Milk polar lipids reduce lipid cardiovascular risk factors in overweight postmenopausal women: towards a gut sphingomyelin-cholesterol interplay

Cécile Vors, Laurie Joumard-Cubizolles, Manon Lecomte, Emmanuel Combe, Lemlih Ouchchane, Jocelyne Drai, Ketsia Raynal, Florent Joffre, Laure Meiller, Mélanie Le Barz, Patrice Gaborit, Aurélie Caille, Monique Sothier, Carla Domingues-Faria, Adeline Blot, Aurélie Wauquier, Emilie Blond, Valérie Sauvinet, Geneviève Gésan-Guiziou, Jean-Pierre Bodin, Philippe Moulin, David Cheillan, Hubert Vidal, Béatrice Morio, Eddy Cotte, Françoise Morel-Laporte, Martine Laville, Annick Bernalier-Donadille, Stéphanie Lambert-Porcheron, Corinne Malpuech-Brugère, Marie-Caroline Michalski, Cécile Vors, Laurie Joumard-Cubizolles, Manon Lecomte, Emmanuel Combe, Lemlih Ouchchane, Jocelyne Drai, Ketsia Raynal, Florent Joffre, Laure Meiller, Mélanie Le Barz, Patrice Gaborit, Aurélie Caille, Monique Sothier, Carla Domingues-Faria, Adeline Blot, Aurélie Wauquier, Emilie Blond, Valérie Sauvinet, Geneviève Gésan-Guiziou, Jean-Pierre Bodin, Philippe Moulin, David Cheillan, Hubert Vidal, Béatrice Morio, Eddy Cotte, Françoise Morel-Laporte, Martine Laville, Annick Bernalier-Donadille, Stéphanie Lambert-Porcheron, Corinne Malpuech-Brugère, Marie-Caroline Michalski

Abstract

Objective: To investigate whether milk polar lipids (PL) impact human intestinal lipid absorption, metabolism, microbiota and associated markers of cardiometabolic health.

Design: A double-blind, randomised controlled 4-week study involving 58 postmenopausal women was used to assess the chronic effects of milk PL consumption (0, 3 or 5 g-PL/day) on lipid metabolism and gut microbiota. The acute effects of milk PL on intestinal absorption and metabolism of cholesterol were assessed in a randomised controlled crossover study using tracers in ileostomy patients.

Results: Over 4 weeks, milk PL significantly reduced fasting and postprandial plasma concentrations of cholesterol and surrogate lipid markers of cardiovascular disease risk, including total/high-density lipoprotein-cholesterol and apolipoprotein (Apo)B/ApoA1 ratios. The highest PL dose preferentially induced a decreased number of intestine-derived chylomicron particles. Also, milk PL increased faecal loss of coprostanol, a gut-derived metabolite of cholesterol, but major bacterial populations and faecal short-chain fatty acids were not affected by milk PL, regardless of the dose. Acute ingestion of milk PL by ileostomy patients shows that milk PL decreased cholesterol absorption and increased cholesterol-ileal efflux, which can be explained by the observed co-excretion with milk sphingomyelin in the gut.

Conclusion: The present data demonstrate for the first time in humans that milk PL can improve the cardiometabolic health by decreasing several lipid cardiovascular markers, notably through a reduced intestinal cholesterol absorption involving specific interactions in the gut, without disturbing the major bacterial phyla of gut microbiota.

Trial registration number: NCT02099032 and NCT02146339; Results.

Keywords: colonic microflora; lipid absorption; lipid metabolism; lipoprotein-cholesterol; nutrition.

