Synergistic effects of leucine and resveratrol on insulin sensitivity and fat metabolism in adipocytes and mice

Antje Bruckbauer, Michael B Zemel, Teresa Thorpe, Murthy R Akula, Alan C Stuckey, Dustin Osborne, Emily B Martin, Stephen Kennel, Jonathan S Wall, Antje Bruckbauer, Michael B Zemel, Teresa Thorpe, Murthy R Akula, Alan C Stuckey, Dustin Osborne, Emily B Martin, Stephen Kennel, Jonathan S Wall

Abstract

Background: Sirtuins are important regulators of glucose and fat metabolism, and sirtuin activation has been proposed as a therapeutic target for insulin resistance and diabetes. We have shown leucine to increase mitochondrial biogenesis and fat oxidation via Sirt1 dependent pathways. Resveratrol is a widely recognized activator of Sirt; however, the biologically-effective high concentrations used in cell and animal studies are generally impractical or difficult to achieve in humans. Accordingly, we sought to determine whether leucine would exhibit synergy with low levels of resveratrol on sirtuin-dependent outcomes in adipocytes and in diet-induced obese (DIO) mice.

Methods: 3T3-L1 mouse adipocytes were treated with Leucine (0.5 mM), β-hydroxy-β-methyl butyrate (HMB) (5 μM) or Resveratrol (200 nM) alone or in combination. In addition, diet-induced obese mice were treated for 6-weeks with low (2 g/kg diet) or high (10 g/kg diet) dose HMB, Leucine (24 g/kg diet; 200% of normal level) or low (12.5 mg/kg diet) or high (225 mg/kg diet) dose resveratrol, alone or as combination with leucine-resveratrol or HMB-resveratrol.

Results: Fatty acid oxidation, AMPK, Sirt1 and Sirt3 activity in 3T3-L1 adipocytes and in muscle cells, were significantly increased by the combinations compared to the individual treatments. Similarly, 6-week feeding of low-dose resveratrol combined with either leucine or its metabolite HMB to DIO mice increased adipose Sirt1 activity, muscle glucose and palmitate uptake (measured via PET/CT), insulin sensitivity (HOMAIR), improved inflammatory stress biomarkers (CRP, IL-6, MCP-1, adiponectin) and reduced adiposity comparable to the effects of high dose resveratrol, while low-dose resveratrol exerted no independent effect.

Conclusion: These data demonstrate that either leucine or its metabolite HMB may be combined with a low concentration of resveratrol to exert synergistic effects on Sirt1-dependent outcomes; this may result in more practical dosing of resveratrol in the management of obesity, insulin-resistance and diabetes.

