db16-0385] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/07/2016] [Indexed: 11/13/2022]
Abstract
White adipose tissue (WAT) is a complex organ with both metabolic and endocrine functions. Dysregulation of all of these functions of WAT, together with low-grade inflammation of the tissue in obese individuals, contributes to the development of insulin resistance and type 2 diabetes. n-3 polyunsaturated fatty acids (PUFAs) of marine origin play an important role in the resolution of inflammation and exert beneficial metabolic effects. Using experiments in mice and overweight/obese patients with type 2 diabetes, we elucidated the structures of novel members of fatty acid esters of hydroxy fatty acids-lipokines derived from docosahexaenoic acid (DHA) and linoleic acid, which were present in serum and WAT after n-3 PUFA supplementation. These compounds contained DHA esterified to 9- and 13-hydroxyoctadecadienoic acid (HLA) or 14-hydroxydocosahexaenoic acid (HDHA), termed 9-DHAHLA, 13-DHAHLA, and 14-DHAHDHA, and were synthesized by adipocytes at concentrations comparable to those of protectins and resolvins derived from DHA in WAT. 13-DHAHLA exerted anti-inflammatory and proresolving properties while reducing macrophage activation by lipopolysaccharides and enhancing the phagocytosis of zymosan particles. Our results document the existence of novel lipid mediators, which are involved in the beneficial anti-inflammatory effects attributed to n-3 PUFAs, in both mice and humans.
Collapse
Affiliation(s)
- Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marie Brezinova
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Rombaldova
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Barbora Slavikova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Posta
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Beier
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Petra Janovska
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Veleba
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Kopecky
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Kudova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Terezie Pelikanova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Kopecky
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
39
|
Chiesa G, Busnelli M, Manzini S, Parolini C. Nutraceuticals and Bioactive Components from Fish for Dyslipidemia and Cardiovascular Risk Reduction. Mar Drugs 2016; 14:md14060113. [PMID: 27338419 PMCID: PMC4926072 DOI: 10.3390/md14060113] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/11/2016] [Accepted: 05/26/2016] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease remains the most common health problem in developed countries, and residual risk after implementing all current therapies is still high. Permanent changes in lifestyle may be hard to achieve and people may not always be motivated enough to make the recommended modifications. Emerging research has explored the application of natural food-based strategies in disease management. In recent years, much focus has been placed on the beneficial effects of fish consumption. Many of the positive effects of fish consumption on dyslipidemia and heart diseases have been attributed to n-3 polyunsaturated fatty acids (n-3 PUFAs, i.e., EPA and DHA); however, fish is also an excellent source of protein and, recently, fish protein hydrolysates containing bioactive peptides have shown promising activities for the prevention/management of cardiovascular disease and associated health complications. The present review will focus on n-3 PUFAs and bioactive peptides effects on cardiovascular disease risk factors. Moreover, since considerable controversy exists regarding the association between n-3 PUFAs and major cardiovascular endpoints, we have also reviewed the main clinical trials supporting or not this association.
Collapse
Affiliation(s)
- Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, Milano 20133, Italy.
| | - Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, Milano 20133, Italy.
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, Milano 20133, Italy.
| | - Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, Milano 20133, Italy.
| |
Collapse
|
40
|
Association of fish consumption and dietary intake of marine n-3 PUFA with myocardial infarction in a prospective Danish cohort study. Br J Nutr 2016; 116:167-77. [PMID: 27189437 DOI: 10.1017/s000711451600180x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Several studies have investigated the potential benefits of marine n-3 PUFA in CVD, generally suggesting a lower risk of CHD. However, recent trials have questioned these results. This study investigated the association of fish consumption with dietary intake of marine n-3 PUFA with incident myocardial infarction (MI). In a Danish cohort study, 57 053 subjects between 50 and 64 years of age were enrolled from 1993 to 1997. From national registries, we identified all cases of incident MI. Dietary fish consumption was assessed using a semi-quantitative food questionnaire, including twenty-six questions regarding fish intake. In addition, we calculated the intake of total and individual marine n-3 PUFA. During a median follow-up of 17·0 years, we identified 3089 cases of incident MI. For both men and women, a high intake of fatty fish was inversely related to incident MI. Thus, when comparing the highest and the lowest quintile of fatty fish intake, we found a 12 % lower relative risk of MI in men (hazard ratio (HR) 0·88; 95 % CI 0·77, 1·00) and a 22 % lower relative risk in women (HR 0·78; 95 % CI 0·63, 0·96) after adjustments. For women, similar associations were observed for individual and total marine n-3 PUFA. In contrast, intake of lean fish was not associated with MI. In conclusion, incident MI was inversely related to a high intake of fatty fish, but not lean fish. However, test for trends across quintiles was not statistically significant. In general, this study supports the view that consumption of fatty fish may protect against MI.