Conflict of interest statement

Competing interests: This work was supported in part by a grant from the French Dairy Interbranch Organization (CNIEL). M-CM has received research funding for other research projects from CNIEL, Danone-Nutricia Research, Sodiaal-Candia R&D. M-CM has consultancy activities for food, fats and oils and dairy companies. M-CM is a member of the scientific advisory board of ITERG, the Industrial Technical Centre for the oils and fats business sector. These activities had no link with the present study. BM has received research funding from Sofiprotéol with no link with the present study. PG is an employee of ACTALIA Produits Laitiers, an Agri-Food Technical Institute, with a strong specialisation in dairy research and development, and food safety. KR was an employee of ACTALIA Produits Laitiers and is now employee of Terra Lacta, a dairy cooperative. J-PB: ENILIA had research funding from dairy companies Laiterie des Fayes and Eurial. These activities had no link with the present study. GG-G has received research funding for other research projects from Savencia, Danone, Sodiaal-Candia R&D, Boccard. She has consultancy activities for dairy companies and filtration equipment providers. These activities had no link with the present study. MLa is a member of the Scientific Committee of Roquette and had research collaborations with Mondelez and Bridor. These activities had no link with the present study. The authors have no additional financial interests. Other authors declared no conflict of interest. There are no awarded or filed patents pertaining to the results presented in the paper.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Impact of 4-week intervention with up to 5 g milk PL in cream cheese on postprandial concentrations of lipid CV risk markers (VALOBAB-C trial). Panel explanation (A) and kinetics of serum total C (B), serum TAG (C), plasma ApoB/ApoA1 ratio (D) and plasma ApoB48 (E) before (V1, dotted line) and after (V2, full line) the daily consumption of 100 g of cheese with or without PL during 4 weeks. Data are represented as mean±SEM. The pgroup and pposthoc are shown for the 8-hour postprandial period (linear mixed model followed by post hoc analyses on ΔV2−V1); ptimexgroup was not significant. Group size: control n=18, 3 g-PL n=18, 5 g-PL n=20. See also online supplementary figure S1 for study design. Apo, apolipoprotein; C, cholesterol; CVD, cardiovascular disease; CV, cardiovascular; PL, polar lipids; TAG, triacylglycerols.
Figure 2
Figure 2
Modulation of postprandial chylomicron parameters after 4-week intervention with control, 3 g-PL or 5 g-PL cream cheese (VALOBAB-C trial). Panel explanation (A) and plasma kinetics of CMRF TAG (B), CMRF C (C) and CMRF size (D) before (V1, dotted line) and after (V2, full line) the daily consumption of 100 g of cheese with or without PL during 4 weeks. Data are represented as mean±SEM. pgroup and pposthoc are shown for postprandial period from 120 to 480 min (linear mixed model followed by post hoc analyses on ΔV2−V1). ptimexgroup was not significant. (B, C): after adjustment on quartiles of volunteer age and waist circumference: pgroup<0.05. For technical reason, analyses performed in centre 1 only, sample size: control n=9, 3 g-PL n=9, 5 g-PL n=10. CMRF, chylomicron-rich fraction; PL, polar lipids; TAG, triacylglycerols.
Figure 3
Figure 3
