Dietary adaptation for weight loss maintenance at Yale (DAWLY): Protocol and predictions for a randomized controlled trial

Xi Fang, Xue Davis, Kyle D Flack, Chavonn Duncan, Fangyong Li, Marney White, Carlos Grilo, Dana M Small, Xi Fang, Xue Davis, Kyle D Flack, Chavonn Duncan, Fangyong Li, Marney White, Carlos Grilo, Dana M Small

Abstract

Background: Current therapies for obesity treatment are effective at producing short-term weight loss, but weight loss maintenance remains a significant challenge. Here we investigate the impact of pre-intervention dietary fat intake on the efficacy of a dietary supplement to support weight loss maintenance. Preclinical work demonstrates that a vagal afferent pathway critical for sensing dietary lipids is blunted by a high-fat diet (HFD), resulting in a reduced preference for a low-fat emulsion and severe blunting of the dopamine (DA) response to the gastric infusion of lipids. Infusion of the gut lipid messenger oleoylethanolamide (OEA), which is also depleted by HFD, immediately reverses this DA blunting and restores preference for the low-fat emulsion. Studies of OEA supplementation for weight loss in humans have had limited success. Given the strong effect of HFD on this pathway, we designed a study to test whether the efficacy of OEA as a weight loss treatment is related to pre-intervention habitual intake of dietary fat.

Methods/design: We employed a randomized, double-blind, placebo-controlled trial in which 100 adults with overweight/obesity (OW/OB) were randomized to receive either OEA or placebo daily for 16 months. Following a baseline evaluation of diet, metabolic health, adiposity, and brain response to a palatable an energy dense food, participants in both groups underwent a 4-month behavioral weight loss intervention (LEARN®) followed by a 1-year maintenance period. The study aims are to (1) determine if pre-intervention dietary fat intake moderates the ability of OEA to improve weight loss and weight loss maintenance after a gold standard behavioral weight loss treatment; (2) identify biomarkers that predict outcome and optimize a stratification strategy; and (3) test a model underlying OEA's effectiveness.

Discussion: Focusing on interventions that target the gut-brain axis is supported by mounting evidence for the role of gut-brain signaling in food choice and the modulation of this circuit by diet. If successful, this work will provide support for targeting the gut-brain pathway for weight loss maintenance using a precision medicine approach that is easy and inexpensive to implement.

Clinical trial registration: [www.ClinicalTrials.gov], identifier [NCT04614233].

Keywords: Riduzone; dietary intake; obesity; oleoylethanolamide; overweight; randomized controlled trial; supplementation; weight loss.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Fang, Davis, Flack, Duncan, Li, White, Grilo and Small.

Figures

FIGURE 1
FIGURE 1
Model of OEA efficacy. The relationship between diet and gut-brain signaling is shown in the context of a low-fat diet (LFD, gray box with lettuce) and a high-fat diet (HFD, red box with hamburger). Following habitual consumption HFD the vagal afferent pathway that ascends from intestine to the brain is blunted (dotted line) and this is associated with a blunted dopamine (DA) response to nutrients (red line on graph) compared to DA response to nutrients in LFD consumers (black line on graph). DA responses are associated with fat concentration preference, depicted in the preference scale above the graph. Preferences are shifted to the right in the context of a HFD. Supplementation with OEA in the HFD consumers is shown in cyan. Supplementation rescues the blunted vagal afferent pathway leading to a recovered DA response to nutrients (cyan line in graph) and a shift in preference toward low fat food. HFD, high-fat diet; LFD, low-fat diet; DA, dopamine; OEA, oleoylethanolamide. Figure created in Biorender.
FIGURE 2
FIGURE 2
Study design. OEA, oleoylethanolamide; FA, full assessment; SA, short assessment.
FIGURE 3
FIGURE 3
fMRI solution presentation. The solution run is 16-min long and consisted of a 1–10 s visual cue (average 3 s), a 1.5 s delivery of either chocolate/strawberry milkshake or tasteless solution, followed by a 6–14 s rest period (average 8 s) during which the subject could swallow. If milkshake is delivered, a 1.5 s rinse of deionized water then occurred and was followed by a 4–12 s rest period (average 6 s). There are 30 repetitions of milkshake and 30 repetitions of tasteless run. MS, milkshake; TLS, tasteless.