Collapse
|
41
|
Rivas E, Wooten JS, Newmire DE, Ben-Ezra V. Omega-3 fatty acid supplementation combined with acute aerobic exercise does not alter the improved post-exercise insulin response in normoglycemic, inactive and overweight men. Eur J Appl Physiol 2016; 116:1255-65. [PMID: 27155848 DOI: 10.1007/s00421-016-3387-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 05/01/2016] [Indexed: 12/16/2022]
Abstract
PURPOSE The aim of this study was to determine if omega-3 (n-3) supplementation combined with acute aerobic exercise would improve glucose and insulin responses in normoglycemic, inactive, overweight men. METHODS In a random order, ten inactive and normoglycemic men (30.6 ± 10 years, 85.4 ± 11 kg, 26.7 ± 4 BMI) completed a rest (R) and exercise trial (EX) without n-3 supplementation. Following 42 days of n-3 supplementation, participants again completed a rest (R + n-3) and exercise trial (EX + n-3) with continued n-3 supplementation. The exercise trial consisted of 3 days of ~70 % VO2peak for 60 min/session. N-3 supplementation entailed 4.55 g/day of n-3 (EPA 2.45 g, DHA 1.61 g). A 75 g oral glucose tolerance (OGTT) test was administered 14-16 h after each trial. RESULTS Relative to R (35,278 ± 9169 pmol/L), EX without n-3 reduced the incremental area under the curve for insulin (iAUCinsulin) during an OGTT by 21.3 % (27765 ± 4925 pmol/L, p = 0.018) and 20.6 % after the EX + n-3 trial (27,999 ± 8370 pmol/L; p = 0.007). In addition, EX (96 ± 21 pmol/L; p = 0.006) reduced C-peptide by 13.5 % when compared to R (111 ± 26 pmol/L). No difference was observed between R and n-3 trials for iAUCinsulin and iAUCC-peptide. Only EX improved insulin sensitivity index by 5.6 % (p = 0.02) when compared to R. CONCLUSIONS These data suggest that n-3 supplementation does not add any additional benefit beyond the exercise induced insulin responses in inactive men. Furthermore, n-3 supplementation alone does not appear to impair insulin action in normoglycemic, inactive, overweight men.
Collapse
Affiliation(s)
- Eric Rivas
- Exercise Physiology and Biochemistry Laboratory, Department of Kinesiology, Texas Woman's University, Denton, TX, USA. .,Institute for Clinical and Translational Science & Department of Pediatrics, The University of California, Irvine, CA, USA.
