Metabolic Determinants of Weight Gain in Humans

Paolo Piaggi, Paolo Piaggi

Abstract

One of the fundamental challenges in obesity research is to identify subjects prone to weight gain so that obesity and its comorbidities can be promptly prevented or treated. The principles of thermodynamics as applied to human body energetics demonstrate that susceptibility to weight gain varies among individuals as a result of interindividual differences in energy expenditure and energy intake, two factors that counterbalance one another and determine daily energy balance and, ultimately, body weight change. This review focuses on the variability among individuals in human metabolism that determines weight change. Conflicting results have been reported about the role of interindividual differences in energy metabolism during energy balance in relation to future weight change. However, recent studies have shown that metabolic responses to acute, short-term dietary interventions that create energy imbalance, such as low-protein overfeeding or fasting for 24 hours, may reveal the underlying metabolic phenotype that determines the degree of resistance to diet-induced weight loss or the propensity to spontaneous weight gain over time. Metabolically "thrifty" individuals, characterized by a predilection for saving energy in settings of undernutrition and dietary protein restriction, display a minimal increase in plasma fibroblast growth factor 21 concentrations in response to a low-protein overfeeding diet and tend to gain more weight over time compared with metabolically "spendthrift" individuals. Similarly, interindividual variability in the causal relationship between energy expenditure and energy intake ("energy sensing") and in the metabolic response to cold exposure (e.g., brown adipose tissue activation) seems, to some extent, to be indicative of individual propensity to weight gain. Thus, an increased understanding and the clinical characterization of phenotypic differences in energy metabolism among individuals (metabolic profile) may lead to new strategies to prevent weight gain or improve weight-loss interventions by targeted therapies on the basis of metabolic phenotype and susceptibility to obesity in individual persons.

Trial registration: ClinicalTrials.gov NCT00523627 NCT00687115.

Conflict of interest statement

Disclosure: The author declared no conflict of interest.

© 2019 The Obesity Society.

Figures

Figure 1.. Definition of thrifty and spendthrift…
Figure 1.. Definition of thrifty and spendthrift metabolic phenotypes.
Human metabolic phenotypes can be revealed by measuring the short-term (24 hours) response to fasting and overfeeding (especially, low-protein overfeeding) from conditions of energy balance. Compared to metabolically spendthrift individuals, subjects with a thrifty metabolism demonstrate a greater decrease in 24-h energy expenditure from energy balance during fasting and smaller increase in energy expenditure during overfeeding (upper-left panel), thus showing relatively lower metabolic rates in both dietary conditions (upper-right panel). Subjects with a thrifty metabolism lose less weight during sustained caloric restriction (lower-left panel) and gain more weight during prolonged overfeeding (lower-right panel).
Figure 2.. Variability in energy sensing and…
Figure 2.. Variability in energy sensing and its effects on weight change.
Positive linear relationship between energy intake and energy expenditure (EE) as determined by energy-sensing mechanisms (left graph). Inter-individual variability in the degree of energy sensing (i.e., over- vs. under-sensing) might explain the degree of susceptibility to weight gain or loss (right graph). Over the entire range of this energy-sensing relationship (e.g., low EE or high EE), there are subjects who normally sense their EE and eat accordingly (black circle), thus they can better maintain their weight over time. At any given point of the energy-sensing relationship, there are also subjects who over-sense their EE (solid square) and, as such, positively misconstrue their energy needs by consuming food as if they had higher EE (open circle), thus overeating relative to their energy needs and being more inclined to gain weight over time. Conversely, subjects that under-sense their EE (open square), eat less food than what they expend, thus are more prone to lose weight over time.