Figures

Figure 1
Figure 1
Leucine and HMB Synergize with Resvetratrol in activation of Sirt1 and Sirt 3 activity. a) Sirt1 in Muscle Cells. C2C12 muscle cells were incubated with indicated treatments under low glucose (5 mM) conditions and Sirt1 activity was measured. Data are expressed as mean ± SE (n = 7 to 9) and are expressed as % change from control; control = 1085 ± 41 AFU/mg protein. * indicates significant difference compared to control (p < 0.04), ** indicates significant difference compared to control, leucine and HMB (p < 0.01). b) Sirt1 in Adipocytes. 3T3-L1 mouse adipocytes were incubated with indicated treatments under low glucose (5 mM) conditions and Sirt1 activity was measured. Data are expressed as mean ± SE (n = 7 to 9) and are expressed as % change from control; control = 759 ± 63 AFU/mg protein. * indicates significant difference compared to control (p < 0.05), ** indicates significant difference compared to control, leucine and HMB (p < 0.01).c) Sirt3 in Muscle Cells. C2C12 muscle cells were incubated with indicated treatments under low glucose (5 mM) conditions and Sirt1 activity was measured. Data are expressed as mean ± SE (n = 7 to 9) and are expressed as % change from control; control = 410 ± 57 AFU/mg protein. * indicates significant difference compared to control (p < 0.03), ** indicates significant difference compared to control, leucine and HMB (p < 0.02). d) Sirt3 in Adipocytes. 3T3-L1 mouse adipocytes were incubated with indicated treatments for 4 hours under low glucose (5 mM) conditions . Mitochondrial protein was isolated and Sirt3 activity was measured. Data are presented as mean ± SE (n = 6) and are expressed as % change from control; control = 507.8 ± 20.5 AFU/mg protein. * indicates significant difference compared to control (p = 0.03).
Figure 2
Figure 2
Leucine and HMB Synergize with Resveratrol to stimulate AMPK activity in 3T3L1 adipocytes. 3T3-L1 mouse adipocytes were incubated with indicated treatments for 24 hours and AMPK activity was measured. Data are presented as mean ± SE (n = 4). a indicates significant difference to control (p < 0.04), b indicates significant difference to control, HMB and leucine (p < 0.03), c indicates significant difference to control and leucine (p < 0.03).
Figure 3
Figure 3
Leucine and HMB Synergize with Resveratrol to Stimulate Fatty Acid Oxidation under a) Low Glucose Conditions and b) High Glucose Conditions. 3T3-L1 mouse adipocytes were incubated with indicated treatments for 4 hours under a) low glucose (5 mM) or b) high glucose (25 mM) conditions. Data are presented as mean ± SE (n = 6) and are expressed as % stimulation over control, where low glucose control = 193 ± 39 cpm/ng DNA and high glucose control = 302 ± 24 cpm/ng DNA. Stars above the bars indicate significant difference * compared to control (p < 0.05), ** compared to control, Leucine, and HMB (p < 0.005).
Figure 4
Figure 4
Resveratrol-HMB synergy in glucose uptake using FDG-PET. Mice were fed a high fat-diet with indicated treatments for 6 weeks. At the end of the treatment period, Fluorine-18-deoxy-glucose (FDG) or palmitate PET/CT scans were performed to measure whole body glucose (a) or fat uptake (b). Representative images of control diet group and resveratrol/low HMB diet group are shown
Figure 5
Figure 5
Effects of resveratrol, leucine and HMB on adipose tissue Sirt1 activity in diet-induced obese mice. Mice were fed a high fat-diet with indicated treatments for 6 weeks. At the end of the treatment period, Sirt1 activity in adipose tissue was measured. Data are presented as means ± SE (n = 9 to 10). Stars above the bars indicate significant difference compared to control (p < 0.02).

