Obesity: an evolutionary context
John R. Speakman, Joel K. Elmquist
Obesity: an evolutionary context
People completely lacking body fat (lipodystrophy/lipoatrophy) and those with severe obesity both show profound metabolic and other health issues. Regulating levels of body fat somewhere between these limits would, therefore, appear to be adaptive. Two different models might be contemplated. More traditional is a set point (SP) where the levels are regulated around a fixed level. Alternatively, dual-intervention point (DIP) is a system that tolerates fairly wide variation but is activated when critically high or low levels are breached. The DIP system seems to fit our experience much better than an SP, and models suggest that it is more likely to have evolved. A DIP system may have evolved because of two contrasting selection pressures. At the lower end, we may have been selected to avoid low levels of fat as a buffer against starvation, to avoid disease-induced anorexia, and to support reproduction. At the upper end, we may have been selected to avoid excess storage because of the elevated risks of predation. This upper limit of control seems to have malfunctioned because some of us deposit large fat stores, with important negative health effects. Why has evolution not protected us against this problem? One possibility is that the protective system slowly fell apart due to random mutations after we dramatically reduced the risk of being predated during our evolutionary history. By chance, it fell apart more in some people than others, and these people are now unable to effectively manage their weight in the face of the modern food glut. To understand the evolutionary context of obesity, it is important to separate the adaptive reason for storing some fat (i.e. the lower intervention point), from the nonadaptive reason for storing lots of fat (a broken upper intervention point). The DIP model has several consequences, showing how we understand the obesity problem and what happens when we attempt to treat it.
obesity / BMI / body fat / adiposity / evolution / selection / adaptive / leptin resistance / metabolic programming / dual-intervention point model / set-point model
[1] |
Lawrence R. Lipodystrophy and hepatomegaly with diabetes, lipaemia, and other metabolic disturbances: a case throwing new light on the action of insulin. Lancet 1946; 247: 724- 31.
|
[2] |
Garg A. Lipodystrophies. Am J Med 2000; 108: 143- 52.
|
[3] |
Gavrilova O, Marcus-Samuels B, Graham D et al. Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice. J Clin Invest 2000; 105: 271- 8.
|
[4] |
Chan JM, Rimm EB, Colditz GA et al. Obesity, fat distribution, and weight gain as risk factors for clinical diabetes in men. Diabetes Care 1994; 17: 961- 9.
|
[5] |
Field AE, Coakley EH, Must A et al. Impact of overweight on the risk of developing common chronic diseases during a 10-year period. Arch Intern Med 2001; 161: 1581- 6.
|
[6] |
Kennedy GC. The role of depot fat in the hypothalamic control of food intake in the rat. Proc R Soc Lond B Biol Sci 1953; 140: 578- 96.
|
[7] |
Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature 1998; 395: 763- 70.
|
[8] |
Levitsky DA. Putting behavior back into feeding behavior: a tribute to George Collier. Appetite 2002; 38: 143- 8.
|
[9] |
Speakman JR. A nonadaptive scenario explaining the genetic predisposition to obesity: the “predation release” hypothesis. Cell Metab 2007; 6: 5- 12.
|
[10] |
Speakman JR, Levitsky DA, Allison DB et al. Set points, settling points and some alternative models: theoretical options to understand how genes and environments combine to regulate body adiposity. Dis Model Mech 2011; 4: 733- 45.
|
[11] |
Allison DB, Kaprio J, Korkeila M et al. The heritability of body mass index among an international sample of monozygotic twins reared apart. Int J Obes Relat Metab Disord 1996; 20: 501- 6.
|
[12] |
Leibel RL, Rosenbaum M, Hirsch J. Changes in energy expenditure resulting from altered body weight. N Engl J Med 1995; 332: 621- 8.
|
[13] |
Schwartz MW, Woods SC, Porte D et al. Central nervous system control of food intake. Nature 2000; 404: 661- 71.
|
[14] |
Morton GJ, Cummings DE, Baskin DG et al. Central nervous system control of food intake and body weight. Nature 2006; 443: 289- 95.
|
[15] |
Zhang Y, Proenca R, Maffei M et al. Positional cloning of the mouse obese gene and its human homologue. Nature 1994; 372: 425- 32.
|
[16] |
Berthoud HR, Albaugh VL, Neuhuber WL. Gut-brain communication and obesity: understanding functions of the vagus nerve. J Clin Invest 2021; 131: e143770.
|
[17] |
Browning KN, Carson KE. Central neurocircuits regulating food intake in response to gut inputs-preclinical evidence. Nutrients 2021; 13: 908.
|
[18] |
Myers MG, Affinati AH, Richardson N et al. Central nervous system regulation of organismal energy and glucose homeostasis. Nat Metab 2021; 3: 1033.
|
[19] |
Tartaglia LA, Dembski M, Weng X et al. Identification and expression cloning of a leptin receptor, OB-R. Cell 1995; 83: 1263- 71.
|
[20] |
Lee GH, Proenca R, Montez JM et al. Abnormal splicing of the leptin receptor in diabetic mice. Nature 1996; 379: 632- 5.
