51
|
Sugimoto K, Hosomi R, Yoshida M, Fukunaga K. Dietary Phospholipids Prepared From Scallop Internal Organs Attenuate the Serum and Liver Cholesterol Contents by Enhancing the Expression of Cholesterol Hydroxylase in the Liver of Mice. Front Nutr 2021; 8:761928. [PMID: 34778346 PMCID: PMC8578998 DOI: 10.3389/fnut.2021.761928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
In this study, we successfully prepared scallop oil (SCO), which contains high levels of phospholipids (PL) and eicosapentaenoic acid (EPA), from the internal organs of the Japanese giant scallop (Patinopecten yessoensis), one of the most important underutilized fishery resources in Japan. The intake of SCO lowers the serum and liver cholesterol contents in mice; however, whether the fatty acids (FA) composition or PL of SCO exhibits any cholesterol-lowering effect remains unknown. To elucidate whether the cholesterol-lowering function is due to FA composition or PL of SCO, and investigate the cholesterol-lowering mechanism by SCO, in the present study, mice were fed SCO's PL fraction (SCO-PL), triglyceride (TG)-type oil with almost the same FA composition as SCO-PL, called SCO's TG fraction (SCO-TG), soybean oil (SOY-TG), and soybean's PL fraction (SOY-PL). Male C57BL/6J mice (5-week-old) were fed high-fat and cholesterol diets containing 3% (w/w) experimental oils (SOY-TG, SOY-PL, SCO-TG, and SCO-PL) for 28 days. The SCO-PL diet significantly decreased the serum and liver cholesterol contents compared with the SOY-TG diet, but the intake of SOY-PL and SCO-TG did not show this effect. This result indicated that the serum and liver cholesterol-lowering effect observed in the SCO intake group was due to the effect of SCO-PL. The cholesterol-lowering effect of SCO-PL was in part related to the promotion of liver cholesterol 7α-hydroxylase (CYP7A1) expression, which is the rate-limiting enzyme for bile acid synthesis. In contrast, the expression levels of the ileum farnesoid X receptor (Fxr) and fibroblast growth factor 15 (Fgf15), which inhibit the expression of liver CYP7A1, were significantly reduced in the SCO-PL group than the SOY-TG group. From these results, the increase in the liver CYP7A1 expression by dietary SCO-PL was in part through the reduction of the ileum Fxr/Fgf15 regulatory pathway. Therefore, this study showed that SCO-PL may be a health-promoting component as it lowers the serum and liver cholesterol contents by increasing the liver CYP7A1 expression, which is not seen in SOY-PL and SCO-TG.
Collapse
Affiliation(s)
- Koki Sugimoto
- Laboratory of Food and Nutritional Sciences, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Japan
| | - Ryota Hosomi
- Laboratory of Food and Nutritional Sciences, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Japan
| | - Munehiro Yoshida
- Laboratory of Food and Nutritional Sciences, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Japan
| | - Kenji Fukunaga
- Laboratory of Food and Nutritional Sciences, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Japan
| |
Collapse
|
52
|
Wilson ML, Davies IG, Waraksa W, Khayyatzadeh SS, Al-Asmakh M, Mazidi M. The Impact of Microbial Composition on Postprandial Glycaemia and Lipidaemia: A Systematic Review of Current Evidence. Nutrients 2021; 13:nu13113887. [PMID: 34836140 PMCID: PMC8625294 DOI: 10.3390/nu13113887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 11/30/2022] Open
Abstract
Postprandial hyperglycaemia is associated with increased risk of cardiovascular disease. Recent studies highlight the role of the gut microbiome in influencing postprandial glycaemic (PPG) and lipidaemic (PPL) responses. The authors of this review sought to address the question: “To what extent does individual gut microbiome diversity and composition contribute to PPG and PPL responses?”. CINAHL Plus, PubMed, Web of Science, and the Cochrane Central Register of Controlled Trials (CENTRAL) databases were searched from January 2010 to June 2020. Following screening, 22 studies were eligible to be included in the current review. All trials reported analysis of gut microbiome diversity and composition and PPG and/or PPL. Results were reported according to the ‘Preferred Reporting Items for Systematic Reviews and Meta-Analysis’ (PRISMA) statement. Individual microbiota structure was found to play a key role in determining postprandial metabolic responses in adults and is attributed to a complex interplay of diet, microbiota composition, and metagenomic activity, which may be predicted by metagenomic analysis. Alterations of gut microbiota, namely relative abundance of bacterial phylum Actinobacteria and Proteobacteria, along with Enterobacteriaceae, were associated with individual variation in postprandial glycaemic response in adults. The findings of the current review present new evidence to support a personalised approach to nutritional recommendations and guidance for optimal health, management, and treatment of common metabolic disorders. In conclusion, personalised nutrition approaches based on individual microbial composition may improve postprandial regulation of glucose and lipids, providing a potential strategy to ameliorate cardiometabolic health outcomes.
Collapse
Affiliation(s)
- Megan L. Wilson
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool L3 3AF, UK; (M.L.W.); (I.G.D.); (W.W.)
| | - Ian G. Davies
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool L3 3AF, UK; (M.L.W.); (I.G.D.); (W.W.)
| | - Weronika Waraksa
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool L3 3AF, UK; (M.L.W.); (I.G.D.); (W.W.)
| | - Sayyed S. Khayyatzadeh
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd 8916188635, Iran;
- Department of Nutrition, Faculty of Health, Shahid Sadoughi University of Medical Sciences, Yazd 8915173143, Iran
| | - Maha Al-Asmakh
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Mohsen Mazidi
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford OX3 7LF, UK
- Clinical Trial Service Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- Department of Twin Research and Genetic Epidemiology, King’s College London, South Wing St Thomas’, London SE1 7EH, UK
- Correspondence:
| |
Collapse
|
53
|
Jiang C, Cheong LZ, Zhang X, Ali AH, Jin Q, Wei W, Wang X. Dietary Sphingomyelin Metabolism and Roles in Gut Health and Cognitive Development. Adv Nutr 2021; 13:S2161-8313(22)00073-4. [PMID: 34549256 PMCID: PMC8970835 DOI: 10.1093/advances/nmab117] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sphingomyelin (SM) is a widely occurring sphingolipid that is a major plasma membrane constituent. Milk and dairy products are rich SM sources, and human milk has high SM content. Numerous studies have evaluated the roles of SM in maintaining cell membrane structure and cellular signal transduction. There has been a growing interest in exploring the role of dietary SM, especially from human milk, in imparting health benefits. This review focuses on recent publications regarding SM content in several dietary sources and dietary SM metabolism. SM digestion and absorption are slow and incomplete and mainly occur in the middle sections of the small intestine. This review also evaluates the effect of dietary SM on gut health and cognitive development. Studies indicate that SM may promote gut health by reducing intestinal cholesterol absorption in adults. However, there has been a lack of data supporting clinical trials. An association between milk SM and neural development is evident before childhood. Hence, additional studies and well-designed randomized controlled trials that incorporate dietary SM evaluation, SM metabolism, and its long-term functions on infants and children are required.