References

    1. Vinales KL, Begaye B, Bogardus C, Walter M, Krakoff J, Piaggi P. FGF21 is a Hormonal Mediator of the Human “Thrifty” Metabolic Phenotype. Diabetes 2018: db180696.
    1. Schlogl M, Piaggi P, Pannacciuli N, Bonfiglio SM, Krakoff J, Thearle MS. Energy Expenditure Responses to Fasting and Overfeeding Identify Phenotypes Associated With Weight Change. Diabetes 2015;64: 3680–3689.
    1. Rynders CA, Bergouignan A, Kealey E, Bessesen DH. Ability to adjust nocturnal fat oxidation in response to overfeeding predicts 5-year weight gain in adults. Obesity (Silver Spring) 2017;25: 873–880.
    1. Piaggi P, Thearle MS, Bogardus C, Krakoff J. Lower energy expenditure predicts long-term increases in weight and fat mass. The Journal of clinical endocrinology and metabolism 2013;98: E703–707.
    1. Vinales KL, Begaye B, Bogardus C, Walter M, Krakoff J, Piaggi P. FGF21 is a Hormonal Mediator of the Human “Thrifty” Metabolic Phenotype. Diabetes 2018.
    1. Lam YY, Ravussin E. Analysis of energy metabolism in humans: A review of methodologies. Molecular metabolism 2016;5: 1057–1071.
    1. Lam YY, Ravussin E. Indirect calorimetry: an indispensable tool to understand and predict obesity. Eur J Clin Nutr 2017;71: 318–322.
    1. Ravussin E, Lillioja S, Anderson TE, Christin L, Bogardus C. Determinants of 24-hour energy expenditure in man. Methods and results using a respiratory chamber. The Journal of clinical investigation 1986;78: 1568–1578.
    1. Weyer C, Snitker S, Rising R, Bogardus C, Ravussin E. Determinants of energy expenditure and fuel utilization in man: effects of body composition, age, sex, ethnicity and glucose tolerance in 916 subjects. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity 1999;23: 715–722.
    1. Keys A, Taylor HL, Grande F. Basal metabolism and age of adult man. Metabolism 1973;22: 579–587.
    1. Bosy-Westphal A, Eichhorn C, Kutzner D, Illner K, Heller M, Muller MJ. The age-related decline in resting energy expenditure in humans is due to the loss of fat-free mass and to alterations in its metabolically active components. The Journal of nutrition 2003;133: 2356–2362.
    1. Johnstone AM, Murison SD, Duncan JS, Rance KA, Speakman JR. Factors influencing variation in basal metabolic rate include fat-free mass, fat mass, age, and circulating thyroxine but not sex, circulating leptin, or triiodothyronine. The American journal of clinical nutrition 2005;82: 941–948.
    1. Toubro S, Sorensen TI, Ronn B, Christensen NJ, Astrup A. Twenty-four-hour energy expenditure: the role of body composition, thyroid status, sympathetic activity, and family membership. The Journal of clinical endocrinology and metabolism 1996;81: 2670–2674.
    1. Astrup A, Buemann B, Christensen NJ, Madsen J, Gluud C, Bennett P, et al. The contribution of body composition, substrates, and hormones to the variability in energy expenditure and substrate utilization in premenopausal women. The Journal of clinical endocrinology and metabolism 1992;74: 279–286.
    1. Piaggi P, Masindova I, Muller YL, Mercader J, Wiessner GB, Chen P, et al. A Genome-Wide Association Study Using a Custom Genotyping Array Identifies Variants in GPR158 Associated With Reduced Energy Expenditure in American Indians. Diabetes 2017;66: 2284–2295.
    1. Bogardus C, Lillioja S, Ravussin E, Abbott W, Zawadzki JK, Young A, et al. Familial dependence of the resting metabolic rate. The New England journal of medicine 1986;315: 96–100.
    1. Ravussin E, Lillioja S, Knowler WC, Christin L, Freymond D, Abbott WG, et al. Reduced rate of energy expenditure as a risk factor for body-weight gain. The New England journal of medicine 1988;318: 467–472.
    1. Bouchard C, Perusse L, Leblanc C, Tremblay A, Theriault G. Inheritance of the amount and distribution of human body fat. International journal of obesity 1988;12: 205–215.
    1. Zurlo F, Lillioja S, Esposito-Del Puente A, Nyomba BL, Raz I, Saad MF, et al. Low ratio of fat to carbohydrate oxidation as predictor of weight gain: study of 24-h RQ. The American journal of physiology 1990;259: E650–657.