References

    1. Hales CM, Fryar CD, Ogden CL. Prevalence of Obesity and Severe Obesity Among Adults: United States, 2017–2018. Hyattsville, MD: National Center for Health Statistics; (2020).
    1. Wadden TA, Tronieri JS, Butryn ML. Lifestyle modification approaches for the treatment of obesity in adults. Am Psychol. (2020) 75:235–51. 10.1037/amp0000517
    1. Wing RR, Phelan S. Long-term weight loss maintenance. Am J Clin Nutr. (2005) 82:222S–5S. 10.1093/ajcn/82.1.222S
    1. Franz MJ, Vanwormer JJ, Crain AL, Boucher JL, Histon T, Caplan W, et al. Weight-loss outcomes: a systematic review and meta-analysis of weight-loss clinical trials with a minimum 1-year follow-up. J Am Diet Assoc. (2007) 107:1755–67. 10.1016/j.jada.2007.07.017
    1. Sawamoto R, Nozaki T, Nishihara T, Furukawa T, Hata T, Komaki G, et al. Predictors of successful long-term weight loss maintenance: a two-year follow-up. Biopsychosoc Med. (2017) 11:14. 10.1186/s13030-017-0099-3
    1. Wing RR, Bray GA, Cassidy-Begay M, Clark JM, Coday M. Look Ahead Research Group Effects of intensive lifestyle intervention on all-cause mortality in older adults with type 2 diabetes and overweight/obesity: results from the look AHEAD study. Diabetes Care. (2022) 45:1252–9. 10.2337/figshare.19196288.v1
    1. Wing RR, Neiberg RH, Bahnson JL, Clark JM, Espeland MA, Hill JO, et al. Weight change during the postintervention follow-up of look AHEAD. Diabetes Care. (2022) 45:1306–14. 10.2337/dc21-1990
    1. O’rahilly S, Farooqi IS. Human obesity: a heritable neurobehavioral disorder that is highly sensitive to environmental conditions. Diabetes. (2008) 57:2905–10. 10.2337/db08-0210
    1. Rossi MA, Stuber GD. Overlapping brain circuits for homeostatic and hedonic feeding. Cell Metab. (2018) 27:42–56. 10.1016/j.cmet.2017.09.021
    1. Han W, Tellez LA, Perkins MH, Perez IO, Qu T, Ferreira J, et al. A Neural circuit for gut-induced reward. Cell. (2018) 175:665–678e623. 10.1016/j.cell.2018.08.049
    1. Tellez LA, Medina S, Han W, Ferreira JG, Licona-Limon P, Ren X, et al. A gut lipid messenger links excess dietary fat to dopamine deficiency. Science. (2013) 341:800–2. 10.1126/science.1239275
    1. Johnson PM, Kenny PJ. Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. Nat Neurosci. (2010) 13:635–41. 10.1038/nn.2519
    1. Mazzone CM, Liang-Guallpa J, Li C, Wolcott NS, Boone MH, Southern M, et al. High-fat food biases hypothalamic and mesolimbic expression of consummatory drives. Nat Neurosci. (2020) 23, 1253–1266. 10.1038/s41593-020-0684-9
    1. Adams WK, Sussman JL, Kaur S, D’souza AM, Kieffer TJ, Winstanley CA. Long-term, calorie-restricted intake of a high-fat diet in rats reduces impulse control and ventral striatal D2 receptor signalling – two markers of addiction vulnerability. Eur J Neurosci. (2015) 42:3095–104. 10.1111/ejn.13117
    1. Hryhorczuk C, Sharma S, Fulton SE. Metabolic disturbances connecting obesity and depression. Front Neurosci. (2013) 7:177. 10.3389/fnins.2013.00177
    1. Hryhorczuk C, Florea M, Rodaros D, Poirier I, Daneault C, Des Rosiers C, et al. Dampened mesolimbic dopamine function and signaling by saturated but not monounsaturated dietary lipids. Neuropsychopharmacology. (2016) 41:811–21. 10.1038/npp.2015.207
    1. Diep TA, Madsen AN, Holst B, Kristiansen MM, Wellner N, Hansen SH, et al. Dietary fat decreases intestinal levels of the anorectic lipids through a fat sensor. FASEB J. (2011) 25:765–74. 10.1096/fj.10-166595
    1. Dipatrizio NV, Piomelli D. Intestinal lipid-derived signals that sense dietary fat. J Clin Invest. (2015) 125:891–8. 10.1172/JCI76302
