Population-based plasma lipidomics reveals developmental changes in metabolism and signatures of obesity risk: a mother-offspring cohort study

Sartaj Ahmad Mir, Li Chen, Satvika Burugupalli, Bo Burla, Shanshan Ji, Adam Alexander T Smith, Kothandaraman Narasimhan, Adaikalavan Ramasamy, Karen Mei-Ling Tan, Kevin Huynh, Corey Giles, Ding Mei, Gerard Wong, Fabian Yap, Kok Hian Tan, Fiona Collier, Richard Saffery, Peter Vuillermin, Anne K Bendt, David Burgner, Anne-Louise Ponsonby, Yung Seng Lee, Yap Seng Chong, Peter D Gluckman, Johan G Eriksson, Peter J Meikle, Markus R Wenk, Neerja Karnani, Sartaj Ahmad Mir, Li Chen, Satvika Burugupalli, Bo Burla, Shanshan Ji, Adam Alexander T Smith, Kothandaraman Narasimhan, Adaikalavan Ramasamy, Karen Mei-Ling Tan, Kevin Huynh, Corey Giles, Ding Mei, Gerard Wong, Fabian Yap, Kok Hian Tan, Fiona Collier, Richard Saffery, Peter Vuillermin, Anne K Bendt, David Burgner, Anne-Louise Ponsonby, Yung Seng Lee, Yap Seng Chong, Peter D Gluckman, Johan G Eriksson, Peter J Meikle, Markus R Wenk, Neerja Karnani

Abstract

Background: Lipids play a vital role in health and disease, but changes to their circulating levels and the link with obesity remain poorly characterized in expecting mothers and their offspring in early childhood.

Methods: LC-MS/MS-based quantitation of 480 lipid species was performed on 2491 plasma samples collected at 4 time points in the mother-offspring Asian cohort GUSTO (Growing Up in Singapore Towards healthy Outcomes). These 4 time points constituted samples collected from mothers at 26-28 weeks of gestation (n=752) and 4-5 years postpartum (n=650), and their offspring at birth (n=751) and 6 years of age (n=338). Linear regression models were used to identify the pregnancy and developmental age-specific variations in the plasma lipidomic profiles, and their association with obesity risk. An independent birth cohort (n=1935), the Barwon Infant Study (BIS), comprising mother-offspring dyads of Caucasian origin was used for validation.

Results: Levels of 36% of the profiled lipids were significantly higher (absolute fold change > 1.5 and Padj < 0.05) in antenatal maternal circulation as compared to the postnatal phase, with phosphatidylethanolamine levels changing the most. Compared to antenatal maternal lipids, cord blood showed lower concentrations of most lipid species (79%) except lysophospholipids and acylcarnitines. Changes in lipid concentrations from birth to 6 years of age were much higher in magnitude (log2FC=-2.10 to 6.25) than the changes observed between a 6-year-old child and an adult (postnatal mother) (log2FC=-0.68 to 1.18). Associations of cord blood lipidomic profiles with birth weight displayed distinct trends compared to the lipidomic profiles associated with child BMI at 6 years. Comparison of the results between the child and adult BMI identified similarities in association with consistent trends (R2=0.75). However, large number of lipids were associated with BMI in adults (67%) compared to the children (29%). Pre-pregnancy BMI was specifically associated with decrease in the levels of phospholipids, sphingomyelin, and several triacylglycerol species in pregnancy.

Conclusions: In summary, our study provides a detailed landscape of the in utero lipid environment provided by the gestating mother to the growing fetus, and the magnitude of changes in plasma lipidomic profiles from birth to early childhood. We identified the effects of adiposity on the circulating lipid levels in pregnant and non-pregnant women as well as offspring at birth and at 6 years of age. Additionally, the pediatric vs maternal overlap of the circulating lipid phenotype of obesity risk provides intergenerational insights and early opportunities to track and intervene the onset of metabolic adversities.

Clinical trial registration: This birth cohort is a prospective observational study, which was registered on 1 July 2010 under the identifier NCT01174875 .

Keywords: Adiposity; Development; Gestation; Intergenerational; Lipidomics; Lipids and fatty acids; Maternal-fetal; Metabolomics.

