Low-dose glucocorticoid treatment affects multiple aspects of intermediary metabolism in healthy humans: a randomised controlled trial

D H van Raalte, M Brands, N J van der Zijl, M H Muskiet, P J W Pouwels, M T Ackermans, H P Sauerwein, M J Serlie, M Diamant, D H van Raalte, M Brands, N J van der Zijl, M H Muskiet, P J W Pouwels, M T Ackermans, H P Sauerwein, M J Serlie, M Diamant

Abstract

Aim/hypothesis: To assess whether low-dose glucocorticoid treatment induces adverse metabolic effects, as is evident for high glucocorticoid doses.

Methods: In a randomised placebo-controlled double-blind (participants and the investigators who performed the studies and assessed the outcomes were blinded) dose-response intervention study, 32 healthy men (age 22 ± 3 years; BMI 22.4 ± 1.7 kg/m(2)) were allocated to prednisolone 7.5 mg once daily (n = 12), prednisolone 30 mg once daily (n = 12), or placebo (n = 8) for 2 weeks using block randomisation. Main outcome measures were glucose, lipid and protein metabolism, measured by stable isotopes, before and at 2 weeks of treatment, in the fasted state and during a two-step hyperinsulinaemic clamp conducted in the Clinical Research Unit of the Academic Medical Centre, Amsterdam, the Netherlands

Results: Prednisolone, compared with placebo, dose dependently and significantly increased fasting plasma glucose levels, whereas only prednisolone 30 mg increased fasting insulin levels (29 ± 15 pmol/l). Prednisolone 7.5 mg and prednisolone 30 mg decreased the ability of insulin to suppress endogenous glucose production (by 17 ± 6% and 46 ± 7%, respectively, vs placebo). Peripheral glucose uptake was not reduced by prednisolone 7.5 mg, but was decreased by prednisolone 30 mg by 34 ± 6% (p < 0.0001). Compared with placebo, prednisolone treatment tended to decrease lipolysis in the fasted state (p = 0.062), but both prednisolone 7.5 mg and prednisolone 30 mg decreased insulin-mediated suppression of lipolysis by 11 ± 5% and 34 ± 6%, respectively. Finally, prednisolone treatment increased whole-body proteolysis during hyperinsulinaemia, which tended to be driven by prednisolone 30 mg (5 ± 2%; p = 0.06). No side effects were reported by the study participants. All participants completed the study and were analysed.

Conclusions/interpretation: Not only at high doses but also at low doses, glucocorticoid therapy impaired intermediary metabolism by interfering with the metabolic actions of insulin on liver and adipose tissue. These data indicate that even low-dose glucocorticoids may impair glucose tolerance when administered chronically.

Trial registration: ISRCTN83991850.

Figures

Fig. 1
Fig. 1
The effects of prednisolone treatment on glucose metabolism. a Prednisolone treatment increased basal EGP. This was driven by prednisolone 30 mg once daily. b EGP during insulin infusion was dose dependently increased by prednisolone treatment. c The rate of glucose disappearance was decreased by prednisolone 30 mg, but not by prednisolone 7.5 mg once daily. Data represent means ± SEM. Black bars, before treatment; white bars, day 14 of treatment. Between-group changes from baseline were tested by Kruskal–Wallis (indicated by top line). Post hoc tests were done by Mann–Whitney U with Bonferroni correction for multiple testing (indicated by line with brackets). **p < 0.01; ***p < 0.001. PLB, placebo
Fig. 2
Fig. 2
The effects of prednisolone treatment on glycerol appearance (Ra). a Prednisolone treatment tended to decrease basal lipolysis, which seemed caused by the prednisolone 30 mg once daily dose. b During insulin infusion targeted at 200 pmol/l, prednisolone treatment dose dependently increased glycerol turnover. This reached significance for prednisolone 30 mg, but remained a trend for prednisolone 7.5 mg once daily. c At higher insulin (600 pmol/l) during step 2 of the clamp, a similar pattern was observed, with a dose-dependent increase of whole-body lipolysis with prednisolone treatment. Data represent means ± SEM. Black bars, before treatment; white bars, day 14 of treatment. Between-group changes from baseline were tested by Kruskal–Wallis (indicated by top line). Post hoc tests were done by Mann–Whitney U with Bonferroni correction for multiple testing (indicated by line with brackets). *p < 0.05; **p < 0.01; †p = 0.062; ‡p = 0.09. PLB, placebo
Fig. 3
Fig. 3
The effect of prednisolone treatment on valine appearance (Ra). a Valine turnover in the basal state was not affected by prednisolone treatment. b Overall, valine turnover was increased during step 1 of the hyperinsulinaemic–euglycaemic clamp by prednisolone treatment. This seemed driven by prednisolone 30 mg in post hoc analysis, but failed to reach statistical significance. c A similar pattern was observed during step 2 of the clamp, where overall prednisolone treatment increased valine turnover; however, in post hoc testing, no significance was reached for either dose compared with placebo. Data represent means ± SEM. Black bars, before treatment; white bars, day 14 of treatment. Between-group changes from baseline were tested by Kruskal–Wallis (indicated by top line). Post hoc tests were done by Mann–Whitney U with Bonferroni correction for multiple testing (indicated by line with brackets). *p < 0.05; †p = 0.06. PLB, placebo

