High-dose resveratrol supplementation in obese men: an investigator-initiated, randomized, placebo-controlled clinical trial of substrate metabolism, insulin sensitivity, and body composition

Morten M Poulsen, Poul F Vestergaard, Berthil F Clasen, Yulia Radko, Lars P Christensen, Hans Stødkilde-Jørgensen, Niels Møller, Niels Jessen, Steen B Pedersen, Jens Otto L Jørgensen, Morten M Poulsen, Poul F Vestergaard, Berthil F Clasen, Yulia Radko, Lars P Christensen, Hans Stødkilde-Jørgensen, Niels Møller, Niels Jessen, Steen B Pedersen, Jens Otto L Jørgensen

Abstract

Obesity, diabetes, hypertension, and hyperlipidemia constitute risk factors for morbidity and premature mortality. Based on animal and in vitro studies, resveratrol reverts these risk factors via stimulation of silent mating type information regulation 2 homolog 1 (SIRT1), but data in human subjects are scarce. The objective of this study was to examine the metabolic effects of high-dose resveratrol in obese human subjects. In a randomized, placebo-controlled, double-blinded, and parallel-group design, 24 obese but otherwise healthy men were randomly assigned to 4 weeks of resveratrol or placebo treatment. Extensive metabolic examinations including assessment of glucose turnover and insulin sensitivity (hyperinsulinemic euglycemic clamp) were performed before and after the treatment. Insulin sensitivity, the primary outcome measure, deteriorated insignificantly in both groups. Endogenous glucose production and the turnover and oxidation rates of glucose remained unchanged. Resveratrol supplementation also had no effect on blood pressure; resting energy expenditure; oxidation rates of lipid; ectopic or visceral fat content; or inflammatory and metabolic biomarkers. The lack of effect disagrees with persuasive data obtained from rodent models and raises doubt about the justification of resveratrol as a human nutritional supplement in metabolic disorders.

Trial registration: ClinicalTrials.gov NCT01150955.