Impact of 4-week intervention with milk polar lipids (PL)-enriched cream cheese on faecal lipids in the VALOBAB-C trial. Faecal loss variations (ΔV2−V1) of total lipids (A), cholesterol (B), coprostanol (C) and coprostanol/cholesterol ratio (D) after daily consumption of 100 g of cheese with or without PL during 4 weeks. Coprostanol/cholesterol ratio before (V1) and after (V2) the 4-week consumption of 100 g of cheese with or without PL (E). Spearman’s correlation between faecal coprostanol/cholesterol ratio and serum low-density lipoprotein-cholesterol (LDL-C) (F) and total cholesterol (C) (G) after (V2) daily consumption of 100 g of cheese with or without PL during 4 weeks (orange: control group, light blue: 3 g-PL group, dark blue: 5 g-PL group). Spearman’s correlation between faecal coprostanol/cholesterol ratio and serum total C (H) and LDL-C (I) at V2 in the 5 g-PL group. Data are indicated as median with IQR. The pgroup is shown to compare intervention effect between the three groups (analysis on ranks on ΔV2−V1); PPL is shown to compare intervention effect between the control group and both PL groups regardless of dose. For technical reasons, analyses performed in centre 1 only, sample size: (A) n=9 (control) to 10 (PL); (B–C) n=4 (control) to 7 (PL); (D–I) n=7 (control) to 9 (PL). NB: regardless of group and visit, the observed faecal losses of cholesterol (range 0.2–1 mg/g faeces) and coprostanol (range 0.45–5.2 mg/g faeces) were consistent with previous published data (cholesterol: 1.88±0.53 to 5.8±1.56 mg/g dry faeces; coprostanol: 3–27.4 mg/g lyophilised faeces, considering that dry matter is ~25% of total faeces weight).
Figure 4
Figure 4
Major phylogenetic groups and bacterial species of gut microbiota after 4-week intervention with up to 5 g milk polar lipids (PL) in cream cheese in the VALOBAB-C trial. Variations (log-fold change V2/V1) of the abundance of the main bacterial groups and species that were measured as log 16S rDNA gene copies per g of faeces: (A) total bacteria, (B) Firmicutes, (C) Bacteroides-Prevotella group, (D) Akkermancia muciniphila, (E) Bifidobacterium spp, (F) Lactobacillus-Leuconostoc-Pediococcus group, (G) Clostridium coccoides group, (H) Clostridium leptum group, (I) Faecalibacterium prausnitzii, (J) Roseburia-Eubacterium rectale group, (K) Veillonella spp, (L) Enterobacteriaceae family, (M) Escherichia coli and (N) Bilophila wadworthia. Data are indicated as median with IQR; pgroup is shown. Group size: control n=18, 3 g-PL n=18, 5 g-PL n=20.
Figure 5
Figure 5
Faecal profile of short-chain fatty acids before (V1) and after (V2) 4-week intervention with up to 5 g/day milk polar lipids (PL) in cream cheese in the VALOBAB-C trial. (A) Acetate, propionate and butyrate, (B) isobutyrate, formate, succinate and lactate. Data are represented as mean±SEM, pgroup are shown. NB: regarding the total amount of major short-chain fatty acids in (A), acetate+propionate+butyrate: pgroup=0.35. For technical reasons, analyses performed in centre 2 only, sample size: control n=8, 3 g-PL n=7, 5 g-PL n=8.
Figure 6
Figure 6
Milk polar lipids (PL) impact on intestinal cholesterol absorption in the VALOBAB-D pilot study in ileostomy volunteers. Panel explanation (A), cumulated enrichment over 0–480 min of [2H]-cholesterol in plasma (B) and chylomicron-rich fraction (CMRF) (C), ileal losses of total cholesterol (D) and sphingomyelin (SM) (E), during the 0–240 min and 240–480 min postprandial periods (see also online supplementary figure S3G for SM composition in ileal efflux). Data are expressed as mean±SEM; n=4 per group. (B) Friedman test (pmeal) followed by Dunn’s post hoc tests for control and 5 g-PL groups. (C) Repeated measures one-way analysis of variance (ANOVA) (pmeal) followed by Tukey’s post hoc tests. (D, E) Repeated measures two-way ANOVA (pmeal, ptime, pmeal×time) followed by Tukey’s post hoc tests. *Pposthoc<0.05 vs control group. The pPL compares the effect of control meal with both PL meals regardless of dose. Sample size: n=4 subjects, crossover design (see online supplementary figure S2). NB: The 8-hour total cholesterol efflux (tracer+tracee in both free and esterified forms) after the control meal (642±57 mg) was consistent with previous reports; here after 3 g-PL meal the 8-hour total cholesterol efflux was 1510±417 and 1752±354 mg after 5 g-PL meal. NB: The proportion vs ingested milk SM of the 8-hour ileal efflux of milk SM after cheese matrix (20%–25% of ingested dose) was similar with 19% reported after pure milk SM in six ileostomy subjects.