References

    1. Bijlsma JW, van der Goes MC, Hoes JN, Jacobs JW, Buttgereit F, Kirwan J. Low-dose glucocorticoid therapy in rheumatoid arthritis: an obligatory therapy. Ann NY Acad Sci. 2010;1193:123–126. doi: 10.1111/j.1749-6632.2009.05342.x.
    1. Hernandez-Rodriguez J, Cid MC, Lopez-Soto A, Espigol-Frigole G, Bosch X. Treatment of polymyalgia rheumatica: a systematic review. Arch Intern Med. 2009;169:1839–1850. doi: 10.1001/archinternmed.2009.352.
    1. Salvarani C, Cantini F, Hunder GG. Polymyalgia rheumatica and giant-cell arteritis. Lancet. 2008;372:234–245. doi: 10.1016/S0140-6736(08)61077-6.
    1. Parker BJ, Bruce IN. High dose methylprednisolone therapy for the treatment of severe systemic lupus erythematosus. Lupus. 2007;16:387–393. doi: 10.1177/0961203307079502.
    1. Lichtenstein GR, Abreu MT, Cohen R, Tremaine W. American Gastroenterological Association Institute medical position statement on corticosteroids, immunomodulators, and infliximab in inflammatory bowel disease. Gastroenterology. 2006;130:935–939. doi: 10.1053/j.gastro.2006.01.047.
    1. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic effects: towards expansion of therapeutic options? Eur. J Clin Invest. 2009;39:81–93. doi: 10.1111/j.1365-2362.2008.02067.x.
    1. Buttgereit F, Da Silva JA, Boers M, et al. Standardised nomenclature for glucocorticoid dosages and glucocorticoid treatment regimens: current questions and tentative answers in rheumatology. Ann Rheum Dis. 2002;61:718–722. doi: 10.1136/ard.61.8.718.
    1. Wajngot A, Giacca A, Grill V, Vranic M, Efendic S. The diabetogenic effects of glucocorticoids are more pronounced in low- than in high-insulin responders. Proc Natl Acad Sci USA. 1992;89:6035–6039. doi: 10.1073/pnas.89.13.6035.
    1. Larsson H, Ahren B. Insulin resistant subjects lack islet adaptation to short-term dexamethasone-induced reduction in insulin sensitivity. Diabetologia. 1999;42:936–943. doi: 10.1007/s001250051251.
    1. Gravholt CH, Dall R, Christiansen JS, Moller N, Schmitz O. Preferential stimulation of abdominal subcutaneous lipolysis after prednisolone exposure in humans. Obes Res. 2002;10:774–781. doi: 10.1038/oby.2002.105.
    1. Taskinen MR, Kuusi T, Yki-Jarvinen H, Nikkila EA. Short-term effects of prednisone on serum lipids and high density lipoprotein subfractions in normolipidemic healthy men. J Clin Endocrinol Metab. 1988;67:291–299. doi: 10.1210/jcem-67-2-291.
    1. Short KR, Bigelow ML, Nair KS. Short-term prednisone use antagonizes insulin’s anabolic effect on muscle protein and glucose metabolism in young healthy people. Am J Physiol Endocrinol Metab. 2009;297:E1260–E1268. doi: 10.1152/ajpendo.00345.2009.
    1. Fernandez-Rodriguez E, Stewart PM, Cooper MS. The pituitary-adrenal axis and body composition. Pituitary. 2009;12:105–115. doi: 10.1007/s11102-008-0098-2.
    1. Gurwitz JH, Bohn RL, Glynn RJ, Monane M, Mogun H, Avorn J. Glucocorticoids and the risk for initiation of hypoglycemic therapy. Arch Intern Med. 1994;154:97–101. doi: 10.1001/archinte.154.1.97.
    1. Gulliford MC, Charlton J, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in a large population. Diabetes Care. 2006;29:2728–2729. doi: 10.2337/dc06-1499.