Collapse
Affiliation(s)
- Chenyu Jiang
- State Key Lab of Food Science and Technology, Jiangnan University, Wuxi, China,Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ling-Zhi Cheong
- Department of Food Science and Engineering, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
| | - Xue Zhang
- State Key Lab of Food Science and Technology, Jiangnan University, Wuxi, China,Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Abdelmoneim H Ali
- State Key Lab of Food Science and Technology, Jiangnan University, Wuxi, China,Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qingzhe Jin
- State Key Lab of Food Science and Technology, Jiangnan University, Wuxi, China,Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Wei
- Address correspondence to WW (e-mail: )
| | - Xingguo Wang
- State Key Lab of Food Science and Technology, Jiangnan University, Wuxi, China,Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
54
|
Chen Z, Ahmed M, Ha V, Jefferson K, Malik V, Ribeiro PAB, Zuchinali P, Drouin-Chartier JP. Dairy Product Consumption and Cardiovascular Health: a Systematic Review and Meta-Analysis of Prospective Cohort Studies. Adv Nutr 2021; 13:S2161-8313(22)00071-0. [PMID: 34550320 PMCID: PMC8970833 DOI: 10.1093/advances/nmab118] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The association between dairy product consumption and cardiovascular health remains highly debated. We quantitatively synthesized prospective cohort evidence on the associations between dairy consumption and risk of hypertension (HTN), coronary heart disease (CHD) and stroke. We systematically searched PubMed, Embase, and Web of Science through August 1st, 2020 to retrieve prospective cohort studies that reported on dairy consumption and risk of HTN, CHD or stroke. We used random-effects models to calculate the pooled relative risk (RR) and 95% confidence interval (CI) for the highest vs the lowest category of intake and for 1 serving/day increase in consumption. We rated the quality of evidence using NutriGrade. Fifty-five studies were included. Total dairy consumption was associated with a lower risk of HTN (RR for highest vs lowest level of intake: 0.91, 95% CI: 0.86-0.95, I2 = 73.5%; RR for 1 serving/day increase: 0.96, 95% CI: 0.94-0.97, I2 = 66.5%), CHD (highest vs lowest level of intake: 0.96, 95% CI: 0.92-1.00, I2 = 46.6%; 1 serving/day increase: 0.98, 95% CI: 0.95-1.00, I2 = 56.7%), and stroke (highest vs lowest level of intake: 0.90, 95% CI: 0.85-0.96, I2 = 60.8%; 1 serving/day increase: 0.96, 95% CI: 0.93-0.99, I2 = 74.7%). Despite moderate to considerable heterogeneity, these associations remained consistent across multiple subgroups. Evidence on the relationship between total dairy and risk of HTN and CHD were of moderate quality and of low quality for stroke. Low-fat dairy consumption was associated with lower risk of HTN and stroke, and high-fat dairy with a lower risk of stroke. Milk, cheese, or yogurt consumption showed inconsistent associations with the cardiovascular outcomes in high vs. low intake and dose-response meta-analyses. Total dairy consumption was associated with a modestly lower risk of hypertension, CHD and stroke. Moderate to considerable heterogeneity was observed in the estimates and the overall quality of the evidence was low to moderate.
Collapse
Affiliation(s)
- Zhangling Chen
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA,Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mavra Ahmed
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada,Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON, Canada
| | - Vanessa Ha
- School of Medicine, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | | | - Vasanti Malik
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA,Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Paula A B Ribeiro
- Montreal Behavioural Medicine Centre, CIUSSS du Nord-de-l’Île-de-Montréal, Montréal, QC, Canada,Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Priccila Zuchinali
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | | |
Collapse
|
55
|
Nilsson Å, Duan RD, Ohlsson L. Digestion and Absorption of Milk Phospholipids in Newborns and Adults. Front Nutr 2021; 8:724006. [PMID: 34490332 PMCID: PMC8417471 DOI: 10.3389/fnut.2021.724006] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/19/2021] [Indexed: 12/25/2022] Open
Abstract
Milk polar lipids provide choline, ethanolamine, and polyunsaturated fatty acids, which are needed for the growth and plasticity of the tissues in a suckling child. They may also inhibit cholesterol absorption by interacting with cholesterol during micelle formation. They may also have beneficial luminal, mucosal, and metabolic effects in both the neonate and the adult. The milk fat globule membrane contains large proportions of sphingomyelin (SM), phosphatidylcholine (PC), and phosphatidylethanolamine (PE), and some phosphatidylserine (PS), phosphatidylinositol (PI), and glycosphingolipids. Large-scale technical procedures are available for the enrichment of milk fat globule membrane (MFGM) in milk replacement formulations and food additives. Pancreatic phospholipase A2 (PLA2) and mucosal phospholipase B digest glycero-phospholipids in the adult. In the neonate, where these enzymes may be poorly expressed, pancreatic lipase-related protein 2 probably has a more important role. Mucosal alkaline SM-ase and ceramidase catalyze the digestion of SM in both the neonate and the adult. In the mucosa, the sphingosine is converted into sphingosine-1-phosphate, which is both an intermediate in the conversion to palmitic acid and a signaling molecule. This reaction sequence also generates ethanolamine. Here, we summarize the pathways by which digestion and absorption may be linked to the biological effects of milk polar lipids. In addition to the inhibition of cholesterol absorption and the generation of lipid signals in the gut, the utilization of absorbed choline and ethanolamine for mucosal and hepatic phospholipid synthesis and the acylation of absorbed lyso-PC with polyunsaturated fatty acids to chylomicron and mucosal phospholipids are important.
Collapse
Affiliation(s)
- Åke Nilsson
- Division of Medicine, Gastroenterology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Rui-Dong Duan
- Gastroenterology and Nutrition Laboratory, Division of Medicine, Department of Clinical Science, Lund University, Lund, Sweden
| | - Lena Ohlsson
- Division of Medicine, Experimental Vascular Medicine, Department of Clinical Science, Lund University, Lund, Sweden
| |
Collapse
|
56
|
Juste C, Gérard P. Cholesterol-to-Coprostanol Conversion by the Gut Microbiota: What We Know, Suspect, and Ignore. Microorganisms 2021; 9:1881. [PMID: 34576776 PMCID: PMC8468837 DOI: 10.3390/microorganisms9091881] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/24/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
Every day, up to 1 g of cholesterol, composed of the unabsorbed dietary cholesterol, the biliary cholesterol secretion, and cholesterol of cells sloughed from the intestinal epithelium, enters the colon. All cholesterol arriving in the large intestine can be metabolized by the colonic bacteria. Cholesterol is mainly converted into coprostanol, a non-absorbable sterol that is excreted in the feces. Interestingly, cholesterol-to-coprostanol conversion in human populations is variable, with a majority of high converters and a minority of low or inefficient converters. Two major pathways have been proposed, one involving the direct stereospecific reduction of the Δ5 double bond direct while the indirect pathway involves the intermediate formation of 4-cholelesten-3-one and coprostanone. Despite the fact that intestinal cholesterol conversion was discovered more than a century ago, only a few cholesterol-to-coprostanol-converting bacterial strains have been isolated and characterized. Moreover, the responsible genes were mainly unknown until recently. Interestingly, cholesterol-to-coprostanol conversion is highly regulated by the diet. Finally, this gut bacterial metabolism has been linked to health and disease, and recent evidence suggests it could contribute to lower blood cholesterol and cardiovascular risks.
