Skip to main content
Top
Published in: Reviews in Endocrine and Metabolic Disorders 1/2022

Open Access 01-02-2022 | Obesity

Leptin as a key regulator of the adipose organ

Authors: Catalina Picó, Mariona Palou, Catalina Amadora Pomar, Ana María Rodríguez, Andreu Palou

Published in: Reviews in Endocrine and Metabolic Disorders | Issue 1/2022

Login to get access

Abstract

Leptin is a hormone primarily produced by the adipose tissue in proportion to the size of fat stores, with a primary function in the control of lipid reserves. Besides adipose tissue, leptin is also produced by other tissues, such as the stomach, placenta, and mammary gland. Altogether, leptin exerts a broad spectrum of short, medium, and long-term regulatory actions at the central and peripheral levels, including metabolic programming effects that condition the proper development and function of the adipose organ, which are relevant for its main role in energy homeostasis. Comprehending how leptin regulates adipose tissue may provide important clues to understand the pathophysiology of obesity and related diseases, such as type 2 diabetes, as well as its prevention and treatment. This review focuses on the physiological and long-lasting regulatory effects of leptin on adipose tissue, the mechanisms and pathways involved, its main outcomes on whole-body physiological homeostasis, and its consequences on chronic diseases.
Literature
1.
go back to reference Zhang Y, et al. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372(6505):425–32.PubMedCrossRef Zhang Y, et al. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372(6505):425–32.PubMedCrossRef
2.
go back to reference Palou A, et al. Obesity: molecular bases of a multifactorial problem. Eur J Nutr. 2000;39(4):127–44.PubMedCrossRef Palou A, et al. Obesity: molecular bases of a multifactorial problem. Eur J Nutr. 2000;39(4):127–44.PubMedCrossRef
3.
4.
go back to reference Cinti S. The adipose organ: endocrine aspects and insights from transgenic models. Eat Weight Disord. 2001;6(3 Suppl):4–8.PubMed Cinti S. The adipose organ: endocrine aspects and insights from transgenic models. Eat Weight Disord. 2001;6(3 Suppl):4–8.PubMed
5.
go back to reference Campfield LA, et al. Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science. 1995;269(5223):546–9.PubMedCrossRef Campfield LA, et al. Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science. 1995;269(5223):546–9.PubMedCrossRef
6.
go back to reference Halaas JL, et al. Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 1995;269(5223):543–6.PubMedCrossRef Halaas JL, et al. Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 1995;269(5223):543–6.PubMedCrossRef
7.
go back to reference Pelleymounter MA, et al. Effects of the obese gene product on body weight regulation in ob/ob mice. Science. 1995;269(5223):540–3.PubMedCrossRef Pelleymounter MA, et al. Effects of the obese gene product on body weight regulation in ob/ob mice. Science. 1995;269(5223):540–3.PubMedCrossRef
8.
go back to reference Montague CT, et al. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 1997;387(6636):903–8.PubMedCrossRef Montague CT, et al. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 1997;387(6636):903–8.PubMedCrossRef
9.
go back to reference Farooqi IS, et al. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med. 1999;341(12):879–84.PubMedCrossRef Farooqi IS, et al. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med. 1999;341(12):879–84.PubMedCrossRef
10.
go back to reference Considine RV, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334(5):292–5.PubMedCrossRef Considine RV, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334(5):292–5.PubMedCrossRef
12.
go back to reference Cinti S, et al. Secretory granules of endocrine and chief cells of human stomach mucosa contain leptin. Int J Obes Relat Metab Disord. 2000;24(6):789–93.PubMedCrossRef Cinti S, et al. Secretory granules of endocrine and chief cells of human stomach mucosa contain leptin. Int J Obes Relat Metab Disord. 2000;24(6):789–93.PubMedCrossRef
14.
go back to reference Pico C, et al. Leptin production by the stomach is up-regulated in obese (fa/fa) Zucker rats. Obes Res. 2002;10(9):932–8.PubMedCrossRef Pico C, et al. Leptin production by the stomach is up-regulated in obese (fa/fa) Zucker rats. Obes Res. 2002;10(9):932–8.PubMedCrossRef
15.
go back to reference Pico C, et al. Gastric leptin: a putative role in the short-term regulation of food intake. Br J Nutr. 2003;90(4):735–41.PubMedCrossRef Pico C, et al. Gastric leptin: a putative role in the short-term regulation of food intake. Br J Nutr. 2003;90(4):735–41.PubMedCrossRef
16.
go back to reference Smith-Kirwin SM, et al. Leptin expression in human mammary epithelial cells and breast milk. J Clin Endocrinol Metab. 1998;83(5):1810–3.PubMedCrossRef Smith-Kirwin SM, et al. Leptin expression in human mammary epithelial cells and breast milk. J Clin Endocrinol Metab. 1998;83(5):1810–3.PubMedCrossRef
17.
go back to reference Masuzaki H, et al. Nonadipose tissue production of leptin: leptin as a novel placenta-derived hormone in humans. Nat Med. 1997;3(9):1029–33.PubMedCrossRef Masuzaki H, et al. Nonadipose tissue production of leptin: leptin as a novel placenta-derived hormone in humans. Nat Med. 1997;3(9):1029–33.PubMedCrossRef
18.
go back to reference Palou M, Pico C, Palou A. Leptin as a breast milk component for the prevention of obesity. Nutr Rev. 2018;76(12):875–92.PubMed Palou M, Pico C, Palou A. Leptin as a breast milk component for the prevention of obesity. Nutr Rev. 2018;76(12):875–92.PubMed
19.
go back to reference Park HK, Ahima RS. Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism. Metabolism. 2015;64(1):24–34.CrossRef Park HK, Ahima RS. Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism. Metabolism. 2015;64(1):24–34.CrossRef
20.
go back to reference Ramos-Lobo AM, Donato J Jr. The role of leptin in health and disease. Temperature (Austin). 2017;4(3):258–91.CrossRef Ramos-Lobo AM, Donato J Jr. The role of leptin in health and disease. Temperature (Austin). 2017;4(3):258–91.CrossRef
21.
go back to reference Lynes MD, Tseng YH. Deciphering adipose tissue heterogeneity. Ann N Y Acad Sci. 2018;1411(1):5–20.PubMedCrossRef Lynes MD, Tseng YH. Deciphering adipose tissue heterogeneity. Ann N Y Acad Sci. 2018;1411(1):5–20.PubMedCrossRef
22.
23.
go back to reference Cinti S. The adipose organ: morphological perspectives of adipose tissues. Proc Nutr Soc. 2001;60(3):319–28.PubMedCrossRef Cinti S. The adipose organ: morphological perspectives of adipose tissues. Proc Nutr Soc. 2001;60(3):319–28.PubMedCrossRef
24.
go back to reference Vazquez-Vela ME, Torres N, Tovar AR. White adipose tissue as endocrine organ and its role in obesity. Arch Med Res. 2008;39(8):715–28.PubMedCrossRef Vazquez-Vela ME, Torres N, Tovar AR. White adipose tissue as endocrine organ and its role in obesity. Arch Med Res. 2008;39(8):715–28.PubMedCrossRef
25.