    1. Hoes JN, Jacobs JW, Verstappen SM, Bijlsma JW, van der Heijden GJ. Adverse events of low- to medium-dose oral glucocorticoids in inflammatory diseases: a meta-analysis. Ann Rheum Dis. 2009;68:1833–1838. doi: 10.1136/ard.2008.100008.
    1. Tushuizen ME, Bunck MC, Pouwels PJ, et al. Pancreatic fat content and beta-cell function in men with and without type 2 diabetes. Diabetes Care. 2007;30:2916–2921. doi: 10.2337/dc07-0326.
    1. Tushuizen ME, Pouwels PJ, Bontemps S, et al. Postprandial lipid and apolipoprotein responses following three consecutive meals associate with liver fat content in type 2 diabetes and the metabolic syndrome. Atherosclerosis. 2010;211:308–314. doi: 10.1016/j.atherosclerosis.2010.02.002.
    1. van Raalte DH, Nofrate V, Bunck MC, et al. Acute and 2-week exposure to prednisolone impair different aspects of beta-cell function in healthy men. Eur J Endocrinol. 2010;162:729–735. doi: 10.1530/EJE-09-1034.
    1. Ackermans MT, Ruiter AF, Endert E. Determination of glycerol concentrations and glycerol isotopic enrichments in human plasma by gas chromatography/mass spectrometry. Anal Biochem. 1998;258:80–86. doi: 10.1006/abio.1997.2535.
    1. Ackermans MT, Pereira Arias AM, Bisschop PH, Endert E, Sauerwein HP, Romijn JA. The quantification of gluconeogenesis in healthy men by (2)H2O and [2-(13)C]glycerol yields different results: rates of gluconeogenesis in healthy men measured with (2)H2O are higher than those measured with [2-(13)C]glycerol. J Clin Endocrinol Metab. 2001;86:2220–2226. doi: 10.1210/jc.86.5.2220.
    1. Geukers VG, Oudshoorn JH, Taminiau JA, et al. Short-term protein intake and stimulation of protein synthesis in stunted children with cystic fibrosis. Am J Clin Nutr. 2005;81:605–610.
    1. Finegood DT, Bergman RN, Vranic M. Estimation of endogenous glucose production during hyperinsulinemic–euglycemic glucose clamps. Comparison of unlabeled and labeled exogenous glucose infusates. Diabetes. 1987;36:914–924. doi: 10.2337/diabetes.36.8.914.
    1. Steele R. Influences of glucose loading and of injected insulin on hepatic glucose output. Ann NY Acad Sci. 1959;82:420–430. doi: 10.1111/j.1749-6632.1959.tb44923.x.
    1. Frayn KN. Calculation of substrate oxidation rates in vivo from gaseous exchange. J Appl Physiol. 1983;55:628–634.
    1. Clore JN, Thurby-Hay L. Glucocorticoid-induced hyperglycemia. Endocr Pract. 2009;15:469–474. doi: 10.4158/EP08331.RAR.
    1. Corpeleijn E, Saris WH, Blaak EE. Metabolic flexibility in the development of insulin resistance and type 2 diabetes: effects of lifestyle. Obes Rev. 2009;10:178–193. doi: 10.1111/j.1467-789X.2008.00544.x.
    1. Henriksen JE, Alford F, Vaag A, Handberg A, Beck-Nielsen H. Intracellular skeletal muscle glucose metabolism is differentially altered by dexamethasone treatment of normoglycemic relatives of type 2 diabetic patients. Metabolism. 1999;48:1128–1135. doi: 10.1016/S0026-0495(99)90126-9.
    1. Ruzzin J, Wagman AS, Jensen J. Glucocorticoid-induced insulin resistance in skeletal muscles: defects in insulin signalling and the effects of a selective glycogen synthase kinase-3 inhibitor. Diabetologia. 2005;48:2119–2130. doi: 10.1007/s00125-005-1886-0.
    1. Djurhuus CB, Gravholt CH, Nielsen S, et al. Effects of cortisol on lipolysis and regional interstitial glycerol levels in humans. Am J Physiol Endocrinol Metab. 2002;283:E172–E177.