    1. Laleh P, Yaser K, Abolfazl B, Shahriar A, Mohammad AJ, Nazila F, et al. Oleoylethanolamide increases the expression of PPAR-alpha and reduces appetite and body weight in obese people: a clinical trial. Appetite. (2018) 128:44–9. 10.1016/j.appet.2018.05.129
    1. Tutunchi H, Ostadrahimi A, Saghafi-Asl M, Roshanravan N, Shakeri-Bavil A, Asghari-Jafarabadi M, et al. Expression of NF-kappaB, IL-6, and IL-10 genes, body composition, and hepatic fibrosis in obese patients with NAFLD-Combined effects of oleoylethanolamide supplementation and calorie restriction: a triple-blind randomized controlled clinical trial. J Cell Physiol. (2021) 236:417–26. 10.1002/jcp.29870
    1. Kazemi M, Lalooha F, Nooshabadi MR, Haghighian HK. Decreased dysmenorrhea pain in girls by reducing oxidative stress and inflammatory biomarkers following supplementation with oleoylethanolamide: a randomized controlled trial. J Obstet Gynaecol Res. (2022) 48:1212–21. 10.1111/jog.15196
    1. Fan A, Wu X, Wu H, Li L, Huang R, Zhu Y, et al. Atheroprotective effect of oleoylethanolamide (OEA) targeting oxidized LDL. PLoS One. (2014) 9:e85337. 10.1371/journal.pone.0085337
    1. Yang L, Guo H, Li Y, Meng X, Yan L, Dan Z, et al. Oleoylethanolamide exerts anti-inflammatory effects on LPS-induced THP-1 cells by enhancing PPARalpha signaling and inhibiting the NF-kappaB and ERK1/2/AP-1/STAT3 pathways. Sci Rep. (2016) 6:34611. 10.1038/srep34611
    1. Tutunchi H, Naeini F, Saghafi-Asl M, Farrin N, Monshikarimi A, Ostadrahimi A. Effects of oleoylethanolamide supplementation on atherogenic indices and hematological parameters in patients with nonalcoholic fatty liver disease: a clinical trial. Health Promot Perspect. (2020) 10:373–82. 10.34172/hpp.2020.56
    1. Rodriguez De Fonseca F, Navarro M, Gomez R, Escuredo L, Nava F, Fu J, et al. An anorexic lipid mediator regulated by feeding. Nature. (2001) 414:209–12. 10.1038/35102582
    1. Begg DP, Woods SC. Hedonic and homeostatic overlap following fat ingestion. Cell Metab. (2013) 18:459–60. 10.1016/j.cmet.2013.09.012
    1. Fu J, Oveisi F, Gaetani S, Lin E, Piomelli D. Oleoylethanolamide, an endogenous PPAR-alpha agonist, lowers body weight and hyperlipidemia in obese rats. Neuropharmacology. (2005) 48:1147–53. 10.1016/j.neuropharm.2005.02.013
    1. Rondanelli M, Opizzi A, Solerte SB, Trotti R, Klersy C, Cazzola R. Administration of a dietary supplement (N-oleyl-phosphatidylethanolamine and epigallocatechin-3-gallate formula) enhances compliance with diet in healthy overweight subjects: a randomized controlled trial. Br J Nutr. (2009) 101:457–64. 10.1017/S0007114508024008
    1. Mangine GT, Gonzalez AM, Wells AJ, Mccormack WP, Fragala MS, Stout JR, et al. The effect of a dietary supplement (N-oleyl-phosphatidyl-ethanolamine and epigallocatechin gallate) on dietary compliance and body fat loss in adults who are overweight: a double-blind, randomized control trial. Lipids Health Dis. (2012) 11:127. 10.1186/1476-511X-11-127
    1. Dumas JA, Bunn JY, Nickerson J, Crain KI, Ebenstein DB, Tarleton EK, et al. Dietary saturated fat and monounsaturated fat have reversible effects on brain function and the secretion of pro-inflammatory cytokines in young women. Metabolism. (2016) 65:1582–8. 10.1016/j.metabol.2016.08.003
    1. Gilbert JR, Burger KS. Neuroadaptive processes associated with palatable food intake: present data and future directions. Curr Opin Behav Sci. (2016) 9:91–6. 10.1016/j.cobeha.2016.02.033