Conflict of interest statement

NK and YSC are part of an academic consortium that has received research funding from Abbott Nutrition, Nestec, EVOLVE Biosystems, DSM, and Danone. All other authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Temporal and developmental alterations to the circulatory lipids in the GUSTO cohort: A Antenatal and postnatal plasma collection time points for mother-offspring dyads in GUSTO cohort. B PCA plot of lipidomics data (n=2491). C Postnatal vs. antenatal changes in maternal lipidomic profiles. D Comparison of maternal antenatal plasma with cord blood (CB) lipidomic profiles. E Changes in child lipidomic profiles between birth and 6 years of age. F Comparison of pediatric (6-year-old child) and adult (postnatal mothers) plasma lipidomes. The most significant lipid species based on adjusted p-values are labeled in c-f. Effect size is shown as log2 of fold change. Error bars indicate 95% confidence interval. Diamond—Padj ≥ 0.05 or |FC|≤1.5 (gray), circle—Padj <0.05 and |FC|>1.5, and square—Padj <1.00E−10 and |FC|>1.5
Fig. 2
Fig. 2
Association of maternal and child adiposity with plasma lipidomic profiles: A Association of pre-pregnancy BMI with antenatal plasma lipidome (pregnant state). B Association of maternal BMI with postnatal plasma lipidome (non-pregnant state). C Association of birth weight (BW) with cord blood plasma lipidome. D Association of child BMI with plasma lipidome at 6 years of age. The top 20 lipid species with the directionality of association (10 positive and 10 negative) with adiposity measures in each study group are shown in volcano plots. The horizontal dotted line indicates Padj=0.05 in each volcano plot. Effect sizes are shown as % change in lipid concentration per unit change in BMI, or per 100 g change in birth weight. Error bars indicate 95% confidence interval. Diamond—Padj ≥ 0.05 (gray), circle—Padj <0.05, and square—Padj <1.00E−5 in forest plots
Fig. 3
Fig. 3
Comparison of plasma lipidomic profiles associated with mother and child adiposity: A Pie chart comparing the percent overlap and directionality of association in the four studies. B Venn-diagram of lipids species that passed significance in the four association studies. C Effect sizes of 41 lipid species that overlapped between the four studies
Fig. 4
Fig. 4
Replication of GUSTO identified ppBMI and BW lipid signatures in Barwon Infant Study (BIS). A Sample collection at two time points. B PCA plot of lipidomics data (n=1935). C Association of pre-pregnancy BMI (ppBMI) with antenatal plasma lipidome. D Scatter plot of effect sizes in GUSTO and BIS for ppBMI study. E Association of birth weight (BW) with cord blood plasma lipidome. The most significant lipid species based on adjusted p-values are labeled in C and E. Effect sizes are shown as % change in lipid concentration per unit change in BMI, or per 100 g change in birth weight. Error bars indicate 95% confidence interval. Diamond—Padj ≥ 0.05 (gray), circle—Padj <0.05, and square—Padj <1.00E−5. F Scatter plot of effect sizes in GUSTO and BIS for BW study. Red—significant in both cohorts, purple—only significant in GUSTO, yellow—only significant in BIS, and gray—not significant in both cohorts