26.
go back to reference Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84(1):277–359.PubMedCrossRef Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84(1):277–359.PubMedCrossRef
27.
go back to reference Trayhurn P, et al. Effects of fasting and refeeding on ob gene expression in white adipose tissue of lean and obese (oblob) mice. FEBS Lett. 1995;368(3):488–90.PubMedCrossRef Trayhurn P, et al. Effects of fasting and refeeding on ob gene expression in white adipose tissue of lean and obese (oblob) mice. FEBS Lett. 1995;368(3):488–90.PubMedCrossRef
28.
go back to reference Hube F, et al. Difference in leptin mRNA levels between omental and subcutaneous abdominal adipose tissue from obese humans. Horm Metab Res. 1996;28(12):690–3.PubMedCrossRef Hube F, et al. Difference in leptin mRNA levels between omental and subcutaneous abdominal adipose tissue from obese humans. Horm Metab Res. 1996;28(12):690–3.PubMedCrossRef
29.
go back to reference Maffei M, et al. Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat Med. 1995;1(11):1155–61.PubMedCrossRef Maffei M, et al. Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat Med. 1995;1(11):1155–61.PubMedCrossRef
30.
go back to reference Saladin R, et al. Transient increase in obese gene expression after food intake or insulin administration. Nature. 1995;377(6549):527–9.PubMedCrossRef Saladin R, et al. Transient increase in obese gene expression after food intake or insulin administration. Nature. 1995;377(6549):527–9.PubMedCrossRef
31.
go back to reference Murakami T, Iida M, Shima K. Dexamethasone regulates obese expression in isolated rat adipocytes. Biochem Biophys Res Commun. 1995;214(3):1260–7.PubMedCrossRef Murakami T, Iida M, Shima K. Dexamethasone regulates obese expression in isolated rat adipocytes. Biochem Biophys Res Commun. 1995;214(3):1260–7.PubMedCrossRef
32.
go back to reference Lee MJ, et al. Acute and chronic regulation of leptin synthesis, storage, and secretion by insulin and dexamethasone in human adipose tissue. Am J Physiol Endocrinol Metab. 2007;292(3):E858–64.PubMedCrossRef Lee MJ, et al. Acute and chronic regulation of leptin synthesis, storage, and secretion by insulin and dexamethasone in human adipose tissue. Am J Physiol Endocrinol Metab. 2007;292(3):E858–64.PubMedCrossRef
33.
34.
go back to reference Nozhenko Y, et al. Milk leptin surge and biological rhythms of leptin and other regulatory proteins in breastmilk. PLoS One. 2015;10(12):e0145376. Nozhenko Y, et al. Milk leptin surge and biological rhythms of leptin and other regulatory proteins in breastmilk. PLoS One. 2015;10(12):e0145376.
35.
37.
go back to reference Elimam A, Marcus C. Meal timing, fasting and glucocorticoids interplay in serum leptin concentrations and diurnal profile. Eur J Endocrinol. 2002;147(2):181–8.PubMedCrossRef Elimam A, Marcus C. Meal timing, fasting and glucocorticoids interplay in serum leptin concentrations and diurnal profile. Eur J Endocrinol. 2002;147(2):181–8.PubMedCrossRef
38.
go back to reference Russell CD, et al. Leptin expression in adipose tissue from obese humans: depot-specific regulation by insulin and dexamethasone. Am J Physiol. 1998;275(3):E507–15.PubMed Russell CD, et al. Leptin expression in adipose tissue from obese humans: depot-specific regulation by insulin and dexamethasone. Am J Physiol. 1998;275(3):E507–15.PubMed
39.
go back to reference Ricci MR, Fried SK, Mittleman KD. Acute cold exposure decreases plasma leptin in women. Metabolism. 2000;49(4):421–3.PubMedCrossRef Ricci MR, Fried SK, Mittleman KD. Acute cold exposure decreases plasma leptin in women. Metabolism. 2000;49(4):421–3.PubMedCrossRef
40.
go back to reference Ricci MR, et al. Isoproterenol decreases leptin release from rat and human adipose tissue through posttranscriptional mechanisms. Am J Physiol Endocrinol Metab. 2005;288(4):E798-804.PubMedCrossRef Ricci MR, et al. Isoproterenol decreases leptin release from rat and human adipose tissue through posttranscriptional mechanisms. Am J Physiol Endocrinol Metab. 2005;288(4):E798-804.PubMedCrossRef
41.
go back to reference Evans BA, Agar L, Summers RJ. The role of the sympathetic nervous system in the regulation of leptin synthesis in C57BL/6 mice. FEBS Lett. 1999;444(2–3):149–54.PubMedCrossRef Evans BA, Agar L, Summers RJ. The role of the sympathetic nervous system in the regulation of leptin synthesis in C57BL/6 mice. FEBS Lett. 1999;444(2–3):149–54.PubMedCrossRef
42.
go back to reference Rayner DV, Trayhurn P. Regulation of leptin production: sympathetic nervous system interactions. J Mol Med (Berl). 2001;79(1):8–20.CrossRef Rayner DV, Trayhurn P. Regulation of leptin production: sympathetic nervous system interactions. J Mol Med (Berl). 2001;79(1):8–20.CrossRef
43.
go back to reference Dallner OS, et al. Dysregulation of a long noncoding RNA reduces leptin leading to a leptin-responsive form of obesity. Nat Med. 2019;25(3):507–16.PubMedCrossRef Dallner OS, et al. Dysregulation of a long noncoding RNA reduces leptin leading to a leptin-responsive form of obesity. Nat Med. 2019;25(3):507–16.PubMedCrossRef
46.
go back to reference Wauman J, Zabeau L, Tavernier J. The leptin receptor complex: heavier than expected? Front Endocrinol (Lausanne). 2017;8:30.CrossRef Wauman J, Zabeau L, Tavernier J. The leptin receptor complex: heavier than expected? Front Endocrinol (Lausanne). 2017;8:30.CrossRef
47.
go back to reference Flak JN, Myers MG Jr. Minireview: CNS mechanisms of leptin action. Mol Endocrinol. 2016;30(1):3–12.PubMedCrossRef Flak JN, Myers MG Jr. Minireview: CNS mechanisms of leptin action. Mol Endocrinol. 2016;30(1):3–12.PubMedCrossRef
48.
go back to reference Millington GW. The role of proopiomelanocortin (POMC) neurones in feeding behaviour. Nutr Metab (Lond). 2007;4:18.CrossRef Millington GW. The role of proopiomelanocortin (POMC) neurones in feeding behaviour. Nutr Metab (Lond). 2007;4:18.CrossRef
49.
go back to reference Baver SB, et al. Leptin modulates the intrinsic excitability of AgRP/NPY neurons in the arcuate nucleus of the hypothalamus. J Neurosci. 2014;34(16):5486–96.PubMedPubMedCentralCrossRef Baver SB, et al. Leptin modulates the intrinsic excitability of AgRP/NPY neurons in the arcuate nucleus of the hypothalamus. J Neurosci. 2014;34(16):5486–96.PubMedPubMedCentralCrossRef
50.
go back to reference Margetic S, et al. Leptin: a review of its peripheral actions and interactions. Int J Obes Relat Metab Disord. 2002;26(11):1407–33.PubMedCrossRef Margetic S, et al. Leptin: a review of its peripheral actions and interactions. Int J Obes Relat Metab Disord. 2002;26(11):1407–33.PubMedCrossRef
51.
go back to reference Sainz N, et al. Leptin resistance and diet-induced obesity: central and peripheral actions of leptin. Metabolism. 2015;64(1):35–46.PubMedCrossRef Sainz N, et al. Leptin resistance and diet-induced obesity: central and peripheral actions of leptin. Metabolism. 2015;64(1):35–46.PubMedCrossRef
53.
go back to reference Ahima RS, et al. Distinct physiologic and neuronal responses to decreased leptin and mild hyperleptinemia. Endocrinology. 1999;140(11):4923–31.PubMedCrossRef Ahima RS, et al. Distinct physiologic and neuronal responses to decreased leptin and mild hyperleptinemia. Endocrinology. 1999;140(11):4923–31.PubMedCrossRef
54.
go back to reference Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature. 1998;395(6704):763–70.PubMedCrossRef Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature. 1998;395(6704):763–70.PubMedCrossRef
55.