|
[21] |
Chua SC Jr, Chung WK, Wu-Peng XS et al. Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (leptin) receptor. Science 1996; 271: 994- 6.
|
[22] |
Mountjoy KG, Robbins LS, Mortrud MT et al. The cloning of a family of genes that encode the melanocortin receptors. Science 1992; 257: 1248- 51.
|
[23] |
Caron A, Dungan Lemko HM, Castorena CM et al. POMC neurons expressing leptin receptors coordinate metabolic responses to fasting via suppression of leptin levels. Elife 2018; 7: e33710.
|
[24] |
Caron A, Michael NJ. New horizons: is obesity a disorder of neurotransmission? J Clin Endocrinol Metab 2021; 106: e4872- 86.
|
[25] |
Cone RD. Anatomy and regulation of the central melanocortin system. Nat Neurosci 2005; 8: 571- 8.
|
[26] |
Adan RA, Tiesjema B, Hillebrand JJ et al. The MC4 receptor and control of appetite. Br J Pharmacol 2006; 149: 815- 27.
|
[27] |
Girardet C, Butler AA. Neural melanocortin receptors in obesity and related metabolic disorders. Biochim Biophys Acta 2014; 1842: 482- 94.
|
[28] |
Krashes MJ, Lowell BB, Garfield AS. Melanocortin-4 receptor-regulated energy homeostasis. Nat Neurosci 2016; 19: 206- 19.
|
[29] |
Barsh GS, Farooqi IS, O’Rahilly S. Genetics of body-weight regulation. Nature 2000; 404: 644- 51.
|
[30] |
Wade KH, Lam BYH, Melvin A et al. Loss-of-function mutations in the melanocortin 4 receptor in a UK birth cohort. Nat Med 2021; 27: 1088- 96.
|
[31] |
Yeo GSH, Chao DHM, Siegert AM et al. The melanocortin pathway and energy homeostasis: From discovery to obesity therapy. Mol Metab 2021; 48: 101206.
|
[32] |
Bultman SJ, Michaud EJ, Woychik RP. Molecular characterization of the mouse agouti locus. Cell 1992; 71: 1195- 204.
|
[33] |
Spanswick D, Smith MA, Groppi VE,
|
[34] |
Cowley MA, Smart JL, Rubinstein M et al. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 2001; 411: 480- 4.
|
[35] |
Schwartz MW, Seeley RJ, Campfield LA et al. Identification of targets of leptin action in rat hypothalamus. J Clin Invest 1996; 98: 1101- 6.
|
[36] |
Gropp E, Shanabrough M, Borok E et al. Agouti-related peptide-expressing neurons are mandatory for feeding. Nat Neurosci 2005; 8: 1289- 91.
|
[37] |
Luquet S, Perez FA, Hnasko TS et al. NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 2005; 310: 683- 5.
|
[38] |
Xu J, Bartolome CL, Low CS et al. Genetic identification of leptin neural circuits in energy and glucose homeostases. Nature 2018; 556: 505- 9.
|
[39] |
Elmquist JK, Bjørbæk C, Ahima RS et al. Distributions of leptin receptor mRNA isoforms in the rat brain. J Comp Neurol 1998; 395: 535- 47.
|
[40] |
Scott MM, Lachey JL, Sternson SM et al. Leptin targets in the mouse brain. J Comp Neurol 2009; 514: 518- 32.
|
[41] |
Shimazu T, Fukuda A, Ban T. Reciprocal influences of the ventromedial and lateral hypothalamic nuclei on blood glucose level and liver glycogen content. Nature 1966; 210: 1178- 9.
|
[42] |
Shimazu T. Innervation of the liver and glucoregulation: roles of the hypothalamus and autonomic nerves. Nutrition 1996; 12: 65- 6.
|
[43] |
Hetherington AW, Ranson SW. Hypothalamic lesions and adiposity in the rat. Anat Rec 1940; 78: 149- 72.
|
[44] |
Caron A, Lee S, Elmquist JK,
|
[45] |
Cheung CC, Kurrasch DM, Liang JK et al. Genetic labeling of steroidogenic factor-1 (SF-1) neurons in mice reveals ventromedial nucleus of the hypothalamus (VMH) circuitry beginning at neurogenesis and development of a separate non-SF-1 neuronal cluster in the ventrolateral VMH. J Comp Neurol 2013; 521: 1268- 88.
|
[46] |
Ikeda Y, Luo X, Abbud R et al. The nuclear receptor steroidogenic factor-1 is essential for the formation of the ventromedial hypothalamic nucleus. Mol Endocrinol 1995; 9: 478- 86.
|
[47] |
Tran PV, Lee MB, Marín O et al. Requirement of the orphan nuclear receptor SF-1 in terminal differentiation of ventromedial hypothalamic neurons. Mol Cell Neurosci 2003; 22: 441- 53.
|
[48] |
Fujikawa T, Castorena CM, Pearson M et al. SF-1 expression in the hypothalamus is required for beneficial metabolic effects of exercise. Elife 2016; 5: e18206.
|
[49] |
Kim KW, Zhao L, Donato J et al. Steroidogenic factor 1 directs programs regulating diet-induced thermogenesis and leptin action in the ventral medial hypothalamic nucleus. Proc Natl Acad Sci USA 2011; 108: 10673- 8.