References

    1. Alvarez JJ, Montelongo A, Iglesias A, Lasuncion MA, Herrera E. Longitudinal study on lipoprotein profile, high density lipoprotein subclass, and postheparin lipases during gestation in women. J Lipid Res. 1996;37(2):299–308. doi: 10.1016/S0022-2275(20)37617-3.
    1. Grimes SB, Wild R. Effect of pregnancy on lipid metabolism and lipoprotein levels. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, et al., editors. Endotext. South Dartmouth (MA)2000.
    1. Herrera E, Ortega-Senovilla H. Disturbances in lipid metabolism in diabetic pregnancy - Are these the cause of the problem? Best Pract Res Clin Endocrinol Metab. 2010;24(4):515–525. doi: 10.1016/j.beem.2010.05.006.
    1. Shen H, Liu X, Chen Y, He B, Cheng W. Associations of lipid levels during gestation with hypertensive disorders of pregnancy and gestational diabetes mellitus: a prospective longitudinal cohort study. BMJ Open. 2016;6(12):e013509. doi: 10.1136/bmjopen-2016-013509.
    1. Kulkarni SR, Kumaran K, Rao SR, Chougule SD, Deokar TM, Bhalerao AJ, et al. Maternal lipids are as important as glucose for fetal growth: findings from the Pune Maternal Nutrition Study. Diabetes Care. 2013;36(9):2706–2713. doi: 10.2337/dc12-2445.
    1. Misra VK, Trudeau S, Perni U. Maternal serum lipids during pregnancy and infant birth weight: the influence of prepregnancy BMI. Obesity (Silver Spring) 2011;19(7):1476–1481. doi: 10.1038/oby.2011.43.
    1. Christensen JJ, Ulven SM, Retterstol K, Narverud I, Bogsrud MP, Henriksen T, et al. Comprehensive lipid and metabolite profiling of children with and without familial hypercholesterolemia: A cross-sectional study. Atherosclerosis. 2017;266:48–57. doi: 10.1016/j.atherosclerosis.2017.09.021.
    1. Geraghty AA, Alberdi G, O'Sullivan EJ, O'Brien EC, Crosbie B, Twomey PJ, et al. Maternal blood lipid profile during pregnancy and associations with child adiposity: findings from the ROLO study. PLoS One. 2016;11(8):e0161206. doi: 10.1371/journal.pone.0161206.
    1. Jin WY, Lin SL, Hou RL, Chen XY, Han T, Jin Y, et al. Associations between maternal lipid profile and pregnancy complications and perinatal outcomes: a population-based study from China. BMC Pregnancy Childbirth. 2016;16:60. doi: 10.1186/s12884-016-0852-9.
    1. Geraghty AA, Alberdi G, O'Sullivan EJ, O'Brien EC, Crosbie B, Twomey PJ, et al. Maternal and fetal blood lipid concentrations during pregnancy differ by maternal body mass index: findings from the ROLO study. BMC Pregnancy Childbirth. 2017;17(1):360. doi: 10.1186/s12884-017-1543-x.
    1. Wang Q, Wurtz P, Auro K, Makinen VP, Kangas AJ, Soininen P, et al. Metabolic profiling of pregnancy: cross-sectional and longitudinal evidence. BMC Med. 2016;14(1):205. doi: 10.1186/s12916-016-0733-0.
    1. Hellmuth C, Lindsay KL, Uhl O, Buss C, Wadhwa PD, Koletzko B, et al. Association of maternal prepregnancy BMI with metabolomic profile across gestation. Int J Obes (Lond) 2017;41(1):159–169. doi: 10.1038/ijo.2016.153.
    1. Rahman ML, Feng YA, Fiehn O, Albert PS, Tsai MY, Zhu Y, et al. Plasma lipidomics profile in pregnancy and gestational diabetes risk: a prospective study in a multiracial/ethnic cohort. BMJ Open Diabetes Res Care. 2021;9(1).
    1. Soh SE, Tint MT, Gluckman PD, Godfrey KM, Rifkin-Graboi A, Chan YH, et al. Cohort profile: Growing Up in Singapore Towards healthy Outcomes (GUSTO) birth cohort study. Int J Epidemiol. 2014;43(5):1401–1409. doi: 10.1093/ije/dyt125.
    1. Vuillermin P, Saffery R, Allen KJ, Carlin JB, Tang ML, Ranganathan S, et al. Cohort Profile: The Barwon Infant Study. Int J Epidemiol. 2015;44(4):1148–1160. doi: 10.1093/ije/dyv026.
