Leptin alters energy intake and fat mass but not energy expenditure in lean subjects

Pavlina Chrysafi, Nikolaos Perakakis, Olivia M Farr, Konstantinos Stefanakis, Natia Peradze, Aleix Sala-Vila, Christos S Mantzoros, Pavlina Chrysafi, Nikolaos Perakakis, Olivia M Farr, Konstantinos Stefanakis, Natia Peradze, Aleix Sala-Vila, Christos S Mantzoros

Abstract

Based on studies in mice, leptin was expected to decrease body weight in obese individuals. However, the majority of the obese are hyperleptinemic and do not respond to leptin treatment, suggesting the presence of leptin tolerance and questioning the role of leptin as regulator of energy balance in humans. We thus performed detailed novel measurements and analyses of samples and data from our clinical trials biobank to investigate leptin effects on mechanisms of weight regulation in lean normo- and mildly hypo-leptinemic individuals without genetic disorders. We demonstrate that short-term leptin administration alters food intake during refeeding after fasting, whereas long-term leptin treatment reduces fat mass and body weight, and transiently alters circulating free fatty acids in lean mildly hypoleptinemic individuals. Leptin levels before treatment initiation and leptin dose do not predict the observed weight loss in lean individuals suggesting a saturable effect of leptin. In contrast to data from animal studies, leptin treatment does not affect energy expenditure, lipid utilization, SNS activity, heart rate, blood pressure or lean body mass.

Trial registration: ClinicalTrials.gov NCT00140231 NCT00140205 NCT00130117.

Conflict of interest statement

C.S.M. is advisor of Ansh Labs LLC, and consultant to Novo Nordisk and grant recipient through BIDMC and has served as an advisor to Aegerion and Visiting Professor to Regeneron. The remaining authors declare no competing interests.

Figures

Fig. 1. Schematic representation of the study…
Fig. 1. Schematic representation of the study designs of the four clinical studies.
Study 1: Eight healthy lean men and seven healthy lean women were studied under three separate Clinical Research Center (CRC)-based conditions for 72 h: one under isocaloric fed state conditions (normoleptinemia) and two during complete fasting state conditions (induced hypoleptinemia) scheduled in a random order and in a double-blind fashion with administration of physiologic replacement leptin doses (fasting + leptin) or placebo (fasting + placebo). Study 2: Five lean men, five men with obesity, and five lean women participated in three fed-normoleptinemic and three fasting-induced hypoleptinemic studies, which were conducted in the CRC, with leptin administration at three different doses (Dose A = 0.01 mg/kg, Dose B = 0.1 mg/kg, Dose C = 0.3 mg/kg). Study 3: Open-label long-term leptin treatment in mildly hypoleptinemic women. Study 4: Placebo-controlled long-term leptin treatment in mildly hypoleptinemic women.
Fig. 2. Leptin effects on weight and…
Fig. 2. Leptin effects on weight and fat mass.
a Cross-over study of lean subjects during 72-h fed state, fasting+placebo and fasting+leptin (study 1; n = 13). Left: baseline leptin levels in each admission. Center: % weight change at the end of each admission. Right: correlation of baseline leptin with % weight change at the end of each admission. Numbers above bars correspond to subject ID. P values of unpaired t test between lean men (LM) vs lean women (LW) and of correlations are reported; R, correlation coefficient. b Cross-over study of LM, LW, and obese men (OM) in 72-h fasting treated with escalating leptin doses (study 2; n = 15). Left: baseline leptin levels in each admission, Center: % weight change at the end of each admission. Right: correlation of baseline leptin with % weight change at the end of each admission. Numbers above bars correspond to subject ID. P values from one-way ANOVA, from post hoc Bonferroni test between LM vs LW vs OW and from correlations are reported. c Open-label (study 3; n = 7) and placebo-controlled long-term leptin treatment study (study 4; n = 19 (leptin = 10; placebo = 9)) in women with mild hypoleptinemia. Left: correlation of baseline leptin with % weight change after 8 weeks of leptin treatment. Subjects of study 3 were combined with leptin-treated subjects of study 4 in one analysis. Center and right: changes of leptin, body weight, and fat mass from baseline (Δ = change from baseline at each timepoint). In