| | - Joshua S Wooten
- Department of Kinesiology and Health Education, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Daniel E Newmire
- Exercise Physiology and Biochemistry Laboratory, Department of Kinesiology, Texas Woman's University, Denton, TX, USA
| | - Vic Ben-Ezra
- Exercise Physiology and Biochemistry Laboratory, Department of Kinesiology, Texas Woman's University, Denton, TX, USA
| |
Collapse
|
42
|
Balfegó M, Canivell S, Hanzu FA, Sala-Vila A, Martínez-Medina M, Murillo S, Mur T, Ruano EG, Linares F, Porras N, Valladares S, Fontalba M, Roura E, Novials A, Hernández C, Aranda G, Sisó-Almirall A, Rojo-Martínez G, Simó R, Gomis R. Effects of sardine-enriched diet on metabolic control, inflammation and gut microbiota in drug-naïve patients with type 2 diabetes: a pilot randomized trial. Lipids Health Dis 2016; 15:78. [PMID: 27090218 PMCID: PMC4836051 DOI: 10.1186/s12944-016-0245-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 04/07/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Nutrition therapy is the cornerstone of treating diabetes mellitus. The inclusion of fish (particularly oily fish) at least two times per week is recommended by current international dietary guidelines for type 2 diabetes. In contrast to a large number of human studies examining the effects of oily fish on different cardiovascular risk factors, little research on this topic is available in patients with type 2 diabetes. The aims of this pilot study were to investigate the effects of a sardine-enriched diet on metabolic control, adiponectin, inflammatory markers, erythrocyte membrane fatty acid (EMFA) composition, and gut microbiota in drug-naïve patients with type 2 diabetes. METHODS 35 drug-naïve patients with type 2 diabetes were randomized to follow either a type 2 diabetes standard diet (control group: CG), or a standard diet enriched with 100 g of sardines 5 days a week (sardine group: SG) for 6 months. Anthropometric, dietary information, fasting glycated hemoglobin, glucose, insulin, adiponectin, inflammatory markers, EMFA and specific bacterial strains were determined before and after intervention. RESULTS There were no significant differences in glycemic control between groups at the end of the study. Both groups decreased plasma insulin (SG: -35.3%, P = 0.01, CG: -22.6%, P = 0.02) and homeostasis model of assessment--insulin resistance (HOMA-IR) (SG: -39.2%, P = 0.007, CG: -21.8%, P = 0.04) at 6-months from baseline. However only SG increased adiponectin in plasma compared to baseline level (+40.7%, P = 0.04). The omega-3 index increased 2.6% in the SG compared to 0.6% in the CG (P = 0.001). Both dietary interventions decreased phylum Firmicutes (SG and CG: P = 0.04) and increased E. coli concentrations (SG: P = 0.01, CG: P = 0.03) at the end of the study from baseline, whereas SG decreased Firmicutes/Bacteroidetes ratio (P = 0.04) and increased Bacteroides-Prevotella (P = 0.004) compared to baseline. CONCLUSIONS Although enriching diet with 100 g of sardines 5 days a week during 6 months to a type 2 diabetes standard diet seems to have neutral effects on glycemic control in drug-naïve patients with type 2 diabetes, this nutritional intervention could have beneficial effects on cardiovascular risk. Furthermore, both dietary interventions decreased HOMA-IR and altered gut microbiota composition of drug-naïve patients with type 2 diabetes. TRIAL REGISTRATION Trial number and name of the registry: NCT02294526, ClinicalTrials.gov.
Collapse
Affiliation(s)
- Mariona Balfegó
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain. .,Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), c/ Roselló 149 planta 5, 08036, Barcelona, Spain.