References

    1. Chassaing B, Koren O, Goodrich JK, et al. . Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature 2015;519:92–6.10.1038/nature14232
    1. Chassaing B, Van de Wiele T, De Bodt J, et al. . Dietary emulsifiers directly alter human microbiota composition and gene expression ex vivo potentiating intestinal inflammation. Gut 2017;66:1414–27.10.1136/gutjnl-2016-313099
    1. Cohn JS, Kamili A, Wat E, et al. . Dietary phospholipids and intestinal cholesterol absorption. Nutrients 2010;2:116–27.10.3390/nu2020116
    1. FAO. Food and Agriculture Organization of the United Nations Gateway to dairy production and products: Milk and milk products, 2018.
    1. Bourlieu C, Michalski MC. Structure-function relationship of the milk fat globule. Curr Opin Clin Nutr Metab Care 2015;18:118–27.10.1097/MCO.0000000000000138
    1. Jiménez-Flores R, Brisson G. The milk fat globule membrane as an ingredient: why, how, when? Dairy Science and Technology 2008;88:5–18.10.1051/dst:2007005
    1. Bourlieu C, Cheillan D, Blot M, et al. . Polar lipid composition of bioactive dairy co-products buttermilk and butterserum: Emphasis on sphingolipid and ceramide isoforms. Food Chem 2018;240:67–74.10.1016/j.foodchem.2017.07.091
    1. Noh SK, Koo SI. Milk sphingomyelin is more effective than egg sphingomyelin in inhibiting intestinal absorption of cholesterol and fat in rats. J Nutr 2004;134:2611–6.10.1093/jn/134.10.2611
    1. Eckhardt ER, Wang DQ, Donovan JM, et al. . Dietary sphingomyelin suppresses intestinal cholesterol absorption by decreasing thermodynamic activity of cholesterol monomers. Gastroenterology 2002;122:948–56.10.1053/gast.2002.32539
    1. Lecomte M, Bourlieu C, Meugnier E, et al. . Milk Polar Lipids Affect In Vitro Digestive Lipolysis and Postprandial Lipid Metabolism in Mice. J Nutr 2015;145:1770–7.10.3945/jn.115.212068
    1. Wat E, Tandy S, Kapera E, et al. . Dietary phospholipid-rich dairy milk extract reduces hepatomegaly, hepatic steatosis and hyperlipidemia in mice fed a high-fat diet. Atherosclerosis 2009;205:144–50.10.1016/j.atherosclerosis.2008.12.004
    1. Kamili A, Wat E, Chung RW, et al. . Hepatic accumulation of intestinal cholesterol is decreased and fecal cholesterol excretion is increased in mice fed a high-fat diet supplemented with milk phospholipids. Nutr Metab 2010;7:90.10.1186/1743-7075-7-90
    1. Demmer E, Van Loan MD, Rivera N, et al. . Addition of a dairy fraction rich in milk fat globule membrane to a high-saturated fat meal reduces the postprandial insulinaemic and inflammatory response in overweight and obese adults. J Nutr Sci 2016;5:5.10.1017/jns.2015.42
    1. Keller S, Malarski A, Reuther C, et al. . Milk phospholipid and plant sterol-dependent modulation of plasma lipids in healthy volunteers. Eur J Nutr 2013;52:1169–79.10.1007/s00394-012-0427-0
    1. Ohlsson L, Burling H, Nilsson A. Long term effects on human plasma lipoproteins of a formulation enriched in butter milk polar lipid. Lipids Health Dis 2009;8:44.10.1186/1476-511X-8-44
    1. Weiland A, Bub A, Barth SW, et al. . Effects of dietary milk- and soya-phospholipids on lipid-parameters and other risk indicators for cardiovascular diseases in overweight or obese men - two double-blind, randomised, controlled, clinical trials. J Nutr Sci 2016;5:5.10.1017/jns.2016.9
    1. David LA, Maurice CF, Carmody RN, et al. . Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014;505:559–63.10.1038/nature12820