References

    1. Stipanuk MH. Leucine and protein synthesis: mTOR and beyond. Nutr Rev. 2007;65(3):122–129. doi: 10.1111/j.1753-4887.2007.tb00289.x.
    1. Zanchi NE, Nicastro H, Lancha AH Jr. Potential antiproteolytic effects of L-leucine: observations of in vitro and in vivo studies. Nutr Metab (Lond) 2008;5:20. doi: 10.1186/1743-7075-5-20.
    1. Lynch CJ, Patson BJ, Anthony J, Vaval A, Jefferson LS, Vary TC. Leucine is a direct-acting nutrient signal that regulates protein synthesis in adipose tissue. Am J Physiol Endocrinol Metab. 2002;283(3):E503–E513.
    1. Sun X, Zemel M. Leucine and calcium regulate fat metabolism and energy partitioning in murine adipocytes and muscle cells. Lipids. 2007;42(4):297–305. doi: 10.1007/s11745-007-3029-5.
    1. Sun X, Zemel MB. Leucine modulation of mitochondrial mass and oxygen consumption in skeletal muscle cells and adipocytes. Nutr Metab (Lond) 2009;6:26. doi: 10.1186/1743-7075-6-26.
    1. Donato J Jr, Pedrosa RG, Cruzat VF, Pires IS, Tirapegui J. Effects of leucine supplementation on the body composition and protein status of rats submitted to food restriction. Nutrition. 2006;22(5):520–527. doi: 10.1016/j.nut.2005.12.008.
    1. Zhang Y, Guo K, LeBlanc RE, Loh D, Schwartz GJ, Yu YH. Increasing dietary leucine intake reduces diet-induced obesity and improves glucose and cholesterol metabolism in mice via multimechanisms. Diabetes. 2007;56(6):1647–1654. doi: 10.2337/db07-0123.
    1. Bruckbauer A, Zemel MB. Effects of dairy consumption on SIRT1 and mitochondrial biogenesis in adipocytes and muscle cells. Nutr Metab (Lond) 2011;8:91. doi: 10.1186/1743-7075-8-91.
    1. Bruckbauer A, Gouffon J, Rekapalli B, Zemel MB. The effects of dairy components on energy partitioning and metabolic risk in mice: a microarray study. J Nutrigenet Nutrigenomics. 2009;2(2):64–77. doi: 10.1159/000205936.
    1. Haigis MC, Guarente LP. Mammalian sirtuins–emerging roles in physiology, aging, and calorie restriction. Genes Dev. 2006;20(21):2913–2921. doi: 10.1101/gad.1467506.
    1. Verdin E, Hirschey MD, Finley LW, Haigis MC. Sirtuin regulation of mitochondria: energy production, apoptosis, and signaling. Trends Biochem Sci. 2010;35(12):669–675. doi: 10.1016/j.tibs.2010.07.003.
    1. Carafa V, Nebbioso A, Altucci L. Sirtuins and disease: the road ahead. Front Pharmacol. 2012;3:4.
    1. Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA, Mayo MW. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004;23(12):2369–2380. doi: 10.1038/sj.emboj.7600244.
    1. Nemoto S, Fergusson MM, Finkel T. SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1{alpha} J Biol Chem. 2005;280(16):16456–16460. doi: 10.1074/jbc.M501485200.
    1. Martinez-Pastor B, Mostoslavsky R. Sirtuins, metabolism, and cancer. Front Pharmacol. 2012;3:22.
    1. Sun C, Zhang F, Ge X, Yan T, Chen X, Shi X, Zhai Q. SIRT1 improves insulin sensitivity under insulin-resistant conditions by repressing PTP1B. Cell Metab. 2007;6(4):307–319. doi: 10.1016/j.cmet.2007.08.014.
    1. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K. et al.Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444(7117):337–342. doi: 10.1038/nature05354.
    1. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, Messadeq N, Milne J, Lambert P, Elliott P. et al.Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006;127(6):1109–1122. doi: 10.1016/j.cell.2006.11.013.
    1. Park SJ, Ahmad F, Philp A, Baar K, Williams T, Luo H, Ke H, Rehmann H, Taussig R, Brown AL. et al.Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases. Cell. 2012;148(3):421–433. doi: 10.1016/j.cell.2012.01.017.