| | - Silvia Canivell
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), c/ Roselló 149 planta 5, 08036, Barcelona, Spain.,Les Corts Primary Health Care Center, Tranverse Group for Research in Primary Care, IDIBAPS, c/ Mejia Lequerica s/n, 08028, Barcelona, Spain.,Present address: Centre Hospitalier Universitaire Vaudois (CHUV), Departement de Endocrinologie, Rue Saint-Martin 3, CH-1003, Lausanne, Switzerland
| | - Felicia A Hanzu
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Department of Endocrinology and Nutrition, Hospital Clínic of Barcelona, c/Villarroel 170 Escala 11 planta 2, 08036, Barcelona, Spain.,University of Barcelona, Facultat de Medicina, c/ Casanova 143, 08036, Barcelona, Spain
| | - Aleix Sala-Vila
- CIBER in Physiopathology of Obesity and Nutrition (CIBERobn), c/Villarroel 170, Edifici Helios, 08036, Barcelona, Spain
| | - Margarita Martínez-Medina
- Laboratory of Molecular Microbiology, Biology Department, University of Girona, Av. Montilivi s/n, E-17071, Girona, Spain
| | - Serafín Murillo
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), c/ Roselló 149 planta 5, 08036, Barcelona, Spain
| | - Teresa Mur
- Terrassa Sud Primary Health Care Center, Mútua de Terrassa, Av. Santa Eulàlia s/n, 08223, Terrassa, Barcelona, Spain
| | - Elena G Ruano
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), c/ Roselló 149 planta 5, 08036, Barcelona, Spain
| | - Francisca Linares
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Endocrinology and Nutrition Department, Hospital Carlos Haya, Biomedical Research Institute of Málaga (IBIMA), Plaza Hospital Civil s/n Sótano Pabellón 1, 29009, Málaga, Spain
| | - Nuria Porras
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Endocrinology and Nutrition Department, Hospital Carlos Haya, Biomedical Research Institute of Málaga (IBIMA), Plaza Hospital Civil s/n Sótano Pabellón 1, 29009, Málaga, Spain
| | - Silvia Valladares
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Vall d'Hebrón Research Institute and Autonomous University of Barcelona, Pg. de la Vall d'Hebrón 119-129 planta 8, Barcelona, Spain
| | - Maria Fontalba
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Endocrinology and Nutrition Department, Hospital Carlos Haya, Biomedical Research Institute of Málaga (IBIMA), Plaza Hospital Civil s/n Sótano Pabellón 1, 29009, Málaga, Spain
| | - Elena Roura
- Alicia Foundation, Camí Sant Benet, 08272 Sant Fruitós de Bages, Barcelona, Spain
| | - Anna Novials
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), c/ Roselló 149 planta 5, 08036, Barcelona, Spain.,Department of Endocrinology and Nutrition, Hospital Clínic of Barcelona, c/Villarroel 170 Escala 11 planta 2, 08036, Barcelona, Spain
| | - Cristina Hernández
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Vall d'Hebrón Research Institute and Autonomous University of Barcelona, Pg. de la Vall d'Hebrón 119-129 planta 8, Barcelona, Spain
| | - Gloria Aranda
- Department of Endocrinology and Nutrition, Hospital Clínic of Barcelona, c/Villarroel 170 Escala 11 planta 2, 08036, Barcelona, Spain
| | - Antoni Sisó-Almirall
- Les Corts Primary Health Care Center, Tranverse Group for Research in Primary Care, IDIBAPS, c/ Mejia Lequerica s/n, 08028, Barcelona, Spain.,University of Barcelona, Facultat de Medicina, c/ Casanova 143, 08036, Barcelona, Spain
| | - Gemma Rojo-Martínez
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Endocrinology and Nutrition Department, Hospital Carlos Haya, Biomedical Research Institute of Málaga (IBIMA), Plaza Hospital Civil s/n Sótano Pabellón 1, 29009, Málaga, Spain
| | - Rafael Simó
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Vall d'Hebrón Research Institute and Autonomous University of Barcelona, Pg. de la Vall d'Hebrón 119-129 planta 8, Barcelona, Spain
| | - Ramon Gomis