study 4, dashed lines correspond to the washout period after 36 weeks of study. In study 3, P values (P) for time effect (i.e., days of treatment) and in study 4, P values of G (group: leptin or placebo), T (time: weeks of treatment), and G*T interaction of mixed models adjusted for baseline are reported. By P < 0.05 (study 3) and G*T < 0.05 (study 4), post hoc Bonferroni test was performed (only significant results are reported). One, two, or three asterisks indicate P < 0.05, <0.01, or <0.001 for the specific timepoint vs baseline in study 3 and for leptin vs placebo in the specific timepoint in study 4. Correlations were performed with Pearson’s or Spearman’s correlation test. Data are presented as means ± SEMs. Exact P values: b, left: leptin at baseline dose 0.3 mg/kg = 0.003 × 10−1; leptin at baseline dose 0.3 mg/kg LM vs LW = 0.003 × 10−1. c Center: leptin = 0.003 × 10−1; weight = 0.001 × 10−3; fat mass = 0.002 × 10−5; leptin post hoc test at 81/2 weeks = 0.031 and at 13 weeks = 0.003 × 10−1. Weight post hoc test at 61/2 weeks = 0.002, at 81/2 weeks = 0.001 × 10−1, at 11 weeks = 0.001 × 10−1, and at 13 weeks = 0.001 × 10−1. Fat mass post hoc test at 61/2 weeks = 0.002, at 81/2 weeks = 0.002 × 10−2, at 11 weeks = 0.005 × 10−3, and at 13 weeks = 0.004 × 10−4. c, Right: leptin G = 0.005 × 10−2; T = 0.007 × 10−5; G*T = 0.002 × 10−5; body weight T = 0.007 × 10−5; fat mass T = 0.009 × 10−5; G*T = 0.001 × 10−2; leptin post hoc test at 4 weeks = 0.039; at 8 weeks = 0.001; at 12 weeks = 0.004 × 10−1; at 16 weeks = 0.005 × 10−3; at 20 weeks = 0.003 × 10−2; at 24 weeks = 0.001 × 10−2; at 28 weeks = 0.002 × 10−3; at 32 weeks = 0.004 × 10−3; at 36 weeks = 0.002 × 10−3. Fat mass post hoc test at 24 weeks = 0.002 × 10−1; at 36 weeks = 0.001 × 10−1.
Fig. 3. Short- and long-term leptin effects…
Fig. 3. Short- and long-term leptin effects on energy intake and expenditure.
a Energy intake after 72-h in fed state, fasting+placebo, or fasting + leptin (study 1, n = 13). P values from repeated measure ANOVA and post hoc Bonferroni test are reported. R, correlation coefficient. b Expected (based on leptin-induced caloric deficit in study 1) vs observed fat mass loss during long-term leptin treatment in studies 3 and 4. As per the protocol, in study 4, if a subject lost >5% of baseline weight, the dose was reduced by 0.04 mg/kg (red arrows). P values of G (group: observed or expected fat mass change), T (days/weeks of treatment), and G*T interaction of mixed models are reported. By G*T < 0.05, post hoc Bonferroni test was performed: two asterisks indicate P < 0.01 for observed vs expected fat mass change at the specific timepoint. c Energy expenditure during 72-h fed state, fasting+ placebo, and fasting + leptin (study 1, n = 13). P values of G (group: fed, fasting + placebo, fasting + leptin), T (days of treatment), and G*T interaction of mixed models adjusted for baseline are reported. Post hoc Bonferroni test was performed between the estimated means of the three groups and between the three groups at each timepoint. Three asterisks indicate P < 0.001 for fed vs fasting + placebo (red) and fed vs fasting + leptin (blue). d Energy expenditure during 72-h fasting treated with escalating leptin doses (study 2, n = 15). P values of D (dose: 0.01, 0.1, and 0.3 mg/kg/d), T (hours of fasting), and D*T interaction of mixed models adjusted for baseline are reported. e Energy expenditure during open-label long-term leptin treatment in mildly hypoleptinemic women (study 3, n = 7). P values of paired t test (RMR and body temperature) and of time effect of mixed models adjusted for baseline (exercise score) are reported. No post hoc test was performed since P > 0.05. f Energy expenditure during placebo-controlled long-term leptin treatment in mildly hypoleptinemic women (study 4, n = 19 (leptin = 10; placebo = 9)). P values of G (group: placebo or leptin), T (weeks of study), and G*T interaction of mixed models adjusted for baseline are reported. No post hoc test was performed since G*T > 0.05. All P values are two-sided. For post hoc Bonferroni test, only significant results are reported. Data are demonstrated as means ± SEMs. Exact P values: a Correlation of food intake with leptin prior to meal = 0.002 × 10−2. b Study 4: post hoc test at 12 weeks = 0.002; at 24 weeks = 0.001; at 36 weeks = 0.003. c Day 3 fed vs Pl = 0.004 × 10−3; fed vs Le = 0.003 × 10−1. d Temperature T = 0.002 × 10−12.