Collapse
Affiliation(s)
| | - Philippe Gérard
- AgroParisTech, Micalis Institute, Université Paris-Saclay, INRAE, 78350 Jouy-en-Josas, France;
| |
Collapse
|
57
|
Zhang Y, Zhang T, Liang Y, Jiang L, Sui X. Dietary Bioactive Lipids: A Review on Absorption, Metabolism, and Health Properties. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:8929-8943. [PMID: 34161727 DOI: 10.1021/acs.jafc.1c01369] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Dietary lipids are an indispensable source of energy and nutrition in human life. Numerous studies have shown that dietary bioactive lipids have many health benefits, including prevention or treatment of chronic diseases. The different chemical compositions and structural characteristics of bioactive lipids not only affect their digestion, absorption, and metabolism but also affect their health properties. In this review, the major dietary bioactive lipids (fatty acids, carotenoids, phytosterols, phenolic lipids, fat-soluble vitamins, and sphingomyelins) in foods are systematically summarized, from the aspects of composition, digestion, absorption, metabolism, source, structural characteristics, and their health properties. In particular, the relationship between the compositional and structural changes of bioactive lipids and their absorption and metabolism is discussed as well as their effect on health properties. This review provides a comprehensive summary toward health properties of dietary bioactive lipids.
Collapse
Affiliation(s)
- Yan Zhang
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
- College of Horticulture and Landscape Architecture, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
- National-Local Joint Engineering Research Center for Development and Utilization of Small Fruits in Cold Regions, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Tianyi Zhang
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Yan Liang
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Lianzhou Jiang
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Xiaonan Sui
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| |
Collapse
|
58
|
Thøgersen R, Lindahl IEI, Khakimov B, Kjølbæk L, Juhl Jensen K, Astrup A, Hammershøj M, Raben A, Bertram HC. Progression of Postprandial Blood Plasma Phospholipids Following Acute Intake of Different Dairy Matrices: A Randomized Crossover Trial. Metabolites 2021; 11:454. [PMID: 34357348 PMCID: PMC8307057 DOI: 10.3390/metabo11070454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 11/17/2022] Open
Abstract
Studies have indicated that the dairy matrix can affect postprandial responses of dairy products, but little is known about the effect on postprandial plasma phospholipid levels. This study investigated postprandial plasma phospholipid levels following consumption of four different dairy products that are similar in micro and macro nutrients, but different in texture and structure: cheddar cheese (Cheese), homogenized cheddar cheese (Hom. Cheese), micellar casein isolate with cream (MCI Drink) or a gel made from the MCI Drink (MCI Gel). The study was an acute randomized, crossover trial in human volunteers with four test days. Blood samples were collected during an 8 h postprandial period and the content of 53 plasma phospholipids was analysed using liquid chromatography-mass spectrometry (LC-MS). No meal-time interactions were revealed; however, for nine of the 53 phospholipids, a meal effect was found. Thus, the results indicated a lower plasma level of specific lyso-phosphatidylethanolamines (LPEs) and lyso-phosphatidylcholines (LPCs) following consumption of the MCI Gel compared to the MCI Drink and Hom. Cheese, which might be attributed to an effect of viscosity. However, further studies are needed in order to reveal more details on the effect of the dairy matrix on postprandial phospholipids.
Collapse
Affiliation(s)
- Rebekka Thøgersen
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark; (I.E.I.L.); (M.H.); (H.C.B.)
| | - Ida Emilie I. Lindahl
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark; (I.E.I.L.); (M.H.); (H.C.B.)
| | - Bekzod Khakimov
- Department of Food Science, University of Copenhagen, DK-1958 Frederiksberg C, Denmark;
| | - Louise Kjølbæk
- Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-1958 Frederiksberg C, Denmark; (L.K.); (A.A.); (A.R.)
| | | | - Arne Astrup
- Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-1958 Frederiksberg C, Denmark; (L.K.); (A.A.); (A.R.)
| | - Marianne Hammershøj
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark; (I.E.I.L.); (M.H.); (H.C.B.)
| | - Anne Raben
- Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-1958 Frederiksberg C, Denmark; (L.K.); (A.A.); (A.R.)
- Steno Diabetes Center Copenhagen, DK-2820 Gentofte, Denmark
| | - Hanne Christine Bertram
- Department of Food Science, Aarhus University, Agro Food Park 48, DK-8200 Aarhus N, Denmark; (I.E.I.L.); (M.H.); (H.C.B.)
| |
Collapse
|
59
|
Schmidt KA, Cromer G, Burhans MS, Kuzma JN, Hagman DK, Fernando I, Murray M, Utzschneider KM, Holte S, Kraft J, Kratz M. Impact of low-fat and full-fat dairy foods on fasting lipid profile and blood pressure: exploratory endpoints of a randomized controlled trial. Am J Clin Nutr 2021; 114:882-892. [PMID: 34258627 PMCID: PMC8408839 DOI: 10.1093/ajcn/nqab131] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/01/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Dietary guidelines traditionally recommend low-fat dairy because dairy's high saturated fat content is thought to promote cardiovascular disease (CVD). However, emerging evidence indicates that dairy fat may not negatively impact CVD risk factors when consumed in foods with a complex matrix. OBJECTIVE The aim was to compare the effects of diets limited in dairy or rich in either low-fat or full-fat dairy on CVD risk factors. METHODS In this randomized controlled trial, 72 participants with metabolic syndrome completed a 4-wk run-in period, limiting their dairy intake to ≤3 servings/wk of nonfat milk. Participants were then randomly assigned to 1 of 3 diets, either continuing the limited-dairy diet or switching to a diet containing 3.3 servings/d of either low-fat or full-fat milk, yogurt, and cheese for 12 wk. Exploratory outcome measures included changes in the fasting lipid profile and blood pressure. RESULTS In the per-protocol analysis (n = 66), there was no intervention effect on fasting serum total, LDL, and HDL cholesterol; triglycerides; free fatty acids; or cholesterol content in 38 isolated plasma lipoprotein fractions (P > 0.1 for all variables in repeated-measures ANOVA). There was also no intervention effect on diastolic blood pressure, but a significant intervention effect for systolic blood pressure (P = 0.048), with a trend for a decrease in the low-fat dairy diet (-1.6 ± 8.6 mm Hg) compared with the limited-dairy diet (+2.5 ± 8.2 mm Hg) in post hoc testing. Intent-to-treat results were consistent for all endpoints, with the exception that systolic blood pressure became nonsignificant (P = 0.08). CONCLUSIONS In men and women with metabolic syndrome, a diet rich in full-fat dairy had no effects on fasting lipid profile or blood pressure compared with diets limited in dairy or rich in low-fat dairy. Therefore, dairy fat, when consumed as part of complex whole foods, does not adversely impact these classic CVD risk factors. This trial was registered at clinicaltrials.gov as NCT02663544.