- CIBER in Diabetes and Associated Metabolic Disorders (CIBERDEM), c/ Monforte de Lemos 3-5 Pabellón 11 planta 0, 28029, Madrid, Spain.,Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), c/ Roselló 149 planta 5, 08036, Barcelona, Spain.,Department of Endocrinology and Nutrition, Hospital Clínic of Barcelona, c/Villarroel 170 Escala 11 planta 2, 08036, Barcelona, Spain.,University of Barcelona, Facultat de Medicina, c/ Casanova 143, 08036, Barcelona, Spain
| |
Collapse
|
43
|
Masquio DCL, de Piano-Ganen A, Oyama LM, Campos RMDS, Santamarina AB, de Souza GIDMH, Gomes AD, Moreira RG, Corgosinho FC, do Nascimento CMO, Tock L, Tufik S, de Mello MT, Dâmaso AR. The role of free fatty acids in the inflammatory and cardiometabolic profile in adolescents with metabolic syndrome engaged in interdisciplinary therapy. J Nutr Biochem 2016; 33:136-44. [PMID: 27155920 DOI: 10.1016/j.jnutbio.2016.03.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 03/15/2016] [Accepted: 03/31/2016] [Indexed: 01/15/2023]
Abstract
The purpose of the present study was to evaluate if interdisciplinary therapy can influence the cardiometabolic and serum free fatty acid profile. The second aim was to evaluate if there is an association between serum free fatty acids, inflammation and cardiometabolic biomarkers in obese adolescents with and without metabolic syndrome submitted to a long-term interdisciplinary therapy. The study involved 108 postpuberty obese adolescents, who were divided according to metabolic syndrome (MetS) diagnosis: MetS (n=32) and Non-MetS (n=76). The interdisciplinary therapy consisted of a 1-year period of nutrition, psychology, physical exercise and clinical support. After therapy, both groups improved metabolic, inflammatory (leptin, adiponectin, leptin/adiponectin ratio, adiponectin/leptin ratio and C-reactive protein) and cardiometabolic profile (PAI-1 and ICAM). Metabolic syndrome prevalence reduced from 28.70% to 12.96%. Both groups reduced myristic acid (C14:0) and increased docosahexaenoic acid (DHA, C22:6n3), heneicosapentaenoic acid (HPA, C21:5n3) and arachidonic acid (C20:4n6). After adjustment for metabolic syndrome and the number of metabolic syndrome parameters, multiple regression analysis showed that changes in VCAM and PAI-1 were negatively associated with changes in cis-linoleic acid (C18:2n6c). Additionally, changes in trans-linoleic acid (C18:2n6t) were also positively associated with these biomarkers. Moreover, leptin and leptin/adiponectin ratio were negatively associated with changes in docosapentaenoic acid (DPA, C22:5n3) and stearidonic acid (SDA, C18:4n3). Adiponectin/leptin ratio was positively associated with docosapentaenoic acid (DPA, C22:5n3). Changes in adiponectin were positively correlated with changes in omega 3, such as heneicosapentaenoic acid (HPA, C21:5n3) and docosapentaenoic acid (DPA, C22:5n3). Results support that interdisciplinary therapy can control inflammatory and cardiometabolic profile in obese adolescents. Moreover, serum fatty acids can be influenced by lifestyle changes and are able to modulate these biomarkers.
Collapse
Affiliation(s)
| | | | - Lila Missae Oyama
- Programa de Pós-Graduação em Nutrição, Universidade Federal de São Paulo (UNIFESP), Brazil; Laboratório de Fisiologia da Nutrição, Universidade Federal de São Paulo (UNIFESP), Brazil
| | | | | | | | - Aline Dal'Olio Gomes
- Departamento de Fisiologia Geral, Instituto de Biociências, Universidade de São Paulo (USP), Brazil
| | - Renata Guimarães Moreira
- Departamento de Fisiologia Geral, Instituto de Biociências, Universidade de São Paulo (USP), Brazil
| | | | - Claudia Maria Oller do Nascimento
- Programa de Pós-Graduação em Nutrição, Universidade Federal de São Paulo (UNIFESP), Brazil; Laboratório de Fisiologia da Nutrição, Universidade Federal de São Paulo (UNIFESP), Brazil
| | | | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - Marco Túlio de Mello
- Programa de Pós-Graduação em Nutrição, Universidade Federal de São Paulo (UNIFESP), Brazil; Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - Ana R Dâmaso
- Programa de Pós-Graduação em Nutrição, Universidade Federal de São Paulo (UNIFESP), Brazil.