Fig. 4. Leptin effects on heart rate…
Fig. 4. Leptin effects on heart rate and blood pressure.
a Seventy-two hours fed state or fasting + leptin or fasting + placebo (study 1, n = 13). P values of G (group: fed or fasting + placebo or fasting + leptin), T (time: days of study), and G*T interaction of mixed models adjusted for baseline are reported. b Seventy-two hours fasting treated with escalating leptin doses (study 2, n = 15). P values of D (dose: 0.01 or 0.1 or 0.3 mg/kg/d), T (time: hours of fasting), and D*T interaction of mixed models adjusted for baseline are reported. c Open-label long-term leptin treatment in mildly hypoleptinemic women (study 3, n = 7). P value (P) of paired t test is reported. d Placebo-controlled long-term leptin treatment in mildly hypoleptinemic women (study 4, n = 19 (leptin = 10; placebo = 9)). P values of G (group: placebo or leptin), T (time: weeks of study), and G*T interaction of mixed models adjusted for baseline are reported. No post hoc Bonnferroni test was performed since G*T > 0.05 (studies 1 and 4) and D*T > 0.05 (study 2). All P values are two-sided. Data are demonstrated as means ± SEMs. Exact P values: b HR T = 0.001 × 10−17; SBP T = 0.003 × 10−1; DBP T = 0.002 × 10−2; MBP T = 0.002 × 10−2.
Fig. 5. Leptin effects on adrenal hormones.
Fig. 5. Leptin effects on adrenal hormones.
a Seventy-two hours fed state or fasting+leptin or fasting + placebo (study 1, n = 13). Left: blood aldosterone and cortisol at the start and completion of the study. Right: 24-h urine cortisol, epinephrine, and norepinephrine collected at the last day of the study. P values of G (group: fed or fasting + placebo or fasting + leptin), T (time: days of study), and G*T interaction of mixed models, adjusted for baseline are reported. For urine catecholamines, P values were calculated with repeated measure ANOVA, since only the group factor existed. By G*T < 0.05 (blood aldosterone and cortisol) and by G < 0.05 (urine catecholamines), post hoc Bonferroni test was performed between the estimated means of the three groups and between the three groups at each timepoint. Three asterisks indicate P < 0.001 for fed vs fasting + placebo (red) and for fed vs fasting + leptin (blue) at the particular timepoint. b Seventy-two hours of fasting treated with escalating leptin doses (study 2, n = 15). P values of D (dose: 0.01 or 0.1 or 0.3 mg/kg/d), T (time: days of fasting), and D*T interaction of mixed models adjusted for baseline are reported. No post hoc Bonnferroni test was performed since D*T > 0.05. c Open-label long-term leptin treatment in mildly hypoleptinemic women (study 3, n = 7). P value (P) of time effect (i.e., days of study) of mixed models adjusted for baseline is reported. By P < 0.05 post hoc Bonferroni’s test for each timepoint compared to baseline was additionally performed and two asterisks indicate P < 0.01 for the specific timepoint vs 0 (baseline). d Placebo-controlled long-term leptin treatment in mildly hypoleptinemic women (study 4, n = 19 (leptin = 10; placebo = 9)). P values of G (group: placebo or leptin), T (time: weeks of study), and G*T interaction of mixed models adjusted for baseline are reported. No post hoc Bonnferroni test was performed since G*T > 0.05. For Bonferroni post hoc tests, only significant results are reported. All P values are two-sided. Data are demonstrated as means ± SEMs. Exact P values: a aldosterone T = 0.001 × 10−3; day 3 post hoc test for fed vs Pl = 0.004 × 10−2; fed vs Le = 0.006 × 10−2. b Aldosterone T = 0.001 × 10−15; renin T = 0.005 × 10−17; urine epinephrine T = 0.004 × 10−1. c Aldosterone post hoc test at 15 days = 0.005.
Fig. 6. Metabolome changes with short- and…
Fig. 6. Metabolome changes with short- and long-term leptin treatment.
ad Effects on metabolite and lipid metabolism of 72-h fed state or fasting treated with leptin or placebo (study 1, n = 13). a Evaluation of metabolites, lipids, and lipoproteins with one-way ANOVA between the three admissions. Red dots indicate parameters significantly different and blue dots parameters not significantly different between groups (fed vs fasting + placebo vs fasting + leptin) with a preset false discovery rate of P < 2.15 × 10−4 (total 68 parameters significant). b Heatmap of the 68 significant parameters according to one-way ANOVA for the three admissions. c sPLS-DA analysis of fasting+leptin vs fasting + placebo: symbols indicate the measurement of component 1 in relation to measurement of component 3 for one subject/on one treatment/on one day of fasting: Blue circles correspond to leptin and red squares to placebo. Increasing color intensity indicates more time (days) of fasting. d The ten parameters that compose components 1 and 3 and their level of contribution (loading) to the component. e Effects on metabolite and lipid metabolism of long-term leptin treatment in mildly hypoleptinemic women (placebo-controlled study 4, n = 19 (leptin = 10; placebo = 9)). Left: sPLS-DA analysis of metabolites and lipoproteins in placebo vs leptin. Symbols indicate the measurement of component 1 in relation to component 2 for one subject/on one treatment/on one day of study: blue circles correspond to leptin and red squares to placebo. Increasing color intensity of symbol indicates more time (weeks) of study. Large oval-colored shapes indicate 95% confidence interval for each group. The observed major overlap between groups suggests no significant differences between placebo and leptin. Right: Evaluation of metabolites and lipoproteins with one-way ANOVA in placebo and leptin-treated subjects for up to 36 weeks. Each dot represents a parameter (blue dot = nonsignificant parameter, red = preset color for significant parameters but no such parameter was detected). NMR-based metabolomics were used to quantify amino acids, metabolites, and lipids bound to lipoproteins. GC/MS-EI was used to quantify fatty acid methyl esters. Le_0, Le_1 etc. indicate day 0 (baseline), 1 etc. of fasting + leptin. Pl_0, Pl_1 etc. indicate day 0 (baseline), 1 etc. of fasting + placebo. For metabolite nomenclature, see Supplementary Data 2.