go back to reference Farooqi IS, et al. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest. 2002;110(8):1093–103.PubMedPubMedCentralCrossRef Farooqi IS, et al. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest. 2002;110(8):1093–103.PubMedPubMedCentralCrossRef
56.
go back to reference Licinio J, et al. Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc Natl Acad Sci U S A. 2004;101(13):4531–6.PubMedPubMedCentralCrossRef Licinio J, et al. Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc Natl Acad Sci U S A. 2004;101(13):4531–6.PubMedPubMedCentralCrossRef
57.
go back to reference Brown RJ, et al. Long-term effectiveness and safety of metreleptin in the treatment of patients with generalized lipodystrophy. Endocrine. 2018;60(3):479–89.PubMedPubMedCentralCrossRef Brown RJ, et al. Long-term effectiveness and safety of metreleptin in the treatment of patients with generalized lipodystrophy. Endocrine. 2018;60(3):479–89.PubMedPubMedCentralCrossRef
58.
go back to reference Cook K, et al. Effect of leptin therapy on survival in generalized and partial lipodystrophy: a matched cohort analysis. J Clin Endocrinol Metab. 2021;106(8):e2953–67.PubMedPubMedCentralCrossRef Cook K, et al. Effect of leptin therapy on survival in generalized and partial lipodystrophy: a matched cohort analysis. J Clin Endocrinol Metab. 2021;106(8):e2953–67.PubMedPubMedCentralCrossRef
60.
go back to reference Maffei M, Giordano A. Leptin, the brain and energy homeostasis: from an apparently simple to a highly complex neuronal system. Rev Endocr Metab Disord. 2021. Maffei M, Giordano A. Leptin, the brain and energy homeostasis: from an apparently simple to a highly complex neuronal system. Rev Endocr Metab Disord. 2021.
62.
go back to reference Caro JF, et al. Decreased cerebrospinal-fluid/serum leptin ratio in obesity: a possible mechanism for leptin resistance. Lancet. 1996;348(9021):159–61.PubMedCrossRef Caro JF, et al. Decreased cerebrospinal-fluid/serum leptin ratio in obesity: a possible mechanism for leptin resistance. Lancet. 1996;348(9021):159–61.PubMedCrossRef
63.
go back to reference Halaas JL, et al. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc Natl Acad Sci U S A. 1997;94(16):8878–83.PubMedPubMedCentralCrossRef Halaas JL, et al. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc Natl Acad Sci U S A. 1997;94(16):8878–83.PubMedPubMedCentralCrossRef
64.
go back to reference Harrison L, et al. Fluorescent blood-brain barrier tracing shows intact leptin transport in obese mice. Int J Obes (Lond). 2019;43(6):1305–18.CrossRef Harrison L, et al. Fluorescent blood-brain barrier tracing shows intact leptin transport in obese mice. Int J Obes (Lond). 2019;43(6):1305–18.CrossRef
65.
go back to reference Kleinert M, et al. Time-resolved hypothalamic open flow micro-perfusion reveals normal leptin transport across the blood-brain barrier in leptin resistant mice. Mol Metab. 2018;13:77–82.PubMedPubMedCentralCrossRef Kleinert M, et al. Time-resolved hypothalamic open flow micro-perfusion reveals normal leptin transport across the blood-brain barrier in leptin resistant mice. Mol Metab. 2018;13:77–82.PubMedPubMedCentralCrossRef
66.
go back to reference Banks WA, Farrell CL. Impaired transport of leptin across the blood-brain barrier in obesity is acquired and reversible. Am J Physiol Endocrinol Metab. 2003;285(1):E10–5.PubMedCrossRef Banks WA, Farrell CL. Impaired transport of leptin across the blood-brain barrier in obesity is acquired and reversible. Am J Physiol Endocrinol Metab. 2003;285(1):E10–5.PubMedCrossRef
68.
go back to reference Prevot V, et al. The versatile tanycyte: a hypothalamic integrator of reproduction and energy metabolism. Endocr Rev. 2018;39(3):333–68.PubMedCrossRef Prevot V, et al. The versatile tanycyte: a hypothalamic integrator of reproduction and energy metabolism. Endocr Rev. 2018;39(3):333–68.PubMedCrossRef
69.
go back to reference Duquenne M, et al. Leptin brain entry via a tanycytic LepR-EGFR shuttle controls lipid metabolism and pancreas function. Nat Metab. 2021. Duquenne M, et al. Leptin brain entry via a tanycytic LepR-EGFR shuttle controls lipid metabolism and pancreas function. Nat Metab. 2021.
70.
go back to reference Baskin DG, et al. Increased expression of mRNA for the long form of the leptin receptor in the hypothalamus is associated with leptin hypersensitivity and fasting. Diabetes. 1998;47(4):538–43.PubMedCrossRef Baskin DG, et al. Increased expression of mRNA for the long form of the leptin receptor in the hypothalamus is associated with leptin hypersensitivity and fasting. Diabetes. 1998;47(4):538–43.PubMedCrossRef
71.
go back to reference Martin RL, et al. Leptin resistance is associated with hypothalamic leptin receptor mRNA and protein downregulation. Metabolism. 2000;49(11):1479–84.PubMedCrossRef Martin RL, et al. Leptin resistance is associated with hypothalamic leptin receptor mRNA and protein downregulation. Metabolism. 2000;49(11):1479–84.PubMedCrossRef
72.
go back to reference Picó C, et al. Metabolic programming of obesity by energy restriction during the perinatal period: different outcomes depending on gender and period, type and severity of restriction. Front Physiol. 2012;3:436.PubMedPubMedCentralCrossRef Picó C, et al. Metabolic programming of obesity by energy restriction during the perinatal period: different outcomes depending on gender and period, type and severity of restriction. Front Physiol. 2012;3:436.PubMedPubMedCentralCrossRef
73.
go back to reference Desai M, et al. Programmed hyperphagia due to reduced anorexigenic mechanisms in intrauterine growth-restricted offspring. Reprod Sci. 2007;14(4):329–37.PubMedCrossRef Desai M, et al. Programmed hyperphagia due to reduced anorexigenic mechanisms in intrauterine growth-restricted offspring. Reprod Sci. 2007;14(4):329–37.PubMedCrossRef
74.
go back to reference Breton C, et al. Maternal prenatal undernutrition alters the response of POMC neurons to energy status variation in adult male rat offspring. Am J Physiol Endocrinol Metab. 2009;296(3):E462–72.PubMedCrossRef Breton C, et al. Maternal prenatal undernutrition alters the response of POMC neurons to energy status variation in adult male rat offspring. Am J Physiol Endocrinol Metab. 2009;296(3):E462–72.PubMedCrossRef
75.
go back to reference Garcia AP, et al. Moderate caloric restriction during gestation results in lower arcuate nucleus NPY- and alphaMSH-neurons and impairs hypothalamic response to fed/fasting conditions in weaned rats. Diabetes Obes Metab. 2010;12(5):403–13.PubMedCrossRef Garcia AP, et al. Moderate caloric restriction during gestation results in lower arcuate nucleus NPY- and alphaMSH-neurons and impairs hypothalamic response to fed/fasting conditions in weaned rats. Diabetes Obes Metab. 2010;12(5):403–13.PubMedCrossRef
76.
go back to reference Palou M, et al. Impaired insulin and leptin sensitivity in the offspring of moderate caloric-restricted dams during gestation is early programmed. J Nutr Biochem. 2012;23(12):1627–39.PubMedCrossRef Palou M, et al. Impaired insulin and leptin sensitivity in the offspring of moderate caloric-restricted dams during gestation is early programmed. J Nutr Biochem. 2012;23(12):1627–39.PubMedCrossRef
77.