    1. Price NL, Gomes AP, Ling AJ, Duarte FV, Martin-Montalvo A, North BJ, Agarwal B, Ye L, Ramadori G, Teodoro JS. et al.SIRT1 Is Required for AMPK Activation and the Beneficial Effects of Resveratrol on Mitochondrial Function. Cell Metab. 2012;15(5):675–690. doi: 10.1016/j.cmet.2012.04.003.
    1. Baur JA, Sinclair DA. Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov. 2006;5(6):493–506. doi: 10.1038/nrd2060.
    1. Crandall JP, Oram V, Trandafirescu G, Reid M, Kishore P, Hawkins M, Cohen HW, Barzilai N. Pilot Study of Resveratrol in Older Adults With Impaired Glucose Tolerance. J Gerontol A Biol Sci Med Sci. 2012.
    1. Mukherjee S, Dudley JI, Das DK. Dose-dependency of resveratrol in providing health benefits. Dose–response. 2010;8(4):478–500. doi: 10.2203/dose-response.09-015.Mukherjee.
    1. Timmer S, Auwerx J, Schrauwen P. The journey of resveratrol from yeast to human. Aging (Albany NY) 2012;4(3):146–158.
    1. Smoliga JM, Vang O, Baur JA. Challenges of translating basic research into therapeutics: resveratrol as an example. J Gerontol A Biol Sci Med Sci. 2011;67(2):158–167.
    1. Smoliga JM, Baur JA, Hausenblas HA. Resveratrol and health–a comprehensive review of human clinical trials. Mol Nutr Food Res. 2011;55(8):1129–1141. doi: 10.1002/mnfr.201100143.
    1. Boocock DJ, Faust GE, Patel KR, Schinas AM, Brown VA, Ducharme MP, Booth TD, Crowell JA, Perloff M, Gescher AJ. et al.Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiol Biomarkers Prev. 2007;16(6):1246–1252. doi: 10.1158/1055-9965.EPI-07-0022.
    1. Brand HS, Jorning GG, Chamuleau RA, Abraham-Inpijn L. Effect of a protein-rich meal on urinary and salivary free amino acid concentrations in human subjects. Clin Chim Acta. 1997;264(1):37–47. doi: 10.1016/S0009-8981(97)00070-3.
    1. Almeida L, Vaz-da-Silva M, Falcao A, Soares E, Costa R, Loureiro AI, Fernandes-Lopes C, Rocha JF, Nunes T, Wright L. et al.Pharmacokinetic and safety profile of trans-resveratrol in a rising multiple-dose study in healthy volunteers. Mol Nutr Food Res. 2009;53(Suppl 1):S7–15.
    1. Barger JL, Kayo T, Vann JM, Arias EB, Wang J, Hacker TA, Wang Y, Raederstorff D, Morrow JD, Leeuwenburgh C. et al.A low dose of dietary resveratrol partially mimics caloric restriction and retards aging parameters in mice. PLoS One. 2008;3(6):e2264. doi: 10.1371/journal.pone.0002264.
    1. Nin V, Escande C, Chini CC, Giri S, Camacho-Pereira J, Matalonga J, Lou Z, Chini EN. Role of Deleted in Breast Cancer 1 (DBC1) Protein in SIRT1 Deacetylase Activation Induced by Protein Kinase A and AMP-activated Protein Kinase. J Biol Chem. 2012;287(28):23489–23501. doi: 10.1074/jbc.M112.365874.
    1. Taylor M, Wallhaus TR, Degrado TR, Russell DC, Stanko P, Nickles RJ, Stone CK. An evaluation of myocardial fatty acid and glucose uptake using PET with [18 F]fluoro-6-thia-heptadecanoic acid and [18 F]FDG in Patients with Congestive Heart Failure. J Nucl Med. 2001;42(1):55–62.
    1. Guiducci L, Gronroos T, Jarvisalo MJ, Kiss J, Viljanen A, Naum AG, Viljanen T, Savunen T, Knuuti J, Ferrannini E. et al.Biodistribution of the fatty acid analogue 18 F-FTHA: plasma and tissue partitioning between lipid pools during fasting and hyperinsulinemia. J Nucl Med. 2007;48(3):455–462.
    1. LeBlanc AK, Akula M, Martin EB, Rowe JA, Galyon GD, Moyers TD, Stuckey AC, Long MJ, Wall JS, Kennel SJ, World Molecular Imaging Congress. Poster Presentation with Abstract, San Diego, CA; 2011. Whole-body distribution of 14(R,S)-[18F]fluoro-6-thia-heptadecanoic acid with Positron Emission Tomography/Computed Tomography (PET/CT) in domestic cats. September 7th - 10th.
    1. Sugawara Y, Zasadny KR, Neuhoff AW, Wahl RL. Reevaluation of the standardized uptake value for FDG: variations with body weight and methods for correction. Radiology. 1999;213(2):521–525.
    1. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature. 2005;434(7029):113–118. doi: 10.1038/nature03354.