Fig. 7. Fatty acid changes with short-…
Fig. 7. Fatty acid changes with short- and long-term leptin treatment.
a, b Seventy-two hours fed state or fasting+leptin or fasting+placebo (study 1, n = 13). a Blood concentrations of fatty acid profile from start and till completion of the study as ratios of the baseline (0 day). GC/MS-EI was used to quantify fatty acid methyl esters in whole plasma. b Blood-free fatty acids (FFA) from start and till completion of the study; mixed model was performed (for FFA adjusted for baseline). P values of G (group: fed or fasting + placebo or fasting + leptin), T (time: days of study), and G*T interaction of mixed models are reported. In panels a and b by G*T < 0.05, post hoc Bonferroni test was performed between the estimated means of the three groups and between the three groups at each timepoint. One, two, or three asterisks indicate P < 0.05, <0.01, or <0.001 for fed vs fasting + placebo (red) and for fed vs fasting + leptin (blue). One, two, or three hash signs indicate P < 0.05, <0.01, or <0.001 for fasting + leptin vs fasting + placebo in the Bonferroni post hoc t test. c Open-label long-term leptin treatment in mildly hypoleptinemic women (study 3, n = 7). Blood FFA concentrations. P value of time effect (i.e., days of study) of mixed models adjusted for baseline is reported. Two asterisks indicate P < 0.01 for the specific timepoint vs 0 (baseline) in the Bonferroni post hoc t test (performed by P < 0.05). d Placebo-controlled long-term leptin treatment in mildly hypoleptinemic women (study 4, n = 19 (leptin = 10; placebo = 9)). P values of G (group: placebo or leptin), T (time: weeks of study), and G*T interaction of mixed models adjusted for baseline are reported. By G*T < 0.05, post hoc Bonferroni test was additionally performed between the two groups at each timepoint. One or two asterisks indicate P < 0.05, or <0.01 for leptin vs placebo for the specific timepoint. For Bonferroni post hoc tests, only significant results are reported. All P values are two-sided. Data are demonstrated as means ± SEMs. Exact P values. a Total FA (ratio) G = 0.008 × 10−4; T = 0.002 × 10−1; fed vs Le = 0.004 × 10−1; day 2 fed vs Pl = 0.008 & fed vs Le = 0.002; day 3 fed vs Le = 0.001 × 10−1 & Le vs Pl = 0.002. SFA (ratio) G = 0.002 × 10−4; T = 0.003 × 10−2; fed vs Le = 0.008 × 10−5; day 2 fed vs Pl = 0.003 & fed vs Le = 0.002; day 3 fed vs Le = 0.001 × 10−1 & Le vs Pl = 0.001. MUFA (ratio) G = 0.002 × 10−6; fed vs Le = 0.008 × 10−7; day 1 fed vs Le = 0.021; day 2 fed vs Pl = 0.001 & fed vs Le = 0.001 × 10−1; day 3 fed vs Le = 0.001 × 10−1 & Le vs Pl = 0.036. PUFA (ratio) T = 0.005 × 10−3. C14:0 (ratio) G = 0.009 × 10−2; Le vs Pl = 0.004 × 10−1; day 3 fed vs Le = 0.002 & Le vs Pl = 0.002. C16:0 (ratio) G = 0.004 × 10−5; T = 0.002 × 10−2; fed vs Le = 0.002 × 10−5; day 2 fed vs Pl = 0.008 × 10−1 & fed vs Le = 0.008 × 10−1; day 3 fed vs Le = 0.001 × 10−1 & Le vs Pl = 0.002. C16:1 (ratio) G = 0.002 × 10−6; T = 0.004 × 10−2; fed vs Le = 0.007 × 10−7; day 1 fed vs Pl = 0.012; day 2 fed vs Pl = 0.009 & fed vs Le = 0.001 × 10−1; day 3 fed vs Le = 0.001 × 10−1 & Le vs Pl = 0.014. C18:0 (ratio) day 3 fed vs Le = 0.001 × 10−1 & Le vs Pl = 0.008. C18:1n9cis (ratio) G = 0.001 × 10−6; fed vs Le = 0.006 × 10−7; day 1 fed vs Le = 0.022; day 2 fed vs Pl = 0.001 & fed vs Le = 0.001 × 10−1; day 3 fed vs Le = 0.001 × 10−1 & Le vs Pl = 0.039. C18:2n6cis (ratio) T = 0.001 × 10−1; C20:4n6 (ratio) G = 0.002 × 10−1; T = 0.002 × 10−1; fed vs Le = 0.001 × 10−1; day 2 fed vs Pl = 0.022 & fed vs Le = 0.004; day 3 fed vs Le = 0.001 × 10−1 & Le vs Pl = 0.008 × 10−1. C20:5n3 (ratio) day 2 fed vs Le = 0.025; day 3 fed vs Le = 0.004 & Le vs Pl = 0.038. C20:3n6 (ratio) T = 0.006 × 10−12; C22:0 (ratio) T = 0.005 × 10−6; C22:6n3 (ratio) T = 0.001 × 10−3; day 2 fed vs Pl = 0.004 & fed vs Le = 0.004; day 3 fed vs Le = 0.003 × 10−1 & Le vs Pl = 0.011. bG = 0.004 × 10−11; T = 0.005 × 10−13; G*T = 0.004 × 10−3; fed vs Le = 0.005 × 10−10; fed vs Pl = 0.003 × 10−6; day 2 fed vs Le 0.002 × 10−7 & fed vs Pl = 0.004 × 10−5; day 3 fed vs Le = 0.008 × 10−9 & fed vs Pl = 0.001 × 10−10. c Post hoc test at 15 days = 0.002. d Post hoc test at 8 weeks = 0.011 and at 16 weeks = 0.002.