    1. Tataranni PA, Harper IT, Snitker S, Del Parigi A, Vozarova B, Bunt J, et al. Body weight gain in free-living Pima Indians: effect of energy intake vs expenditure. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity 2003;27: 1578–1583.
    1. Roberts SB, Savage J, Coward WA, Chew B, Lucas A. Energy expenditure and intake in infants born to lean and overweight mothers. The New England journal of medicine 1988;318: 461–466.
    1. Griffiths M, Payne PR, Stunkard AJ, Rivers JP, Cox M. Metabolic rate and physical development in children at risk of obesity. Lancet 1990;336: 76–78.
    1. Hohenadel MG, Hollstein T, Thearle M, Reinhardt M, Piaggi P, Salbe AD, et al. A low resting metabolic rate in late childhood is associated with weight gain in adolescence. Metabolism: clinical and experimental 2019.
    1. Buscemi S, Verga S, Caimi G, Cerasola G. Low relative resting metabolic rate and body weight gain in adult Caucasian Italians. Int J Obes (Lond) 2005;29: 287–291.
    1. Luke A, Durazo-Arvizu R, Cao G, Adeyemo A, Tayo B, Cooper R. Positive association between resting energy expenditure and weight gain in a lean adult population. The American journal of clinical nutrition 2006;83: 1076–1081.
    1. Treuth MS, Butte NF, Sorkin JD. Predictors of body fat gain in nonobese girls with a familial predisposition to obesity. The American journal of clinical nutrition 2003;78: 1212–1218.
    1. Seidell JC, Muller DC, Sorkin JD, Andres R. Fasting respiratory exchange ratio and resting metabolic rate as predictors of weight gain: the Baltimore Longitudinal Study on Aging. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity 1992;16: 667–674.
    1. Weinsier RL, Nelson KM, Hensrud DD, Darnell BE, Hunter GR, Schutz Y. Metabolic predictors of obesity. Contribution of resting energy expenditure, thermic effect of food, and fuel utilization to four-year weight gain of post-obese and never-obese women. The Journal of clinical investigation 1995;95: 980–985.
    1. Anthanont P, Jensen MD. Does basal metabolic rate predict weight gain? The American journal of clinical nutrition 2016;104: 959–963.
    1. Amatruda JM, Statt MC, Welle SL. Total and resting energy expenditure in obese women reduced to ideal body weight. The Journal of clinical investigation 1993;92: 1236–1242.
    1. Marra M, Scalfi L, Covino A, Esposito-Del Puente A, Contaldo F. Fasting respiratory quotient as a predictor of weight changes in non-obese women. International journal of obesity 1998;22: 601–603.
    1. Roberts SB. Abnormalities of energy expenditure and the development of obesity. Obesity research 1995;3 Suppl 2: 155s–163s.
    1. Hall KD, Heymsfield SB, Kemnitz JW, Klein S, Schoeller DA, Speakman JR. Energy balance and its components: implications for body weight regulation. The American journal of clinical nutrition 2012;95: 989–994.
    1. Piaggi P, Vinales KL, Basolo A, Santini F, Krakoff J. Energy expenditure in the etiology of human obesity: spendthrift and thrifty metabolic phenotypes and energy-sensing mechanisms. Journal of endocrinological investigation 2018;41: 83–89.
    1. Schmidt SL, Kealey EH, Horton TJ, VonKaenel S, Bessesen DH. The effects of short-term overfeeding on energy expenditure and nutrient oxidation in obesity-prone and obesity-resistant individuals. Int J Obes (Lond) 2013;37: 1192–1197.
    1. Thearle MS, Pannacciulli N, Bonfiglio S, Pacak K, Krakoff J. Extent and determinants of thermogenic responses to 24 hours of fasting, energy balance, and five different overfeeding diets in humans. The Journal of clinical endocrinology and metabolism 2013;98: 2791–2799.
    1. Ravussin E, Lillioja S, Anderson TE, Christin L, Bogardus C. Determinants of 24-Hour Energy-Expenditure in Man - Methods and Results Using a Respiratory Chamber. Journal of Clinical Investigation 1986;78: 1568–1578.