    1. Djurhuus CB, Gravholt CH, Nielsen S, Pedersen SB, Moller N, Schmitz O. Additive effects of cortisol and growth hormone on regional and systemic lipolysis in humans. Am J Physiol Endocrinol Metab. 2004;286:E488–E494. doi: 10.1152/ajpendo.00199.2003.
    1. Macfarlane DP, Forbes S, Walker BR. Glucocorticoids and fatty acid metabolism in humans: fuelling fat redistribution in the metabolic syndrome. J Endocrinol. 2008;197:189–204. doi: 10.1677/JOE-08-0054.
    1. Birkenhager JC, Timmermans HA, Lamberts SW. Depressed plasma FFA turnover rate in Cushing’s syndrome. J Clin Endocrinol Metab. 1976;42:28–32. doi: 10.1210/jcem-42-1-28.
    1. Miyoshi H, Shulman GI, Peters EJ, Wolfe MH, Elahi D, Wolfe RR. Hormonal control of substrate cycling in humans. J Clin Invest. 1988;81:1545–1555. doi: 10.1172/JCI113487.
    1. Bonadonna RC, Groop LC, Zych K, Shank M, DeFronzo RA. Dose-dependent effect of insulin on plasma free fatty acid turnover and oxidation in humans. Am J Physiol. 1990;259:E736–E750.
    1. Gelding SV, Coldham N, Anyaoku V, Heslop K, Halliday D, Johnston DG. Differential metabolic actions of biosynthetic insulin analogues in normal man assessed by stable isotopic tracers. Diabet Med. 1993;10:470–476. doi: 10.1111/j.1464-5491.1993.tb00101.x.
    1. Nakada MT, Stadel JM, Poksay KS, Crooke ST. Glucocorticoid regulation of beta-adrenergic receptors in 3T3-L1 preadipocytes. Mol Pharmacol. 1987;31:377–384.
    1. Rojas FA, Hirata AE, Saad MJ. Regulation of insulin receptor substrate-2 tyrosine phosphorylation in animal models of insulin resistance. Endocrinology. 2003;21:115–122.
    1. Lundgren M, Buren J, Ruge T, Myrnas T, Eriksson JW. Glucocorticoids down-regulate glucose uptake capacity and insulin-signaling proteins in omental but not subcutaneous human adipocytes. J Clin Endocrinol Metab. 2004;89:2989–2997. doi: 10.1210/jc.2003-031157.
    1. Saad MJ, Folli F, Kahn JA, Kahn CR. Modulation of insulin receptor, insulin receptor substrate-1, and phosphatidylinositol 3-kinase in liver and muscle of dexamethasone-treated rats. J Clin Invest. 1993;92:2065–2072. doi: 10.1172/JCI116803.
    1. Vegiopoulos A, Herzig S. Glucocorticoids, metabolism and metabolic diseases. Mol Cell Endocrinol. 2007;275:43–61. doi: 10.1016/j.mce.2007.05.015.
    1. Slavin BG, Ong JM, Kern PA. Hormonal regulation of hormone-sensitive lipase activity and mRNA levels in isolated rat adipocytes. J Lipid Res. 1994;35:1535–1541.
    1. Schakman O, Gilson H, Thissen JP. Mechanisms of glucocorticoid-induced myopathy. J Endocrinol. 2008;197:1–10. doi: 10.1677/JOE-07-0606.
    1. Dessein PH, Joffe BI. Insulin resistance and impaired beta cell function in rheumatoid arthritis. Arthritis Rheum. 2006;54:2765–2775. doi: 10.1002/art.22053.
    1. Willi SM, Kennedy A, Wallace P, Ganaway E, Rogers NL, Garvey WT. Troglitazone antagonizes metabolic effects of glucocorticoids in humans: effects on glucose tolerance, insulin sensitivity, suppression of free fatty acids, and leptin. Diabetes. 2002;51:2895–2902. doi: 10.2337/diabetes.51.10.2895.
    1. Rosen J, Miner JN. The search for safer glucocorticoid receptor ligands. Endocr Rev. 2005;26:452–464. doi: 10.1210/er.2005-0002.

Source: PubMed

3
S'abonner