|
[50] |
Majdic G, Young M, Gomez-Sanchez E et al. Knockout mice lacking steroidogenic factor 1 are a novel genetic model of hypothalamic obesity. Endocrinology 2002; 143: 607- 14.
|
[51] |
Coutinho EA, Okamoto S, Ishikawa AW et al. Activation of SF1 neurons in the ventromedial hypothalamus by DREADD technology increases insulin sensitivity in peripheral tissues. Diabetes 2017; 66: 2372- 86.
|
[52] |
Haque MS, Minokoshi Y, Hamai M et al. Role of the sympathetic nervous system and insulin in enhancing glucose uptake in peripheral tissues after intrahypothalamic injection of leptin in rats. Diabetes 1999; 48: 1706- 12.
|
[53] |
Labbé SM, Caron A, Lanfray D et al. Hypothalamic control of brown adipose tissue thermogenesis. Front Syst Neurosci 2015; 9: 150.
|
[54] |
Minokoshi Y, Haque MS, Shimazu T. Microinjection of leptin into the ventromedial hypothalamus increases glucose uptake in peripheral tissues in rats. Diabetes 1999; 48: 287- 91.
|
[55] |
Xu Y, Lu Y, Xu P et al. VMAT2-mediated neurotransmission from midbrain leptin receptor neurons in feeding regulation. eNeuro 2017; 4: ENEURO.0083- 17.2017.
|
[56] |
Bingham NC, Anderson KK, Reuter AL et al. Selective loss of leptin receptors in the ventromedial hypothalamic nucleus results in increased adiposity and a metabolic syndrome. Endocrinology 2008; 149: 2138- 48.
|
[57] |
Dhillon H, Zigman JM, Ye C et al. Leptin directly activates SF1 neurons in the VMH, and this action by leptin is required for normal body-weight homeostasis. Neuron 2006; 49: 191- 203.
|
[58] |
Yu KB, Hsiao EY. Roles for the gut microbiota in regulating neuronal feeding circuits. J Clin Invest 2021; 131: e143772.
|
[59] |
Ahima RS, Flier JS. Leptin. Annu Rev Physiol 2000; 62: 413- 37.
|
[60] |
Ahima RS, Kelly J, Elmquist JK et al. Distinct physiologic and neuronal responses to decreased leptin and mild hyperleptinemia. Endocrinology 1999; 140: 4923- 31.
|
[61] |
Ahima RS, Prabakaran D, Mantzoros C et al. Role of leptin in the neuroendocrine response to fasting. Nature 1996; 382: 250- 2.
|
[62] |
Hummel KP, Dickie MM, Coleman DL. Diabetes, a new mutation in mouse. Science 1966; 153: 1127- 8.
|
[63] |
Ingalls AM, Dickie MM, Snell GD. Obese, a new mutation in the house mouse. J Hered 1950; 41: 317- 8.
|
[64] |
Montague CT, Farooqi IS, Whitehead JP et al. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 1997; 387: 903- 8.
|
[65] |
Campfield LA, Smith FJ, Burn P. The OB protein (leptin) pathway—a link between adipose tissue mass and central neural networks. Horm Metab Res 1996; 28: 619- 32.
|
[66] |
Halaas JL, Gajiwala KS, Maffei M et al. Weight-reducing effects of the plasma protein encoded by the obese gene. Science 1995; 269: 543- 6.
|
[67] |
Pelleymounter MA, Cullen MJ, Baker MB et al. Effects of the obese gene-product on body-weight regulation in ob/ob mice. Science 1995; 269: 540- 3.
|
[68] |
Farooqi IS, Jebb SA, Langmack G et al. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. New Engl J Med 1999; 341: 879- 84.
|
[69] |
Halaas JL, Boozer C, Blair-West J et al. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc Natl Acad Sci USA 1997; 94: 8878- 83.
|
[70] |
Heymsfield SB, Greenberg AS, Fujioka K et al. Recombinant leptin for weight loss in obese and lean adults—A randomized, controlled, dose-escalation trial. JAMA 1999; 282: 1568- 75.
|
[71] |
Bjørbæk C, Elmquist JK, Frantz JD et al. Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol Cell 1998; 1: 619- 25.
|
[72] |
Myers MG, Cowley MA, Münzberg H. Mechanisms of leptin action and leptin resistance. Annu Rev Physiol 2008; 70: 537- 56.
|
[73] |
Friedman JM. Leptin and the endocrine control of energy balance. Nat Metab 2019; 1: 754- 64.
|
[74] |
Friedman JM. The function of leptin in nutrition, weight, and physiology. Nutr Rev 2002; 60: S1- 14.
|
[75] |
Zhao S, Kusminski CM, Elmquist JK et al. Leptin: less is more. Diabetes 2020; 69: 823- 9.
|
[76] |
Yanovski JA, Yanovski SZ, Sovik KN et al. A prospective study of holiday weight gain. New Engl J Med 2000; 342: 861- 7.
|
[77] |
Visscher TLS, Seidell JC. Time trends (1993-1997) and seasonal variation in body mass index and waist circumference in the Netherlands. Int J Obes Relat Metab Disord 2004; 28: 1309- 16.