| |
Collapse
|
44
|
Chrysohoou C, Metallinos G, Georgiopoulos G, Mendrinos D, Papanikolaou A, Magkas N, Pitsavos C, Vyssoulis G, Stefanadis C, Tousoulis D. Short term omega-3 polyunsaturated fatty acid supplementation induces favorable changes in right ventricle function and diastolic filling pressure in patients with chronic heart failure; A randomized clinical trial. Vascul Pharmacol 2016; 79:43-50. [PMID: 26807502 DOI: 10.1016/j.vph.2016.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/21/2015] [Accepted: 01/21/2016] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Omega-3 polyunsaturated fatty acids (omega 3-PUFAs) seem to favorably affect cardiac hemodynamics and may benefit the clinical course of heart failure patients. The role of omega 3-PUFAs supplementation on the left and right ventricular function of patients with chronic compensated systolic heart failure, under optimal treatment, was studied. METHODS 205 consecutive patients with chronic compensated heart failure, due to ischemic (IHF) or dilated cardiomyopathy (DCM)-NYHA classification I-III, under optimal medical treatment, were enrolled. Participants were 1-to-1 randomized on 1000 mg omega 3-PUFA supplementation or no supplementation, in a non-blinded fashion. Echocardiographic assessment was performed at first visit and 6 months after. Plasma BNP and serum creatinine levels were also measured. RESULTS As compared with the control group, BNP levels in omega 3-PUFA intervention group were 34.6% lower (p=0.001); end-diastolic and end-systolic left ventricle dimensions were decreased by 2.5% (p=0.047) and 3.7% (p=0.01), maximum diameter of left atrium was decreased by 8.4% (p=0.004), left atrium ejection fraction was ameliorated by 6.03% (p=0.021) and as regards tissue Doppler parameters, TDI_Etv/Atv was decreased in omega 3-PUFA intervention group by 6.3% (p=0.038). Moreover, improvement in diastolic indices was more prominent in subjects with DCM as compared to IHF patients. CONCLUSION Omega 3-PUFA supplementation was associated with improved left diastolic function and decreased BNP levels in patients with chronic heart failure. These findings suggest a beneficial role of omega 3-PUFAs on the hemodynamic course of patients with systolic heart failure.
Collapse
Affiliation(s)
| | - George Metallinos
- 1st Cardiology Clinic, University of Athens, Hippokratio Hospital, Greece
| | | | | | | | - Nikolaos Magkas
- 1st Cardiology Clinic, University of Athens, Hippokratio Hospital, Greece
| | - Christos Pitsavos
- 1st Cardiology Clinic, University of Athens, Hippokratio Hospital, Greece
| | - Gregory Vyssoulis
- 1st Cardiology Clinic, University of Athens, Hippokratio Hospital, Greece
| | | | - Dimitris Tousoulis
- 1st Cardiology Clinic, University of Athens, Hippokratio Hospital, Greece
| |
Collapse
|
45
|
A randomized-controlled clinical trial investigating the effect of omega-3 fatty acids and vitamin E co-supplementation on markers of insulin metabolism and lipid profiles in gestational diabetes. J Clin Lipidol 2016; 10:386-93. [PMID: 27055970 DOI: 10.1016/j.jacl.2015.12.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/02/2015] [Accepted: 12/21/2015] [Indexed: 01/17/2023]
|
46
|
Consumption of a healthy dietary pattern results in significant reductions in C-reactive protein levels in adults: a meta-analysis. Nutr Res 2016; 36:391-401. [PMID: 27101757 DOI: 10.1016/j.nutres.2016.02.009] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/17/2016] [Accepted: 02/19/2016] [Indexed: 12/14/2022]
Abstract
Consumption of healthy dietary patterns has been associated with reduced risk of cardiovascular disease and metabolic syndrome. Dietary intervention targets disease prevention, so studies increasingly use biomarkers of underlying inflammation and metabolic syndrome progression to examine the diet-health relationship. The extent to which these biomarkers contribute to the body of evidence on healthy dietary patterns is unknown. The aim of this meta-analysis was to determine the effect of healthy dietary patterns on biomarkers associated with adiposity, insulin resistance, and inflammation in adults. A systematic search of Scopus, PubMed, Web of Science, and Cochrane Central Register of Controlled Trials (all years to April 2015) was conducted. Inclusion criteria were randomized controlled trials; effects of dietary patterns assessed on C-reactive protein (CRP), total adiponectin, high-molecular-weight adiponectin, tumor necrosis factor-α, adiponectin:leptin, resistin, or retinol binding protein 4. Random effects meta-analyses were conducted to assess the weighted mean differences in change or final mean values for each outcome. Seventeen studies were included in the review. These reflected research on dietary patterns associated with the Mediterranean diet, Nordic diet, Tibetan diet, and the Dietary Approaches to Stop Hypertension diet. Consumption of a healthy dietary pattern was associated with significant reductions in CRP (weighted mean difference, -0.75 [-1.16, -0.35]; P = .0003). Non-significant changes were found for all other biomarkers. This analysis found evidence for favorable effects of healthy dietary patterns on CRP, with limited evidence for other biomarkers. Future research should include additional randomized controlled trials incorporating a greater range of dietary patterns and biomarkers.