    1. Ravussin E, Bogardus C. Relationship of genetics, age, and physical fitness to daily energy expenditure and fuel utilization. The American journal of clinical nutrition 1989;49: 968–975.
    1. Piaggi P, Krakoff J, Bogardus C, Thearle MS. Lower “awake and fed thermogenesis” predicts future weight gain in subjects with abdominal adiposity. Diabetes 2013: DB_130785.
    1. Secor SM. Specific dynamic action: a review of the postprandial metabolic response. J Comp Physiol B 2009;179: 1–56.
    1. Reed GW, Hill JO. Measuring the thermic effect of food. The American journal of clinical nutrition 1996;63: 164–169.
    1. de Jonge L, Bray GA. The thermic effect of food and obesity: a critical review. Obesity research 1997;5: 622–631.
    1. Schutz Y, Bessard T, Jequier E. Diet-induced thermogenesis measured over a whole day in obese and nonobese women. The American journal of clinical nutrition 1984;40: 542–552.
    1. D’Alessio DA, Kavle EC, Mozzoli MA, Smalley KJ, Polansky M, Kendrick ZV, et al. Thermic effect of food in lean and obese men. The Journal of clinical investigation 1988;81: 1781–1789.
    1. Kinabo JL, Durnin JV. Thermic effect of food in man: effect of meal composition, and energy content. The British journal of nutrition 1990;64: 37–44.
    1. Vinales KL, Schlogl M, Piaggi P, Hohenadel M, Graham A, Bonfiglio S, et al. The Consistency in Macronutrient Oxidation and the Role for Epinephrine in the Response to Fasting and Overfeeding. The Journal of clinical endocrinology and metabolism 2017;102: 279–289.
    1. Dulloo AG, Jacquet J. Low-protein overfeeding: a tool to unmask susceptibility to obesity in humans. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity 1999;23: 1118–1121.
    1. Bray GA, Smith SR, de Jonge L, Xie H, Rood J, Martin CK, et al. Effect of dietary protein content on weight gain, energy expenditure, and body composition during overeating: a randomized controlled trial. JAMA 2012;307: 47–55.
    1. Stock MJ. Gluttony and thermogenesis revisited. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity 1999;23: 1105–1117.
    1. Dulloo AG, Jacquet J, Miles-Chan JL, Schutz Y. Passive and active roles of fat-free mass in the control of energy intake and body composition regulation. Eur J Clin Nutr 2017;71: 353–357.
    1. Dulloo AG, Jacquet J, Girardier L. Poststarvation hyperphagia and body fat overshooting in humans: a role for feedback signals from lean and fat tissues. The American journal of clinical nutrition 1997;65: 717–723.
    1. Weyer C, Vozarova B, Ravussin E, Tataranni PA. Changes in energy metabolism in response to 48 h of overfeeding and fasting in Caucasians and Pima Indians. International journal of obesity and related metabolic disorders : journal of the International Association for the Study of Obesity 2001;25: 593–600.
    1. Reinhardt M, Thearle MS, Ibrahim M, Hohenadel MG, Bogardus C, Krakoff J, et al. A Human Thrifty Phenotype Associated With Less Weight Loss During Caloric Restriction. Diabetes 2015;64: 2859–2867.
    1. Schlogl M, Piaggi P, Thiyyagura P, Reiman EM, Chen K, Lutrin C, et al. Overfeeding over 24 hours does not activate brown adipose tissue in humans. The Journal of clinical endocrinology and metabolism 2013;98: E1956–1960.
    1. Hollstein T, Ando T, Basolo A, Krakoff J, Votruba SB, Piaggi P. Metabolic Response to Fasting Predicts Weight Gain During Low-Protein Overfeeding: Further Evidence for Spendthrift and Thrifty Metabolic Phenotypes. doi: 10.2139/ssrn.3275280.
    1. Laeger T, Henagan TM, Albarado DC, Redman LM, Bray GA, Noland RC, et al. FGF21 is an endocrine signal of protein restriction. The Journal of clinical investigation 2014;124: 3913–3922.
    1. Hill CM, Laeger T, Albarado DC, McDougal DH, Berthoud HR, Munzberg H, et al. Low protein-induced increases in FGF21 drive UCP1-dependent metabolic but not thermoregulatory endpoints. Scientific reports 2017;7: 8209.