|
[78] |
MacKay EM, Bergman H. The relation between glycogen and water storage in the liver. J Biol Chem 1932; 96: 373- 80.
|
[79] |
Vidal-Puig A. Adipose tissue expandability, lipotoxicity and the metabolic syndrome. Endocrinol Nutr 2013; 60: 39- 43.
|
[80] |
Ghaben AL, Scherer PE. Adipogenesis and metabolic health. Nat Rev Mol Cell Biol 2019; 20: 242- 58.
|
[81] |
Kim JY, van de Wall E, Laplante M et al. Obesity-associated improvements in metabolic profile through expansion of adipose tissue. J Clin Invest 2007; 117: 2621- 37.
|
[82] |
Neel JV. Diabetes mellitus: a “thrifty” genotype rendered detrimental by “progress”? Am J Hum Genet 1962; 14: 353- 62.
|
[83] |
Speakman JR. Thrifty genes for obesity, an attractive but flawed idea, and an alternative perspective: the ‘drifty gene’ hypothesis. Int J Obes (Lond) 2008; 32: 1611- 7.
|
[84] |
Neel JV. The “thrifty genotype” in 1998. Nutr Rev 1999; 57: S2- S9.
|
[85] |
Krugman P. Arguing with Zombies: Economics, Politics, and the Fight for a Better Future. New York: WW Norton & Company, 2020.
|
[86] |
Speakman JR. The genetics of obesity: five fundamental problems with the famine hypothesis. In: Fantuzzi G, Mazzone T (eds) Adipose Tissue and Adipokines in Health and Disease. New York: Humana Press, 2006.
|
[87] |
Speakman JR. Gene environment interactions and the origin of the modern obesity epidemic: a novel ‘non-adaptive’ scenario. In: Kirkman TC, Cooper SJ (eds) Appetite and Body Weight: Integrative Systems and the Development of Anti-obesity Drugs. New York: Elsevier, 2006.
|
[88] |
Speakman JR. Evolutionary perspectives on the obesity epidemic: adaptive, maladaptive, and neutral viewpoints. Annu Rev Nutr 2013; 33: 289- 317.
|
[89] |
Speakman JR. If body fatness is under physiological regulation, then how come we have an obesity epidemic? Physiology 2014; 29: 88- 98.
|
[90] |
Speakman JR. The evolution of body fatness: trading off disease and predation risk. J Exp Biol 2018; 221: jeb167254.
|
[91] |
Ben-Dor M, Gopher A, Hershkovitz I et al. Man the fat hunter: the demise of Homo erectus and the emergence of a new hominin lineage in the Middle Pleistocene (ca. 400 kyr) levant. PLoS One 2011; 6: e28689.
|
[92] |
Ben-Dor M, Sirtoli R, Barkai R. The evolution of the human trophic level during the Pleistocene. Am J Phys Anthropol 2021; 175: 27- 56.
|
[93] |
Martin PS, Klein R. Prehistoric overkill: the global model. In: Martin PS, Klein R (eds) Quaternary Extinctions: a Prehistoric Revolution. Arizona: University of Arizona Press, 1984, 354- 403.
|
[94] |
Surovell T, Waguespack N, Brantingham PJ. Global archaeological evidence for proboscidean overkill. Proc Natl Acad Sci USA 2005; 102: 6231- 6.
|
[95] |
Carmody RN, Wrangham RW. The energetic significance of cooking. J Hum Evol 2009; 57: 379- 91.
|
[96] |
Debruyne R, Chu G, King CE et al. Out of America: ancient DNA evidence for a new world origin of late quaternary woolly mammoths. Curr Biol 2008; 18: 1320- 6.
|
[97] |
Benyshek DC, Watson JT. Exploring the thrifty genotype’s food-shortage assumptions: a cross-cultural comparison of ethnographic accounts of food security among foraging and agricultural societies. Am J Phys Anthropol 2006; 131: 120- 6.
|
[98] |
Elia M. Hunger disease. Clin Nutr 2000; 19: 379- 86.
|
[99] |
Prentice AM. Obesity and its potential mechanistic basis. Br Med Bull 2001; 60: 51- 67.
|
[100] |
Prentice AM, Hennig BJ, Fulford AJ. Evolutionary origins of the obesity epidemic: natural selection of thrifty genes or genetic drift following predation release? Int J Obes (Lond) 2008; 32: 1607- 10.
|
[101] |
Bersaglieri T, Sabeti PC, Patterson N et al. Genetic signatures of strong recent positive selection at the lactase gene. Am J Hum Genet 2004; 74: 1111- 20.
|
[102] |
Speakman JR. Sex- and age-related mortality profiles during famine: testing the ‘body fat’ hypothesis. J Biosoc Sci 2013; 45: 823- 40.
|
[103] |
Bindon JR, Baker PT. Bergmann’s rule and the thrifty genotype. Am J Phys Anthropol 1997; 104: 201- 10.
|
[104] |
Gosling AL, Buckley HR, Matisoo-Smith E et al. Pacific populations, metabolic disease and ‘just-so stories’: a critique of the ‘thrifty genotype’ hypothesis in Oceania. Ann Hum Genet 2015; 79: 470- 80.