References

    1. Frederich RC, et al. Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin action. Nat. Med. 1995;1:1311–1314. doi: 10.1038/nm1295-1311.
    1. Considine RV, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N. Engl. J. Med. 1996;334:292–295. doi: 10.1056/NEJM199602013340503.
    1. Boden G, Chen X, Mozzoli M, Ryan I. Effect of fasting on serum leptin in normal human subjects. J. Clin. Endocrinol. Metab. 1996;81:3419–3423.
    1. Wu B, et al. Effect of repeated fasting/refeeding on body weight control and energy balance regulation in rats. Wei Sheng Yan Jiu. 2010;39:601–605.
    1. Audi L, et al. Leptin in relation to resumption of menses in women with anorexia nervosa. Mol. Psychiatry. 1998;3:544–547. doi: 10.1038/sj.mp.4000418.
    1. Halaas JL, et al. Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 1995;269:543–546. doi: 10.1126/science.7624777.
    1. Singh A, et al. Leptin-mediated changes in hepatic mitochondrial metabolism, structure, and protein levels. Proc. Natl Acad. Sci. USA. 2009;106:13100–13105. doi: 10.1073/pnas.0903723106.
    1. Zeng W, et al. Sympathetic neuro-adipose connections mediate leptin-driven lipolysis. Cell. 2015;163:84–94. doi: 10.1016/j.cell.2015.08.055.
    1. Halaas JL, et al. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc. Natl Acad. Sci. USA. 1997;94:8878–8883. doi: 10.1073/pnas.94.16.8878.
    1. Brown RJ, et al. Long-term effectiveness and safety of metreleptin in the treatment of patients with generalized lipodystrophy. Endocrine. 2018;60:479–489. doi: 10.1007/s12020-018-1589-1.
    1. Farooqi IS, et al. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J. Clin. Investig. 2002;110:1093–1103. doi: 10.1172/JCI0215693.
    1. Lee JH, Chan JL, Sourlas E, Raptopoulos V, Mantzoros CS. Recombinant methionyl human leptin therapy in replacement doses improves insulin resistance and metabolic profile in patients with lipoatrophy and metabolic syndrome induced by the highly active antiretroviral therapy. J. Clin. Endocrinol. Metab. 2006;91:2605–2611. doi: 10.1210/jc.2005-1545.
    1. Oral EA, et al. Long-term effectiveness and safety of metreleptin in the treatment of patients with partial lipodystrophy. Endocrine. 2019;64:500–511. doi: 10.1007/s12020-019-01862-8.
    1. Heymsfield SB, et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. J. Am. Med. Assoc. 1999;282:1568–1575. doi: 10.1001/jama.282.16.1568.
    1. Mittendorfer B, et al. Recombinant human leptin treatment does not improve insulin action in obese subjects with type 2 diabetes. Diabetes. 2011;60:1474–1477. doi: 10.2337/db10-1302.
    1. Moon HS, et al. Efficacy of metreleptin in obese patients with type 2 diabetes: cellular and molecular pathways underlying leptin tolerance. Diabetes. 2011;60:1647–1656. doi: 10.2337/db10-1791.
    1. Shetty GK, et al. Leptin administration to overweight and obese subjects for 6 months increases free leptin concentrations but does not alter circulating hormones of the thyroid and IGF axes during weight loss induced by a mild hypocaloric diet. Eur. J. Endocrinol. 2011;165:249–254. doi: 10.1530/EJE-11-0252.