Collapse
Affiliation(s)
- Kelsey A Schmidt
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Nutritional Sciences Program, School of Public Health, University of Washington, Seattle, WA, USA
| | - Gail Cromer
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Maggie S Burhans
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jessica N Kuzma
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Derek K Hagman
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Imashi Fernando
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Nutritional Sciences Program, School of Public Health, University of Washington, Seattle, WA, USA
| | - Merideth Murray
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Nutritional Sciences Program, School of Public Health, University of Washington, Seattle, WA, USA
| | - Kristina M Utzschneider
- VA Puget Sound Health Care System, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Sarah Holte
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jana Kraft
- College of Agriculture and Life Sciences, University of Vermont, Burlington, VT, USA
| | | |
Collapse
|
60
|
Molecular mechanism associated with the use of magnetic fermentation in modulating the dietary lipid composition and nutritional quality of goat milk. Food Chem 2021; 366:130554. [PMID: 34284188 DOI: 10.1016/j.foodchem.2021.130554] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/13/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Standard fermentation (SF) mainly affected the metabolism of glycerophospholipid and sphingolipid, and increased the total lipid content of goat milk. Content of total lipid was decreased by magnetic fermentation compared with SF, mainly due to triacylglycerol and diacylglycerol. Comprehensive characteristic of lipids dynamic changes during standard and magnetic fermentation was performed using high-throughput quantitative lipidomics. Totally, 488 lipid molecular species covering 12 subclasses were detected, and triacylglycerol was the highest levels, followed by diacylglycerol and phosphoethanolamine in the whole fermentation stage. Specifically, except for ceramide and simple Glc series, the content of all polar lipids in SF was dropped and neutral lipids subjoined. Compared with SF, the decrease of triacylglycerol (1752.47 to 784.78 μg/mL), diacylglycerol (60.36 to 24.89 μg/mL) and simple Glc series (4.36 to 2.40 μg/mL) were observed, while ceramide (6.54 to 25.87 μg/mL) increased, suggesting magnetic fermentation as effective approach to potentially improve the nutritional of goat milk.
Collapse
|
61
|
Ferraris Q, Qian MC. Direct ethanolic extraction of polar lipids and fractional crystallization from whey protein phospholipid concentrate. JDS COMMUNICATIONS 2021; 2:177-181. [PMID: 36338445 PMCID: PMC9623725 DOI: 10.3168/jdsc.2021-0076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/13/2021] [Indexed: 06/13/2023]
Abstract
Polar lipids, including sphingomyelin (SM) and other sphingolipids, have nutritional implications in many important metabolic pathways. Milk and whey are valuable sources of these bioactive lipids, differing in SM content compared with other plant-based lecithins in the food industry. Dairy lipids were extracted from whey protein phospholipid concentrate powder by direct exposure to a food-grade solvent, ethanol. A total of 20 mass equivalents of solvent was required to completely recover all lipid material of the original whey protein phospholipid concentrate sample. Using a minimal apparatus, absolute ethanol, and heating to a boiling point, a final total lipid extract composed of 37.7% phospholipids (PL) by mass was achieved. The method was developed with potential pilot-scale and industry application in mind. Investigations into fractional crystallization showed limited success at purifying PL from triglyceride content (<1% increase in percent PL by mass). However, the relative percentage of SM was increased in the fractionated sample compared with the total lipid extract. Fatty acid analysis of the PL fraction reported a composition of 33.3% monounsaturated fatty acids and 9.8% polyunsaturated fatty acids. Additional work is needed to investigate different conditions for lipid recovery, PL recovery, and PL purification by fractional crystallization in ethanol.
Collapse
|
62
|
Zhang Q, Ye L, Xin F, Zhou J, Cao B, Dong Y, Qian L. Milk Fat Globule Membrane Supplementation During Suckling Ameliorates Maternal High Fat Diet-Induced Hepatic Steatosis in Adult Male Offspring of Mice. J Nutr 2021; 151:1487-1496. [PMID: 33693864 DOI: 10.1093/jn/nxab026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/09/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Exposure to a maternal high-fat diet (HFD) predisposes offspring to nonalcoholic fatty liver disease. OBJECTIVES The aim of this study was to explore whether milk fat globule membrane (MFGM) supplementation during suckling exerts a long-term protective effect on hepatic lipid metabolism in adult offspring exposed to maternal HFD. METHODS We fed 5-week-old female C57BL/6J mice either a HFD (60% kcal fat) or control diet (CD; 16.7% kcal fat) for 3 weeks before mating, as well as throughout gestation and lactation. After delivery, male offspring from HFD dams were supplemented with 1 g/(kg body weight·day) MFGM (HFD + MFGM group) or the same volume of vehicle (HFD group) during suckling. Male offspring from CD dams were also supplemented with vehicle during suckling (CD group). All offspring were weaned onto CD for 8 weeks. Histopathology, metabolic parameters, lipogenic level, oxidative stress, and mitochondria function in the liver were analyzed. A 1-way ANOVA and a Kruskal-Wallis test were used for multi-group comparisons. RESULTS As compared to the CD group, the HFD group had more lipid droplets in livers, and exhibited ∼100% higher serum triglycerides, ∼38% higher hepatic triglycerides, ∼75% higher serum aspartate aminotransferase, and ∼130% higher fasting blood glucose (P < 0.05). The changes of these metabolic parameters were normalized in the HFD + MFGM group. Phosphorylated mammalian targets of rapamycin and AKT were downregulated, but phosphorylated adenosine monophosphate-activated protein kinase was upregulated in the HFD + MFGM group as compared to the HFD group (P < 0.05). As compared to the CD group, the HFD group showed an ∼80% higher malondialdehyde level, and ∼20% lower superoxide dismutase activity (P < 0.05), which were normalized in the HFD + MFGM group. Additionally, mitochondria function was also impaired in the HFD group and normalized in the HFD + MFGM group. CONCLUSIONS MFGM supplementation during suckling ameliorates maternal HFD-induced hepatic steatosis in mice via suppressing de novo lipogenesis, reinforcing antioxidant defenses and improving mitochondrial function.