go back to reference Palou M, et al. Sexual dimorphism in the lasting effects of moderate caloric restriction during gestation on energy homeostasis in rats is related with fetal programming of insulin and leptin resistance. Nutr Metab. 2010;7:69.CrossRef Palou M, et al. Sexual dimorphism in the lasting effects of moderate caloric restriction during gestation on energy homeostasis in rats is related with fetal programming of insulin and leptin resistance. Nutr Metab. 2010;7:69.CrossRef
78.
go back to reference Bjorbak C, et al. SOCS3 mediates feedback inhibition of the leptin receptor via Tyr985. J Biol Chem. 2000;275(51):40649–57.PubMedCrossRef Bjorbak C, et al. SOCS3 mediates feedback inhibition of the leptin receptor via Tyr985. J Biol Chem. 2000;275(51):40649–57.PubMedCrossRef
79.
go back to reference Zhang ZY, Dodd GT, Tiganis T. Protein tyrosine phosphatases in hypothalamic insulin and leptin signaling. Trends Pharmacol Sci. 2015;36(10):661–74.PubMedCrossRef Zhang ZY, Dodd GT, Tiganis T. Protein tyrosine phosphatases in hypothalamic insulin and leptin signaling. Trends Pharmacol Sci. 2015;36(10):661–74.PubMedCrossRef
80.
go back to reference Howard JK, et al. Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of Socs3. Nat Med. 2004;10(7):734–8.PubMedCrossRef Howard JK, et al. Enhanced leptin sensitivity and attenuation of diet-induced obesity in mice with haploinsufficiency of Socs3. Nat Med. 2004;10(7):734–8.PubMedCrossRef
81.
go back to reference Briancon N, et al. Combined neural inactivation of suppressor of cytokine signaling-3 and protein-tyrosine phosphatase-1B reveals additive, synergistic, and factor-specific roles in the regulation of body energy balance. Diabetes. 2010;59(12):3074–84.PubMedPubMedCentralCrossRef Briancon N, et al. Combined neural inactivation of suppressor of cytokine signaling-3 and protein-tyrosine phosphatase-1B reveals additive, synergistic, and factor-specific roles in the regulation of body energy balance. Diabetes. 2010;59(12):3074–84.PubMedPubMedCentralCrossRef
83.
go back to reference Tsou RC, et al. Deficiency of PTP1B in leptin receptor-expressing neurons leads to decreased body weight and adiposity in mice. Endocrinology. 2012;153(9):4227–37.PubMedPubMedCentralCrossRef Tsou RC, et al. Deficiency of PTP1B in leptin receptor-expressing neurons leads to decreased body weight and adiposity in mice. Endocrinology. 2012;153(9):4227–37.PubMedPubMedCentralCrossRef
84.
go back to reference Ozcan L, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9(1):35–51.PubMedCrossRef Ozcan L, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9(1):35–51.PubMedCrossRef
85.
go back to reference Andreoli MF, et al. Leptin resensitisation: a reversion of leptin-resistant states. J Endocrinol. 2019;241(3):R81–96.PubMedCrossRef Andreoli MF, et al. Leptin resensitisation: a reversion of leptin-resistant states. J Endocrinol. 2019;241(3):R81–96.PubMedCrossRef
86.
go back to reference Cakir I, et al. Obesity induces hypothalamic endoplasmic reticulum stress and impairs proopiomelanocortin (POMC) post-translational processing. J Biol Chem. 2013;288(24):17675–88.PubMedPubMedCentralCrossRef Cakir I, et al. Obesity induces hypothalamic endoplasmic reticulum stress and impairs proopiomelanocortin (POMC) post-translational processing. J Biol Chem. 2013;288(24):17675–88.PubMedPubMedCentralCrossRef
88.
go back to reference Martinez-Sanchez N. There and back again: leptin actions in white adipose tissue. Int J Mol Sci. 2020;21(17). Martinez-Sanchez N. There and back again: leptin actions in white adipose tissue. Int J Mol Sci. 2020;21(17).
89.
go back to reference William Jr. WN, Ceddia RB, Curi R. Leptin controls the fate of fatty acids in isolated rat white adipocytes. J Endocrinol. 2002;175(3):735–44. William Jr. WN, Ceddia RB, Curi R. Leptin controls the fate of fatty acids in isolated rat white adipocytes. J Endocrinol. 2002;175(3):735–44.
90.
go back to reference Koltes DA, Spurlock ME, Spurlock DM. Adipose triglyceride lipase protein abundance and translocation to the lipid droplet increase during leptin-induced lipolysis in bovine adipocytes. Domest Anim Endocrinol. 2017;61:62–76.PubMedCrossRef Koltes DA, Spurlock ME, Spurlock DM. Adipose triglyceride lipase protein abundance and translocation to the lipid droplet increase during leptin-induced lipolysis in bovine adipocytes. Domest Anim Endocrinol. 2017;61:62–76.PubMedCrossRef
91.
go back to reference Zeng W, et al. Sympathetic neuro-adipose connections mediate leptin-driven lipolysis. Cell. 2015;163(1):84–94.PubMedCrossRef Zeng W, et al. Sympathetic neuro-adipose connections mediate leptin-driven lipolysis. Cell. 2015;163(1):84–94.PubMedCrossRef
92.
go back to reference Shen J, et al. In vivo effects of leptin on autonomic nerve activity and lipolysis in rats. Neurosci Lett. 2007;416(2):193–7.PubMedCrossRef Shen J, et al. In vivo effects of leptin on autonomic nerve activity and lipolysis in rats. Neurosci Lett. 2007;416(2):193–7.PubMedCrossRef
93.
go back to reference Bai Y, et al. Obese gene expression alters the ability of 30A5 preadipocytes to respond to lipogenic hormones. J Biol Chem. 1996;271(24):13939–42.PubMedCrossRef Bai Y, et al. Obese gene expression alters the ability of 30A5 preadipocytes to respond to lipogenic hormones. J Biol Chem. 1996;271(24):13939–42.PubMedCrossRef
94.
go back to reference Wang MY, Lee Y, Unger RH. Novel form of lipolysis induced by leptin. J Biol Chem. 1999;274(25):17541–4.PubMedCrossRef Wang MY, Lee Y, Unger RH. Novel form of lipolysis induced by leptin. J Biol Chem. 1999;274(25):17541–4.PubMedCrossRef
95.
go back to reference Jaubert AM, et al. Rapid nitration of adipocyte phosphoenolpyruvate carboxykinase by leptin reduces glyceroneogenesis and induces fatty acid release. PLoS One. 2012;7(7):e40650. Jaubert AM, et al. Rapid nitration of adipocyte phosphoenolpyruvate carboxykinase by leptin reduces glyceroneogenesis and induces fatty acid release. PLoS One. 2012;7(7):e40650.