|
[105] |
Diamond J. The double puzzle of diabetes.Nature 2003; 423: 599- 602.
|
[106] |
Houghton P. People of the great ocean, aspects of human biology of the early Pacific. Cambridge: Cambridge University Press, 1996.
|
[107] |
Furusawa T, Naka I, Yamauchi T et al. The Q223R polymorphism in LEPR is associated with obesity in Pacific Islanders. Hum Genet 2010; 127: 287- 94.
|
[108] |
Minster RL, Hawley NL, Su CT et al. A thrifty variant in CREBRF strongly influences body mass index in Samoans. Nat Genetics 2016; 48: 1049- 54.
|
[109] |
Hanson RL, Safabakhsh S, Curtis JM et al. Association of CREBRF variants with obesity and diabetes in Pacific Islanders from Guam and Saipan. Diabetologia 2019; 62: 1647- 52.
|
[110] |
Arslanian KJ, Fidow UT, Atanoa T et al. A missense variant in CREBRF, rs373863828, is associated with fat-free mass, not fat mass in Samoan infants. Int J Obes (Lond) 2021; 45: 45- 55.
|
[111] |
Foley C. Don’t eat for Winter: Unlock Nature’s Secret to Reveal Your True Body. Upthedeise Enterprises, 2017.
|
[112] |
Speakman, JR. The cost of living: field metabolic rates of small mammals. Adv Ecol Res 1999; 30: 177- 297.
|
[113] |
Humphries MM, Boutin S, Thomas DW et al. Expenditure freeze: the metabolic response of small mammals to cold environments. Ecol Lett 2005; 8: 1326- 33.
|
[114] |
Speakman JR, Chi Q, Oldakowski L et al. Surviving winter on the Qinghai-Tibetan Plateau: Pikas suppress energy demands and exploit yak feces to survive winter. Proc Natl Acad Sci USA 2021; 118: e2100707118.
|
[115] |
Trondrud LM, Pigeon G, Albon S et al. Determinants of heart rate in Svalbard reindeer reveal mechanisms of seasonal energy management. Philos Trans R Soc Lond B Biol Sci 2021; 376: 20200215.
|
[116] |
Król E, Johnson MS, Speakman JR. Limits to sustained energy intake VIII. Resting metabolic rate and organ morphology of laboratory mice lactating at thermoneutrality. J Exp Biol 2003; 206: 4283- 91.
|
[117] |
Krol E, Redman P, Thomson PJ et al. Effect of photoperiod on body mass, food intake and body composition in the field vole, Microtus agrestis. J Exp Biol 2005; 208: 571- 84.
|
[118] |
Król E, Speakman JR. Regulation of body mass and adiposity in the field vole, Microtus agrestis: a model of leptin resistance. J Endocrinol 2007; 192: 271- 8.
|
[119] |
Zinkel SRJ, Moe M 3rd, Stern EA et al. Comparison of total energy expenditure between school and summer months. Pediatr Obes 2013; 8: 404- 10.
|
[120] |
Westerterp KR. Seasonal variation in body mass, body composition and activity-induced energy expenditure: a long-term study. Eur J Clin Nutr 2020; 74: 135- 40.
|
[121] |
Mellars P, French JC. Tenfold population increase in Western Europe at the Neandertal-to-modern human transition. Science 2011; 333: 623- 7.
|
[122] |
Flegal KM, Carroll MD, Ogden CL et al. Prevalence and trends in obesity among US adults, 1999-2008. JAMA 2010; 303: 235- 41.
|
[123] |
Humphries MM, Kramer DL, Thomas DW. The role of energy availability in mammalian hibernation: an experimental test in free-ranging eastern chipmunks.Physiol Biochem Zool 2003; 76: 180- 6.
|
[124] |
Pauls RW. Body temperature dynamics of the red squirrel (Tamiasciurus-Hudsonicus): adaptations for energy conservation. Can J Zool 1979; 57: 1349- 54.
|
[125] |
Dearing MD. The function of haypiles of pikas (Ochotona princeps). J Mammal 1997; 78: 1156- 63.
|
[126] |
Fletcher QE, Landry-Cuerrier M, Boutin S et al. Reproductive timing and reliance on hoarded capital resources by lactating red squirrels. Oecologia 2013; 173: 1203- 15.
|
[127] |
Corrales-Medina VF, Valayam J, Serpa JA et al. The obesity paradox in community-acquired bacterial pneumonia. Int J Infect Dis 2011; 15: e54- 7.
|
[128] |
Hanrahan CF, Golub JE, Mohapi L et al. Body mass index and risk of tuberculosis and death. AIDS 2010; 24: 1501- 8.
|
[129] |
van der Sande MAB, Schim van der Loeff MF, Aveika AA et al. Body mass index at time of HIV diagnosis—a strong and independent predictor of survival. J Acquir Immune Defic Syndr 2004; 37: 1288- 94.
|
[130] |
Popkin BM, Du S, Green WD et al. Individuals with obesity and COVID-19: a global perspective on the epidemiology and biological relationships. Obes Rev 2020; 21: e13128.