Figures

FIG. 1.
FIG. 1.
Insulin sensitivity and glucose metabolism. Glucose metabolism was examined before and after 4 weeks of resveratrol or placebo supplementation. Black bars and black dots indicate placebo group (n = 12) and white bars and white dots indicate the resveratrol group (n = 12). Results are presented as group means ± SEM. Insulin sensitivity was assessed by a hyperinsulinemic euglycemic clamp. The participants were clamped at a blood glucose level of ∼5 mmol/L with an insulin infusion of 0.5 mU/kg/min. A and B: The GIR before and after intervention, respectively. P values reflect between-group differences assessed by two-way repeated-measures ANOVA. C: The corresponding whole-body insulin sensitivity, the M value, defined as the mean GIR during the last 30 min of the clamp. P value reflects the potential treatment effect analyzed by two-way repeated-measures ANOVA. D: Glucose Rd. E: EGP. F: Nonoxidative glucose disposal. *A statistically significant within-group effect of the insulin stimulation evaluated by a paired t test. Overall comparisons of potential treatment effects were performed by two-way repeated-measures ANOVA in the basal and clamp situations, respectively. NS, nonsignificant.
FIG. 2.
FIG. 2.
Blood pressure. Ambulatory blood pressure was measured automatically by a portable device every 20th minute over 24 h. Results are presented as box plots of the absolute changes from baseline after 4 weeks of placebo (n = 12) or resveratrol (n = 12) supplementation, respectively. The box boundaries indicate the 25th–75th percentile range, and error bars represent the 5th–95th percentiles. Solid and dotted lines represent median and mean, respectively. Resveratrol supplementation tends to increase both systolic and diastolic blood pressure; however, the elevations are not significant when compared with the placebo group by two-way repeated-measures ANOVA (systolic, P = 0.39; diastolic, P = 0.36).
FIG. 3.
FIG. 3.
Body composition. Absolute changes in essential body composition parameters after 4 weeks of placebo (n = 12) or resveratrol supplementation (n = 12). The box boundaries indicate the 25th–75th percentile range, and error bars represent the 5th–95th percentiles. Solid and dotted lines represent median and mean, respectively. A–D were assessed by whole-body dual-energy X-ray absorptiometry; E and F were quantified by MR imaging based on repetitive axial slices from the proximal border of the left kidney to the femur neck; nine were evaluable in each group. Two-way repeated-measures ANOVA revealed no statistically significant differences between groups.
FIG. 4.
FIG. 4.
Ectopic lipid. Relative changes in ectopic lipid deposition from baseline after 4 weeks of placebo or resveratrol supplementation, respectively. Results are generated by 1H-MR spectroscopy and raw data processing provides an estimate of the ratio of lipid to water (LWR) within the tissue. The box boundaries indicate the 25th–75th percentile range, and error bars represent the 5th–95th percentiles. Solid and dotted lines represent median and mean, respectively. Two-way repeated-measures ANOVA revealed no statistically significant differences between groups. A: Changes in hepatic lipid content; 11 were evaluable in each group. B: Changes in intramyocellular lipid (IMCL) content; 10 were evaluable in each group.
FIG. 5.
FIG. 5.
Gene expression and protein phosphorylation. Intracellular protein levels and relative mRNA expression in muscle (A–D) and adipose (E and F) tissue biopsies taken before and after 4 weeks' treatment with placebo (n = 12) or resveratrol (n = 12). Biopsies were taken before and during (20 min after initiation) a hyperinsulinemic euglycemic clamp. Black bars indicate the placebo group and white bars indicate the resveratrol group. Results are presented as group means ± SEM, and overall comparisons of potential treatment effects were performed by two-way repeated-measures ANOVA in the basal and clamp situations, respectively. A: Phosphorylation of the intracellular kinase AMP-activated protein kinase (AMPK) assessed by Western blot analysis in muscle tissue. B: Total acetylation of lysine residues assessed by Western blot analysis in muscle tissue. C: Relative GLUT4 mRNA expression in muscle tissue assessed by RT-PCR. D: Relative peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α) mRNA expression in muscle tissue assessed by RT-PCR. E: Relative tumor necrosis factor (TNF)-α mRNA expression in subcutaneous adipose tissue assessed by RT-PCR. F: Relative nuclear factor (NF)-κB mRNA expression in subcutaneous adipose tissue assessed by RT-PCR. *In C, we found an overall treatment effect in the clamp situation, and a post hoc test revealed a statistically significant (P < 0.05) decreased expression of GLUT4 in the resveratrol group.