    1. Tindall AM, Petersen KS, Kris-Etherton PM. Dietary Patterns Affect the Gut Microbiome-The Link to Risk of Cardiometabolic Diseases. J Nutr 2018;148:1402–7.10.1093/jn/nxy141
    1. Martinez-Guryn K, Hubert N, Frazier K, et al. . Small Intestine Microbiota Regulate Host Digestive and Absorptive Adaptive Responses to Dietary Lipids. Cell Host Microbe 2018;23:458–69.10.1016/j.chom.2018.03.011
    1. Cuevas-Tena M, Bermúdez JD, Silvestre RLÁ, et al. . Impact of colonic fermentation on sterols after the intake of a plant sterol-enriched beverage: a randomized, double-blind crossover trial. Clin Nutr 201810.1016/j.clnu.2018.08.012
    1. Stampfer MJ, Colditz GA, Willett WC. Menopause and heart disease. A review. Ann N Y Acad Sci 1990;592:193–203. discussion 57-62.
    1. Grundy SM. Summary of the second report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel II). JAMA 1993;269:3015–23.10.1001/jama.1993.03500230097036
    1. Marchi R, Dell’Agnolo CM, Lopes TCR, et al. . Prevalence of metabolic syndrome in pre- and postmenopausal women. Arch Endocrinol Metab 2017;61:160–6.10.1590/2359-3997000000253
    1. Perk J, De Backer G, Gohlke H, et al. . European Association for Cardiovascular Prevention & Rehabilitation (EACPR) ESC Committee for Practice Guidelines (CPG). European Guidelines on cardiovascular disease prevention in clinical practice (version 2012). The Fifth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of nine societies and by invited experts). Eur Heart J 2012;33:1635–701.10.1093/eurheartj/ehs092
    1. Gassi JY, Blot M, Beaucher E, et al. . Preparation and characterisation of a milk polar lipids enriched ingredient from fresh industrial liquid butter serum: Combination of physico-chemical modifications and technological treatments. Int Dairy J 2016;52:26–34.10.1016/j.idairyj.2015.08.012
    1. Conway V, Couture P, Richard C, et al. . Impact of buttermilk consumption on plasma lipids and surrogate markers of cholesterol homeostasis in men and women. Nutr Metab Cardiovasc Dis 2013;23:1255–62.10.1016/j.numecd.2013.03.003
    1. Telle-Hansen VH, Holven KB, Ulven SM. Impact of a Healthy Dietary Pattern on Gut Microbiota and Systemic Inflammation in Humans. Nutrients 2018;10:1783.10.3390/nu10111783
    1. Salonen A, de Vos WM. Impact of diet on human intestinal microbiota and health. Annu Rev Food Sci Technol 2014;5:239–62.10.1146/annurev-food-030212-182554
    1. Ohlsson L, Hertervig E, Jönsson BA, et al. . Sphingolipids in human ileostomy content after meals containing milk sphingomyelin. Am J Clin Nutr 2010;91:672–8.10.3945/ajcn.2009.28311
    1. Lopez C, Madec M-N, Jimenez-Flores R. Lipid rafts in the bovine milk fat globule membrane revealed by the lateral segregation of phospholipids and heterogeneous distribution of glycoproteins. Food Chem 2010;120:22–33.10.1016/j.foodchem.2009.09.065
    1. Pearson TA, Mensah GA, Alexander RW, et al. . Centers for Disease Control and Prevention American Heart Association. Markers of inflammation and cardiovascular disease: application to clinical and public health practice: A statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation 2003;107:499–511.
    1. McQueen MJ, Hawken S, Wang X, et al. . INTERHEART study investigators. Lipids, lipoproteins, and apolipoproteins as risk markers of myocardial infarction in 52 countries (the INTERHEART study): a case-control study. Lancet 2008;372:224–33.10.1016/S0140-6736(08)61076-4