    1. Alshehry ZH, Barlow CK, Weir JM, Zhou Y, McConville MJ, Meikle PJ. An efficient single phase method for the extraction of plasma lipids. Metabolites. 2015;5(2):389–403. doi: 10.3390/metabo5020389.
    1. Huynh K, Barlow CK, Jayawardana KS, Weir JM, Mellett NA, Cinel M, et al. High-throughput plasma lipidomics: detailed mapping of the associations with cardiometabolic risk factors. Cell. Chem Biol. 2019;26(1):71–84 e4.
    1. Weir JM, Wong G, Barlow CK, Greeve MA, Kowalczyk A, Almasy L, et al. Plasma lipid profiling in a large population-based cohort. J Lipid Res. 2013;54(10):2898–2908. doi: 10.1194/jlr.P035808.
    1. Dunn WB, Broadhurst D, Begley P, Zelena E, Francis-McIntyre S, Anderson N, et al. Procedures for large-scale metabolic profiling of serum and plasma using gas chromatography and liquid chromatography coupled to mass spectrometry. Nat Protoc. 2011;6(7):1060–1083. doi: 10.1038/nprot.2011.335.
    1. Pettitt DJ, Jovanovic L. Birth weight as a predictor of type 2 diabetes mellitus: the U-shaped curve. Curr Diab Rep. 2001;1(1):78–81. doi: 10.1007/s11892-001-0014-x.
    1. Lee CH, Cook S, Lee JS, Han B. Comparison of two meta-analysis methods: inverse-variance-weighted average and weighted sum of Z-scores. Genomics Inform. 2016;14(4):173–180. doi: 10.5808/GI.2016.14.4.173.
    1. Tyrrell J, Richmond RC, Palmer TM, Feenstra B, Rangarajan J, Metrustry S, et al. Genetic evidence for causal relationships between maternal obesity-related traits and birth weight. JAMA. 2016;315(11):1129–1140. doi: 10.1001/jama.2016.1975.
    1. Darmady JM, Postle AD. Lipid metabolism in pregnancy. Br J Obstet Gynaecol. 1982;89(3):211–215. doi: 10.1111/j.1471-0528.1982.tb03616.x.
    1. Herrera E. Lipid metabolism in pregnancy and its consequences in the fetus and newborn. Endocrine. 2002;19(1):43–55. doi: 10.1385/ENDO:19:1:43.
    1. Kim MS, Pinto SM, Getnet D, Nirujogi RS, Manda SS, Chaerkady R, et al. A draft map of the human proteome. Nature. 2014;509(7502):575–581. doi: 10.1038/nature13302.
    1. Wallentin L, Fahraeus L. Cholesterol esterification rate and its relation to lipoprotein levels in plasma in normal human pregnancy. J Lab Clin Med. 1986;107(3):216–220.
    1. Larque E, Demmelmair H, Gil-Sanchez A, Prieto-Sanchez MT, Blanco JE, Pagan A, et al. Placental transfer of fatty acids and fetal implications. Am J Clin Nutr. 2011;94(6 Suppl):1908S–1913S. doi: 10.3945/ajcn.110.001230.
    1. Lindegaard ML, Olivecrona G, Christoffersen C, Kratky D, Hannibal J, Petersen BL, et al. Endothelial and lipoprotein lipases in human and mouse placenta. J Lipid Res. 2005;46(11):2339–2346. doi: 10.1194/jlr.M500277-JLR200.
    1. Prieto-Sanchez MT, Ruiz-Palacios M, Blanco-Carnero JE, Pagan A, Hellmuth C, Uhl O, et al. Placental MFSD2a transporter is related to decreased DHA in cord blood of women with treated gestational diabetes. Clin Nutr. 2017;36(2):513–521. doi: 10.1016/j.clnu.2016.01.014.
    1. Nguyen LN, Ma D, Shui G, Wong P, Cazenave-Gassiot A, Zhang X, et al. Mfsd2a is a transporter for the essential omega-3 fatty acid docosahexaenoic acid. Nature. 2014;509(7501):503–506. doi: 10.1038/nature13241.
    1. Grayson DS, Kroenke CD, Neuringer M, Fair DA. Dietary omega-3 fatty acids modulate large-scale systems organization in the rhesus macaque brain. J Neurosci. 2014;34(6):2065–2074. doi: 10.1523/JNEUROSCI.3038-13.2014.
    1. SanGiovanni JP, Chew EY. The role of omega-3 long-chain polyunsaturated fatty acids in health and disease of the retina. Prog Retin Eye Res. 2005;24(1):87–138. doi: 10.1016/j.preteyeres.2004.06.002.
    1. Alessandri JM, Guesnet P, Vancassel S, Astorg P, Denis I, Langelier B, et al. Polyunsaturated fatty acids in the central nervous system: evolution of concepts and nutritional implications throughout life. Reprod Nutr Dev. 2004;44(6):509–538. doi: 10.1051/rnd:2004063.