    1. Fogteloo AJ, Pijl H, Frolich M, McCamish M, Meinders AE. Effects of recombinant human leptin treatment as an adjunct of moderate energy restriction on body weight, resting energy expenditure and energy intake in obese humans. Diabetes, Nutr. Metab. 2003;16:109–114.
    1. Hukshorn CJ, et al. Weekly subcutaneous pegylated recombinant native human leptin (PEG-OB) administration in obese men. J. Clin. Endocrinol. Metab. 2000;85:4003–4009. doi: 10.1210/jcem.85.11.6955.
    1. Hukshorn CJ, et al. The effect of pegylated recombinant human leptin (PEG-OB) on weight loss and inflammatory status in obese subjects. Int. J. Obes. Relat. Metab. Disord.: J. Int. Assoc. Study Obes. 2002;26:504–509. doi: 10.1038/sj.ijo.0801952.
    1. Zelissen PM, et al. Effect of three treatment schedules of recombinant methionyl human leptin on body weight in obese adults: a randomized, placebo-controlled trial. Diabetes, Obes. Metab. 2005;7:755–761. doi: 10.1111/j.1463-1326.2005.00468.x.
    1. Lejeune MP, Hukshorn CJ, Saris WH, Westerterp-Plantenga MS. Effect of dietary restraint during and following pegylated recombinant leptin (PEG-OB) treatment of overweight men. Int. J. Obes. Relat. Metab. Disord.: J. Int. Assoc. Study Obes. 2003;27:1494–1499. doi: 10.1038/sj.ijo.0802431.
    1. DePaoli A, Long A, Fine GM, Stewart M, O’Rahilly S. Efficacy of metreleptin for weight loss in overweight and obese adults with low leptin levels. Diabetes. 2018;67:296. doi: 10.2337/db18-296-LB.
    1. Flier JS, Maratos-Flier E. Leptin’s physiologic role: does the emperor of energy balance have no clothes? Cell Metab. 2017;26:24–26. doi: 10.1016/j.cmet.2017.05.013.
    1. Friedman JM. Leptin and the endocrine control of energy balance. Nat. Metab. 2019;1:754–764. doi: 10.1038/s42255-019-0095-y.
    1. Chan JL, Heist K, DePaoli AM, Veldhuis JD, Mantzoros CS. The role of falling leptin levels in the neuroendocrine and metabolic adaptation to short-term starvation in healthy men. J. Clin. Investig. 2003;111:1409–1421. doi: 10.1172/JCI200317490.
    1. Chan JL, et al. Differential regulation of metabolic, neuroendocrine, and immune function by leptin in humans. Proc. Natl Acad. Sci. USA. 2006;103:8481–8486. doi: 10.1073/pnas.0505429103.
    1. Chou SH, et al. Leptin is an effective treatment for hypothalamic amenorrhea. Proc. Natl Acad. Sci. USA. 2011;108:6585–6590. doi: 10.1073/pnas.1015674108.
    1. Welt CK, et al. Recombinant human leptin in women with hypothalamic amenorrhea. N. Engl. J. Med. 2004;351:987–997. doi: 10.1056/NEJMoa040388.
    1. Chan JL, Wong SL, Mantzoros CS. Pharmacokinetics of subcutaneous recombinant methionyl human leptin administration in healthy subjects in the fed and fasting states: regulation by gender and adiposity. Clin. Pharmacokinetics. 2008;47:753–764. doi: 10.2165/00003088-200847110-00006.
    1. Oral EA, et al. Leptin-replacement therapy for lipodystrophy. N. Engl. J. Med. 2002;346:570–578. doi: 10.1056/NEJMoa012437.
    1. Mackowiak PA, Wasserman SS, Levine MM. A critical appraisal of 98.6 degrees F, the upper limit of the normal body temperature, and other legacies of Carl Reinhold August Wunderlich. J. Am. Med. Assoc. 1992;268:1578–1580. doi: 10.1001/jama.1992.03490120092034.
    1. Gilgen-Ammann R, et al. Energy expenditure estimation from respiration variables. Sci. Rep. 2017;7:15995. doi: 10.1038/s41598-017-16135-5.
    1. Steinhauser ML, et al. The circulating metabolome of human starvation. JCI insight. 2018;3:e121434. doi: 10.1172/jci.insight.121434.
    1. Perry RJ, et al. Leptin mediates a glucose-fatty acid cycle to maintain glucose homeostasis in starvation. Cell. 2018;172:234–248. doi: 10.1016/j.cell.2017.12.001.
    1. Rosenbaum M, Leibel RL. Physiological responses to leptin levels in lipodystrophy: a model for other hypoleptinemias? J. Clin. Investig. 2018;128:3237–3239. doi: 10.1172/JCI122042.
    1. Pelleymounter MA, et al. Effects of the obese gene product on body weight regulation in ob/ob mice. Science. 1995;269:540–543. doi: 10.1126/science.7624776.