96.
go back to reference Fruhbeck G, Aguado M, Martinez JA. In vitro lipolytic effect of leptin on mouse adipocytes: evidence for a possible autocrine/paracrine role of leptin. Biochem Biophys Res Commun. 1997;240(3):590–4.PubMedCrossRef Fruhbeck G, Aguado M, Martinez JA. In vitro lipolytic effect of leptin on mouse adipocytes: evidence for a possible autocrine/paracrine role of leptin. Biochem Biophys Res Commun. 1997;240(3):590–4.PubMedCrossRef
97.
go back to reference Kakuma T, et al. Role of leptin in peroxisome proliferator-activated receptor gamma coactivator-1 expression. Endocrinology. 2000;141(12):4576–82.PubMedCrossRef Kakuma T, et al. Role of leptin in peroxisome proliferator-activated receptor gamma coactivator-1 expression. Endocrinology. 2000;141(12):4576–82.PubMedCrossRef
98.
go back to reference Ceddia RB, et al. Leptin inhibits insulin-stimulated incorporation of glucose into lipids and stimulates glucose decarboxylation in isolated rat adipocytes. J Endocrinol. 1998;158(3):R7-9.PubMedCrossRef Ceddia RB, et al. Leptin inhibits insulin-stimulated incorporation of glucose into lipids and stimulates glucose decarboxylation in isolated rat adipocytes. J Endocrinol. 1998;158(3):R7-9.PubMedCrossRef
99.
go back to reference Ceddia RB, et al. Leptin stimulates uncoupling protein-2 mRNA expression and Krebs cycle activity and inhibits lipid synthesis in isolated rat white adipocytes. Eur J Biochem. 2000;267(19):5952–8.PubMedCrossRef Ceddia RB, et al. Leptin stimulates uncoupling protein-2 mRNA expression and Krebs cycle activity and inhibits lipid synthesis in isolated rat white adipocytes. Eur J Biochem. 2000;267(19):5952–8.PubMedCrossRef
100.
go back to reference Li YC, et al. Regulation of ATGL expression mediated by leptin in vitro in porcine adipocyte lipolysis. Mol Cell Biochem. 2010;333(1–2):121–8.PubMedCrossRef Li YC, et al. Regulation of ATGL expression mediated by leptin in vitro in porcine adipocyte lipolysis. Mol Cell Biochem. 2010;333(1–2):121–8.PubMedCrossRef
101.
go back to reference Sarmiento U, et al. Morphologic and molecular changes induced by recombinant human leptin in the white and brown adipose tissues of C57BL/6 mice. Lab Invest. 1997;77(3):243–56.PubMed Sarmiento U, et al. Morphologic and molecular changes induced by recombinant human leptin in the white and brown adipose tissues of C57BL/6 mice. Lab Invest. 1997;77(3):243–56.PubMed
103.
104.
go back to reference Sarjeant K, Stephens JM. Adipogenesis. Cold Spring Harb Perspect Biol. 2012;4(9):a008417. Sarjeant K, Stephens JM. Adipogenesis. Cold Spring Harb Perspect Biol. 2012;4(9):a008417.
105.
go back to reference Machinal-Quelin F, et al. Proadipogenic effect of leptin on rat preadipocytes in vitro: activation of MAPK and STAT3 signaling pathways. Am J Physiol Cell Physiol. 2002;282(4):C853–63.PubMedCrossRef Machinal-Quelin F, et al. Proadipogenic effect of leptin on rat preadipocytes in vitro: activation of MAPK and STAT3 signaling pathways. Am J Physiol Cell Physiol. 2002;282(4):C853–63.PubMedCrossRef
106.
go back to reference Palhinha L, et al. Leptin induces proadipogenic and proinflammatory signaling in adipocytes. Front Endocrinol (Lausanne). 2019;10:841.CrossRef Palhinha L, et al. Leptin induces proadipogenic and proinflammatory signaling in adipocytes. Front Endocrinol (Lausanne). 2019;10:841.CrossRef
107.
go back to reference Wagoner B, Hausman DB, Harris RB. Direct and indirect effects of leptin on preadipocyte proliferation and differentiation. Am J Physiol Regul Integr Comp Physiol. 2006;290(6):R1557–64.PubMedCrossRef Wagoner B, Hausman DB, Harris RB. Direct and indirect effects of leptin on preadipocyte proliferation and differentiation. Am J Physiol Regul Integr Comp Physiol. 2006;290(6):R1557–64.PubMedCrossRef
108.
go back to reference Ramsay TG. Porcine preadipocyte proliferation and differentiation: a role for leptin? J Anim Sci. 2005;83(9):2066–74.PubMedCrossRef Ramsay TG. Porcine preadipocyte proliferation and differentiation: a role for leptin? J Anim Sci. 2005;83(9):2066–74.PubMedCrossRef
109.
go back to reference Qian H, et al. Brain administration of leptin causes deletion of adipocytes by apoptosis. Endocrinology. 1998;139(2):791–4.PubMedCrossRef Qian H, et al. Brain administration of leptin causes deletion of adipocytes by apoptosis. Endocrinology. 1998;139(2):791–4.PubMedCrossRef
110.
go back to reference Gullicksen PS, et al. Adipose tissue cellularity and apoptosis after intracerebroventricular injections of leptin and 21 days of recovery in rats. Int J Obes Relat Metab Disord. 2003;27(3):302–12.PubMedCrossRef Gullicksen PS, et al. Adipose tissue cellularity and apoptosis after intracerebroventricular injections of leptin and 21 days of recovery in rats. Int J Obes Relat Metab Disord. 2003;27(3):302–12.PubMedCrossRef
111.
go back to reference Hamrick MW, et al. Injections of leptin into rat ventromedial hypothalamus increase adipocyte apoptosis in peripheral fat and in bone marrow. Cell Tissue Res. 2007;327(1):133–41.PubMedCrossRef Hamrick MW, et al. Injections of leptin into rat ventromedial hypothalamus increase adipocyte apoptosis in peripheral fat and in bone marrow. Cell Tissue Res. 2007;327(1):133–41.PubMedCrossRef
112.
go back to reference Della-Fera MA, et al. Sensitivity of ob/ob mice to leptin-induced adipose tissue apoptosis. Obes Res. 2005;13(9):1540–7.PubMedCrossRef Della-Fera MA, et al. Sensitivity of ob/ob mice to leptin-induced adipose tissue apoptosis. Obes Res. 2005;13(9):1540–7.PubMedCrossRef
113.
go back to reference Page KA, et al. beta-adrenergic receptor agonists increase apoptosis of adipose tissue in mice. Domest Anim Endocrinol. 2004;26(1):23–31.PubMedCrossRef Page KA, et al. beta-adrenergic receptor agonists increase apoptosis of adipose tissue in mice. Domest Anim Endocrinol. 2004;26(1):23–31.PubMedCrossRef
114.
go back to reference Cohen B, et al. Leptin induces angiopoietin-2 expression in adipose tissues. J Biol Chem. 2001;276(11):7697–700.PubMedCrossRef Cohen B, et al. Leptin induces angiopoietin-2 expression in adipose tissues. J Biol Chem. 2001;276(11):7697–700.PubMedCrossRef
116.
go back to reference Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in adult humans. Am J Physiol Endocrinol Metab. 2007;293(2):E444–52.PubMedCrossRef Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in adult humans. Am J Physiol Endocrinol Metab. 2007;293(2):E444–52.PubMedCrossRef
117.
go back to reference Petrovic N, et al. Chronic peroxisome proliferator-activated receptor gamma (PPARgamma) activation of epididymally derived white adipocyte cultures reveals a population of thermogenically competent, UCP1-containing adipocytes molecularly distinct from classic brown adipocytes. J Biol Chem. 2010;285(10):7153–64.PubMedCrossRef Petrovic N, et al. Chronic peroxisome proliferator-activated receptor gamma (PPARgamma) activation of epididymally derived white adipocyte cultures reveals a population of thermogenically competent, UCP1-containing adipocytes molecularly distinct from classic brown adipocytes. J Biol Chem. 2010;285(10):7153–64.PubMedCrossRef
119.
go back to reference Palou A, Pico C, Bonet ML. Nutritional potential of metabolic remodelling of white adipose tissue. Curr Opin Clin Nutr Metab Care. 2013;16(6):650–6.PubMedCrossRef Palou A, Pico C, Bonet ML. Nutritional potential of metabolic remodelling of white adipose tissue. Curr Opin Clin Nutr Metab Care. 2013;16(6):650–6.PubMedCrossRef
120.