Collapse
|
47
|
Abstract
Suboptimal nutrition is a leading cause of poor health. Nutrition and policy science have advanced rapidly, creating confusion yet also providing powerful opportunities to reduce the adverse health and economic impacts of poor diets. This review considers the history, new evidence, controversies, and corresponding lessons for modern dietary and policy priorities for cardiovascular diseases, obesity, and diabetes mellitus. Major identified themes include the importance of evaluating the full diversity of diet-related risk pathways, not only blood lipids or obesity; focusing on foods and overall diet patterns, rather than single isolated nutrients; recognizing the complex influences of different foods on long-term weight regulation, rather than simply counting calories; and characterizing and implementing evidence-based strategies, including policy approaches, for lifestyle change. Evidence-informed dietary priorities include increased fruits, nonstarchy vegetables, nuts, legumes, fish, vegetable oils, yogurt, and minimally processed whole grains; and fewer red meats, processed (eg, sodium-preserved) meats, and foods rich in refined grains, starch, added sugars, salt, and trans fat. More investigation is needed on the cardiometabolic effects of phenolics, dairy fat, probiotics, fermentation, coffee, tea, cocoa, eggs, specific vegetable and tropical oils, vitamin D, individual fatty acids, and diet-microbiome interactions. Little evidence to date supports the cardiometabolic relevance of other popular priorities: eg, local, organic, grass-fed, farmed/wild, or non-genetically modified. Evidence-based personalized nutrition appears to depend more on nongenetic characteristics (eg, physical activity, abdominal adiposity, gender, socioeconomic status, culture) than genetic factors. Food choices must be strongly supported by clinical behavior change efforts, health systems reforms, novel technologies, and robust policy strategies targeting economic incentives, schools and workplaces, neighborhood environments, and the food system. Scientific advances provide crucial new insights on optimal targets and best practices to reduce the burdens of diet-related cardiometabolic diseases.
Collapse
Affiliation(s)
- Dariush Mozaffarian
- From Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA.
| |
Collapse
|
48
|
De Boer AA, Monk JM, Liddle DM, Power KA, Ma DWL, Robinson LE. Fish Oil-Derived Long-Chain n-3 Polyunsaturated Fatty Acids Reduce Expression of M1-Associated Macrophage Markers in an ex vivo Adipose Tissue Culture Model, in Part through Adiponectin. Front Nutr 2015; 2:31. [PMID: 26528480 PMCID: PMC4602148 DOI: 10.3389/fnut.2015.00031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/28/2015] [Indexed: 01/03/2023] Open
Abstract
Adipose tissue (AT) macrophages (ATM) play a key role in obesity-associated pathologies, and their phenotype can be influenced by the local tissue microenvironment. Interestingly, long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) and the LC n-3 PUFA-upregulated adipokine, adiponectin (Ad), may mitigate excessive ATM inflammatory M1-polarization responses. However, to what extent LC n-3 PUFA and Ad work in concert to affect macrophage phenotype has not been examined. Thus, we used an established ex vivo AT organ culture model using visceral AT from mice fed a control (CON; 10% w/w safflower oil) n-6 PUFA-rich diet or an isocaloric fish oil (FO; 3% w/w menhaden oil + 7% w/w safflower oil)-derived LC n-3 PUFA-rich diet to generate AT conditioned media (ACM). We then evaluated if CON or FO ACM affected macrophage polarization markers in a model designed to mimic acute [18 h ACM plus lipopolysaccharide (LPS) for the last 6 h] or chronic (macrophages treated with LPS-challenged CON or FO ACM for 24 h) inflammation ± Ad-neutralizing antibody and the LPS-neutralizing agent, polymyxin B. In the acute inflammation model, macrophages treated with FO ACM had decreased lipid uptake and mRNA expression of M1 markers (Nos2, Nfκb, Il6, Il18, Ccl2, and Ccl5) compared with CON ACM (p ≤ 0.05); however, these effects were largely attenuated when Ad was neutralized (p > 0.05). Furthermore, in the chronic inflammation model, macrophages treated with FO ACM had decreased mRNA expression of M1 markers (Nos2, Tnfα, Ccl2, and Il1β) and IL-6 and CCL2 secretion (p ≤ 0.05); however, some of these effects were lost when Ad was neutralized, and were further exacerbated when both Ad and LPS were neutralized. Taken together, this work shows that LC n-3 PUFA and Ad work in concert to suppress certain M1 macrophage responses. Thus, future strategies to modulate the ATM phenotype should consider the role of both LC n-3 PUFA and Ad in mitigating obese AT inflammation.