    1. Tang GB, Tang XF, Li K, Wang DH. Intracerebroventricular administration of leptin increase physical activity but has no effect on thermogenesis in cold-acclimated rats. Sci. Rep. 2015;5:11189. doi: 10.1038/srep11189.
    1. Pandit R, Beerens S, Adan RAH. Role of leptin in energy expenditure: the hypothalamic perspective. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2017;312:R938–R947. doi: 10.1152/ajpregu.00045.2016.
    1. Correia ML, Morgan DA, Sivitz WI, Mark AL, Haynes WG. Leptin acts in the central nervous system to produce dose-dependent changes in arterial pressure. Hypertension. 2001;37:936–942. doi: 10.1161/01.HYP.37.3.936.
    1. Machleidt F, et al. Experimental hyperleptinemia acutely increases vasoconstrictory sympathetic nerve activity in healthy humans. J. Clin. Endocrinol. Metab. 2013;98:E491–E496. doi: 10.1210/jc.2012-3009.
    1. Morton GJ, et al. Identification of a physiological role for leptin in the regulation of ambulatory activity and wheel running in mice. Am. J. Physiol. Endocrinol. Metab. 2011;300:E392–E401. doi: 10.1152/ajpendo.00546.2010.
    1. Satoh N, et al. Sympathetic activation of leptin via the ventromedial hypothalamus: leptin-induced increase in catecholamine secretion. Diabetes. 1999;48:1787–1793. doi: 10.2337/diabetes.48.9.1787.
    1. Shibuya I, et al. Regulation of catecholamine synthesis by leptin. Ann. N. Y Acad. Sci. 2002;971:522–527. doi: 10.1111/j.1749-6632.2002.tb04517.x.
    1. Korner J, et al. Randomized double-blind placebo-controlled study of leptin administration after gastric bypass. Obesity. 2013;21:951–956. doi: 10.1002/oby.20433.
    1. Kissileff HR, et al. Leptin reverses declines in satiation in weight-reduced obese humans. Am. J. Clin. Nutr. 2012;95:309–317. doi: 10.3945/ajcn.111.012385.
    1. Rosenbaum M, et al. Low-dose leptin reverses skeletal muscle, autonomic, and neuroendocrine adaptations to maintenance of reduced weight. J. Clin. Investig. 2005;115:3579–3586. doi: 10.1172/JCI25977.
    1. Rosenbaum M, Sy M, Pavlovich K, Leibel RL, Hirsch J. Leptin reverses weight loss-induced changes in regional neural activity responses to visual food stimuli. J. Clin. Investig. 2008;118:2583–2591.
    1. Ahima RS, et al. Role of leptin in the neuroendocrine response to fasting. Nature. 1996;382:250–252. doi: 10.1038/382250a0.
    1. Hwa JJ, et al. Leptin increases energy expenditure and selectively promotes fat metabolism in ob/ob mice. Am. J. Physiol. 1997;272:R1204–R1209.
    1. Lawler, K. et al. Leptin-mediated changes in the human metabolome. J. Clin. Endocrinol. Metab.105, dgaa251 (2020).
    1. Grewal S, et al. Metabolomic analysis of the effects of leptin replacement therapy in patients with lipodystrophy. J. Endocr. Soc. 2020;4:bvz022. doi: 10.1210/jendso/bvz022.
    1. Clemmons DR. Metabolic actions of insulin-like growth factor-I in normal physiology and diabetes. Endocrinol. Metab. Clin. North Am. 2012;41:425–443. doi: 10.1016/j.ecl.2012.04.017.
    1. Kaklamani VG, et al. Dietary fat and carbohydrates are independently associated with circulating insulin-like growth factor 1 and insulin-like growth factor-binding protein 3 concentrations in healthy adults. J. Clin. Oncol. 1999;17:3291–3298. doi: 10.1200/JCO.1999.17.10.3291.
    1. Kirsten R, et al. Effects of aldosterone on lipid metabolism and renal oxygen consumption in the rat. Pflug. Arch. 1977;368:189–194. doi: 10.1007/BF00585195.
    1. Shimomura I, Hammer RE, Ikemoto S, Brown MS, Goldstein JL. Leptin reverses insulin resistance and diabetes mellitus in mice with congenital lipodystrophy. Nature. 1999;401:73–76. doi: 10.1038/43448.
    1. Puschel J, et al. Beneficial effects of leptin substitution on impaired eating behavior in lipodystrophy are sustained beyond 150 weeks of treatment. Cytokine. 2019;113:400–404. doi: 10.1016/j.cyto.2018.10.012.
    1. Schlogl H, et al. Leptin substitution in patients with lipodystrophy: neural correlates for long-term success in the normalization of eating behavior. Diabetes. 2016;65:2179–2186. doi: 10.2337/db15-1550.
    1. Farr OM, et al. Leptin therapy alters appetite and neural responses to food stimuli in brain areas of leptin-sensitive subjects without altering brain structure. J. Clin. Endocrinol. Metab. 2014;99:E2529–E2538. doi: 10.1210/jc.2014-2774.