    1. van den Bosch H, Schrakamp G, Hardeman D, Zomer AW, Wanders RJ, Schutgens RB. Ether lipid synthesis and its deficiency in peroxisomal disorders. Biochimie. 1993;75(3-4):183–189. doi: 10.1016/0300-9084(93)90076-5.
    1. Munn NJ, Arnio E, Liu D, Zoeller RA, Liscum L. Deficiency in ethanolamine plasmalogen leads to altered cholesterol transport. J Lipid Res. 2003;44(1):182–192. doi: 10.1194/jlr.M200363-JLR200.
    1. Honsho M, Abe Y, Fujiki Y. Dysregulation of plasmalogen homeostasis impairs cholesterol biosynthesis. J Biol Chem. 2015;290(48):28822–28833. doi: 10.1074/jbc.M115.656983.
    1. Koberlin MS, Snijder B, Heinz LX, Baumann CL, Fauster A, Vladimer GI, et al. A conserved circular network of coregulated lipids modulates innate immune responses. Cell. 2015;162(1):170–183. doi: 10.1016/j.cell.2015.05.051.
    1. Jimenez-Rojo N, Leonetti MD, Zoni V, Colom A, Feng S, Iyengar NR, et al. Conserved functions of ether lipids and sphingolipids in the early secretory pathway. Curr Biol. 2020;30(19):3775–87 e7. doi: 10.1016/j.cub.2020.07.059.
    1. Schoderbeck M, Auer B, Legenstein E, Genger H, Sevelda P, Salzer H, et al. Pregnancy-related changes of carnitine and acylcarnitine concentrations of plasma and erythrocytes. J Perinat Med. 1995;23(6):477–485. doi: 10.1515/jpme.1995.23.6.477.
    1. Mitro SD, Wu J, Rahman ML, Cao Y, Zhu Y, Chen Z, et al. Longitudinal plasma metabolomics profile in pregnancy-a study in an ethnically diverse U.S. pregnancy cohort. Nutrients. 2021;13(9).
    1. Lindsay KL, Hellmuth C, Uhl O, Buss C, Wadhwa PD, Koletzko B, et al. Longitudinal metabolomic profiling of amino acids and lipids across healthy pregnancy. PLoS One. 2015;10(12):e0145794. doi: 10.1371/journal.pone.0145794.
    1. Matey-Hernandez ML, Williams FMK, Potter T, Valdes AM, Spector TD, Menni C. Genetic and microbiome influence on lipid metabolism and dyslipidemia. Physiol Genomics. 2018;50(2):117–126. doi: 10.1152/physiolgenomics.00053.2017.
    1. Schoeler M, Caesar R. Dietary lipids, gut microbiota and lipid metabolism. Rev Endocr Metab Disord. 2019;20(4):461–472. doi: 10.1007/s11154-019-09512-0.
    1. Cuevas-Sierra A, Ramos-Lopez O, Riezu-Boj JI, Milagro FI, Martinez JA. Diet, gut microbiota, and obesity: links with host genetics and epigenetics and potential applications. Adv Nutr. 2019;10(suppl_1):S17–S30. doi: 10.1093/advances/nmy078.
    1. Szapary PO, Bloedon LT, Foster GD. Physical activity and its effects on lipids. Curr Cardiol Rep. 2003;5(6):488–492. doi: 10.1007/s11886-003-0112-2.
    1. Clark E, Isler C, Strickland D, McMillan AG, Fang X, Kuehn D, et al. Influence of aerobic exercise on maternal lipid levels and offspring morphometrics. Int J Obes (Lond) 2019;43(3):594–602. doi: 10.1038/s41366-018-0258-z.
    1. Ramirez-Velez R, Lobelo F, Aguilar-de Plata AC, Izquierdo M, Garcia-Hermoso A. Exercise during pregnancy on maternal lipids: a secondary analysis of randomized controlled trial. BMC Pregnancy Childbirth. 2017;17(1):396. doi: 10.1186/s12884-017-1571-6.
    1. Lundqvist A, Johansson I, Wennberg A, Hultdin J, Hogberg U, Hamberg K, et al. Reported dietary intake in early pregnant compared to non-pregnant women - a cross-sectional study. BMC Pregnancy Childbirth. 2014;14:373. doi: 10.1186/s12884-014-0373-3.
    1. Oken E, Osterdal ML, Gillman MW, Knudsen VK, Halldorsson TI, Strom M, et al. Associations of maternal fish intake during pregnancy and breastfeeding duration with attainment of developmental milestones in early childhood: a study from the Danish National Birth Cohort. Am J Clin Nutr. 2008;88(3):789–796. doi: 10.1093/ajcn/88.3.789.