|
[131] |
Subbarao K, Mahanty S. Respiratory virus infections: understanding COVID-19. Immunity 2020; 52: 905- 9.
|
[132] |
Arentz M, Yim E, Klaff L et al. Characteristics and outcomes of 21 critically ill patients with COVID-19 in Washington State. JAMA 2020; 323: 1612- 4.
|
[133] |
Grasselli G, Pesenti A, Cecconi M. Critical care utilization for the COVID-19 outbreak in Lombardy, Italy: early experience and forecast during an emergency response. JAMA 2020; 323: 1545- 6.
|
[134] |
Guan W, Ni Z, Hu Y et al. Clinical characteristics of Coronavirus disease 2019 in China. N Engl J Med 2020; 382: 1708- 20.
|
[135] |
Koutsakos M, Kedzierska K. A race to determine what drives COVID-19 severity. Nature 2020; 583: 366- 8.
|
[136] |
Onder G, Rezza G, Brusaferro S. Case-fatality rate and characteristics of patients dying in relation to COVID-19 in Italy. JAMA 2020; 323: 1775- 6.
|
[137] |
Porcheddu R, Serra C, Kelvin D et al. Similarity in case fatality rates (CFR) of COVID-19/SARS-COV-2 in Italy and China. J Infect Dev Ctries 2020; 14: 125- 8.
|
[138] |
Ruan Q, Yang K, Wang W et al. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med 2020; 46: 1294- 7.
|
[139] |
Williamson EJ, Walker AJ, Bhaskaran K et al. Factors associated with COVID-19-related death using OpenSAFELY.Nature 2020; 584: 430- 6.
|
[140] |
Wu C, Chen X, Cai Y. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med 2020; 180: 934- 43.
|
[141] |
Wang S, Ma P, Zhang S et al. Fasting blood glucose at admission is an independent predictor for 28-day mortality in patients with COVID-19 without previous diagnosis of diabetes: a multi-centre retrospective study. Diabetologia 2020; 63: 2102- 11.
|
[142] |
Kim F, Pham M, Maloney E et al. Vascular inflammation, insulin resistance, and reduced nitric oxide production precede the onset of peripheral insulin resistance. Arterioscler Thromb Vasc 2008; 28: 1982- 8.
|
[143] |
Fadini GP, Morieri ML, Longato E et al. Prevalence and impact of diabetes among people infected with SARS-CoV-2. J Endocrinol Invest 2020; 43: 867- 9.
|
[144] |
Gao M, Piernas C, Astbury NM et al. Associations between bodymass index and COVID-19 severity in 6.9 million people in England: a prospective, community-based, cohort study. Lancet Diabetes Endocrinol 2021; 9: 350- 9.
|
[145] |
Zihlman AL, Bolter DR. Body composition in Pan paniscus compared with Homo sapiens has implications for changes during human evolution. Proc Natl Acad Sci USA 2015; 112: 7466- 71.
|
[146] |
Araújo PM, Viegas I, Rocha AD et al. Understanding how birds rebuild fat stores during migration: insights from an experimental study. Sci Rep 2019; 9: 10065.
|
[147] |
Speakman JR. The physiological costs of reproduction in small mammals. Philos Trans R Soc Lond B Biol Sci 2008; 363: 375- 98.
|
[148] |
Chehab FF, Lim ME, Lu R. Correction of the sterility defect in homozygous obese female mice by treatment with the human recombinant leptin. Nat Genet 1996; 12: 318- 20.
|
[149] |
Chan JL, Mantzoros CS. Role of leptin in energy-deprivation states: normal human physiology and clinical implications for hypothalamic amenorrhoea and anorexia nervosa. Lancet 2005; 366: 74- 85.
|
[150] |
Welt CK, Chan JL, Bullen J et al. Recombinant human leptin in women with hypothalamic amenorrhea. New Engl J Med 2004; 351: 987- 97.
|
[151] |
Heldstab SA, van Schaik CP, Isler K. Being fat and smart: A comparative analysis of the fat-brain trade-off in mammals. J Hum Evol 2016; 100: 25- 34.
|
[152] |
Pontzer H, Yamada Y, Sagayama H et al. Daily energy expenditure through the human life course. Science 2021; 373: 808- 12.
|
[153] |
Speakman JR, Westerterp KR. A mathematical model of weight loss under total starvation: evidence against the thrifty-gene hypothesis. Dis Model Mech 2013; 6: 236- 51.
|
[154] |
Houston AI, Mcnamara JM. A theoretical investigation of the fat reserves and mortality levels of small birds in winter. Ornis Scand 1993; 24: 205- 19.
|
[155] |
Houston AI, Mcnamara JM, Hutchinson JMC. General results concerning the trade-off between gaining energy and avoiding predation. Phil Trans R Soc Lond B 1993; 341: 375- 97.
|
[156] |
Lima SL. Predation risk and unpredictable feeding conditions—determinants of body-mass in birds. Ecology 1986; 67: 377- 85.
|
[157] |
Cresswell W. Diurnal and seasonal mass variation in blackbirds Turdus merula: consequences for mass-dependent predation risk. J Anim Ecol 1998; 67: 78- 90.