References

    1. World Health Organization. 2012. Obesity and overweight. Fact sheet no. 311 [report online]. Available from Accessed 12 November 2012
    1. Wing RR, Hill JO. Successful weight loss maintenance. Annu Rev Nutr 2001;21:323–341
    1. Smoliga JM, Baur JA, Hausenblas HA. Resveratrol and health—a comprehensive review of human clinical trials. Mol Nutr Food Res 2011;55:1129–1141
    1. Stervbo U, Vang O, Bonnesen C. A review of the content of the putative chemopreventive phytoalexin resveratrol in red wine. Food Chem 2007;101:449–457
    1. Barger JL, Kayo T, Vann JM, et al. A low dose of dietary resveratrol partially mimics caloric restriction and retards aging parameters in mice. PLoS One 2008;3:e2264.
    1. Agarwal B, Baur JA. Resveratrol and life extension. Ann N Y Acad Sci 2011;1215:138–143
    1. Baur JA, Pearson KJ, Price NL, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006;444:337–342
    1. Lagouge M, Argmann C, Gerhart-Hines Z, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell 2006;127:1109–1122
    1. Pearson KJ, Baur JA, Lewis KN, et al. Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab 2008;8:157–168
    1. Vang O, Ahmad N, Baile CA, et al. What is new for an old molecule? Systematic review and recommendations on the use of resveratrol. PLoS One 2011;6:e19881.
    1. Chachay VS, Kirkpatrick CM, Hickman IJ, Ferguson M, Prins JB, Martin JH. Resveratrol—pills to replace a healthy diet? Br J Clin Pharmacol 2011;72:27–38
    1. Kang W, Hong HJ, Guan J, et al. Resveratrol improves insulin signaling in a tissue-specific manner under insulin-resistant conditions only: in vitro and in vivo experiments in rodents. Metabolism 2012;61:424–433
    1. Andersen G, Burkon A, Sulzmaier FJ, et al. High dose of dietary resveratrol enhances insulin sensitivity in healthy rats but does not lead to metabolite concentrations effective for SIRT1 expression. Mol Nutr Food Res 2011;55:1197–1206
    1. Marchal J, Blanc S, Epelbaum J, Aujard F, Pifferi F. Effects of chronic calorie restriction or dietary resveratrol supplementation on insulin sensitivity markers in a primate, Microcebus murinus. PLoS One 2012;7:e34289.
    1. Rivera L, Morón R, Zarzuelo A, Galisteo M. Long-term resveratrol administration reduces metabolic disturbances and lowers blood pressure in obese Zucker rats. Biochem Pharmacol 2009;77:1053–1063
    1. Cui X, Jin Y, Hofseth AB, et al. Resveratrol suppresses colitis and colon cancer associated with colitis. Cancer Prev Res (Phila) 2010;3:549–559
    1. Bujanda L, Hijona E, Larzabal M, et al. Resveratrol inhibits nonalcoholic fatty liver disease in rats. BMC Gastroenterol 2008;8:40.
    1. Jang M, Cai L, Udeani GO, et al. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997;275:218–220
    1. Provinciali M, Re F, Donnini A, et al. Effect of resveratrol on the development of spontaneous mammary tumors in HER-2/neu transgenic mice. Int J Cancer 2005;115:36–45
    1. Sengottuvelan M, Nalini N. Dietary supplementation of resveratrol suppresses colonic tumour incidence in 1,2-dimethylhydrazine-treated rats by modulating biotransforming enzymes and aberrant crypt foci development. Br J Nutr 2006;96:145–153
    1. Narayanan NK, Nargi D, Randolph C, Narayanan BA. Liposome encapsulation of curcumin and resveratrol in combination reduces prostate cancer incidence in PTEN knockout mice. Int J Cancer 2009;125:1–8
    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;108:2166–2175
    1. Gomez-Zorita S, Fernandez-Quintela A, Macarulla MT, et al. Resveratrol attenuates steatosis in obese Zucker rats by decreasing fatty acid availability and reducing oxidative stress. Br J Nutr 2012;107:202–210
    1. Shang J, Chen LL, Xiao FX, Sun H, Ding HC, Xiao H. Resveratrol improves non-alcoholic fatty liver disease by activating AMP-activated protein kinase. Acta Pharmacol Sin 2008;29:698–706
    1. Dal-Pan A, Blanc S, Aujard F. Resveratrol suppresses body mass gain in a seasonal non-human primate model of obesity. BMC Physiol 2010;10:11.
    1. Howitz KT, Bitterman KJ, Cohen HY, et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 2003;425:191–196
    1. Wood JG, Rogina B, Lavu S, et al. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature 2004;430:686–689