    1. Dalenberg JR, Patel BP, Denis R, Veldhuizen MG, Nakamura Y, Vinke PC, et al. Short-term consumption of sucralose with, but not without, carbohydrate impairs neural and metabolic sensitivity to sugar in humans. Cell Metab. (2020) 31:493–502e497. 10.1016/j.cmet.2020.01.014
    1. Thompson FE, Kirkpatrick SI, Subar AF, Reedy J, Schap TE, Wilson MM, et al. The national cancer institute’s dietary assessment primer: a resource for diet research. J Acad Nutr Diet. (2015) 115:1986–95. 10.1016/j.jand.2015.08.016
    1. Francis H, Stevenson R. Validity and test-retest reliability of a short dietary questionnaire to assess intake of saturated fat and free sugars: a preliminary study. J Hum Nutr Diet. (2013) 26:234–42. 10.1111/jhn.12008
    1. Ortega RM, Perez-Rodrigo C, Lopez-Sobaler AM. Dietary assessment methods: dietary records. Nutr Hosp. (2015) 31(Suppl. 3):38–45.
    1. Hovens IB, Dalenberg JR, Small DM. A brief neuropsychological battery for measuring cognitive functions associated with obesity. Obesity (Silver Spring). (2019) 27:1988–96. 10.1002/oby.22644
    1. D’ardenne K, Savage CR, Small D, Vainik U, Stoeckel LE. Core neuropsychological measures for obesity and diabetes trials: initial report. Front Psychol (2020) 11:554127. 10.3389/fpsyg.2020.554127
    1. Small DM. Dopamine adaptations as a common pathway for neurocognitive impairment in diabetes and obesity: a neuropsychological perspective. Front Neurosci. (2017) 11:134. 10.3389/fnins.2017.00134
    1. Sun X, Luquet S, Small DM. DRD2: bridging the genome and ingestive behavior. Trends Cogn Sci. (2017) 21:372–84. 10.1016/j.tics.2017.03.004
    1. Kehagia AA, Barker RA, Robbins TW. Cognitive impairment in Parkinson’s disease: the dual syndrome hypothesis. Neurodegener Dis. (2013) 11:79–92. 10.1159/000341998
    1. Lange KW, Robbins TW, Marsden CD, James M, Owen AM, Paul GM. L-dopa withdrawal in Parkinson’s disease selectively impairs cognitive performance in tests sensitive to frontal lobe dysfunction. Psychopharmacology (Berl). (1992) 107:394–404. 10.1007/BF02245167
    1. Elliott R, Sahakian BJ, Matthews K, Bannerjea A, Rimmer J, Robbins TW. Effects of methylphenidate on spatial working memory and planning in healthy young adults. Psychopharmacology (Berl). (1997) 131:196–206. 10.1007/s002130050284
    1. Waltz JA, Frank MJ, Wiecki TV, Gold JM. Altered probabilistic learning and response biases in schizophrenia: behavioral evidence and neurocomputational modeling. Neuropsychology. (2011) 25:86–97. 10.1037/a0020882
    1. Compher C, Frankenfield D, Keim N, Roth-Yousey L. Evidence Analysis Working Group. Best practice methods to apply to measurement of resting metabolic rate in adults: a systematic review. J Am Diet Assoc. (2006) 106:881–903. 10.1016/j.jada.2006.02.009
    1. Becker GM, Degroot MH, Marschak J. Measuring utility by a single-response sequential method. Behav Sci. (1964) 9:226–32. 10.1002/bs.3830090304
    1. Perszyk EE, Hutelin Z, Trinh J, Kanyamibwa A, Fromm S, Davis XS, et al. Fat and carbohydrate interact to potentiate food reward in healthy weight but not in overweight or obesity. Nutrients. (2021) 13:1203. 10.3390/nu13041203
    1. Difeliceantonio AG, Coppin G, Rigoux L, Edwin Thanarajah S, Dagher A, Tittgemeyer M, et al. Supra-additive effects of combining fat and carbohydrate on food reward. Cell Metab. (2018) 28:,33–44e33. 10.1016/j.cmet.2018.05.018
    1. Mason AE, Vainik U, Acree M, Tomiyama AJ, Dagher A, Epel ES, et al. Improving assessment of the spectrum of reward-related eating: the RED-13. Front Psychol. (2017) 8:795. 10.3389/fpsyg.2017.00795