|
[158] |
Gentle LK, Gosler AG. Fat reserves and perceived predation risk in the great tit, Parus major. Proc Biol Sci 2001; 268: 487- 91.
|
[159] |
Gosler AG, Greenwood JJ, Perrins C. Predation risk and the cost of being fat. Nature 1995; 377: 621- 3.
|
[160] |
Monarca RI, Mathias Mda L, Speakman JR. Behavioural and physiological responses of wood mice (Apodemus sylvaticus) to experimental manipulations of predation and starvation risk. Physiol Behav 2015; 149: 331- 9.
|
[161] |
Monarca RI, Mathias MD, Wang DH et al. Predation risk modulates diet-induced obesity in male C57BL/6 mice. Obesity 2015; 23: 2059- 65.
|
[162] |
Tidhar WL, Bonier F, Speakman JR. Sex- and concentration-dependent effects of predator feces on seasonal regulation of body mass in the bank vole Clethrionomys glareolus. Horm Behav 2007; 52: 436- 44.
|
[163] |
Genné-Bacon EA, Trinko JR, DiLeone RJ. Innate fear-induced weight regulation in the C57BL/6J mouse. Front Behav Neurosci 2016; 10: 132.
|
[164] |
Speakman JR. Why lipostatic set point systems are unlikely to evolve. Mol Metab 2018; 7: 147- 54.
|
[165] |
Higginson AD, McNamara JM, Houston AI. Fatness and fitness: exposing the logic of evolutionary explanations for obesity. Proc Biol Sci 2016; 283: 20152443.
|
[166] |
Tam J, Fukumura D, Jain RK. A mathematical model of murine metabolic regulation by leptin: energy balance and defense of a stable body weight. Cell Metab 2009; 9: 52- 63.
|
[167] |
Hall KD, Guo J. Obesity energetics: body weight regulation and the effects of diet composition. Gastroenterology 2017; 152: 1718- 1727.e3.
|
[168] |
Pontzer H. Hotter and sicker: External energy expenditure and the tangled evolutionary roots of anthropogenic climate change and chronic disease. Am J Hum Biol 2021; 33: e23579.
|
[169] |
Runcie J, Thomson TJ. Prolonged starvation—a dangerous procedure? Br Med J 1970; 3: 432- 5.
|
[170] |
Stewart WK, Fleming LW. Features of a successful therapeutic fast of 382 days’ duration. Postgrad Med J 1973; 49: 203- 9.
|
[171] |
Thomson TJ, Runcie J, Miller V. Treatment of obesity by total fasting for up to 249 days. Lancet 1966; 2: 992- 6.
|
[172] |
Kramer KL, Greaves RD. Synchrony between growth and reproductive patterns in human females: early investment in growth among Pume foragers. Am J Phys Anthropol 2010; 141: 235- 44.
|
[173] |
Allison DB, Neale MC, Kezis MI et al. Assortative mating for relative weight: genetic implications. Behav Genet 1996; 26: 103- 11.
|
[174] |
Speakman JR, Djafarian K, Stewart J et al. Assortative mating for obesity. Am J Clin Nutr 2007; 86: 316- 23.
|
[175] |
Lucas A, Baker BA, Desai M et al. Nutrition in pregnant or lactating rats programs lipid metabolism in the offspring. Brit J Nutr 1996; 76: 605- 12.
|
[176] |
Bouret S, Levin BE, Ozanne SE. Gene-environment interactions controlling energy and glucose homeostasis and the developmental origins of obesity. Physiol Rev 2015; 95: 47- 82.
|
[177] |
Huang Y, Mendoza JO, Hambly C et al. Limits to sustained energy intake. XXXI. Effect of graded levels of dietary fat on lactation performance in Swiss mice. J Exp Biol 2020; 223: jeb221911.
|
[178] |
Peters CR. Nut-like oil seeds—food for monkeys, chimpanzees, humans, and probably ape-men. Am J Phys Anthropol 1987; 73: 333- 63.
|
[179] |
Ayub Q, Moutsianas L, Chen Y et al. Revisiting the thrifty gene hypothesis via 65 loci associated with susceptibility to type 2 diabetes. Am J Hum Genet 2014; 94: 176- 85.
|
[180] |
Koh XH, Liu X, Teo YY. Can evidence from genome-wide association studies and positive natural selection surveys be used to evaluate the thrifty gene hypothesis in East Asians? PLoS One 2014; 9: e110974.
|
[181] |
Southam L, Soranzo N, Montgomery SB et al. Is the thrifty genotype hypothesis supported by evidence based on confirmed type 2 diabetes- and obesity-susceptibility variants? Diabetologia 2009; 52: 1846- 51.
|
[182] |
Wang G, Speakman JR. Analysis of positive selection at single nucleotide polymorphisms associated with body mass index does not support the “thrifty gene” hypothesis. Cell Metab 2016; 24: 531- 41.
|
[183] |
Wang G, Li N, Chang S et al. A prospective follow-up study of the relationship between c-reactive protein and human cancer risk in the Chinese Kailuan Female Cohort. Cancer Epidemiol Biomarkers Prev 2015; 24: 459- 65.