Collapse
Affiliation(s)
- Anna A De Boer
- Department of Human Health and Nutritional Sciences, University of Guelph , Guelph, ON , Canada
| | - Jennifer M Monk
- Department of Human Health and Nutritional Sciences, University of Guelph , Guelph, ON , Canada ; Guelph Food Research Centre, Agriculture and Agri-Food Canada , Guelph, ON , Canada
| | - Danyelle M Liddle
- Department of Human Health and Nutritional Sciences, University of Guelph , Guelph, ON , Canada
| | - Krista A Power
- Guelph Food Research Centre, Agriculture and Agri-Food Canada , Guelph, ON , Canada
| | - David W L Ma
- Department of Human Health and Nutritional Sciences, University of Guelph , Guelph, ON , Canada
| | - Lindsay E Robinson
- Department of Human Health and Nutritional Sciences, University of Guelph , Guelph, ON , Canada
| |
Collapse
|
49
|
DeClercq V, d'Eon B, McLeod RS. Fatty acids increase adiponectin secretion through both classical and exosome pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1123-33. [DOI: 10.1016/j.bbalip.2015.04.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/06/2015] [Accepted: 04/13/2015] [Indexed: 11/26/2022]
|
50
|
Zanetti M, Grillo A, Losurdo P, Panizon E, Mearelli F, Cattin L, Barazzoni R, Carretta R. Omega-3 Polyunsaturated Fatty Acids: Structural and Functional Effects on the Vascular Wall. BIOMED RESEARCH INTERNATIONAL 2015; 2015:791978. [PMID: 26301252 PMCID: PMC4537737 DOI: 10.1155/2015/791978] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/26/2015] [Indexed: 01/24/2023]
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFA) consumption is associated with reduced cardiovascular disease risk. Increasing evidence demonstrating a beneficial effect of n-3 PUFA on arterial wall properties is progressively emerging. We reviewed the recent available evidence for the cardiovascular effects of n-3 PUFA focusing on structural and functional properties of the vascular wall. In experimental studies and clinical trials n-3 PUFA have shown the ability to improve arterial hemodynamics by reducing arterial stiffness, thus explaining some of its cardioprotective properties. Recent studies suggest beneficial effects of n-3 PUFA on endothelial activation, which are likely to improve vascular function. Several molecular, cellular, and physiological pathways influenced by n-3 PUFA can affect arterial wall properties and therefore interfere with the atherosclerotic process. Although the relative weight of different physiological and molecular mechanisms and the dose-response on arterial wall properties have yet to be determined, n-3 PUFA have the potential to beneficially impact arterial wall remodeling and cardiovascular outcomes by targeting arterial wall stiffening and endothelial dysfunction.
Collapse
Affiliation(s)
- Michela Zanetti
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Andrea Grillo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Pasquale Losurdo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Emiliano Panizon
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Filippo Mearelli
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Luigi Cattin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Rocco Barazzoni
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Renzo Carretta
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| |
Collapse
|