Collapse
Affiliation(s)
- Qianren Zhang
- Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Lin Ye
- Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Fengzhi Xin
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jiefei Zhou
- Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Baige Cao
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yan Dong
- Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Linxi Qian
- Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| |
Collapse
|
63
|
Le Barz M, Vors C, Combe E, Joumard-Cubizolles L, Lecomte M, Joffre F, Trauchessec M, Pesenti S, Loizon E, Breyton AE, Meugnier E, Bertrand K, Drai J, Robert C, Durand A, Cuerq C, Gaborit P, Leconte N, Bernalier-Donadille A, Cotte E, Laville M, Lambert-Porcheron S, Ouchchane L, Vidal H, Malpuech-Brugère C, Cheillan D, Michalski MC. Milk polar lipids favorably alter circulating and intestinal ceramide and sphingomyelin species in postmenopausal women. JCI Insight 2021; 6:146161. [PMID: 33857018 PMCID: PMC8262315 DOI: 10.1172/jci.insight.146161] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/09/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND High circulating levels of ceramides (Cer) and sphingomyelins (SM) are associated with cardiometabolic diseases. The consumption of whole fat dairy products, naturally containing such polar lipids (PL), is associated with health benefits, but the impact on sphingolipidome remains unknown. METHODS In a 4-week randomized controlled trial, 58 postmenopausal women daily consumed milk PL-enriched cream cheese (0, 3, or 5 g of milk PL). Postprandial metabolic explorations were performed before and after supplementation. Analyses included SM and Cer species in serum, chylomicrons, and feces. The ileal contents of 4 ileostomy patients were also explored after acute milk PL intake. RESULTS Milk PL decreased serum atherogenic C24:1 Cer, C16:1 SM, and C18:1 SM species (Pgroup < 0.05). Changes in serum C16+18 SM species were positively correlated with the reduction of cholesterol (r = 0.706), LDL-C (r = 0.666), and ApoB (r = 0.705) (P < 0.001). Milk PL decreased chylomicron content in total SM and C24:1 Cer (Pgroup < 0.001), parallel to a marked increase in total Cer in feces (Pgroup < 0.001). Milk PL modulated some specific SM and Cer species in both ileal efflux and feces, suggesting differential absorption and metabolization processes in the gut. CONCLUSION Milk PL supplementation decreased atherogenic SM and Cer species associated with the improvement of cardiovascular risk markers. Our findings bring insights on sphingolipid metabolism in the gut, especially Cer, as signaling molecules potentially participating in the beneficial effects of milk PL. TRIAL REGISTRATION ClinicalTrials.gov, NCT02099032, NCT02146339. FUNDING ANR-11-ALID-007-01; PHRCI-2014: VALOBAB, no. 14-007; CNIEL; GLN 2018-11-07; HCL (sponsor).
Collapse
Affiliation(s)
- Mélanie Le Barz
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France
| | - Cécile Vors
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France.,TCentre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310, Pierre-Bénite, France
| | - Emmanuel Combe
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France
| | - Laurie Joumard-Cubizolles
- Université Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH Auvergne, 63000, Clermont-Ferrand, France
| | - Manon Lecomte
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France.,TCentre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310, Pierre-Bénite, France
| | - Florent Joffre
- ITERG, ZA Pessac-Canéjan, 11 Rue Gaspard Monge, 33610, Canéjan, France
| | - Michèle Trauchessec
- Hospices Civils de Lyon, 69000, Lyon, France.,Unité Maladies Héréditaires du Métabolisme, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 69677, Bron, France
| | - Sandra Pesenti
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France
| | - Emmanuelle Loizon
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France
| | - Anne-Esther Breyton
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France.,TCentre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310, Pierre-Bénite, France
| | - Emmanuelle Meugnier
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France
| | - Karène Bertrand
- ITERG, ZA Pessac-Canéjan, 11 Rue Gaspard Monge, 33610, Canéjan, France
| | - Jocelyne Drai
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France.,Hospices Civils de Lyon, 69000, Lyon, France.,Unité de Nutrition Endocrinologie Métabolisme, Service de Biochimie, Centre de Biologie et de Pathologie Sud, Hospices Civils de Lyon, 69495, Pierre-Bénite, France
| | - Chloé Robert
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France.,TCentre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310, Pierre-Bénite, France
| | - Annie Durand
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France
| | - Charlotte Cuerq
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France.,Hospices Civils de Lyon, 69000, Lyon, France.,Unité de Nutrition Endocrinologie Métabolisme, Service de Biochimie, Centre de Biologie et de Pathologie Sud, Hospices Civils de Lyon, 69495, Pierre-Bénite, France
| | - Patrice Gaborit
- ACTALIA Dairy Products and Technologies, Avenue François Mitterrand, BP49, 17700, Surgères, France.,ENILIA ENSMIC, Avenue François Mitterrand, 17700, Surgères, France
| | - Nadine Leconte
- INRAE, Institut Agro, STLO (Science et Technologie du Lait et de l'Œuf), 35042, Rennes, France
| | | | - Eddy Cotte
- Hospices Civils de Lyon, 69000, Lyon, France.,Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Service de chirurgie digestive, 69310, Pierre-Bénite, France.,Université Claude Bernard Lyon 1, Faculté de médecine Lyon-Sud-Charles Mérieux, EMR 3738, 69600, Oullins, France
| | - Martine Laville
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France.,TCentre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310, Pierre-Bénite, France.,Hospices Civils de Lyon, 69000, Lyon, France.,Université Claude Bernard Lyon 1, Faculté de médecine Lyon-Sud-Charles Mérieux, EMR 3738, 69600, Oullins, France
| | - Stéphanie Lambert-Porcheron
- TCentre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310, Pierre-Bénite, France.,Hospices Civils de Lyon, 69000, Lyon, France
| | - Lemlih Ouchchane
- Université Clermont Auvergne, CNRS, SIGMA Clermont, Institut Pascal, 63000, Clermont-Ferrand, France.,CHU Clermont-Ferrand, Unité de Biostatistique-Informatique Médicale, 63000, Clermont-Ferrand, France
| | - Hubert Vidal
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France
| | - Corinne Malpuech-Brugère
- Université Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH Auvergne, 63000, Clermont-Ferrand, France
| | - David Cheillan
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France.,Hospices Civils de Lyon, 69000, Lyon, France.,Unité Maladies Héréditaires du Métabolisme, Service de Biochimie et Biologie Moléculaire Grand Est, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, 69677, Bron, France
| | - Marie-Caroline Michalski
- Univ Lyon, CarMeN laboratory, INSERM, INRAE, INSA Lyon, Université Claude Bernard Lyon 1, Charles Mérieux Medical School, 69310, Pierre-Bénite, France.,TCentre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310, Pierre-Bénite, France
| |
Collapse
|
64
|
Rohrhofer J, Zwirzitz B, Selberherr E, Untersmayr E. The Impact of Dietary Sphingolipids on Intestinal Microbiota and Gastrointestinal Immune Homeostasis. Front Immunol 2021; 12:635704. [PMID: 34054805 PMCID: PMC8160510 DOI: 10.3389/fimmu.2021.635704] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
The large surfaces of gastrointestinal (GI) organs are well adapted to their diverse tasks of selective nutritional uptake and defense against the external environment. To maintain a functional balance, a vast number of immune cells is located within the mucosa. A strictly regulated immune response is required to impede constant inflammation and to maintain barrier function. An increasing prevalence of GI diseases has been reported in Western societies over the past decades. This surge in GI disorders has been linked to dietary changes followed by an imbalance of the gut microbiome, leading to a chronic, low grade inflammation of the gut epithelium. To counteract the increasing health care costs associated with diseases, it is paramount to understand the mechanisms driving immuno-nutrition, the associations between nutritional compounds, the commensal gut microbiota, and the host immune response. Dietary compounds such as lipids, play a central role in GI barrier function. Bioactive sphingolipids (SLs), e.g. sphingomyelin (SM), sphingosine (Sph), ceramide (Cer), sphingosine-1- phosphate (S1P) and ceramide-1-phosphate (C1P) may derive from dietary SLs ingested through the diet. They are not only integral components of cell membranes, they additionally modulate cell trafficking and are precursors for mediators and second messenger molecules. By regulating intracellular calcium levels, cell motility, cell proliferation and apoptosis, SL metabolites have been described to influence GI immune homeostasis positively and detrimentally. Furthermore, dietary SLs are suggested to induce a shift in the gut microbiota. Modes of action range from competing with the commensal bacteria for intestinal cell attachment to prevention from pathogen invasion by regulating innate and immediate defense mechanisms. SL metabolites can also be produced by gut microorganisms, directly impacting host metabolic pathways. This review aims to summarize recent findings on SL signaling and functional variations of dietary SLs. We highlight novel insights in SL homeostasis and SL impact on GI barrier function, which is directly linked to changes of the intestinal microbiota. Knowledge gaps in current literature will be discussed to address questions relevant for understanding the pivotal role of dietary SLs on chronic, low grade inflammation and to define a balanced and healthy diet for disease prevention and treatment.