    1. Laeger T, Albarado DC, Burke SJ, Trosclair L, Hedgepeth JW, Berthoud HR, et al. Metabolic Responses to Dietary Protein Restriction Require an Increase in FGF21 that Is Delayed by the Absence of GCN2. Cell Rep 2016;16: 707–716.
    1. Maida A, Zota A, Sjoberg KA, Schumacher J, Sijmonsma TP, Pfenninger A, et al. A liver stress-endocrine nexus promotes metabolic integrity during dietary protein dilution. The Journal of clinical investigation 2016;126: 3263–3278.
    1. Kharitonenkov A, Shiyanova TL, Koester A, Ford AM, Micanovic R, Galbreath EJ, et al. FGF-21 as a novel metabolic regulator. The Journal of clinical investigation 2005;115: 1627–1635.
    1. Ge X, Chen C, Hui X, Wang Y, Lam KS, Xu A. Fibroblast growth factor 21 induces glucose transporter-1 expression through activation of the serum response factor/Ets-like protein-1 in adipocytes. J Biol Chem 2011;286: 34533–34541.
    1. Heinitz S, Piaggi P, Yang S, Bonfiglio S, Steel J, Krakoff J, et al. Response of skeletal muscle UCP2-expression during metabolic adaptation to caloric restriction. Int J Obes (Lond) 2018;42: 974–984.
    1. Galgani JE, Moro C, Ravussin E. Metabolic flexibility and insulin resistance. American journal of physiology Endocrinology and metabolism 2008;295: E1009–1017.
    1. Goodpaster BH, Sparks LM. Metabolic Flexibility in Health and Disease. Cell metabolism 2017;25: 1027–1036.
    1. Kelley DE, Goodpaster B, Wing RR, Simoneau JA. Skeletal muscle fatty acid metabolism in association with insulin resistance, obesity, and weight loss. The American journal of physiology 1999;277: E1130–1141.
    1. Smith RL, Soeters MR, Wust RCI, Houtkooper RH. Metabolic flexibility as an adaptation to energy resources and requirements in health and disease. Endocrine reviews 2018.
    1. Galgani J, Ravussin E. Energy metabolism, fuel selection and body weight regulation. Int J Obes (Lond) 2008;32 Suppl 7: S109–119.
    1. Abbott WG, Howard BV, Christin L, Freymond D, Lillioja S, Boyce VL, et al. Short-term energy balance: relationship with protein, carbohydrate, and fat balances. The American journal of physiology 1988;255: E332–337.
    1. Begaye B, Vinales K, Ando T, Krakoff J, Piaggi P. Impaired Metabolic Flexibility to High-Fat Overfeeding Predicts Future Weight Gain in Humans. 2018 ObesityWeek: Nashville, 2018.
    1. Chen KY, Brychta RJ, Linderman JD, Smith S, Courville A, Dieckmann W, et al. Brown fat activation mediates cold-induced thermogenesis in adult humans in response to a mild decrease in ambient temperature. The Journal of clinical endocrinology and metabolism 2013;98: E1218–1223.
    1. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JM, Kemerink GJ, Bouvy ND, et al. Cold-activated brown adipose tissue in healthy men. The New England journal of medicine 2009;360: 1500–1508.
    1. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, et al. Identification and importance of brown adipose tissue in adult humans. The New England journal of medicine 2009;360: 1509–1517.
    1. Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren R, Niemi T, et al. Functional brown adipose tissue in healthy adults. The New England journal of medicine 2009;360: 1518–1525.
    1. Zingaretti MC, Crosta F, Vitali A, Guerrieri M, Frontini A, Cannon B, et al. The presence of UCP1 demonstrates that metabolically active adipose tissue in the neck of adult humans truly represents brown adipose tissue. Faseb J 2009;23: 3113–3120.
    1. Saito M, Okamatsu-Ogura Y, Matsushita M, Watanabe K, Yoneshiro T, Nio-Kobayashi J, et al. High incidence of metabolically active brown adipose tissue in healthy adult humans: effects of cold exposure and adiposity. Diabetes 2009;58: 1526–1531.
    1. Wijers SL, Saris WH, van Marken Lichtenbelt WD. Individual thermogenic responses to mild cold and overfeeding are closely related. The Journal of clinical endocrinology and metabolism 2007;92: 4299–4305.
    1. Hibi M, Oishi S, Matsushita M, Yoneshiro T, Yamaguchi T, Usui C, et al. Brown adipose tissue is involved in diet-induced thermogenesis and whole-body fat utilization in healthy humans. International journal of obesity 2016;40: 1655–1661.