|
[184] |
Wang GL, Ekeleme-Egedigwe CA, El Hamdouchi A et al. Beauty and the body of the beholder: raters’ BMI has only limited association with ratings of attractiveness of the opposite sex. Obesity (Silver Spring) 2018; 26: 522- 30.
|
[185] |
Gloy VL, Lutz TA, Langhans W et al. Basal plasma levels of insulin, leptin, ghrelin, and amylin do not signal adiposity in rats recovering from forced overweight. Endocrinology 2010; 151: 4280- 8.
|
[186] |
Flier JS, Maratos-Flier E. Leptin’s physiologic role: does the emperor of energy balance have no clothes? Cell Metab 2017; 26: 24- 6.
|
[187] |
Harris RB. In vivo evidence for unidentified leptin-induced circulating factors that control white fat mass. Am J Physiol Regul Integr Comp Physiol 2015; 309: R1499- 511.
|
[188] |
Lund J, Lund C, Morville T et al. The unidentified hormonal defense against weight gain. PLoS Biol 2020; 18: e3000629.
|
[189] |
Ravussin Y, Edwin E, Gallop M et al. Evidence for a non-leptin system that defends against weight gain in overfeeding. Cell Metab 2018; 28: 289- 99.e5.
|
[190] |
Ravussin Y, Leibel RL, Ferrante AW. A missing link in body weight homeostasis: the catabolic signal of the overfed state. Cell Metab 2014; 20: 565- 72.
|
[191] |
Frühbeck G, Gómez-Ambrosi J. Rationale for the existence of additional adipostatic hormones. FASEB J 2001; 15: 1996- 2006.
|
[192] |
Haynes WG, Morgan DA, Djalali A et al. Interactions between the melanocortin system and leptin in control of sympathetic nerve traffic. Hypertension 1999; 33: 542- 7.
|
[193] |
Satoh N, Ogawa Y, Katsuura G et al. Satiety effect and sympathetic activation of leptin are mediated by hypothalamic melanocortin system. Neurosci Lett 1998; 249: 107- 10.
|
[194] |
Satoh N, Ogawa Y, Katsuura G et al. Sympathetic activation of leptin via the ventromedial hypothalamus—leptin-induced increase in catecholamine secretion. Diabetes 1999; 48: 1787- 93.
|
[195] |
Williams DL, Bowers RR, Bartness TJ et al. Brainstem melanocortin 3/4 receptor stimulation increases uncoupling protein gene expression in brown fat. Endocrinology 2003; 144: 4692- 7.
|
[196] |
Voss-Andreae A, Murphy JG, Ellacott KL et al. Role of the central melanocortin circuitry in adaptive thermogenesis of brown adipose tissue. Endocrinology 2007; 148: 1550- 60.
|
[197] |
Song CK, Vaughan CH, Keen-Rhinehart E et al. Melanocortin-4 receptor mRNA expressed in sympathetic outflow neurons to brown adipose tissue: neuroanatomical and functional evidence. Am J Physiol Regul Integr Comp Physiol 2008; 295: R417- 28.
|
[198] |
Wang G, Speakman JR. Analysis of signatures of selection at single nucleotide polymorphisms (SNPs) associated with body mass index (BMI) does not support the ‘Thrifty gene’ hypothesis. Cell Metab 2016; 24: 531- 41.
|
[199] |
Balthasar N, Dalgaard LT, Lee CE et al. Divergence of melanocortin pathways in the control of food intake and energy expenditure. Cell 2005; 123: 493- 505.
|
[200] |
Berglund ED, Liu T, Kong X et al. Melanocortin 4 receptors in autonomic neurons regulate thermogenesis and glycemia. Nat Neurosci 2014; 17: 911- 3.
|
[201] |
Henry FE, Sugino K, Tozer A et al. Cell type-specific transcriptomics of hypothalamic energy-sensing neuron responses to weight-loss. Elife 2015; 4: e09800.
|
[202] |
Lam BYH Cimino I Polex-Wolf J et al. Heterogeneity of hypothalamic pro-opiomelanocortin-expressing neurons revealed by single-cell RNA sequencing. Mol Metab 2017; 6: 383- 92.
|
[203] |
Williams KW, Liu T, Kong X et al. Xbp1s in Pomc neurons connects ER stress with energy balance and glucose homeostasis. Cell Metab 2014; 20: 471- 82.
|
[204] |
Zhu Y, Gao Y, Tao C et al. Connexin 43 mediates white adipose tissue beiging by facilitating the propagation of sympathetic neuronal signals. Cell Metab 2016; 24: 420- 33.
|
[205] |
Dodd GT, Decherf S, Loh K et al. Leptin and insulin act on POMC neurons to promote the browning of white fat. Cell 2015; 160: 88- 104.
|
[206] |
Yano W, Kubota N, Itoh S et al. Molecular mechanism of moderate insulin resistance in adiponectin-knockout mice. Endocr J 2008; 55: 515- 22.
|
[207] |
King NA, Hopkins M, Caudwell P et al. Individual variability following 12 weeks of supervised exercise: identification and characterization of compensation for exercise-induced weight loss. Int J Obes (Lond) 2008; 32: 177- 84.
|
/
〈 | 〉 |