    1. Baigent C, Keech A, Kearney PM, et al. . Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet 2005;366:1267–78.10.1016/S0140-6736(05)67394-1
    1. Ference BA. Causal effect of lipids and lipoproteins on atherosclerosis. Cardiol Clin 2018;36:203–11.10.1016/j.ccl.2017.12.001
    1. Cariou B, Le May C, Costet P. Clinical aspects of PCSK9. Atherosclerosis 2011;216:258–65.10.1016/j.atherosclerosis.2011.04.018
    1. Ramprasath VR, Jones PJ, Buckley DD, et al. . Effect of dietary sphingomyelin on absorption and fractional synthetic rate of cholesterol and serum lipid profile in humans. Lipids Health Dis 2013;12:125.10.1186/1476-511X-12-125
    1. Baumgartner S, Kelly ER, van der Made S, et al. . The influence of consuming an egg or an egg-yolk buttermilk drink for 12 wk on serum lipids, inflammation, and liver function markers in human volunteers. Nutrition 2013;29:1237–44.10.1016/j.nut.2013.03.020
    1. Rosqvist F, Smedman A, Lindmark-Månsson H, et al. . Potential role of milk fat globule membrane in modulating plasma lipoproteins, gene expression, and cholesterol metabolism in humans: a randomized study. Am J Clin Nutr 2015;102:20–30.10.3945/ajcn.115.107045
    1. Morifuji M, Higashi S, Oba C, et al. . Milk Phospholipids Enhance Lymphatic Absorption of Dietary Sphingomyelin in Lymph-Cannulated Rats. Lipids 2015;50:987–96.10.1007/s11745-015-4054-4
    1. Mensink RP, Katan MB. Effect of dietary fatty acids on serum lipids and lipoproteins. A meta-analysis of 27 trials. Arterioscler Thromb 1992;12:911–9.10.1161/01.ATV.12.8.911
    1. Nordestgaard BG, Langsted A, Freiberg JJ. Nonfasting hyperlipidemia and cardiovascular disease. Curr Drug Targets 2009;10:328–35.10.2174/138945009787846434
    1. Chapman MJ, Ginsberg HN, Amarenco P, et al. . Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high risk of cardiovascular disease: evidence and guidance for management. Eur Heart J 2011;32:1345–61.10.1093/eurheartj/ehr112
    1. Walldius G, Jungner I, Holme I, et al. . High apolipoprotein B, low apolipoprotein A-I, and improvement in the prediction of fatal myocardial infarction (AMORIS study): a prospective study. Lancet 2001;358:2026–33.10.1016/S0140-6736(01)07098-2
    1. Tomkin GH, Owens D. The chylomicron: relationship to atherosclerosis. Int J Vasc Med 2012;2012:1–13.10.1155/2012/784536
    1. Ohlsson L, Burling H, Duan RD, et al. . Effects of a sphingolipid-enriched dairy formulation on postprandial lipid concentrations. Eur J Clin Nutr 2010;64:1344–9.10.1038/ejcn.2010.164
    1. Gérard P. Metabolism of Cholesterol and Bile Acids by the Gut Microbiota. Pathogens 2014;3:14–24.10.3390/pathogens3010014
    1. Mosele JI, Gosalbes MJ, Macià A, et al. . Effect of daily intake of pomegranate juice on fecal microbiota and feces metabolites from healthy volunteers. Mol Nutr Food Res 2015;59:1942–53.10.1002/mnfr.201500227
    1. Cuevas-Tena M, Alegría A, Lagarda MJ. Determination of Fecal Sterols Following a Diet with and without Plant Sterols. Lipids 2017;52:871–84.10.1007/s11745-017-4286-6
    1. Durack J, Lynch SV. The gut microbiome: Relationships with disease and opportunities for therapy. J Exp Med 2019;216:20–40.10.1084/jem.20180448
    1. den Besten G, van Eunen K, Groen AK, et al. . The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 2013;54:2325–40.10.1194/jlr.R036012