go back to reference Scarpace PJ, et al. Leptin increases uncoupling protein expression and energy expenditure. Am J Physiol. 1997;273(1 Pt 1):E226–30.PubMed Scarpace PJ, et al. Leptin increases uncoupling protein expression and energy expenditure. Am J Physiol. 1997;273(1 Pt 1):E226–30.PubMed
121.
go back to reference Scarpace PJ, Matheny M. Leptin induction of UCP1 gene expression is dependent on sympathetic innervation. Am J Physiol. 1998;275(2):E259–64.PubMed Scarpace PJ, Matheny M. Leptin induction of UCP1 gene expression is dependent on sympathetic innervation. Am J Physiol. 1998;275(2):E259–64.PubMed
122.
go back to reference Gullicksen PS, et al. Energy metabolism and expression of uncoupling proteins 1, 2, and 3 after 21 days of recovery from intracerebroventricular mouse leptin in rats. Physiol Behav. 2002;75(4):473–82.PubMedCrossRef Gullicksen PS, et al. Energy metabolism and expression of uncoupling proteins 1, 2, and 3 after 21 days of recovery from intracerebroventricular mouse leptin in rats. Physiol Behav. 2002;75(4):473–82.PubMedCrossRef
123.
go back to reference Commins SP, et al. Induction of uncoupling protein expression in brown and white adipose tissue by leptin. Endocrinology. 1999;140(1):292–300.PubMedCrossRef Commins SP, et al. Induction of uncoupling protein expression in brown and white adipose tissue by leptin. Endocrinology. 1999;140(1):292–300.PubMedCrossRef
124.
go back to reference Enriori PJ, et al. Leptin action in the dorsomedial hypothalamus increases sympathetic tone to brown adipose tissue in spite of systemic leptin resistance. J Neurosci. 2011;31(34):12189–97.PubMedPubMedCentralCrossRef Enriori PJ, et al. Leptin action in the dorsomedial hypothalamus increases sympathetic tone to brown adipose tissue in spite of systemic leptin resistance. J Neurosci. 2011;31(34):12189–97.PubMedPubMedCentralCrossRef
125.
go back to reference Okamatsu-Ogura Y, et al. Uncoupling protein 1 contributes to fat-reducing effect of leptin. Obes Res Clin Pract. 2007;1(4):223–90.PubMedCrossRef Okamatsu-Ogura Y, et al. Uncoupling protein 1 contributes to fat-reducing effect of leptin. Obes Res Clin Pract. 2007;1(4):223–90.PubMedCrossRef
126.
go back to reference Cousin B, et al. Occurrence of brown adipocytes in rat white adipose tissue: molecular and morphological characterization. J Cell Sci. 1992;103(Pt 4):931–42.PubMedCrossRef Cousin B, et al. Occurrence of brown adipocytes in rat white adipose tissue: molecular and morphological characterization. J Cell Sci. 1992;103(Pt 4):931–42.PubMedCrossRef
127.
go back to reference Rosenwald M, Wolfrum C. The origin and definition of brite versus white and classical brown adipocytes. Adipocyte. 2014;3(1):4–9.PubMedCrossRef Rosenwald M, Wolfrum C. The origin and definition of brite versus white and classical brown adipocytes. Adipocyte. 2014;3(1):4–9.PubMedCrossRef
128.
go back to reference Castro E, Silva TEO, Festuccia WT. Critical review of beige adipocyte thermogenic activation and contribution to whole-body energy expenditure. Horm Mol Biol Clin Investig. 2017;31(2). Castro E, Silva TEO, Festuccia WT. Critical review of beige adipocyte thermogenic activation and contribution to whole-body energy expenditure. Horm Mol Biol Clin Investig. 2017;31(2).
129.
go back to reference Plum L, et al. Enhanced leptin-stimulated Pi3k activation in the CNS promotes white adipose tissue transdifferentiation. Cell Metab. 2007;6(6):431–45.PubMedCrossRef Plum L, et al. Enhanced leptin-stimulated Pi3k activation in the CNS promotes white adipose tissue transdifferentiation. Cell Metab. 2007;6(6):431–45.PubMedCrossRef
131.
go back to reference Kiernan K, MacIver NJ. The role of the adipokine leptin in immune cell function in health and disease. Front Immunol. 2020;11:622468. Kiernan K, MacIver NJ. The role of the adipokine leptin in immune cell function in health and disease. Front Immunol. 2020;11:622468.
132.
134.
go back to reference Curat CA, et al. From blood monocytes to adipose tissue-resident macrophages: induction of diapedesis by human mature adipocytes. Diabetes. 2004;53(5):1285–92.PubMedCrossRef Curat CA, et al. From blood monocytes to adipose tissue-resident macrophages: induction of diapedesis by human mature adipocytes. Diabetes. 2004;53(5):1285–92.PubMedCrossRef
135.
go back to reference Monteiro L, et al. Leptin in the regulation of the immunometabolism of adipose tissue-macrophages. J Leukoc Biol. 2019;106(3):703–16.PubMedCrossRef Monteiro L, et al. Leptin in the regulation of the immunometabolism of adipose tissue-macrophages. J Leukoc Biol. 2019;106(3):703–16.PubMedCrossRef
137.
go back to reference Tilg H, Moschen AR. Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol. 2006;6(10):772–83.PubMedCrossRef Tilg H, Moschen AR. Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol. 2006;6(10):772–83.PubMedCrossRef
138.
go back to reference Hoffmann A, et al. Leptin decreases circulating inflammatory IL-6 and MCP-1 in mice. BioFactors. 2019;45(1):43–8.PubMedCrossRef Hoffmann A, et al. Leptin decreases circulating inflammatory IL-6 and MCP-1 in mice. BioFactors. 2019;45(1):43–8.PubMedCrossRef
139.
go back to reference Luan B, et al. Leptin-mediated increases in catecholamine signaling reduce adipose tissue inflammation via activation of macrophage HDAC4. Cell Metab. 2014;19(6):1058–65.PubMedPubMedCentralCrossRef Luan B, et al. Leptin-mediated increases in catecholamine signaling reduce adipose tissue inflammation via activation of macrophage HDAC4. Cell Metab. 2014;19(6):1058–65.PubMedPubMedCentralCrossRef
140.
go back to reference Zhao S, et al. Partial leptin deficiency confers resistance to diet-induced obesity in mice. Mol Metab. 2020;37:100995. Zhao S, et al. Partial leptin deficiency confers resistance to diet-induced obesity in mice. Mol Metab. 2020;37:100995.
142.
go back to reference Rodgers A, Sferruzzi-Perri AN. Developmental programming of offspring adipose tissue biology and obesity risk. Int J Obes (Lond). 2021. Rodgers A, Sferruzzi-Perri AN. Developmental programming of offspring adipose tissue biology and obesity risk. Int J Obes (Lond). 2021.
143.
go back to reference Garcia AP, et al. Moderate caloric restriction during gestation in rats alters adipose tissue sympathetic innervation and later adiposity in offspring. PLoS One. 2011;6(2):e17313. Garcia AP, et al. Moderate caloric restriction during gestation in rats alters adipose tissue sympathetic innervation and later adiposity in offspring. PLoS One. 2011;6(2):e17313.