Collapse
Affiliation(s)
- Johanna Rohrhofer
- Gastrointestinal Immunology Group, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Benjamin Zwirzitz
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Evelyne Selberherr
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Eva Untersmayr
- Gastrointestinal Immunology Group, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
65
|
Raza GS, Herzig KH, Leppäluoto J. Invited review: Milk fat globule membrane-A possible panacea for neurodevelopment, infections, cardiometabolic diseases, and frailty. J Dairy Sci 2021; 104:7345-7363. [PMID: 33896625 DOI: 10.3168/jds.2020-19649] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/15/2021] [Indexed: 12/23/2022]
Abstract
Milk is an evolutionary benefit for humans. For infants, it offers optimal nutrients for normal growth, neural development, and protection from harmful microbes. Humans are the only mammals who drink milk throughout their life. Lipids in colostrum originate mostly from milk fat globule membrane (MFGM) droplets extruded from the mammary gland. The MFGM gained much interest as a potential nutraceutical, due to their high phospholipid (PL), ganglioside (GD), and protein contents. In this review, we focused on health effects of MFGM ingredients and dairy food across the life span, especially on neurodevelopment, cardiometabolic health, and frailty in older adults. The MFGM supplements to infants and children reduced gastrointestinal and respiratory tract infections and improved neurodevelopment due to the higher content of protein, PL, and GD in MFGM. The MFGM formulas containing PL and GD improved brain myelination and fastened nerve conduction speed, resulting in improved behavioral developments. Administration of MFGM-rich ingredients improved insulin sensitivity and decreased inflammatory markers, LDL-cholesterol, and triglycerides by lowering intestinal absorption of cholesterol and increasing its fecal excretion. The MFGM supplements, together with exercise, improved ambulatory activities, leg muscle mass, and muscle fiber velocity in older adults. There are great variations in the composition of lipids and proteins in MFGM products, which make comparisons of the different studies impossible. In addition, investigations of the individual MFGM components are required to evaluate their specific effects and molecular mechanisms. Although we are currently only beginning to understand the possible health effects of MFGM products, the current MFGM supplementation trials as presented in this review have shown significant clinical health benefits across the human life span, which are worth further investigation.
Collapse
Affiliation(s)
- Ghulam Shere Raza
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland; Oulu University Hospital, 90220 Oulu, Finland; Pediatric Institute, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Juhani Leppäluoto
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland.
| |
Collapse
|
66
|
Feeney EL, Lamichhane P, Sheehan JJ. The cheese matrix: Understanding the impact of cheese structure on aspects of cardiovascular health – A food science and a human nutrition perspective. INT J DAIRY TECHNOL 2021. [DOI: 10.1111/1471-0307.12755] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Emma L Feeney
- Institute of Food and Health University College Dublin 2.16a Science Centre South Dublin 4Ireland
- Food for Health Ireland (FHI) S2.09 Science Centre South Belfield, Dublin 4Ireland
| | - Prabin Lamichhane
- Teagasc Food Research Centre Moorepark Fermoy, Cork P61 C996 Ireland
| | - Jeremiah J Sheehan
- Food for Health Ireland (FHI) S2.09 Science Centre South Belfield, Dublin 4Ireland
- Teagasc Food Research Centre Moorepark Fermoy, Cork P61 C996 Ireland
| |
Collapse
|
67
|
Pu Z, Yang F, Wang L, Diao Y, Chen D. Advancements of compounds targeting Wnt and Notch signalling pathways in the treatment of inflammatory bowel disease and colon cancer. J Drug Target 2020; 29:507-519. [PMID: 33307848 DOI: 10.1080/1061186x.2020.1864741] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Wnt and Notch signalling pathways are important for maintenance of intestinal epithelial barrier integrity by intestinal stem cells (ISCs). Dysfunction of these pathways is implicated in inflammatory bowel disease (IBD) and colon cancer. The objective of this review is to summarise advancements of drugs that regulate Wnt and Notch in the treatment of IBD and colon cancer. The compositions and biological effects of Wnt and Notch modulators in both ISCs and non-ISCs are discussed. The drugs, including phytochemicals, plant extracts, probiotics and synthetic compounds, have been found to regulate Wnt and Notch signalling pathways by targeting regulatory factors (including secreted frizzled-related proteins or pathway proteins such as β-catenin and γ-secretase) to alleviate IBD and colon cancer. This review highlights the potential for targeting Wnt and Notch pathways to treat IBD and colon cancer.
Collapse
Affiliation(s)
- Zhuonan Pu
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Fang Yang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Liang Wang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Yunpeng Diao
- Colleage of Pharmacy, Dalian Medical University, Dalian, Liaoning, China
| | - Dapeng Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
68
|
Poppitt SD. Cow's Milk and Dairy Consumption: Is There Now Consensus for Cardiometabolic Health? Front Nutr 2020; 7:574725. [PMID: 33364249 PMCID: PMC7753100 DOI: 10.3389/fnut.2020.574725] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/02/2020] [Indexed: 12/18/2022] Open
Abstract
Cow's milk and dairy products derived from this complex food source have long been proposed as beneficial to human health, yet underlying clinical evidence of direct benefit continues to raise controversy. Limited evidence supports positive cardiometabolic effects of a number of dairy macro- and micronutrient components including whey protein and casein, unsaturated fats, milk fat globule membrane (MFGM) and polar phospholipids, vitamin D and calcium, in addition to non-bovine components including bacterial and yeast probiotics. More controversial remain lipid components trans fats, including trans vaccenic acid, trans palmitoleic acid, and conjugated cis trans linoleic acid (CLA), plus medium-chain and odd-chain dairy fats. New evidence is rapidly identifying multiple pathways by which these dairy nutrients may effect health. Processing, including fermentation and homogenization, may also have positive effects. Conversely, the high saturated fat content of dairy has long raised concern, aligned with international guidelines to minimize dietary intake of animal-origin saturated fatty acids (SFA) to achieve better cardiometabolic health. However, led in part by observational studies and meta-analyses showing dairy to have no or even an inverse association with cardiometabolic health, evidence from randomized controlled trials (RCTs) has been scrutinized over the last 5 years, and focus on low-fat dairy has been challenged. Recent evidence supports the hypothesis that adverse effects of SFAs on metabolic health may be ameliorated when these fats are consumed within a complex matrix such as milk, cheese or yogurt, and that dairy food categories may influence outcomes as much as total fat content. For example, yogurt and high-fat, high-SFA cheese have a negative association with risk of type 2 diabetes (T2D) in many, not all, published trials. However, large sample dairy RCTs of long duration with CVD or T2D incidence as primary endpoints are lacking. This is a clear research gap, with these clinical studies required if a causative link between dairy and improved cardiometabolic health is to be confirmed and in turn promoted through dietary guidelines. Current advisories from national guidance groups such as American Heart Association (AHA) and European Society of Cardiology (ESC) continue to promote consumption of low-fat dairy products, whilst liquid milk and yogurt remain part of nutrition guidelines from joint American Diabetes Association (ADA)/European Association for Study of Diabetes (EASD) reports, and as part of a "no-one-size-fits-all" answer to diet and T2D by the ADA in their most recent 2019 Consensus Report.