    1. Mijatovic-Vukas J, Capling L, Cheng S, Stamatakis E, Louie J, Cheung NW, et al. Associations of diet and physical activity with risk for gestational diabetes mellitus: a systematic review and meta-analysis. Nutrients. 2018;10(6).
    1. Middleton P, Gomersall JC, Gould JF, Shepherd E, Olsen SF, Makrides M. Omega-3 fatty acid addition during pregnancy. Cochrane Database Syst Rev 2018;11:CD003402.
    1. Wijendran V, Lawrence P, Diau GY, Boehm G, Nathanielsz PW, Brenna JT. Significant utilization of dietary arachidonic acid is for brain adrenic acid in baboon neonates. J Lipid Res. 2002;43(5):762–767. doi: 10.1016/S0022-2275(20)30118-8.
    1. Han X, Holtzman DM, McKeel DW., Jr Plasmalogen deficiency in early Alzheimer’s disease subjects and in animal models: molecular characterization using electrospray ionization mass spectrometry. J Neurochem. 2001;77(4):1168–1180. doi: 10.1046/j.1471-4159.2001.00332.x.
    1. Murphy RC. Free-radical-induced oxidation of arachidonoyl plasmalogen phospholipids: antioxidant mechanism and precursor pathway for bioactive eicosanoids. Chem Res Toxicol. 2001;14(5):463–472. doi: 10.1021/tx000250t.
    1. Chew WS, Torta F, Ji S, Choi H, Begum H, Sim X, et al. Large-scale lipidomics identifies associations between plasma sphingolipids and T2DM incidence. JCI. Insight. 2019;5.
    1. Beyene HB, Olshansky G, AA TS, Giles C, Huynh K, Cinel M, et al. High-coverage plasma lipidomics reveals novel sex-specific lipidomic fingerprints of age and BMI: Evidence from two large population cohort studies. PLoS Biol 2020;18(9):e3000870.
    1. Dabadie H, Motta C, Peuchant E, LeRuyet P, Mendy F. Variations in daily intakes of myristic and alpha-linolenic acids in sn-2 position modify lipid profile and red blood cell membrane fluidity. Br J Nutr. 2006;96(2):283–289. doi: 10.1079/BJN20061813.
    1. Dabadie H, Peuchant E, Bernard M, LeRuyet P, Mendy F. Moderate intake of myristic acid in sn-2 position has beneficial lipidic effects and enhances DHA of cholesteryl esters in an interventional study. J Nutr Biochem. 2005;16(6):375–382. doi: 10.1016/j.jnutbio.2005.01.010.
    1. Schwab U, Seppanen-Laakso T, Yetukuri L, Agren J, Kolehmainen M, Laaksonen DE, et al. Triacylglycerol fatty acid composition in diet-induced weight loss in subjects with abnormal glucose metabolism--the GENOBIN study. PLoS One. 2008;3(7):e2630. doi: 10.1371/journal.pone.0002630.
    1. Sonagra AD, Biradar SM, K D, Murthy DSJ. Normal pregnancy- a state of insulin resistance. J Clin Diagn Res. 2014;8(11):CC01–CC03.
    1. LaBarre JL, Puttabyatappa M, Song PXK, Goodrich JM, Zhou L, Rajendiran TM, et al. Maternal lipid levels across pregnancy impact the umbilical cord blood lipidome and infant birth weight. Sci Rep. 2020;10(1):14209. doi: 10.1038/s41598-020-71081-z.
    1. Lu YP, Reichetzeder C, Prehn C, Yin LH, Yun C, Zeng S, et al. Cord blood lysophosphatidylcholine 16: 1 is positively associated with birth weight. Cell Physiol Biochem. 2018;45(2):614–624. doi: 10.1159/000487118.
    1. Burugupalli S, Smith AAT, Oshlensky G, Huynh K, Giles C, Wang T, et al. Ontogeny of circulating lipid metabolism in pregnancy and early childhood: a longitudinal population study. Elife. 2022;11.
    1. Eissing L, Scherer T, Todter K, Knippschild U, Greve JW, Buurman WA, et al. De novo lipogenesis in human fat and liver is linked to ChREBP-beta and metabolic health. Nat Commun. 2013;4:1528. doi: 10.1038/ncomms2537.
    1. Lambert JE, Ramos-Roman MA, Browning JD, Parks EJ. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology. 2014;146(3):726–735. doi: 10.1053/j.gastro.2013.11.049.

Source: PubMed

3
Abonnieren