144.
go back to reference Palou M, et al. Moderate calorie restriction during gestation programs offspring for lower BAT thermogenic capacity driven by thyroid and sympathetic signaling. Int J Obes (Lond). 2015;39(2):339–45.CrossRef Palou M, et al. Moderate calorie restriction during gestation programs offspring for lower BAT thermogenic capacity driven by thyroid and sympathetic signaling. Int J Obes (Lond). 2015;39(2):339–45.CrossRef
145.
go back to reference Lakshmy R. Metabolic syndrome: role of maternal undernutrition and fetal programming. Rev Endocr Metab Disord. 2013;14(3):229–40.PubMedCrossRef Lakshmy R. Metabolic syndrome: role of maternal undernutrition and fetal programming. Rev Endocr Metab Disord. 2013;14(3):229–40.PubMedCrossRef
147.
go back to reference Bouret SG, Draper SJ, Simerly RB. Trophic action of leptin on hypothalamic neurons that regulate feeding. Science. 2004;304(5667):108–10.PubMedCrossRef Bouret SG, Draper SJ, Simerly RB. Trophic action of leptin on hypothalamic neurons that regulate feeding. Science. 2004;304(5667):108–10.PubMedCrossRef
149.
go back to reference Cardoso RC, et al. Developmental programming: impact of gestational steroid and metabolic milieus on adiposity and insulin sensitivity in prenatal testosterone-treated female sheep. Endocrinology. 2016;157(2):522–35.PubMedCrossRef Cardoso RC, et al. Developmental programming: impact of gestational steroid and metabolic milieus on adiposity and insulin sensitivity in prenatal testosterone-treated female sheep. Endocrinology. 2016;157(2):522–35.PubMedCrossRef
151.
go back to reference Greenwood MR, Hirsch J. Postnatal development of adipocyte cellularity in the normal rat. J Lipid Res. 1974;15(5):474–83.PubMedCrossRef Greenwood MR, Hirsch J. Postnatal development of adipocyte cellularity in the normal rat. J Lipid Res. 1974;15(5):474–83.PubMedCrossRef
152.
go back to reference Moreno-Mendez E, et al. Early-life programming of adipose tissue. Nutr Res Rev. 2020;33(2):244–59.PubMedCrossRef Moreno-Mendez E, et al. Early-life programming of adipose tissue. Nutr Res Rev. 2020;33(2):244–59.PubMedCrossRef
153.
go back to reference Arroyo-Jousse V, et al. Adipokines underlie the early origins of obesity and associated metabolic comorbidities in the offspring of women with pregestational obesity. Biochim Biophys Acta Mol Basis Dis. 2020;1866(2):165558. Arroyo-Jousse V, et al. Adipokines underlie the early origins of obesity and associated metabolic comorbidities in the offspring of women with pregestational obesity. Biochim Biophys Acta Mol Basis Dis. 2020;1866(2):165558.
154.
go back to reference Meyer DM, et al. Leptin in maternal plasma and cord blood as a predictor of offspring adiposity at 5 years: a follow-up study. Obesity (Silver Spring). 2018;26(2):279–83.CrossRef Meyer DM, et al. Leptin in maternal plasma and cord blood as a predictor of offspring adiposity at 5 years: a follow-up study. Obesity (Silver Spring). 2018;26(2):279–83.CrossRef
155.
go back to reference Telschow A, et al. High maternal and low cord blood leptin are associated with BMI-SDS gain in the first year of life. Obes Facts. 2019;12(5):575–85.PubMedPubMedCentralCrossRef Telschow A, et al. High maternal and low cord blood leptin are associated with BMI-SDS gain in the first year of life. Obes Facts. 2019;12(5):575–85.PubMedPubMedCentralCrossRef
156.
go back to reference Hinkle SN, et al. Maternal adipokines longitudinally measured across pregnancy and their associations with neonatal size, length, and adiposity. Int J Obes (Lond). 2019;43(7):1422–34.CrossRef Hinkle SN, et al. Maternal adipokines longitudinally measured across pregnancy and their associations with neonatal size, length, and adiposity. Int J Obes (Lond). 2019;43(7):1422–34.CrossRef
157.
go back to reference Castro NP, et al. The relationship between maternal plasma leptin and adiponectin concentrations and newborn adiposity. Nutrients. 2017;9(3). Castro NP, et al. The relationship between maternal plasma leptin and adiponectin concentrations and newborn adiposity. Nutrients. 2017;9(3).
158.
go back to reference Bi S, et al. Identification of a placental enhancer for the human leptin gene. J Biol Chem. 1997;272(48):30583–8.PubMedCrossRef Bi S, et al. Identification of a placental enhancer for the human leptin gene. J Biol Chem. 1997;272(48):30583–8.PubMedCrossRef
159.
160.
go back to reference Tung WK, et al. Association of cord plasma leptin with birth size in term newborns. Pediatr Neonatol. 2009;50(6):255–60.PubMedCrossRef Tung WK, et al. Association of cord plasma leptin with birth size in term newborns. Pediatr Neonatol. 2009;50(6):255–60.PubMedCrossRef
161.
go back to reference Mantzoros CS, et al. Cord blood leptin and adiponectin as predictors of adiposity in children at 3 years of age: a prospective cohort study. Pediatrics. 2009;123(2):682–9.PubMedCrossRef Mantzoros CS, et al. Cord blood leptin and adiponectin as predictors of adiposity in children at 3 years of age: a prospective cohort study. Pediatrics. 2009;123(2):682–9.PubMedCrossRef
162.
go back to reference Karakosta P, et al. Cord blood leptin levels in relation to child growth trajectories. Metabolism. 2016;65(6):874–82.PubMedCrossRef Karakosta P, et al. Cord blood leptin levels in relation to child growth trajectories. Metabolism. 2016;65(6):874–82.PubMedCrossRef
163.
go back to reference Chaoimh CN, et al. Cord blood leptin and gains in body weight and fat mass during infancy. Eur J Endocrinol. 2016;175(5):403–10.PubMedCrossRef Chaoimh CN, et al. Cord blood leptin and gains in body weight and fat mass during infancy. Eur J Endocrinol. 2016;175(5):403–10.PubMedCrossRef
164.
go back to reference Boeke CE, et al. Differential associations of leptin with adiposity across early childhood. Obesity (Silver Spring). 2013;21(7):1430–7.CrossRef Boeke CE, et al. Differential associations of leptin with adiposity across early childhood. Obesity (Silver Spring). 2013;21(7):1430–7.CrossRef
165.
go back to reference Simpson J, et al. Programming of adiposity in childhood and adolescence: associations with birth weight and cord blood adipokines. J Clin Endocrinol Metab. 2017;102(2):499–506.PubMed Simpson J, et al. Programming of adiposity in childhood and adolescence: associations with birth weight and cord blood adipokines. J Clin Endocrinol Metab. 2017;102(2):499–506.PubMed
166.
go back to reference Udagawa J, et al. Roles of leptin in prenatal and perinatal brain development. Congenit Anom (Kyoto). 2007;47(3):77–83.CrossRef Udagawa J, et al. Roles of leptin in prenatal and perinatal brain development. Congenit Anom (Kyoto). 2007;47(3):77–83.CrossRef
167.
168.
go back to reference Yau-Qiu ZX, et al. Leptin distribution in rat foetal and extraembryonic tissues in late gestation: a physiological view of amniotic fluid leptin. Nutrients. 2020;12(9). Yau-Qiu ZX, et al. Leptin distribution in rat foetal and extraembryonic tissues in late gestation: a physiological view of amniotic fluid leptin. Nutrients. 2020;12(9).
169.
go back to reference Ip S, et al. A summary of the agency for healthcare research and quality’s evidence report on breastfeeding in developed countries. Breastfeed Med. 2009;4(Suppl 1):S17-30.PubMedCrossRef Ip S, et al. A summary of the agency for healthcare research and quality’s evidence report on breastfeeding in developed countries. Breastfeed Med. 2009;4(Suppl 1):S17-30.PubMedCrossRef
170.
go back to reference von Kries R, et al. Breast feeding and obesity: cross sectional study. BMJ. 1999;319(7203):147–50.CrossRef von Kries R, et al. Breast feeding and obesity: cross sectional study. BMJ. 1999;319(7203):147–50.CrossRef
171.
go back to reference Horta BL, Loret de Mola C, Victora CG. Long-term consequences of breastfeeding on cholesterol, obesity, systolic blood pressure and type 2 diabetes: a systematic review and meta-analysis. Acta Paediatr. 2015;104(467):30–7. Horta BL, Loret de Mola C, Victora CG. Long-term consequences of breastfeeding on cholesterol, obesity, systolic blood pressure and type 2 diabetes: a systematic review and meta-analysis. Acta Paediatr. 2015;104(467):30–7.