    1. Westerterp-Plantenga MS, Saris WH, Hukshorn CJ, Campfield LA. Effects of weekly administration of pegylated recombinant human OB protein on appetite profile and energy metabolism in obese men. Am. J. Clin. Nutr. 2001;74:426–434. doi: 10.1093/ajcn/74.4.426.
    1. Dallner OS, et al. Dysregulation of a long noncoding RNA reduces leptin leading to a leptin-responsive form of obesity. Nat. Med. 2019;25:507–516. doi: 10.1038/s41591-019-0370-1.
    1. Chan JL, Mietus JE, Raciti PM, Goldberger AL, Mantzoros CS. Short-term fasting-induced autonomic activation and changes in catecholamine levels are not mediated by changes in leptin levels in healthy humans. Clin. Endocrinol. 2007;66:49–57.
    1. Ainsworth BE, et al. 2011 Compendium of physical activities: a second update of codes and MET values. Med. Sci. sports Exerc. 2011;43:1575–1581. doi: 10.1249/MSS.0b013e31821ece12.
    1. Sumner LW, et al. Proposed minimum reporting standards for chemical analysis Chemical Analysis Working Group (CAWG) Metabolomics Standards Initiative (MSI) Metabolomics. 2007;3:211–221. doi: 10.1007/s11306-007-0082-2.
    1. Soininen P, Kangas AJ, Wurtz P, Suna T, Ala-Korpela M. Quantitative serum nuclear magnetic resonance metabolomics in cardiovascular epidemiology and genetics. Circ. Cardiovasc. Genet. 2015;8:192–206. doi: 10.1161/CIRCGENETICS.114.000216.
    1. Soininen P, et al. High-throughput serum NMR metabonomics for cost-effective holistic studies on systemic metabolism. Analyst. 2009;134:1781–1785. doi: 10.1039/b910205a.
    1. Inouye M, et al. Metabonomic, transcriptomic, and genomic variation of a population cohort. Mol. Syst. Biol. 2010;6:441. doi: 10.1038/msb.2010.93.
    1. Zuber V, Colijn JM, Klaver C, Burgess S. Selecting likely causal risk factors from high-throughput experiments using multivariable Mendelian randomization. Nat. Commun. 2020;11:29. doi: 10.1038/s41467-019-13870-3.
    1. Hagenbeek FA, et al. Heritability estimates for 361 blood metabolites across 40 genome-wide association studies. Nat. Commun. 2020;11:39. doi: 10.1038/s41467-019-13770-6.
    1. Vojinovic D, et al. Relationship between gut microbiota and circulating metabolites in population-based cohorts. Nat. Commun. 2019;10:5813. doi: 10.1038/s41467-019-13721-1.
    1. Gallois A, et al. A comprehensive study of metabolite genetics reveals strong pleiotropy and heterogeneity across time and context. Nat. Commun. 2019;10:4788. doi: 10.1038/s41467-019-12703-7.
    1. Deelen J, et al. A metabolic profile of all-cause mortality risk identified in an observational study of 44,168 individuals. Nat. Commun. 2019;10:3346. doi: 10.1038/s41467-019-11311-9.
    1. Kettunen J, et al. Genome-wide study for circulating metabolites identifies 62 loci and reveals novel systemic effects of LPA. Nat. Commun. 2016;7:11122. doi: 10.1038/ncomms11122.
    1. Holmes MV, et al. Lipids, lipoproteins, and metabolites and risk of myocardial infarction and stroke. J. Am. Coll. Cardiol. 2018;71:620–632. doi: 10.1016/j.jacc.2017.12.006.
    1. Wurtz P, Soininen P. Reply to: “Methodological issues regarding: “a third of nonfasting plasma cholesterol is in remnant lipoproteins: lipoprotein subclass profiling in 9293 individuals””. Atherosclerosis. 2020;302:59–61. doi: 10.1016/j.atherosclerosis.2020.03.028.
    1. Liu J, et al. Integration of epidemiologic, pharmacologic, genetic and gut microbiome data in a drug-metabolite atlas. Nat. Med. 2020;26:110–117. doi: 10.1038/s41591-019-0722-x.
    1. Julkunen, H., Cichonska, A., Slagboom, P. E. & Würtz, P. Blood biomarker score identifies individuals at high risk for severe COVID-19 a decade prior to diagnosis: metabolic profiling of 105,000 adults in the UK Biobank. Preprint at 10.1101/2020.07.02.20143685 (2020).
    1. Agren JJ, Julkunen A, Penttila I. Rapid separation of serum lipids for fatty acid analysis by a single aminopropyl column. J. Lipid Res. 1992;33:1871–1876.
    1. Chong J, Xia J. MetaboAnalystR: an R package for flexible and reproducible analysis of metabolomics data. Bioinformatics. 2018;34:4313–4314. doi: 10.1093/bioinformatics/bty528.

Source: PubMed

3
Subscribe