    1. Peterson CM, Lecoultre V, Frost EA, Simmons J, Redman LM, Ravussin E. The thermogenic responses to overfeeding and cold are differentially regulated. Obesity (Silver Spring) 2016;24: 96–101.
    1. Marlatt KL, Ravussin E. Brown Adipose Tissue: an Update on Recent Findings. Curr Obes Rep 2017;6: 389–396.
    1. Blundell JE, Caudwell P, Gibbons C, Hopkins M, Naslund E, King N, et al. Role of resting metabolic rate and energy expenditure in hunger and appetite control: a new formulation. Disease models & mechanisms 2012;5: 608–613.
    1. Economos CD, Hatfield DP, King AC, Ayala GX, Pentz MA. Food and physical activity environments: an energy balance approach for research and practice. American journal of preventive medicine 2015;48: 620–629.
    1. Piaggi P, Vinales KL, Basolo A, Santini F, Krakoff J. Energy expenditure in the etiology of human obesity: spendthrift and thrifty metabolic phenotypes and energy-sensing mechanisms. Journal of endocrinological investigation 2017.
    1. Blundell JE, Dalton M, Gibbons C. Food intake and appetite in the aetiology of obesity. Advanced Nutrition and Dietetics in Obesity 2017: 97.
    1. Hall KD, Guo J. Obesity Energetics: Body Weight Regulation and the Effects of Diet Composition. Gastroenterology 2017;152: 1718–1727 e1713.
    1. Caudwell P, Finlayson G, Gibbons C, Hopkins M, King N, Naslund E, et al. Resting metabolic rate is associated with hunger, self-determined meal size, and daily energy intake and may represent a marker for appetite. The American journal of clinical nutrition 2013;97: 7–14.
    1. Weise CM, Hohenadel MG, Krakoff J, Votruba SB. Body composition and energy expenditure predict ad-libitum food and macronutrient intake in humans. Int J Obes (Lond) 2014;38: 243–251.
    1. McNeil J, Lamothe G, Cameron JD, Riou ME, Cadieux S, Lafreniere J, et al. Investigating predictors of eating: is resting metabolic rate really the strongest proxy of energy intake? The American journal of clinical nutrition 2017;106: 1206–1212.
    1. Piaggi P, Thearle MS, Krakoff J, Votruba SB. Higher Daily Energy Expenditure and Respiratory Quotient, Rather Than Fat-Free Mass, Independently Determine Greater ad Libitum Overeating. The Journal of clinical endocrinology and metabolism 2015;100: 3011–3020.
    1. Lam YY, Ravussin E. Variations in energy intake: it is more complicated than we think. The American journal of clinical nutrition 2017;106: 1169–1170.
    1. Edholm OG, Fletcher JG, Widdowson EM, McCance RA. The energy expenditure and food intake of individual men. The British journal of nutrition 1955;9: 286–300.
    1. Mayer J, Roy P, Mitra KP. Relation between caloric intake, body weight, and physical work: studies in an industrial male population in West Bengal. The American journal of clinical nutrition 1956;4: 169–175.
    1. Edholm OG, Adam JM, Healy MJ, Wolff HS, Goldsmith R, Best TW. Food intake and energy expenditure of army recruits. The British journal of nutrition 1970;24: 1091–1107.
    1. Edholm OG. Energy balance in man studies carried out by the Division of Human Physiology, National Institute for Medical Research. J Hum Nutr 1977;31: 413–431.
    1. Blundell JE, Caudwell P, Gibbons C, Hopkins M, Naslund E, King NA, et al. Body composition and appetite: fat-free mass (but not fat mass or BMI) is positively associated with self-determined meal size and daily energy intake in humans. The British journal of nutrition 2012;107: 445–449.
    1. Hopkins M, Finlayson G, Duarte C, Whybrow S, Ritz P, Horgan GW, et al. Modelling the associations between fat-free mass, resting metabolic rate and energy intake in the context of total energy balance. Int J Obes (Lond) 2016;40: 312–318.
    1. Basolo A, Votruba SB, Heinitz S, Krakoff J, Piaggi P. Deviations in energy sensing predict long-term weight change in overweight Native Americans. Metabolism: clinical and experimental 2018;82: 65–71.

Source: PubMed

3
Prenumerera