Collapse
Affiliation(s)
- Sally D. Poppitt
- Human Nutrition Unit, Department of Medicine, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
69
|
Vors C, Le Barz M, Bourlieu C, Michalski MC. Dietary lipids and cardiometabolic health: a new vision of structure-activity relationship. Curr Opin Clin Nutr Metab Care 2020; 23:451-459. [PMID: 32889824 DOI: 10.1097/mco.0000000000000693] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The impact of dietary lipids on cardiometabolic health was mainly studied considering their fatty acid composition. This review aims to present the recent change in paradigm whereby the food matrix, the molecular and supramolecular structures of dietary lipids modulate their digestive fate and cardiometabolic impact. RECENT FINDINGS Epidemiological studies have reported that the metabolic impact of full-fat dairy products is better than predictable upon saturated fatty acid richness. Milk polar lipid supplementation reduced adiposity and inflammation in rodents by modulating gut microbiota and barrier, and decreased lipid markers of cardiovascular disease risk in humans by lowering cholesterol absorption. The metabolic importance of the structure of lipid molecules carrying omega-3 (molecular carrier) has also been documented. Plant lipids exhibit specific assemblies, membrane and molecular structures with potential health benefits. Lipid emulsifiers used to stabilize fats in processed foods are not mere bystanders of lipid effects and can induce both beneficial and adverse health effects. SUMMARY These findings open new clinical research questions aiming to further characterize the cardiometabolic fate of lipids, from digestion to bioactive metabolites, according to the food source or molecular carrier. This should be useful to elaborate food formulations for target populations and personalized dietary recommendations.
Collapse
Affiliation(s)
- Cécile Vors
- Université de Lyon, CarMeN laboratory, INRAE, INSERM, Université Claude Bernard Lyon 1, INSA-Lyon
- CRNH Rhône-Alpes, CENS, Pierre-Bénite
| | - Mélanie Le Barz
- Université de Lyon, CarMeN laboratory, INRAE, INSERM, Université Claude Bernard Lyon 1, INSA-Lyon
| | - Claire Bourlieu
- UMR IATE 1208, INRAE/CIRAD/UM/Institut Agro, Montpellier, France
| | - Marie-Caroline Michalski
- Université de Lyon, CarMeN laboratory, INRAE, INSERM, Université Claude Bernard Lyon 1, INSA-Lyon
- CRNH Rhône-Alpes, CENS, Pierre-Bénite
| |
Collapse
|
70
|
Abd El‐Salam MH, El‐Shibiny S. Milk fat globule membrane: An overview with particular emphasis on its nutritional and health benefits. INT J DAIRY TECHNOL 2020. [DOI: 10.1111/1471-0307.12730] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | - Safinaz El‐Shibiny
- Dairy Department National Research Centre El‐Behous St Dokki Cairo Egypt
| |
Collapse
|
71
|
Quarles WR, Pokala A, Shaw EL, Ortega-Anaya J, Hillmann L, Jimenez-Flores R, Bruno RS. Alleviation of Metabolic Endotoxemia by Milk Fat Globule Membrane: Rationale, Design, and Methods of a Double-Blind, Randomized, Controlled, Crossover Dietary Intervention in Adults with Metabolic Syndrome. Curr Dev Nutr 2020; 4:nzaa130. [PMID: 32885133 PMCID: PMC7456308 DOI: 10.1093/cdn/nzaa130] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Milk fat globule membrane (MFGM) is a phospholipid-rich component of dairy fat that might explain the benefits of full-fat dairy products on cardiometabolic risk. Preclinical studies support that MFGM decreases gut permeability, which could attenuate gut-derived endotoxin translocation and consequent inflammatory responses that impair cardiometabolic health. OBJECTIVES To describe the rationale, study design, and planned outcomes that will evaluate the efficacy of MFGM-enriched milk compared with a comparator beverage on health-promoting gut barrier functions in persons with metabolic syndrome (MetS). METHODS We plan a double-blind, randomized, crossover trial in which people with MetS will receive a rigorously controlled eucaloric diet for 2 wk that contains 3 daily servings of an MFGM-enriched bovine milk beverage or a comparator beverage that is formulated with nonfat dairy powder, coconut and palm oils, and soy phospholipids. Compliance will be monitored by assessing urinary para-aminobenzoic acid that is added to all test beverages. After the intervention, participants will ingest a high-fat/high-carbohydrate meal challenge to assess metabolic excursions at 30-min intervals for 3 h. Nondigestible sugar probes also will be ingested prior to collecting 24-h urine to assess region-specific gut permeability. Intervention efficacy will be determined based on circulating endotoxin (primary outcome) and glycemia (secondary outcome). Tertiary outcomes include: gut and systemic inflammatory responses, microbiota composition and SCFAs, gut permeability, and circulating insulin and incretins. EXPECTED RESULTS MFGM is expected to decrease circulating endotoxin and glycemia without altering body mass. These improvements are anticipated to be accompanied by decreased gut permeability, decreased intestinal and circulating biomarkers of inflammation, increased circulating incretins, and beneficial antimicrobial and prebiotic effects in the gut microbiome. CONCLUSIONS Demonstration of improvements in gut barrier functions that limit endotoxemia and glycemia could help to establish direct evidence that full-fat dairy lowers cardiometabolic risk, especially in people with MetS.The clinical trial associated with this article has been registered at clinicaltrials.gov (NCT03860584).