    1. Valenzano DR, Terzibasi E, Genade T, Cattaneo A, Domenici L, Cellerino A. Resveratrol prolongs lifespan and retards the onset of age-related markers in a short-lived vertebrate. Curr Biol 2006;16:296–300
    1. Timmers S, Konings E, Bilet L, et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab 2011;14:612–622
    1. Brasnyó P, Molnár GA, Mohás M, et al. Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients. Br J Nutr 2011;106:383–389
    1. Crandall JP, Oram V, Trandafirescu G, et al. Pilot study of resveratrol in older adults with impaired glucose tolerance. J Gerontol A Biol Sci Med Sci 2012;67:1307–1312
    1. Magyar K, Halmosi R, Palfi A, et al. Cardioprotection by resveratrol: a human clinical trial in patients with stable coronary artery disease. Clin Hemorheol Microcirc 2012;50:179–187
    1. Møller N, Jørgensen JO, Schmitz O, et al. Effects of a growth hormone pulse on total and forearm substrate fluxes in humans. Am J Physiol 1990;258:E86–E91
    1. Steele R. Influences of glucose loading and of injected insulin on hepatic glucose output. Ann N Y Acad Sci 1959;82:420–430
    1. Ferrannini E. The theoretical bases of indirect calorimetry: a review. Metabolism 1988;37:287–301
    1. Friedewald WT, Levy RI, Fredrickson DS. Estimation of the concentration of low-density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge. Clin Chem 1972;18:499–502
    1. Orskov H, Thomsen HG, Yde H. Wick chromatography for rapid and reliable immunoassay of insulin, glucagon and growth hormone. Nature 1968;219:193–195
    1. Frystyk J, Tarnow L, Hansen TK, Parving HH, Flyvbjerg A. Increased serum adiponectin levels in type 1 diabetic patients with microvascular complications. Diabetologia 2005;48:1911–1918
    1. Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 1985;28:412–419
    1. Moller L, Stodkilde-Jorgensen H, Jensen FT, Jorgensen JO. Fasting in healthy subjects is associated with intrahepatic accumulation of lipids as assessed by 1H-magnetic resonance spectroscopy. Clin Sci (Lond) 2008;114:547–552
    1. Provencher SW. Estimation of metabolite concentrations from localized in vivo proton NMR spectra. Magn Reson Med 1993;30:672–679
    1. Positano V, Gastaldelli A, Sironi AM, Santarelli MF, Lombardi M, Landini L. An accurate and robust method for unsupervised assessment of abdominal fat by MRI. J Magn Reson Imaging 2004;20:684–689
    1. Nielsen C, Gormsen LC, Jessen N, et al. Growth hormone signaling in vivo in human muscle and adipose tissue: impact of insulin, substrate background, and growth hormone receptor blockade. J Clin Endocrinol Metab 2008;93:2842–2850
    1. Brown VA, Patel KR, Viskaduraki M, et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res 2010;70:9003–9011
    1. Boocock DJ, Faust GE, Patel KR, et al. Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiol Biomarkers Prev 2007;16:1246–1252
    1. Walle T, Hsieh F, DeLegge MH, Oatis JE, Jr, Walle UK. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab Dispos 2004;32:1377–1382
    1. Almeida L, Vaz-da-Silva M, Falcão A, 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–S15
    1. Nunes T, Almeida L, Rocha JF, et al. Pharmacokinetics of trans-resveratrol following repeated administration in healthy elderly and young subjects. J Clin Pharmacol 2009;49:1477–1482
    1. Olholm J, Paulsen SK, Cullberg KB, Richelsen B, Pedersen SB. Anti-inflammatory effect of resveratrol on adipokine expression and secretion in human adipose tissue explants. Int J Obes (Lond) 2010;34:1546–1553
    1. Pedersen SB, Ølholm J, Paulsen SK, Bennetzen MF, Richelsen B. Low Sirt1 expression, which is upregulated by fasting, in human adipose tissue from obese women. Int J Obes (Lond) 2008;32:1250–1255
    1. Bonora E, Kiechl S, Willeit J, et al. Prevalence of insulin resistance in metabolic disorders: the Bruneck Study. Diabetes 1998;47:1643–1649
    1. Gormsen LC, Nielsen C, Jessen N, Jørgensen JO, Møller N. Time-course effects of physiological free fatty acid surges on insulin sensitivity in humans. Acta Physiol (Oxf) 2011;201:349–356
    1. Park SJ, Ahmad F, Philp A, et al. Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases. Cell 2012;148:421–433

Source: PubMed

3
Subskrybuj