    1. Mennella JA, Finkbeiner S, Reed DR. The proof is in the pudding: children prefer lower fat but higher sugar than do mothers. Int J Obes (Lond). (2012) 36:1285–91. 10.1038/ijo.2012.51
    1. Green BG, Dalton P, Cowart B, Shaffer G, Rankin K, Higgins J. Evaluating the ‘labeled magnitude scale’ for measuring sensations of taste and smell. Chem Senses. (1996) 21:323–34. 10.1093/chemse/21.3.323
    1. Lim J, Wood A, Green BG. Derivation and evaluation of a labeled hedonic scale. Chem Senses. (2009) 34:739–51. 10.1093/chemse/bjp054
    1. Beck AT, Steer RA, Ball R, Ranieri W. Comparison of Beck depression inventories -IA and -II in psychiatric outpatients. J Pers Assess. (1996) 67:588–97. 10.1207/s15327752jpa6703_13
    1. Craig CL, Marshall AL, Sjostrom M, Bauman AE, Booth ML, Ainsworth BE, et al. International physical activity questionnaire: 12-country reliability and validity. Med Sci Sports Exerc. (2003) 35:1381–95. 10.1249/01.MSS.0000078924.61453.FB
    1. Tutunchi H, Ostadrahimi A, Saghafi-Asl M, Hosseinzadeh-Attar MJ, Shakeri A, Asghari-Jafarabadi M, et al. Oleoylethanolamide supplementation in obese patients newly diagnosed with non-alcoholic fatty liver disease: effects on metabolic parameters, anthropometric indices, and expression of PPAR-alpha, UCP1, and UCP2 genes. Pharmacol Res. (2020) 156:104770. 10.1016/j.phrs.2020.104770
    1. Oveisi F, Gaetani S, Eng KT, Piomelli D. Oleoylethanolamide inhibits food intake in free-feeding rats after oral administration. Pharmacol Res. (2004) 49:461–6. 10.1016/j.phrs.2003.12.006
    1. Thabuis C, Destaillats F, Lambert DM, Muccioli GG, Maillot M, Harach T, et al. Lipid transport function is the main target of oral oleoylethanolamide to reduce adiposity in high-fat-fed mice. J Lipid Res. (2011) 52:1373–82. 10.1194/jlr.M013391
    1. Brownell KD. LEARN Program for Weight Management 2000. Dallas, TX: American Health Publishing Company; (2000).
    1. Wadden TA, Berkowitz RI, Womble LG, Sarwer DB, Phelan S, Cato RK, et al. Randomized trial of lifestyle modification and pharmacotherapy for obesity. N Engl J Med. (2005) 353:2111–20. 10.1056/NEJMoa050156
    1. Grilo CM, White MA, Masheb RM, Ivezaj V, Morgan PT, Gueorguieva R. Randomized controlled trial testing the effectiveness of adaptive “SMART” stepped-care treatment for adults with binge-eating disorder comorbid with obesity. Am Psychol. (2020) 75:204–18. 10.1037/amp0000534
    1. Babbs RK, Sun X, Felsted J, Chouinard-Decorte F, Veldhuizen MG, Small DM. Decreased caudate response to milkshake is associated with higher body mass index and greater impulsivity. Physiol Behav. (2013) 121:103–11. 10.1016/j.physbeh.2013.03.025
    1. Sun X, Kroemer NB, Veldhuizen MG, Babbs AE, De Araujo IE, Gitelman DR, et al. Basolateral amygdala response to food cues in the absence of hunger is associated with weight gain susceptibility. J Neurosci. (2015) 35:7964–76. 10.1523/JNEUROSCI.3884-14.2015
    1. Stice E, Spoor S, Bohon C, Small DM. Relation between obesity and blunted striatal response to food is moderated by TaqIA A1 allele. Science. (2008) 322:449–52. 10.1126/science.1161550
    1. Geha PY, Aschenbrenner K, Felsted J, O’malley SS, Small DM. Altered hypothalamic response to food in smokers. Am J Clin Nutr. (2013) 97:15–22. 10.3945/ajcn.112.043307
    1. Scheinost D, Noble S, Horien C, Greene AS, Lake EM, Salehi M, et al. Ten simple rules for predictive modeling of individual differences in neuroimaging. Neuroimage. (2019) 193:35–45. 10.1016/j.neuroimage.2019.02.057