    1. Lukovac S, Belzer C, Pellis L, et al. . Differential modulation by Akkermansia muciniphila and Faecalibacterium prausnitzii of host peripheral lipid metabolism and histone acetylation in mouse gut organoids. MBio 2014;510.1128/mBio.01438-14
    1. Norris GH, Jiang C, Ryan J, et al. . Milk sphingomyelin improves lipid metabolism and alters gut microbiota in high fat diet-fed mice. J Nutr Biochem 2016;30:93–101.10.1016/j.jnutbio.2015.12.003
    1. Milard M, Laugerette F, Durand A, et al. . Milk Polar Lipids in a High-Fat Diet Can Prevent Body Weight Gain: Modulated Abundance of Gut Bacteria in Relation with Fecal Loss of Specific Fatty Acids. Mol Nutr Food Res 2019;63:e180107810.1002/mnfr.201801078
    1. Cani PD, Possemiers S, Van de Wiele T, et al. . Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009;58:1091–103.10.1136/gut.2008.165886
    1. Kriaa A, Bourgin M, Potiron A, et al. . Microbial impact on cholesterol and bile acid metabolism: current status and future prospects. J Lipid Res 2019;60:323–32.10.1194/jlr.R088989
    1. Lia A, Andersson H, Mekki N, et al. . Postprandial lipemia in relation to sterol and fat excretion in ileostomy subjects given oat-bran and wheat test meals. Am J Clin Nutr 1997;66:357–65.10.1093/ajcn/66.2.357
    1. Nyberg L, Duan RD, Nilsson A. A mutual inhibitory effect on absorption of sphingomyelin and cholesterol. J Nutr Biochem 2000;11:244–9.10.1016/S0955-2863(00)00069-3
    1. Beil FU, Grundy SM. Studies on plasma lipoproteins during absorption of exogenous lecithin in man. J Lipid Res 1980;21:525–36.
    1. Le May C, Berger JM, Lespine A, et al. . Transintestinal cholesterol excretion is an active metabolic process modulated by PCSK9 and statin involving ABCB1. Arterioscler Thromb Vasc Biol 2013;33:1484–93.10.1161/ATVBAHA.112.300263
    1. Jakulj L, van Dijk TH, de Boer JF, et al. . Transintestinal Cholesterol Transport Is Active in Mice and Humans and Controls Ezetimibe-Induced Fecal Neutral Sterol Excretion. Cell Metab 2016;24:783–94.10.1016/j.cmet.2016.10.001
    1. Miller M, Stone NJ, Ballantyne C, et al. . Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation 2011;123:2292–333.10.1161/CIR.0b013e3182160726
    1. Collins R, Reith C, Emberson J, et al. . Interpretation of the evidence for the efficacy and safety of statin therapy. Lancet 2016;388:2532–61.10.1016/S0140-6736(16)31357-5
    1. EAS. European Atherosclerosis Society: Commentary on Phytosterol-added Foods Focus on lifestyle: EAS Consensus Panel Position Statement on Phytosterol-added Foods, 2015.
    1. Drouin-Chartier JP, Côté JA, Labonté MÈ, et al. . Comprehensive Review of the Impact of Dairy Foods and Dairy Fat on Cardiometabolic Risk. Adv Nutr 2016;7:1041–51.10.3945/an.115.011619
    1. Thorning TK, Bertram HC, Bonjour JP, et al. . Whole dairy matrix or single nutrients in assessment of health effects: current evidence and knowledge gaps. Am J Clin Nutr 2017;105:1033–45.10.3945/ajcn.116.151548
    1. World Health Organization W. Waist Circumference and Waist–Hip Ratio: Report of a WHO Expert Consultation. Geneva: WHO, 2008.
    1. Alberti KG, Zimmet P, Shaw J. Metabolic syndrome–a new world-wide definition. A Consensus Statement from the International Diabetes Federation. Diabet Med 2006;23:469–80.10.1111/j.1464-5491.2006.01858.x
    1. National Cholesterol Education Program (NCEP). Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III), 2002.

Source: PubMed

3
Abonnere