Collapse
Affiliation(s)
- William R Quarles
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Avi Pokala
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Emily L Shaw
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Joana Ortega-Anaya
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA
| | - Lisa Hillmann
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Rafael Jimenez-Flores
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA
| | - Richard S Bruno
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
72
|
Trichia E, Luben R, Khaw KT, Wareham NJ, Imamura F, Forouhi NG. The associations of longitudinal changes in consumption of total and types of dairy products and markers of metabolic risk and adiposity: findings from the European Investigation into Cancer and Nutrition (EPIC)-Norfolk study, United Kingdom. Am J Clin Nutr 2020; 111:1018-1026. [PMID: 31915813 PMCID: PMC7198306 DOI: 10.1093/ajcn/nqz335] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/17/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The consumption of some types of dairy products has been associated with lower cardiometabolic disease incidence. Knowledge remains limited about habitual dairy consumption and the pathways to cardiometabolic risk. OBJECTIVE We aimed to investigate associations of habitual consumption of total and types of dairy products with markers of metabolic risk and adiposity among adults in the United Kingdom. METHODS We examined associations of changes in dairy consumption (assessed with a food-frequency questionnaire) with parallel changes in cardiometabolic markers using multiple linear regression among 15,612 adults aged 40-78 y at baseline (1993-1997) and followed up over 1998-2000 (mean ± SD: 3.7±0.7 y) in the European Prospective Investigation into Cancer and Nutrition (EPIC)-Norfolk study. RESULTS For adiposity, an increase in fermented dairy products [yogurt (total or low-fat) or low-fat cheese] consumption was associated with a lower increase in body weight and body mass index (BMI). For example, over 3.7 y, increasing yogurt consumption by 1 serving/d was associated with a smaller increase in body weight by 0.23 kg (95% CI: -0.46, -0.01 kg). An increase in full-fat milk, high-fat cheese, and total high-fat dairy was associated with greater increases in body weight and BMI [e.g., for high-fat dairy: β = 0.13 (0.05, 0.21) kg and 0.04 (0.01, 0.07) kg/m2, respectively]. For lipids, an increase in milk (total and low-fat) or yogurt consumption was positively associated with HDL cholesterol. An increase in total low-fat dairy was negatively associated with LDL cholesterol (-0.03 mmol/L; -0.05, -0.01 mmol/L), whereas high-fat dairy (total, butter, and high-fat cheese) consumption was positively associated [e.g., 0.04 (0.02, 0.06) mmol/L for total high-fat dairy]. For glycemia, increasing full-fat milk consumption was associated with a higher increase in glycated hemoglobin (P = 0.027). CONCLUSIONS The habitual consumption of different dairy subtypes may differently influence cardiometabolic risk through adiposity and lipid pathways.
Collapse
Affiliation(s)
- Eirini Trichia
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom,Address correspondence to ET (e-mail: )
| | - Robert Luben
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Nicholas J Wareham
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Fumiaki Imamura
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Nita G Forouhi
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom,Address correspondence to NGF (e-mail: )
| |
Collapse
|
73
|
Anto L, Warykas SW, Torres-Gonzalez M, Blesso CN. Milk Polar Lipids: Underappreciated Lipids with Emerging Health Benefits. Nutrients 2020; 12:E1001. [PMID: 32260440 PMCID: PMC7230917 DOI: 10.3390/nu12041001] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/24/2022] Open
Abstract
Milk fat is encased in a polar lipid-containing tri-layer milk fat globule membrane (MFGM), composed of phospholipids (PLs) and sphingolipids (SLs). Milk PLs and SLs comprise about 1% of total milk lipids. The surfactant properties of PLs are important for dairy products; however, dairy products vary considerably in their polar lipid to total lipid content due to the existence of dairy foods with different fat content. Recent basic science and clinical research examining food sources and health effects of milk polar lipids suggest they may beneficially influence dysfunctional lipid metabolism, gut dysbiosis, inflammation, cardiovascular disease, gut health, and neurodevelopment. However, more research is warranted in clinical studies to confirm these effects in humans. Overall, there are a number of potential effects of consuming milk polar lipids, and they should be considered as food matrix factors that may directly confer health benefits and/or impact effects of other dietary lipids, with implications for full-fat vs. reduced-fat dairy.
Collapse
Affiliation(s)
- Liya Anto
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA; (L.A.); (S.W.W.)
| | - Sarah Wen Warykas
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA; (L.A.); (S.W.W.)
| | | | - Christopher N. Blesso
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA; (L.A.); (S.W.W.)
| |
Collapse
|
74
|
Mozaffarian D. Dietary and policy priorities to reduce the global crises of obesity and diabetes. ACTA ACUST UNITED AC 2020. [DOI: 10.1038/s43016-019-0013-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
75
|
Magkos F, Tetens I, Bügel SG, Felby C, Schacht SR, Hill JO, Ravussin E, Astrup A. A Perspective on the Transition to Plant-Based Diets: a Diet Change May Attenuate Climate Change, but Can It Also Attenuate Obesity and Chronic Disease Risk? Adv Nutr 2020; 11:1-9. [PMID: 31504086 PMCID: PMC7442415 DOI: 10.1093/advances/nmz090] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/28/2019] [Accepted: 07/26/2019] [Indexed: 12/17/2022] Open
Abstract
Current dietary guidelines advocate more plant-based, sustainable diets on the basis of scientific evidence about diet-health relations but also to address environmental concerns. Here, we critically review the effects of plant-based diets on the prevalence of obesity and other health outcomes. Plant-based diets per se have limited efficacy for the prevention and treatment of obesity, but most have beneficial effects in terms of chronic disease risk. However, with the considerable possibilities of translating plant-based diets into various types of dietary patterns, our analysis suggests that potential adverse health effects should also be considered in relation to vulnerable groups of the population. A transition to more plant-based diets may exert beneficial effects on the environment, but is unlikely to affect obesity, and may also have adverse health effects if this change is made without careful consideration of the nutritional needs of the individual relative to the adequacy of the dietary intake.
Collapse
Affiliation(s)
- Faidon Magkos
- Department of Nutrition, Exercise, and Sports (NEXS), University of Copenhagen, Copenhagen, Denmark,Address correspondence to FM (E-mail: )
| | - Inge Tetens
- Department of Nutrition, Exercise, and Sports (NEXS), University of Copenhagen, Copenhagen, Denmark
| | - Susanne Gjedsted Bügel
- Department of Nutrition, Exercise, and Sports (NEXS), University of Copenhagen, Copenhagen, Denmark
| | - Claus Felby
- Section for Forest, Nature, and Biomass, Department of Geosciences and Natural Resource Management, University of Copenhagen, Copenhagen, Denmark
| | - Simon Rønnow Schacht
- Department of Nutrition, Exercise, and Sports (NEXS), University of Copenhagen, Copenhagen, Denmark
| | - James O Hill
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Arne Astrup
- Department of Nutrition, Exercise, and Sports (NEXS), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
76
|
Mozaffarian D. Dairy foods, dairy fat, diabetes, and death: what can be learned from 3 large new investigations? Am J Clin Nutr 2019; 110:1053-1054. [PMID: 31562511 PMCID: PMC6821539 DOI: 10.1093/ajcn/nqz250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| |
Collapse
|
77
|
Protective properties of milk sphingomyelin against dysfunctional lipid metabolism, gut dysbiosis, and inflammation. J Nutr Biochem 2019; 73:108224. [DOI: 10.1016/j.jnutbio.2019.108224] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/25/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022]
|