    1. Allison PD. Handling Missing Data by Maximum Likelihood. Haverford, PA: Statistical Horizons; (2012).
    1. Smith SM. Fast robust automated brain extraction. Hum Brain Mapp. (2002) 17:143–55. 10.1002/hbm.10062
    1. Jenkinson M, Bannister P, Brady M, Smith S. Improved optimization for the robust and accurate linear registration and motion correction of brain images. Neuroimage. (2002) 17:825–41. 10.1016/s1053-8119(02)91132-8
    1. Smith SM, Jenkinson M, Woolrich MW, Beckmann CF, Behrens TE, Johansen-Berg H, et al. Advances in functional and structural MR image analysis and implementation as FSL. Neuroimage. (2004) 23(Suppl. 1):S208–19. 10.1016/j.neuroimage.2004.07.051
    1. Tzourio-Mazoyer N, Landeau B, Papathanassiou D, Crivello F, Etard O, Delcroix N, et al. Automated anatomical labeling of activations in SPM using a macroscopic anatomical parcellation of the MNI MRI single-subject brain. Neuroimage. (2002) 15:273–89.
    1. Shen X, Tokoglu F, Papademetris X, Constable RT. Groupwise whole-brain parcellation from resting-state fMRI data for network node identification. Neuroimage. (2013) 82:403–15. 10.1016/j.neuroimage.2013.05.081
    1. Joshi A, Scheinost D, Okuda H, Belhachemi D, Murphy I, Staib LH, et al. Unified framework for development, deployment and robust testing of neuroimaging algorithms. Neuroinformatics. (2011) 9:69–84. 10.1007/s12021-010-9092-8
    1. Shen X, Finn ES, Scheinost D, Rosenberg MD, Chun MM, Papademetris X, et al. Using connectome-based predictive modeling to predict individual behavior from brain connectivity. Nat Protoc. (2017) 12:506–18.
    1. Wager TD, Davidson ML, Hughes BL, Lindquist MA, Ochsner KN. Prefrontal-subcortical pathways mediating successful emotion regulation. Neuron. (2008) 59:1037–50. 10.1016/j.neuron.2008.09.006
    1. Leblanc ES, Patnode CD, Webber EM, Redmond N, Rushkin M, O’connor EA. Behavioral and pharmacotherapy weight loss interventions to prevent obesity-related morbidity and mortality in adults: updated evidence report and systematic review for the US preventive services task force. JAMA. (2018) 320:1172–91.
    1. Cazzola R, Rondanelli M. N-Oleoyl-phosphatidyl-ethanolamine and epigallo catechin-3-gallate mitigate oxidative stress in overweight and class I obese people on a low-calorie diet. J Med Food. (2020) 23:319–25. 10.1089/jmf.2019.0145
    1. Lam TKT. Neuronal regulation of homeostasis by nutrient sensing. Nat Med. (2010) 16:392–5. 10.1038/nm0410-392
    1. Bilbao A, Serrano A, Cippitelli A, Pavon FJ, Giuffrida A, Suarez J, et al. Role of the satiety factor oleoylethanolamide in alcoholism. Addict Biol. (2016) 21:859–72. 10.1111/adb.12276
    1. Grosshans M, Schwarz E, Bumb JM, Schaefer C, Rohleder C, Vollmert C, et al. Oleoylethanolamide and human neural responses to food stimuli in obesity. JAMA Psychiatry. (2014) 71:1254–61. 10.1001/jamapsychiatry.2014.1215
    1. Guzman M, Lo Verme J, Fu J, Oveisi F, Blazquez C, Piomelli D. Oleoylethanolamide stimulates lipolysis by activating the nuclear receptor peroxisome proliferator-activated receptor alpha (PPAR-alpha). J Biol Chem. (2004) 279:27849–54. 10.1074/jbc.M404087200
    1. Caillon A, Duszka K, Wahli W, Rohner-Jeanrenaud F, Altirriba J. The OEA effect on food intake is independent from the presence of PPARalpha in the intestine and the nodose ganglion, while the impact of OEA on energy expenditure requires the presence of PPARalpha in mice. Metabolism. (2018) 87:13–7. 10.1016/j.metabol.2018.06.005

Source: PubMed

3
Suscribir