172.
go back to reference Pico C, et al. Benefits of breastfeeding in infant health: a role for milk signaling peptides in molecular nutrition: mother and infant. In: Vinciguerra M, Sanchez PC, editors. Academic Press. ISBN; 2021. p. 29–56. Pico C, et al. Benefits of breastfeeding in infant health: a role for milk signaling peptides in molecular nutrition: mother and infant. In: Vinciguerra M, Sanchez PC, editors. Academic Press. ISBN; 2021. p. 29–56.
173.
go back to reference Badillo-Suarez PA, Rodriguez-Cruz M, Nieves-Morales X. Impact of metabolic hormones secreted in human breast milk on nutritional programming in childhood obesity. J Mammary Gland Biol Neoplasia. 2017;22(3):171–91.PubMedCrossRef Badillo-Suarez PA, Rodriguez-Cruz M, Nieves-Morales X. Impact of metabolic hormones secreted in human breast milk on nutritional programming in childhood obesity. J Mammary Gland Biol Neoplasia. 2017;22(3):171–91.PubMedCrossRef
174.
go back to reference Kratzsch J, Bae YJ, Kiess W. Adipokines in human breast milk. Best Pract Res Clin Endocrinol Metab. 2018;32(1):27–38.PubMedCrossRef Kratzsch J, Bae YJ, Kiess W. Adipokines in human breast milk. Best Pract Res Clin Endocrinol Metab. 2018;32(1):27–38.PubMedCrossRef
175.
go back to reference Palou A, et al. Universitat de les Illes Balears, assignee. Use of leptin for the prevention of excess body weight and composition containing leptin. WIPO patent WO2006089987 A1. February 23, 2005. 2005. Palou A, et al. Universitat de les Illes Balears, assignee. Use of leptin for the prevention of excess body weight and composition containing leptin. WIPO patent WO2006089987 A1. February 23, 2005. 2005.
176.
go back to reference Pico C, et al. The intake of physiological doses of leptin during lactation in rats prevents obesity in later life. Int J Obes. 2007;31(8):1199–209.CrossRef Pico C, et al. The intake of physiological doses of leptin during lactation in rats prevents obesity in later life. Int J Obes. 2007;31(8):1199–209.CrossRef
177.
go back to reference Sanchez J, et al. Oral supplementation with physiological doses of leptin during lactation in rats improves insulin sensitivity and affects food preferences later in life. Endocrinology. 2008;149(2):733–40.PubMedCrossRef Sanchez J, et al. Oral supplementation with physiological doses of leptin during lactation in rats improves insulin sensitivity and affects food preferences later in life. Endocrinology. 2008;149(2):733–40.PubMedCrossRef
178.
go back to reference Priego T, et al. Leptin intake during the suckling period improves the metabolic response of adipose tissue to a high-fat diet. Int J Obes (Lond). 2010;34(5):809–19.CrossRef Priego T, et al. Leptin intake during the suckling period improves the metabolic response of adipose tissue to a high-fat diet. Int J Obes (Lond). 2010;34(5):809–19.CrossRef
179.
go back to reference Casabiell X, et al. Presence of leptin in colostrum and/or breast milk from lactating mothers: a potential role in the regulation of neonatal food intake. J Clin Endocrinol Metab. 1997;82(12):4270–3.PubMedCrossRef Casabiell X, et al. Presence of leptin in colostrum and/or breast milk from lactating mothers: a potential role in the regulation of neonatal food intake. J Clin Endocrinol Metab. 1997;82(12):4270–3.PubMedCrossRef
180.
go back to reference Houseknecht KL, et al. Leptin is present in human milk and is related to maternal plasma leptin concentration and adiposity. Biochem Biophys Res Commun. 1997;240(3):742–7.PubMedCrossRef Houseknecht KL, et al. Leptin is present in human milk and is related to maternal plasma leptin concentration and adiposity. Biochem Biophys Res Commun. 1997;240(3):742–7.PubMedCrossRef
181.
go back to reference O’Connor D, et al. Leptin is not present in infant formulas. J Endocrinol Invest. 2003;26(5):490.PubMedCrossRef O’Connor D, et al. Leptin is not present in infant formulas. J Endocrinol Invest. 2003;26(5):490.PubMedCrossRef
182.
go back to reference Miralles O, et al. A physiological role of breast milk leptin in body weight control in developing infants. Obesity (Silver Spring). 2006;14(8):1371–7.CrossRef Miralles O, et al. A physiological role of breast milk leptin in body weight control in developing infants. Obesity (Silver Spring). 2006;14(8):1371–7.CrossRef
183.
go back to reference Konieczna J, et al. Oral leptin treatment in suckling rats ameliorates detrimental effects in hypothalamic structure and function caused by maternal caloric restriction during gestation. PLoS One. 2013;8(11):e81906. Konieczna J, et al. Oral leptin treatment in suckling rats ameliorates detrimental effects in hypothalamic structure and function caused by maternal caloric restriction during gestation. PLoS One. 2013;8(11):e81906.
184.
go back to reference Szostaczuk N, et al. Oral leptin supplementation throughout lactation in rats prevents later metabolic alterations caused by gestational calorie restriction. Int J Obes (Lond). 2017;41(3):360–71.CrossRef Szostaczuk N, et al. Oral leptin supplementation throughout lactation in rats prevents later metabolic alterations caused by gestational calorie restriction. Int J Obes (Lond). 2017;41(3):360–71.CrossRef
185.
go back to reference Proulx K, Richard D, Walker CD. Leptin regulates appetite-related neuropeptides in the hypothalamus of developing rats without affecting food intake. Endocrinology. 2002;143(12):4683–92.PubMedCrossRef Proulx K, Richard D, Walker CD. Leptin regulates appetite-related neuropeptides in the hypothalamus of developing rats without affecting food intake. Endocrinology. 2002;143(12):4683–92.PubMedCrossRef
186.
go back to reference Konieczna J, et al. Leptin intake in suckling rats restores altered T3 levels and markers of adipose tissue sympathetic drive and function caused by gestational calorie restriction. Int J Obes. 2015;39(6):959–66.CrossRef Konieczna J, et al. Leptin intake in suckling rats restores altered T3 levels and markers of adipose tissue sympathetic drive and function caused by gestational calorie restriction. Int J Obes. 2015;39(6):959–66.CrossRef
187.
go back to reference Pucci E, Chiovato L, Pinchera A. Thyroid and lipid metabolism. Int J Obes Relat Metab Disord. 2000;24(Suppl 2):S109–12.PubMedCrossRef Pucci E, Chiovato L, Pinchera A. Thyroid and lipid metabolism. Int J Obes Relat Metab Disord. 2000;24(Suppl 2):S109–12.PubMedCrossRef
Metadata
Title
Leptin as a key regulator of the adipose organ
Authors
Catalina Picó
Mariona Palou
Catalina Amadora Pomar
Ana María Rodríguez
Andreu Palou
Publication date
01-02-2022
Publisher
Springer US
Keywords
Obesity
Obesity
Published in
Reviews in Endocrine and Metabolic Disorders / Issue 1/2022
Print ISSN: 1389-9155
Electronic ISSN: 1573-2606
DOI
https://doi.org/10.1007/s11154-021-09687-5

Other articles of this Issue 1/2022

Reviews in Endocrine and Metabolic Disorders 1/2022 Go to the issue