    1. Yu J, Auwerx J. The role of sirtuins in the control of metabolic homeostasis. Ann N Y Acad Sci. 2009;1173(Suppl 1):E10–19.
    1. Yoshizaki T, Schenk S, Imamura T, Babendure JL, Sonoda N, Bae EJ, Oh DY, Lu M, Milne JC, Westphal C. et al.SIRT1 inhibits inflammatory pathways in macrophages and modulates insulin sensitivity. Am J Physiol Endocrinol Metab. 2010;298(3):E419–428. doi: 10.1152/ajpendo.00417.2009.
    1. Milne JC, Lambert PD, Schenk S, Carney DP, Smith JJ, Gagne DJ, Jin L, Boss O, Perni RB, Vu CB. et al.Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature. 2007;450(7170):712–716. doi: 10.1038/nature06261.
    1. Pearson KJ, Baur JA, Lewis KN, Peshkin L, Price NL, Labinskyy N, Swindell WR, Kamara D, Minor RK, Perez E. et al.Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab. 2008;8(2):157–168. doi: 10.1016/j.cmet.2008.06.011.
    1. Kang W, Hong HJ, Guan J, Kim DG, Yang EJ, Koh G, Park D, Han CH, Lee YJ, Lee DH. Resveratrol improves insulin signaling in a tissue-specific manner under insulin-resistant conditions only: in vitro and in vivo experiments in rodents. Metabolism. 2012;61(3):424–433. doi: 10.1016/j.metabol.2011.08.003.
    1. Robich MP, Osipov RM, Chu LM, Han Y, Feng J, Nezafat R, Clements RT, Manning WJ, Sellke FW. Resveratrol modifies risk factors for coronary artery disease in swine with metabolic syndrome and myocardial ischemia. Eur J Pharmacol. 2011;664(1–3):45–53.
    1. Brasnyo P, Molnar GA, Mohas M, Marko L, Laczy B, Cseh J, Mikolas E, Szijarto IA, Merei A, Halmai R. et al.Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients. Br J Nutr. 2011;106(3):383–389. doi: 10.1017/S0007114511000316.
    1. Calabrese EJ, Mattson MP, Calabrese V. Resveratrol commonly displays hormesis: occurrence and biomedical significance. Hum Exp Toxicol. 2010;29(12):980–1015. doi: 10.1177/0960327110383625.
    1. Cho SJ, Jung UJ, Choi MS. Differential effects of low-dose resveratrol on adiposity and hepatic steatosis in diet-induced obese mice. Br J Nutr. 2012. pp. 1–10.
    1. Layman DK, Walker DA. Potential importance of leucine in treatment of obesity and the metabolic syndrome. J Nutr. 2006;136(1 Suppl):319S–323S.
    1. Layman DK. The role of leucine in weight loss diets and glucose homeostasis. J Nutr. 2003;133(1):261S–267S.
    1. Macotela Y, Emanuelli B, Bang AM, Espinoza DO, Boucher J, Beebe K, Gall W, Kahn R. Dietary Leucine - An Envrionmental Modifier of Insulin Resistance Acting on Multiple Levels of Metabolism. PLoS One. 2011;6(6):e21187. doi: 10.1371/journal.pone.0021187.
    1. Guo K, Yu YH, Hou J, Zhang Y. Chronic leucine supplementation improves glycemic control in etiologically distinct mouse models of obesity and diabetes mellitus. Nutr Metab (Lond) 2010;7:57. doi: 10.1186/1743-7075-7-57.
    1. Freudenberg A, Petzke KJ, Klaus S. Comparison of high-protein diets and leucine supplementation in the prevention of metabolic syndrome and related disorders in mice. J Nutr Biochem. 2012. doi:10.1016/j.jnutbio.2011.10.005.
    1. Salminen A, Hyttinen JM, Kaarniranta K. AMP-activated protein kinase inhibits NF-kappaB signaling and inflammation: impact on healthspan and lifespan. J Mol Med (Berl) 2011;89(7):667–676. doi: 10.1007/s00109-011-0748-0.
    1. Canto C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L, Milne JC, Elliott PJ, Puigserver P, Auwerx J. AMPK regulates energy expenditure by modulating NAD + metabolism and SIRT1 activity. Nature. 2009;458(7241):1056–1060. doi: 10.1038/nature07813.
    1. Yamauchi T, Kamon J, Minokoshi Y, Ito Y, Waki H, Uchida S, Yamashita S, Noda M, Kita S, Ueki K. et al.Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med. 2002;8(11):1288–1295. doi: 10.1038/nm788.

Source: PubMed

Подписаться